Poster Session 1
Session Details
The titles of the abstracts in this session will be posted on Tuesday, October 30. The text of the abstracts will be posted on Monday, November 24.
Presentation numberPS1-01-01
Evaluating the use, self-reported outcomes, and cost of using scalp cooling in a prospective survey study of young women with early-stage breast cancer
Simona Krasnegor, UCSF, San Francisco, CA
S. Krasnegor1, A. Paciorek1, K. H. Natsuhara1, K. Kuwahara2, K. Blevins3, K. Blum1, M. Kim1, H. S. Rugo4, M. Melisko1, M. Majure1, L. A. Huppert1, H. Batra-Sharma1, L. J. Esserman1, A. Chien1; 1Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, 2Geisel School of Medicine, Dartmouth College, Hanover, NH, 3Anschutz School of Medicine, University of Colorado, Aurora, CO, 4Duarte Cancer Center, City of Hope, Duarte, CA.
Background: Hair loss is a significant quality of life issue for women with breast cancer (BC) and may be particularly distressing for young women. Scalp cooling (SC) is effective at preventing chemotherapy (CT)-induced alopecia, and studies suggest improved quality of life in patients who have undergone successful SC. However, SC is rarely covered by insurance. The decision to use SC is personal. We aimed to understand how young women navigate the decision to use SC during CT, and self-reported estimates of hair loss, hair recovery, and cost at 1-year follow-up (FU). Methods: This is a single-institution prospective registry of young women diagnosed with early BC <45 yrs of age. Patients (pts) diagnosed within 6 months were eligible for study entry. This analysis included pts who received CT. Electronic surveys were administered at baseline (BL) and 1-year FU, which included questions about SC use, efficacy, satisfaction, and cost. Anxiety and depression were assessed through validated PROMIS measures. Pearson chi-squared, Kruskal-Wallis, and linear regressions statistics were used to test for associations between SC, PROMIS scores, and pt characteristics. SC devices included Penguin, DigniCap, or other systems. Results: Between May 2018 and May 2025, 265 pts who received neo/adjuvant CT were enrolled in the Young Women’s Registry. Of the 265 pts, 202 completed BL and/or 1-year FU surveys. Median age was 38 years; 60% White, 23% Asian, 3% Black, 8% Hispanic/Latina. 45% of pts had HR+/HER2- tumors, 23% TNBC, and 32% HER2+. 14% were single, 56% had children. 143 (71%) had an annual income >$100,000, 180 (89%) held college or higher degrees. Of pts that used SC, 31% received AC-containing and 69% receivednon-AC-containing CT. Pts who received non-AC regimens were more likely to use SC (p=0.046). Of 202 pts, 106 (52%) used SC. SC use was not associated with age, income, education level, having children, relationship status, or race. Depression and anxiety were assessed at BL using PROMIS measures in 129 pts. Pts that planned to use SC had higher anxiety (p=0.046), but no association with depression scores was evident (p=0.169). 75 of 106 (71%) pts who used SC completed a 1-year FU survey. 6% of pts reported losing <10% of their hair during CT, 19% reported 10-24%, 15% reported 25-49%, 32% reported 50-74%, 22% reported 75-99%, and 6% reported losing 100% of their hair (3/5 pts had received AC). When asked about satisfaction with SC use, 26% stated they were extremely satisfied, 38% were satisfied, 22% were neutral, and 14% were dissatisfied. 79% reported they would use SC again, 21% said they would not. At 1-year FU, 43% reported that their hair was back to BL, 37% recovered most, 13% recovered some, and 7% recovered a little of their hair. No pts reported that they had not recovered any hair. Pts with less hair loss were more likely to report satisfaction with SC use (p<0.001). PROMIS Anxiety and Depression scores at 1-year FU were not associated with degree of self-reported hair loss or recovery. Pts were asked about out-of-pocket SC expenses. 15% reported spending < $1,000, 23% spent $1,000-1,999, 30% spent $2,000-2,999, 18% spent $3,000-3,999, 7% spent $4,000-4,999, and 7% spent > $5,000. There was no association between out-of-pocket expense and satisfaction with SC at 1-year FU. Conclusions: In this prospective, single institution young women’s study, 52% chose to use SC. Higher rates of BL anxiety were associated with a plan to use SC. Despite SC use, some pts reported significant hair loss, with 28% reporting loss of > 75% of their hair. At 1-year FU, the majority (68%) were satisfied with their SC experience, however hair loss, hair regrowth, and cost varied widely. SC is an important option for young pts. These data can improve shared decision-making around the use of SC. Techniques to improve efficacy are still needed.
Presentation numberPS1-01-02
Acupuncture for Preventing Progression of Taxane-Induced Peripheral Neuropathy (ATP) Beyond Cryotherapy: A Phase II Randomized, Double-Blinded, Sham-Controlled Trial
Iris Zhi, NYU Grossman Long Island School of Medicine Perlmutter Cancer Center – Long Island, Minoela, NY
I. Zhi1, E. Kwag2, R. Baser3, S. Li3, L. Taylor3, D. Kim3, J. Mao3, T. Bao4; 1Medical Oncology and Hematology, NYU Grossman Long Island School of Medicine Perlmutter Cancer Center – Long Island, Minoela, NY, 2Integrative Medicine Service, Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 3Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 4Zakim Center for Integrative Therapies and Healthy Living, Dana-Farber Cancer Institute, Boston, MA.
Background: Chemotherapy-induced peripheral neuropathy (CIPN) affects up to 80% of breast cancer patients treated with neurotoxic chemotherapy, with nearly 50% requiring dose reduction or discontinuation. While cryotherapy has shown promise in CIPN prevention, many patients still develop symptoms. This study evaluated the effectiveness of real versus sham acupuncture in early-stage breast cancer patients who developed CIPN despite using cryotherapy during taxane-based chemotherapy.Method: Chemotherapy included paclitaxel or nab-paclitaxel, with co-administered agents (e.g., carboplatin, pembrolizumab, trastuzumab, pertuzumab) permitted. Patients were randomized in a 1:1 to receive either real acupuncture (RA) or sham acupuncture (SA) weekly for a minimum of four sessions during chemotherapy. The primary outcome was neuropathy pain by the Neuropathic Pain Scale (NPS) after 4 weeks of intervention. Secondary outcomes included the relative dose intensity (RDI) of taxane chemotherapy, defined as the ratio of delivered dose to planned dose. Licensed acupuncturists delivered semi-standardized protocols. All continuous outcomes were analyzed using constrained linear mixed models adjusting for randomization stratification variables; binary RDI was analyzed using logistic regression.Results: Eighty female patients were enrolled (median age, 51.0 years; range, 24.0-80.9). Paclitaxel was used in 75%, nab-paclitaxel in 25%, and cryotherapy in 94.6%. Baseline characteristics were balanced. The retention rate was 28.8%. No acupuncture-related adverse events occurred. After 4 weeks, both RA and SA groups showed statistically significant NPS reductions (RA: -6.07, P = .039; SA: -6.98, P = .021), sustained at 12-week follow-up (RA: -12.68, P < .001; SA: -10.52, P 80% maintaining ≥85% RDI (P = .80). Numerically, more RA patients achieved ≥30% NPS reduction (15 vs 11), and fewer worsened (6 vs 11) than SA, though differences were not significant (P = .20).Conclusion: This is the first sham acupuncture-controlled study to evaluate the effectiveness of acupuncture in preventing CIPN progression after cryotherapy in patients with breast cancer. Both RA and SA were associated with statistically significant reductions in neuropathy pain and high chemotherapy delivery. However, the absence of significant differences between real and sham acupuncture suggests that acupuncture may not provide specific therapeutic benefits beyond placebo or attention effects. The continuous improvement observed during and after chemotherapy exceeded the typical trajectory of CIPN, suggesting potential therapeutic effects of both RA and SA warrant further investigation.
| Patient characteristics | Overall (N=80) | Real (N=40) | Sham (N=40) | Treatment characteristics | Overall (N=80) | Real (N=40) | Sham (N=40) | |
| Age, median (range) | 51.0 (24.0-80.9) | 49.1 (24.0-70.0) | 51.2 (32.7-80.9) | Cryotherapy used | 70 (94.6%) | 35 (87.5%) | 35 (87.5%) | |
| BMI, mean (SD) | 28.19 (5.86) | 28.27 (5.93) | 28.10 (5.86) | More than 4 acupuncture session received | 62 (77.5%) | 31 (77.5%) | 31 (77.5%) | |
| Race | Outcome summary | Real | Sham | P (between) | ||||
| White | 46 (57.5%) | 21 (52.5%) | 25 (62.5%) | Primary outcome: Change in NPS | ||||
| Black | 15 (18.8%) | 9 (22.5%) | 6 (15.0%) | Timepoint | Real (mean Δ, 95% CI) | Sham (mean Δ, 95% CI) | P (between) | |
| Asian | 13 (16.3%) | 8 (20.0%) | 5 (12.5%) | Week 4 (primary endpoint) |
-6.07 (-11.82, -0.32)
P=0.039*
|
-6.98 (-12.89, -1.07)
P=0.021*
|
0.82 | |
| Multiracial | 6 (7.5%) | 2 (5.0%) | 4 (10.0%) | 12 weeks after chemotherapy completion |
-12.68 (-18.73, -6.63)
P <0.001*
|
-10.52 (-16.53, -4.51)
P <0.001*
|
0.6 | |
| Ethnicity | Secondary Outcome: RDI | |||||||
| Hispanic | 9 (11.3%) | 3 (7.5%) | 6 (15.0%) | Real | Sham | P (between) | ||
| Non-Hispanic | 71 (88.8%) | 37 (92.5%) | 34 (85.0%) | Mean (SD) | 0.94 (0.12) | 0.94 (0.09) | 0.4 | |
| Acupuncture expectancy (mean) | 13.13 (3.56) | 13.06 (3.40) | 13.19 (3.75) | ≥85%, n (%) | 33 (82.5%) | 34 (85.0%) | 0.8 | |
| Disease characteristics | Overall (N=80) | Real (N=40) | Sham (N=40) | Exploratory Outcome: Clinically meaningful change in NPS | ||||
| Breast Cancer stage | Real | Sham | P (between) | |||||
| I | 38 (47.5%) | 17 (42.5%) | 21 (52.5%) | ≥30% reduction from baseline | 15 (51.7%) | 11 (36.7%) | 0.2 | |
| II | 26 (32.5%) | 15 (37.5%) | 11 (27.5%) | NPS worsened at week 4 | 6 (20.7%) | 11 (36.7%) | 0.2 | |
| III | 16 (20.0%) | 8 (20.0%) | 8 (20.0%) | |||||
| Chemotherapy received | ||||||||
| Paclitaxel | 60 (75.0%) | 31 (77.5%) | 29 (72.5%) | |||||
| Nab-Paclitaxel | 20 (25.0%) | 9 (22.6%) | 11 (27.5%) | |||||
| CIPN grade 1 by CTCAE 5.0 | 79 (98.8%) | 40 (100.0%) | 39 (97.5%) |
Presentation numberPS1-01-03
Impact of Dexamethasone Premedication Timing on Infusion Reactions in HER2 Positive Breast Cancer
Ryoichi Matsunuma, Shizuoka General Hospital, Shizuoka, Japan
R. Matsunuma1, S. Nakagaki2, E. Nakatani3, M. Kikuchi4, N. Wada5, K. Yonezawa6, T. Isono7, R. Hayami1, M. Kaga8, M. Tsuneizumi1; 1Breast Surgery, Shizuoka General Hospital, Shizuoka, JAPAN, 2Pharmacy, Shizuoka General Hospital, Shizuoka, JAPAN, 3Biostatistics and Health Data Science, Graduate School of Medical Science Nagoya City University, Aichi, JAPAN, 4Surgery, Japanese Red Cross Shizuoka Hospital, Shizuoka, JAPAN, 5Clinical Oncology, Tokyo Dental College Ichikawa General Hospital, Ichikawa, JAPAN, 6Surgery, Shizuoka City Hospital, Shizuoka, Shizuoka, JAPAN, 7Surgery, Shimada General Medical Center, Shimada, JAPAN, 8Pharmacy, Tokyo Dental College Ichikawa General Hospital, Shizuoka, JAPAN.
Background: Infusion reactions (IRs) are frequently observed adverse events associated with HER2-targeted monoclonal antibodies, particularly trastuzumab and pertuzumab. These reactions can impair treatment tolerability and necessitate prolonged observation, delay, or discontinuation of therapy. Although dexamethasone is routinely administered as premedication to mitigate docetaxel-induced hypersensitivity, the optimal timing of dexamethasone administration relative to HER2-targeted therapy remains undefined. This study aimed to evaluate whether administering dexamethasone prior to HER2-targeted agents can reduce the incidence of IRs in patients with HER2-positive early breast cancer receiving standard trastuzumab, pertuzumab, and docetaxel therapy. Methods: This randomized, open-label, multicenter trial enrolled 100 patients with HER2-positive stage I-III breast cancer across multiple institutions. Patients were randomly assigned (1:1) to receive dexamethasone either before (experimental group) or after (control group) the infusion of trastuzumab and pertuzumab during the first treatment cycle. All patients received standard chemotherapy with docetaxel (75 mg/m2), trastuzumab (8 mg/kg loading dose), and pertuzumab (840 mg loading dose). Dexamethasone was administered intravenously at 9.9 mg either immediately prior to HER2-targeted therapy or after completion, according to group assignment. The primary endpoint was the incidence of any-grade IRs during the first cycle. Secondary endpoints included the incidence of grade >=3 IRs, IRs during cycle 2, and overall treatment-related adverse events (AEs). The time of IR onset relative to dexamethasone administration was also analyzed. Results: Of the 100 patients enrolled, 50 were allocated to each group. The incidence of IRs during cycle 1 was significantly lower in the experimental group (24.0%) compared to the control group (60.0%) (p =3 IRs occurred in either group. During cycle 2, IR incidence was low and did not significantly differ between the experimental and control groups (8.0% vs. 10.2%, p = 0.703), suggesting that immune tolerance may develop after initial exposure. The frequency of treatment-related AEs, including hematologic and non-hematologic toxicities, was comparable between groups (98.0% vs. 100.0%; p > 0.999). A time-course analysis showed that most IRs in the control group occurred prior to dexamethasone administration, indicating a potential protective effect of early corticosteroid exposure. Conclusions: Premedication with dexamethasone administered prior to HER2-targeted agents significantly reduced the incidence of IRs during the first treatment cycle without introducing additional toxicity. This simple, cost-effective modification to standard premedication protocols improves the safety and tolerability of trastuzumab and pertuzumab combination therapy. Given the widespread use of monoclonal antibodies in oncology, our findings may have broader implications for optimizing premedication strategies across various therapeutic settings.
Presentation numberPS1-01-04
Efficacy of cryotherapy using frozen gloves and socks to prevent docetaxel-induced onycholysis in early breast cancer patients undergoing sequential (neo)adjuvant (F)EC and docetaxel treatment: The Banquise randomized controlled trial.
Laurent Mathiot, Institut de Cancérologie de l’Ouest, Saint-Herblain, France
L. Mathiot1, C. Poiraud2, J. Dimet3, L. Ropers3, M. Fenot2, E. Bourbouloux1, S. Abadie-Lacourtoisie4, C. El Kouri5, V. Delecroix6, R. Lamy7, M. Noblecourt8, O. Cojocarasu9, F. Scotté10, J. Frenel1, F. Priou11; 1Department of Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 2Department of Dermatology, Centre Hospitalier Departemental Vendée, La Roche sur Yon, FRANCE, 3Clinical Research Centre, Centre Hospitalier Departemental Vendée, La Roche sur Yon, FRANCE, 4Department of Medical Oncology, Institut de Cancérolologie de l’Ouest, Angers, FRANCE, 5Department of Medical Oncology, Centre Catherine de Sienne, Nantes, FRANCE, 6Department of Medical Oncology, Clinique Mutualiste de l’Estuaire, Saint-Nazaire, FRANCE, 7Department of Medical Oncology, Centre Hospitalier Bretagne Sud, Lorient, FRANCE, 8Department of Medical Oncology, Centre Hospitalier de Cholet, Cholet, FRANCE, 9Department of Medical Oncology, Centre Hospitalier du Mans, Le Mans, FRANCE, 10Interdisciplinary Cancer Course Department, Gustave Roussy Cancer Institute, Villejuif, FRANCE, 11Department of Medical Oncology, Centre Hospitalier Departemental Vendée, La Roche sur Yon, FRANCE.
Background: The use of docetaxel in the (neo)adjuvant treatment of early-stage breast cancer (eBC) is frequently linked to notable nail toxicities, which can negatively impact quality of life (QoL) and potentially lead to treatment delays. Cryotherapy, involving the use of frozen gloves and socks, is used as a strategy to decrease docetaxel-induced nail toxicity. Strong evidence supporting its efficacy at lower cumulative doses of docetaxel and its overall tolerability remains limited. Methods: BANQUISE trial was a randomized, open-label, multicenter study investigating the efficacy of cryotherapy in eBC patients receiving three cycles of (neo)adjuvant docetaxel (100 mg/m²) following three cycles of (F)EC 100. Participants were randomized after completing three cycles of (F)EC to either the cryotherapy group, with frozen gloves and socks applied 15 minutes before and removed 15 minutes after the docetaxel infusion, or to the control group. The primary endpoint was the incidence of nail toxicity grade ≥ 2 between the first dose of docetaxel and eight weeks after the last dose of docetaxel, assessed using CTCAE v4.0 criteria by investigators. Secondary endpoints included a docetaxel nail toxicity (DNT) score assessed by a blinded dermatologist at weeks 8 and 24 using photographs. This score integrates the CTCAE V4.0 grade and the number of affected fingers, assigning 1 point per digit for grade 1 toxicity and 5 points per digit for grade 2 toxicity. Cryotherapy tolerability, and patient-reported outcomes (PROs) were measured at each cycle and week 8 and 24 after last dose of docetaxel by the EORTC QLQ-C30 and BR23 questionnaires. Statistical analyses used chi-squared and t-tests, with significance set at p<0.05. Results: From January 2014 to March 2019, a total of 280 patients were randomized to either the cryotherapy arm (n=141) or the control arm (n=139). The median age was 54 years (range 27-78). The mean cumulative dose of docetaxel was 471 mg (range: 150-660 mg) in the cryotherapy arm and 467 mg (range: 176-600 mg) in the control arm. Docetaxel dose reduction was required in 28 patients (21.9%) in the cryotherapy group and 30 patients (24.6%) in the control group. Frozen gloves were worn by 119 patients (84%) during all three courses of docetaxel, by 126 patients (89%) during two courses, and by 139 patients (99%) during one course. In the intention-to-treat population, cryotherapy significantly reduced the incidence of grade ≥ 2 nail toxicity at week 8 after the final docetaxel dose (18% vs. 37%, p < 0.001), as assessed by the investigators. This decrease was observed in both hand (13% vs. 29%, p = 0.002) and foot (12% vs. 24%, p = 0.011) nail toxicities. These results were supported by evaluations from a dermatologist blinded to treatment, showing a significant reduction in the DNT score for the cryotherapy group at week 8 (29.4 [95% CI, 17 to 41.8] vs. 54.9 [95% CI, 42.7 to 67.2]; p < 0.001) and at week 24 (9.5 [95% CI, 3.1 to 22] vs. 22.8 [95% CI, 10.3 to 35.3]; p = 0.025). Severe discomfort and pain were reported by 41% and 35% of patients wearing frozen gloves, and by 43% and 42% of patients using frozen socks, respectively. PROs revealed no significant differences in quality of life between the treatment arms at weeks 8 and 24. Discussion: Cryotherapy effectively reduces the incidence and severity of nail toxicity in eBC patients receiving 3 courses of (neo)adjuvant docetaxel. Although well-tolerated overall, discomfort remains a notable limitation. These findings support cryotherapy as a scalable intervention to improve chemotherapy tolerability.
Presentation numberPS1-01-05
Pneumonitis but not other immune related adverse events (irAEs) associate with pre-treatment immune signatures in early-stage breast cancer patients receiving neoadjuvant immunotherapy (IO): results from 5 IO arms in I-SPY2
Denise M Wolf, University of California, San Francisco, San Francisco, CA
D. M. Wolf1, H. Rugo2, R. Nanda3, S. Umashankar4, C. Yau4, M. Campbell1, R. Sayaman1, M. Magbanua1, L. Brown-Swigart1, G. Hirst1, A. Glas5, I-SPY Investigators, L. Huppert2, F. Symmans6, S. Borowsky7, P. Pohlmann8, A. Clark9, E. Stringer-Reasor10, M. Liu11, H. Han12, H. Soliman12, J. Chien2, R. Shatsky13, C. Isaacs14, D. Yee15, A. DeMichele9, J. Perlmutter16, L. Pusztai17, Z. Quandt2, R. Cohen18, L. van ‘t Veer1, L. Esserman19, A. Basu19; 1Laboratory Medicine, University of California, San Francisco, San Francisco, CA, 2Hellen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, 3Breast Oncology Program, University of Chicago, Chicago, CA, 4Department of Surgery, University of California, San Francisco, San Francisco, CA, 5Biomarkers and data, Quantum Leap Healthcare Collaborative, San Francisco, CA, 6Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 7Pathology, University of California, Davis; school of medicine, Sacramento, CA, 8Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 9Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, 10Hematology & Oncology, University of Alabama, Birmingham, AL, 11Chief Medical Officer, Natera, San Carlos, CA, 12Medical Oncology, Moffit Cancer Center, Tampa, FL, 13Medical Oncology, University of California, San Diego, San Diego, CA, 14Medical Oncology, Georgetown University, Washington, DC, CA, 15Masonic Cancer Center, University of Minessota, Minneapolis, MN, 16Breast Cancer Patient Advocacy Core, Gemini Group, Ann Arbor, MI, 17School of Medicine, Yale University, New Haven, CT, 18Biological Sciences, University of Chicago, Chicago, IL, 19Surgery, University of California, San Francisco, San Francisco, CA.
Background Neoadjuvant immunotherapy has become standard of care for triple negative (TN) breast cancer; and I-SPY2 and other trials have shown efficacy of IO in HR+HER2-, especially in immune+ subsets. However, IO agents carry risk of immune-related toxicities, some long-term. Previously we developed Imprint, an IDE-enabled immune classifier predicting response to IO now being used in I-SPY2.2 as part of the Response Predictive Subtypes. Here we pool irAE data from 5 IO arms in I-SPY2, where both TN and HR+HER2- patients enrolled, to assess whether irAE development may be predicted by gene expression signatures in pre-treatment tumor biopsies. Long-term, the goal is to develop clinical decision support tools to help balance likelihood of response to IO with likelihood of irAE development. Methods 356 patients (206 HR+HER2- and 150 TN) across 5 IO arms (69 pembrolizumab (Pembro), 76 Pembro/SD101, 73 durvalumab/olaparib, 62 cemiplimab (Cemi), and 78 Cemi/LAG3) with irAE and pCR data were analyzed. irAEs considered included adrenal insufficiency (10%; n=36), colitis (2.3%; n=8), diabetes mellitus (0.3%; n=1), hyperthyroidism (4.2%; n=15), hypothyroidism (15%; n=53), hypophysitis (1% ; n=4), pneumotitis (5.3%; n=19), and thyroiditis (1.4%; n=5) (unresolved grade 1 or grades 2/3). 32 mostly-immune (7 immune checkpoints, 14 immune cells, 3 TcClassII, 4 chemokine/cytokine, 1 interferon, 1 TGFB, 1 ESR1/PGR, and 1 proliferation) genes/signatures were evaluated from pre-treatment mRNA from Agilent 44K arrays for 344 patients. Associations between irAEs (overall/any and individual) and signatures were assessed using logistic regression, overall and within HR+HER2-, TN, and ImPrint+/- subsets. P-values were adjusted for multiple hypothesis testing using the Benjamini-Hochberg method (significance BH p<0.05). Results 31% (109/356) patients over the 5 IO arms had at least one irAE; and some patients had 2 (7%; 24/356), 3 (2%; 7/356), or 4 irAEs (1/356) (150 irAEs were distributed over 109 patients). irAE prevalence ranged from 19% to 44% by IO arm, with the highest prevalence in dual-IO arms. Prevalence of irAEs did not differ significantly between HR+HER2- and TN (OR=1.2 [0.7-2]); nor between ImPrint- and ImPrint+ (OR=1.01 [0.6-1.7]). irAEs (any: yes/no) also did not differ between responders and non-responders (OR=1.14 [0.7-1.84]). Pneumonitis, but not other irAEs nor the composite irAE vector, significantly associated with multiple signatures. 19/32 signatures, all immune, significantly associated with pneumonitis in the population as a whole (BH p<0.05). These include higher levels of immune checkpoints (e.g., CD274 (PDL1) and PDCD1 (PD1)), cell populations (e.g., T-cells and B-cells), TcClassII (e.g., ICS5 and Module4_Immune), and chemokine/cytokine signatures (CK12, TIS, and Geparsixto); and lower levels of TGFB (all BH p<0.05). Subset analysis revealed these associations to be subtype specific, restricted to patients with ImPrint+ tumors (38%). PD1 mRNA levels predict pneumonitis in this group; with an AUC=0.84, sensitivity=67% and specificity=91%. For patients with ImPrint- tumors, there were no associations between irAEs and any tested signatures. Conclusion ~30% of patients treated with neoadjuvant IO and standard chemotherapy developed an irAE. irAEs did not associate with subtype or IO response. Pneumonitis, but no other irAE, associated with pre-treatment immune signatures. This association was observed exclusively in patients with ImPrint+ tumors, where PD1 mRNA levels predicted pneumonitis. mRNA signatures may help enable early identification and treatment of IO patients likely to develop pneumonitis.
Presentation numberPS1-01-07
Tamoxifen use increases endometrial cancer risk in premenopausal breast cancer patients
Zoë Molinari, UZ Leuven, Leuven, Belgium
Z. Molinari1, N. Van Damme2, J. Verbeeck2, A. Laenen3, S. Han1, M. Van Houdt1, D. Timmerman4, A. Smeets5, I. Nevelsteen5, Y. Van Herck6, F. Derouane6, H. Janssen7, A. Baten7, J. Verhoeven7, A. Van Rompuy8, P. Berteloot1, T. Van Gorp1, T. Baert1, F. Amant1, E. Van Nieuwenhuysen1, H. Wildiers6, P. Neven1; 1Department of Gynaecological Oncology, UZ Leuven, Leuven, BELGIUM, 2Oncology, Belgian Cancer Registry, Brussel, BELGIUM, 3Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, BELGIUM, 4Department of Obstetrics & Gynaecology, UZ Leuven, Leuven, BELGIUM, 5Department of Surgical Oncology, UZ Leuven, Leuven, BELGIUM, 6Department of General Medical Oncology, UZ Leuven, Leuven, BELGIUM, 7Department of Radiotherapy, UZ Leuven, Leuven, BELGIUM, 8Department of Imaging and Pathology, UZ Leuven, Leuven, BELGIUM.
Tamoxifen use increases endometrial cancer risk in premenopausal breast cancer patients. Z. Molinari, N. Van Damme, J. Verbeeck, A. Laenen, S. Han, M. Van Houdt, D. Timmerman, A. Smeets, I. Nevelsteen, Y. Van Herck, F. Derouane, H. Janssen, A. Baten, J. Verhoeven, A.S. Van Rompuy, P. Berteloot, T. Van Gorp, T. Baert, F. Amant, E. Van Nieuwenhuysen, H. Wildiers, P. NevenBackgroundA paradigm challenging retrospective study suggested that premenopausal breast cancer (BC) patients, were 3.77 times more likely to develop endometrial cancer (ECa) when treated with tamoxifen. Given that many patients may not have been truly premenopausal, we aimed to validate these results in a similar, large-scale nationwide study.MethodsWe conducted a retrospective cohort study of all diagnoses of ECa following tamoxifen initiation for non-metastatic BC in women aged 18–53 between 2008 and 2020 in Belgium. Data were obtained from the Belgian Cancer Registry and administrative databases. Definitive menopausal status could not be verified based on these data. The primary endpoint was time to ECa diagnosis up to 31/12/2022. Continuous variables were summarized as mean or median, and categorical variables as frequencies (%). The tamoxifen and non-tamoxifen cohorts were compared by year of incidence using the Mann-Whitney U test (continuous variables) and Chi-square or Fisher’s exact test (categorical variables). Patients who did not develop ECa were censored at the date of last follow-up. A Cox proportional hazards model assessed the primary endpoint, with treatment group as the main variable and patient/tumor characteristics as covariates. Hazard ratios (HR) with 95% confidence intervals (CI) were calculated using SAS software version 9.4, with significance set at p < 0.05.ResultsWe identified 38,857 early BC patients: 15,568 patients (40%; median age 47 years; 12.9% with cardiovascular risk; 3.2% diabetic) in the non-tamoxifen group and 23,289 (60%; median age 47 years; 10.9% cardiovascular risk; 2.3% diabetic) in the tamoxifen group. Tamoxifen users were more likely to have low-grade and lobular tumors, and less likely to receive (neo)adjuvant or targeted (chemo)therapy. During follow-up, 27 ECa events occurred in the non-tamoxifen group (0.17%) and 96 in the tamoxifen group (0.41%). This translated to 0.22% and 0.51% of survivors, respectively. The difference was statistically significant (HR 2.05, CI: 1.34-3.15). After adjusting for potential confounders—diabetes, cardiovascular profile, tumor grade, pT, pN, chemotherapy, and targeted therapy—the risk remained significantly elevated in the tamoxifen group (adjusted HR 1.93, CI: 1.14-3.28; p = 0.0141). ConclusionIn this Belgian cohort, tamoxifen use in women under the age of 54 with unknown menopausal status and early-stage invasive BC was associated with statistically significant increased risk of developing ECa, independent of other risk factors.
Presentation numberPS1-01-09
Real-time monitoring for drug-induced interstitial lung disease with Trastuzumab Deruxtecan and Rilvegostomig combination therapy prevents persistent, symptomatic disease: Safety insights from the I-SPY2.2 platform trial
Ciara C OSullivan, Mayo Clinic Comprehensive Cancer Center, Rochester, MN
T. Kulkarni1, R. Nanda2, H. S. Rugo3, C. C. OSullivan4, K. M. Kalinsky5, J. C. Boughey6, P. R. Pohlmann7, J. Perlmutter8, D. Yee9, A. Borowsky10, F. Symmans11, N. Hylton12, L. Van ‘t Veer13, C. Yau14, L. Esserman15; 1Medicine, The University of Alabama at Birmingham, Birmingham, AL, 2Medicine, University of Chicago, Chicago, IL, 3Medicine, City of Hope National Medical Center, Duarte, CA, 4Medicine, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 5Medicine, Emory University Winship Cancer Institute, Atlanta, GA, 6Surgery, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 7Medicine, UT MDACC, Houston, TX, 8N/A, Gemini Group, Ann Arbor, MI, 9Medicine, University of Minnesota, Minneapolis, MN, 10Pathology, University of California Davis, Sacramento, CA, 11Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 12Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 13Laboratory Medicine, University of California San Francisco, San Francisco, CA, 14Surgery, University of California San Francisco, San Francisco, CA, 15Surgery, University of California San Francisco, San Francisco, CA.
Background: Combinations of antibody-drug conjugates and immunotherapy, such as trastuzumab deruxtecan (T-DXd) and rilvegostomig (rilve), respectively, offer promising efficacy but are associated with a heightened risk of drug-induced interstitial lung disease (ILD)/pneumonitis. Early detection and management are critical for optimizing patient safety, yet there are no standard protocols for real-time evaluation. We hypothesized that vigilant monitoring would enable early detection, prevent symptoms, and minimize the chance of treatment delays while testing novel therapeutic combinations that have a potential risk of ILD. Methods: We tested the combination of T-DXd and rilve administered every 3 weeks in the I-SPY2.2 phase 2 platform trial (NCT01042379), which enrolls patients with clinical stage II/III breast cancer commencing neoadjuvant systemic therapy. Given the potential risk of overlapping pulmonary toxicity of these two agents, we added rigorous monitoring with pulmonary function tests (PFTs), six-minute walk test (6MWT), and high-resolution chest CT (HRCT) scanning pretreatment and every 6 weeks thereafter. A real-time safety workflow was implemented, with pre-specified criteria for halting treatment and escalation to the next block of treatment. All abnormal findings reported on chest CT by site radiologist were centrally reviewed in real time by the study pulmonologist (T.K.) and cases of suspected ILD were adjudicated promptly by a safety committee consisting of the pulmonologist and several medical oncologists, enabling early study drug interruption and therapeutic interventions. Toxicity data were evaluated in the first 51 patients treated with T-DXd plus rilve, and a preplanned hold was initiated until safety was ascertained by the data and safety monitoring board. Results: Safety data were evaluated in the first 51 patients receiving T-DXd plus rilve. Eight ILD events were confirmed (15.68% incidence): 5 grade 1, 2 grade 2, and 1 grade 3. One patient had an early chest CT scan because of dyspnea. All cases were identified by HRCT scans; PFTs and 6MWT did not enhance early detection and were subsequently dropped from the monitoring protocol. No patient was symptomatic for more than 24 hours. Radiographic findings resolved within 14-55 days. Patients with grade 2 & 3 ILD were treated with corticosteroids. No cases of ILD required intensive care or escalated therapy beyond steroids. Early treatment escalation to the subsequent block of therapy occurred in 3 patients; all 3 patients were subsequently able to receive appropriate therapy. At the meeting, safety data from all 100 patients treated with this combination will be reported. Conclusion: Real-time, centralized monitoring and adjudication allowed for early detection and prompt management of ILD, which both mitigated severity and supported timely therapeutic management. Among the monitoring tools studied, HRCT scanning performed every 6 weeks proved most effective for early ILD identification; PFT and 6MWT did not enhance early detection. This approach offers a potential framework for future studies assessing pulmonary safety in regimens combining agents that have a known risk for inducing ILD.
Presentation numberPS1-01-10
Chronic Immune-Related Adverse Events after neoadjuvant pembrolizumab in early-stage triple-negative breast cancer: A Real-World cohort analysis
Delphine Loirat, Institut Curie – Institute of Women’s Cancers, Paris, France
P. Matte1, I. Hacini2, P. H. Cottu1, C. Helal1, M. Mirabel3, F. Coussy1, F. Lerebours4, E. Laas5, E. Romano1, L. Djerroudi6, S. Hescot7, P. Charles8, B. Buecher1, J. Pierga1, L. Cabel1, D. Loirat1; 1Medical Oncology department, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 2Department of nuclear medicine and endocrinology,, Institut Curie – Institute of Women’s Cancers, Saint-Cloud, FRANCE, 3Department of Cardiology, Institut Mutualiste Monsouris,, Paris, FRANCE, 4Medical Oncology department, Institut Curie – Institute of Women’s Cancers, Saint-Cloud, FRANCE, 5Department of Surgery, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 6Department of Diagnostic and Theranostic Medicine, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 7Department of nuclear medicine and endocrinology, Institut Curie – Institute of Women’s Cancers, Saint-Cloud, FRANCE, 8Department of internal medicine, Institut Mutualiste Monsouris,, Paris, FRANCE.
Background: In early-stage triple-negative breast cancer (TNBC), the addition of immune checkpoint inhibitors (ICIs) such as pembrolizumab to (neo)adjuvant chemotherapy has significantly improved clinical outcomes, as demonstrated in the KEYNOTE-522 trial. This approach is now the standard of care in this population. However, the increasing use of ICIs in real-world settings raises concerns about chronic immune-related adverse events (irAEs), defined as toxicities persisting more than 12 weeks after treatment discontinuation. While chronic irAEs are well documented in melanoma and lung cancer, data remain scarce in early-stage TNBC, particularly in the curative setting. Methods: We conducted a retrospective, bicentric study (Institut Curie Paris and Saint-Cloud, France) involving patients with early-stage TNBC treated with pembrolizumab according to the KEYNOTE-522 regimen. All patients received at least one dose of neoadjuvant pembrolizumab. Chronic irAEs were assessed using CTCAE v5.0 criteria and defined as immune-related toxicities persisting beyond 12 weeks after pembrolizumab discontinuation, regardless of grade, starting from grade ≥2 (i.e., requiring medical intervention or impacting daily life). The primary endpoints were the incidence, type, duration, and management of chronic irAEs. Results: A total of 203 patients with stage II/III early TNBC were consecutively included between January 2021 and November 2023. The median age was 49 years (range: 22-74). Germline BRCA1 or BRCA2 mutations were identified in 14.8% of patients (n = 30), and 92.1% (n = 187) had invasive carcinoma of no special type. Postoperatively, 59% (n = 120) received at least one cycle of adjuvant pembrolizumab, while 41% (n = 83) did not, primarily due to limiting toxicities or the presence of residual disease. On average, patients received a total of 10 cycles of pembrolizumab (range: 1-17). The pathological complete response (pCR) rate was 70%. Chronic irAEs were reported in 36.5% of patients (n = 74). Endocrine toxicity was the most frequent, accounting for 79% (n = 58) of patients. Among these, hypothyroidism was observed in 62% (n = 36) and corticotropic insufficiency in 45% (n = 26); four patients experienced both. All endocrine events required long-term hormone replacement therapy, with no cases of functional recovery observed. Cardiac toxicity (myocarditis) was reported in 4.9% of patients (n = 10) and was managed with corticosteroids and standard cardioprotective therapies; no patient required second-line immunosuppression. Other chronic irAEs included gastrointestinal events in 2.0% (n = 4; 2 pancreatitis, 1 colitis, 1 gastritis), renal toxicity in 1.5% (n = 3; all interstitial nephritis), rheumatologic and neurologic events in 1.0% each (n = 2), pulmonary toxicity in 0.5% (n = 1; interstitial lung disease), and sicca syndrome in 1.0% (n = 2). Ten patients (4.9%) experienced multiple chronic irAEs. No deaths related to immune-mediated toxicity were observed.Conclusion: This real-world analysis reveals a high incidence of chronic irAEs among patients receiving curative-intent pembrolizumab for high-risk early-stage TNBC. These chronic irAEs can significantly impact quality of life, highlighting the urgent need for prospective studies and survivorship care planning for this growing population of breast cancer survivors. Funding: Collectif Triplettes Roses grant
Presentation numberPS1-01-11
Outcomes and Toxicity of Combination Chemotherapy and HER2-Targeted Therapy Versus HER2-Targeted Therapy Alone in Elderly Patients with HER2-Positive Breast Cancer
Maha Zafar, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY
F. Khan1, M. Zafar1, B. Singeltary2; 1Hospice and Palliative Medicine, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Hematology and Oncology, TriHealth Good Samaritan Hospital, Cincinnati, OH.
Background: The optimal approach for treating elderly patients (age ≥70) with HER2-positive breast cancer is uncertain, especially when balancing the benefits of combination chemotherapy with HER2-targeted agents versus targeted therapy alone. Given the increased risk of treatment-related toxicities in older adults, it is crucial to understand real-world outcomes and risks associated with these regimens in this population. Methods: We conducted a retrospective, real-world cohort study using the TriNetX platform. We identified two cohorts of patients aged ≥70 years with HER2-positive breast cancer. Cohort A (n=4,178) included patients who received HER2-targeted therapy (trastuzumab or trastuzumab deruxtecan) without concurrent chemotherapy (excluding docetaxel/paclitaxel). Cohort B (n=7,607) included those treated with HER2-targeted therapy in combination with chemotherapy (docetaxel or paclitaxel). Propensity score matching was performed, yielding 3,595 patients per cohort for comparative analyses. Outcomes were assessed with Kaplan-Meier survival curves and hazard ratios (HR) for mortality and major toxicities. Results: After 1:1 propensity score matching, 3,595 patients were included in each cohort. Mortality was slightly higher in the HER2-only group (25.96%) compared to the combination group (23.91%) (HR 1.136, 95% CI: 1.036-1.247, p=0.007). Hospitalization occurred more frequently in the combination group (36.2%) compared to the HER2-only group (30.3%; HR 0.830, 95% CI: 0.749-0.919, p=0.001). Outpatient visits were more frequent in the HER2-only group (56.1% vs. 32.6%; HR 1.649, 95% CI: 1.072-2.537, p=0.016). Rates of cancer progression were similar between groups (24.6% combination vs. 23.8% HER2-only; HR 1.004, p=0.951). Cardiomyopathy was more frequent in the combination group (12.7% vs. 10.4%; HR 0.834, p=0.012), while heart failure showed a non-significant trend towards higher frequency in the combination group (15.5% vs. 13.6%; HR 0.886, p=0.060). Treatment-related toxicities were consistently higher in the combination group, including neutropenia (16.3% vs. 6.0%; HR 0.359, p<0.001), anemia (35.1% vs. 24.9%; HR 0.661, p<0.001), thrombocytopenia (12.3% vs. 10.0%; HR 0.829, p=0.010), peripheral neuropathy (32.3% vs. 16.2%; HR 0.461, p<0.001), nausea/vomiting (33.0% vs. 21.7%; HR 0.608, p<0.001), mucositis (8.2% vs. 3.9%; HR 0.485, p<0.001), and diarrhea (32.1% vs. 20.9%; HR 0.598, p<0.001). Infections such as pneumonia, as well as heart failure with reduced ejection fraction (HFrEF) and severe sepsis, were also more frequent in the combination group, although not all differences reached statistical significance.The combination therapy cohort featured more complex, multi-agent treatment pathways, with nearly all patients receiving both HER2-targeted therapy and chemotherapy either concurrently or sequentially, and a small minority lacking documented treatment sequences. The HER2-only cohort showed simple, single-agent HER2-targeted therapy pathways. Conclusions: Among elderly patients with HER2-positive breast cancer, combination chemotherapy plus HER2-targeted therapy provided a modest but statistically significant improvement in survival compared to HER2-targeted therapy alone. However, this benefit was accompanied by substantially higher rates of hematologic, cardiac, neurologic, and gastrointestinal toxicities. Importantly, the risk of cancer progression was not significantly different between regimens. These results suggest that HER2-targeted therapy alone may be a reasonable and safer alternative in select elderly patients who are frail and have significant comorbidities, offering similar cancer control with significantly less toxicity.
Presentation numberPS1-01-12
Preventing Chemotherapy-Induced Peripheral Neuropathy with Acupuncture: Preliminary Results of a Pooled Analysis of Three Parallel Randomized Trials
Weidong Lu, Dana-Farber Cancer Institute, Boston, MA
W. Lu1, A. Giobbie-Hurder2, A. Tanasijevic1, S. Baedorf Kassis3, E. Diederich1, A. J. Sahraoui1, Y. J. Jeong4, I. H. Shin5, C. Yao6, X. Zhang6, N. Lee4, Y. Yao6, C. Bao7, T. Bao1, E. Yang8, R. Knoerl9, B. E. Bierer3, J. A. Ligibel1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 3The Multi-Regional Clinical Trials Center, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, 4Department of Surgery, Daegu Catholic University, School of Medicine, Daegu, KOREA, REPUBLIC OF, 5Department of Medical Statistics & Informatics, Daegu Catholic University, School of Medicine, Daegu, KOREA, REPUBLIC OF, 6Department of Breast Diseases, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, CHINA, 7Department of Acupuncture, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, CHINA, 8Osher Center for Integrative Health, Brigham and Women’s Hospital and Harvard Medical School, Brookline, MA, 9Health Behavior and Clinical Sciences, University of Michigan School of Nursing, Ann Arbor, MI.
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a common, debilitating side effect, adversely impacting quality of life (QoL) and treatment outcomes in breast cancer patients. Preventive strategies for CIPN remain limited. Acupuncture has shown promise in mitigating CIPN symptoms, but its role in prevention requires further investigation. This study evaluates acupuncture’s effectiveness in preventing CIPN in early-stage breast cancer patients through a pooled analysis of three coordinated international trials. Methods: Patients with stage I-III breast cancer, with no prior neuropathic symptoms, scheduled for taxane-based chemotherapy were randomized 1:1 to acupuncture or relaxation exercises with nature videos as the control in three parallel trials in the USA, China, and South Korea. Participants in the acupuncture group received 14 standardized sessions over 12 weeks, starting before the second chemotherapy cycle. Participants in the control group received nature video relaxation during chemotherapy infusions. Follow-up extended for an additional 12 weeks. The primary endpoint was the change in CIPN severity using EORTC CIPN-20 sensory subscale at 12 weeks. Secondary endpoints included CIPN incidence, pain intensity, and QoL at week 12 and week 24. Data were pooled and analyzed using longitudinal linear mixed-effects model, stratified by site and chemotherapy schedule (weekly vs. non-weekly). All analysis were intention to treat. Results: Of 129 patients who consented, 128 were randomized (USA: 88, China: 20, South Korea: 20) and 127 were analyzed (63 acupuncture, 64 control). Median age was 46 years (range: 30-80) in the acupuncture arm and 49 years (range: 26-77) in the control arm, with 58.2% White and 38.6% Asian. At baseline, patients reported similar CIPN-20 sensory scores between study arms [0.44 points (SE 0.77) vs. 1.29 (0.77), p=0.33]. At week 12, patients in both arms reported a statistically significant increase of mean sensory change score [7.23 (1.75), 95%CI: 3.76-10.70, p<0.0001; and 8.16 (1.67), 95%CI: 4.86-11.47, p<0.0001, respectively]. There are no statistically significant differences between the acupuncture arm vs. the control arm at week 12 and week 24 (Table). No acupuncture-related serious adverse events were observed. Conclusions: Acupuncture was safe and well-tolerated but did not significantly reduce the risk and severity of CIPN when compared to the nature video control in this pooled cohort. Secondary analyses are in progress and will be reported separately. Funding Source: The Ministry of Health & Welfare, Republic of Korea, Comprehensive and Integrative Medicine R&D project through the Korea Health Industry Development Institute (KHIDI) Grant Number: HI20C1753 Clinical Trial Registry Numbers: NCT05528263, ChiCTR2200066714, KCT0008470
| Acupuncture | Nature Video Control | ||||||||||||||
| Measurements | Time point | N | Mean (SE) | N | Mean (SE) | Difference in Change Scores (Acupuncture vs. Nature Video Control ) Estimate (95%CI) | p-value | ||||||||
| EORTC CIPN-20 Sensory subscale | Baseline | 63 | 0.44 (0.77) | 64 | 1.29 (0.77) | ||||||||||
| Change at Week 12 | 56 | 7.23 (1.75) | 62 | 8.16 (1.67) | -0.93 (2.42) (-5.72 to 3.86) | 0.7 | |||||||||
| Change at Week 24 | 55 | 6.93 (1.91) | 59 | 8.70 (1.84) | -1.77 (2.65) (-7.03 to 3.48) | 0.51 |
Presentation numberPS1-01-14
Comparing Functional Measures and Their Associations with Toxicity and Survival in Older Adults with Early Breast Cancer: Results from the Prospective Multicenter HOPE Study
Yuliya Zektser, UCLA David Geffen School of Medicine, Los Angeles, CA
Y. Zektser1, C. Sun2, W. Dale2, J. Ji1, N. V. Baclig1, H. J. Cohen3, H. Kim2, V. Katheria2, C. Lee1, M. Sedrak1; 1Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, 2Department of Supportive Care, Beckman Research Institute of City of Hope, City of Hope, Duarte, CA, 3Department of Medicine, Duke School of Medicine, Durham, NC.
Background: Clinicians infrequently use geriatric assessment in routine care, despite its superior ability to identify at-risk older adults with early breast cancer compared to clinician-rated Karnofsky Performance Status (cKPS). Asking patients to rate their own performance status may offer a more scalable alternative, but the relationship between patient-reported KPS (pKPS) and treatment outcomes is unknown. We hypothesized that pKPS would show stronger associations with treatment outcomes than cKPS but similar associations as geriatric assessment-based frailty. Methods: The HOPE study enrolled 499 women aged >65y with stage I-III breast cancer across 16 US sites who were planned to start neo/adjuvant chemotherapy. At baseline, we obtained a geriatric assessment-based Deficit Accumulation Frailty Index (DAFI; categorized as robust [DAFI <0.2] or prefrail/frail [DAFI >0.2]). cKPS and pKPS were collected and categorized as high [KPS>=80] or low [KPS<70] based on cutoffs consistent with prior studies. Our primary endpoint was grade 3+ toxicity. Secondary endpoints included treatment modifications (dose reductions, dose delays, early discontinuation) and survival (non-breast cancer-specific survival [non-BCSS]). Kappa statistics were used to evaluate agreement between cKPS and pKPS. Adjusting for age, race/ethnicity, stage, and regimen, multivariable logistic regression was used to evaluate associations of DAFI, cKPS, and pKPS with grade 3+ toxicity and treatment modifications, while Cox proportional and Fine-Gray regression were used to evaluate relationships with non-BCSS. Results: Of the 499 participants (median age 70 [65-86]), 21% were prefrail/frail, 4% had low cKPS, and 10% had low pKPS. cKPS and pKPS correlated weakly (kappa=0.16). In the adjusted multivariable model, DAFI was strongly associated with all treatment outcomes: grade 3+ toxicity, treatment modifications, and non-BCSS (Table). cKPS was only associated with toxicity and non-BCSS. pKPS was not associated with any outcomes. Table. Multivariable Associations Between Baseline Functional Status and the Odds of Toxicity and Survival Across Distinct Measures of Function
| Functional Measure |
Total N=499 No. (%) |
Grade 3+ Toxicity OR (95% CI) |
Dose Reduction OR (95% CI) |
Dose Delay OR (95% CI) |
Early Discont. OR (95% CI) |
Non-BCSS HR (95% CI) |
| DAFI | ||||||
| Robust (<0.2) | 394 (79) | 1.00 | 1.00 | 1.00 | 1.00 | 1.00 |
| Pre/frail (>0.2) | 105 (21) | 2.70 (1.66-4.40)* | 1.87 (1.12-3.15)* | 1.85 (1.10-3.13)* | 1.76 (1.05-2.95)* | 2.56 (1.08-6.05)* |
| cKPS | ||||||
| High (80+) | 477 (96) | 1.00 | 1.00 | 1.00 | 1.00 | 1.00 |
| Low (<70) | 22 (4) | 2.80 (1.08-7.26)* | 2.53 (0.98-6.55) | 1.68 (0.62-4.57) | 1.21 (0.44-3.33) | 4.35 (1.25-15.12)* |
| pKPS | ||||||
| High (80+) | 443 (90) | 1.00 | 1.00 | 1.00 | 1.00 | 1.00 |
| Low (<70) | 49 (10) | 1.73 (0.88-3.39) | 1.18 (0.58-2.44) | 1.38 (0.68-2.84) | 1.37 (0.67-2.81) | 1.14 (0.35-3.74) |
Adjusted for age, race/ethnicity, stage, and regimen. *P-value<0.05 Conclusion: Although pKPS offers a simpler, more scalable alternative to geriatric assessment, it was not associated with treatment outcomes in this study. Geriatric assessment-derived frailty remained the most robust measure, showing consistent associations with toxicity, treatment modifications, and survival, while cKPS showed limited associations. These findings underscore the need to prioritize the implementation of geriatric assessments in routine oncology practice to improve risk stratification and guide treatment decisions for older adults with breast cancer.
Presentation numberPS1-01-15
A real-world experience of treatment rechallenge after grade 1 trastuzumab-deruxtecan related interstitial lung disease: clinical outcomes and blinded imaging review
Kelsey H Natsuhara, University of California San Francisco, San Francisco, CA
K. H. Natsuhara1, M. Vella2, S. C. Behr2, N. Reddy3, A. J. Chien1, M. E. Melisko1, M. Majure1, G. C. Buckle1, E. J. Walker1, L. Al Rabadi1, A. Ko1, M. L. Cheng1, A. Algazi1, H. Batra-Sharma1, L. N. Quintal4, L. A. Huppert1, H. S. Rugo5; 1Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, 2Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 3Department of Surgery, University of California San Francisco, San Francisco, CA, 4Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, 5Breast Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA.
Background: Trastuzumab-deruxtecan (T-DXd) is approved for advanced HER2+, low, and ultra-low advanced breast cancer, and multiple other solid tumors. T-DXd carries a rare but serious risk of ILD (incidence 12-15%), requiring frequent imaging and symptom evaluation. For > grade (G) 2 ILD, guidelines recommend permanent drug discontinuation. For asymptomatic G1 ILD, drug is held with the option for rechallenge (RC) after resolution of imaging findings. Limited data exist on T-DXd related ILD imaging findings and clinical outcomes after RC in patients (pts) treated in the real-world setting. Methods: We analyzed all pts treated with T-DXd at our institution from Sept 2018-June 2024. We identified pts with radiographic evidence of ILD by chart review. Adjudication of T-DXd related ILD was by treating provider and graded via CTCAE v5. A chest radiologist conducted a blinded review of baseline and follow-up chest CTs (up to 4 mos post T-DXd) for all pts treated with T-DXd, to identify possible cases of drug-related ILD and assess imaging improvement over time. Results: Of 214 pts treated with T-DXd, 44 (21%) developed ILD (27-G1, 8-G2, 4-G3, 3-G4, 2-G5). Median (med) ILD onset was 116 days (d) after 1st dose [range (r:) 9-833]. Among pts with ILD, 37 had breast, 5 had GI, 1 had lung, and 1 had head and neck cancer, with a med age of 59 yrs (r: 36-78), med 3 prior therapy lines for advanced disease (r: 0-13); 8/44 pts (18%) had renal impairment (CrCl<60mL/min). Among pts with G1 ILD, 21/27 (78%) received steroids for a med of 49d (r: 15-149). Radiographic improvement was seen at a med of 23d (r: 9-79) for pts treated with steroids vs 66d (r: 28-82) without steroids. Of 27 pts with G1 ILD, 22 (81%) were RC with T-DXd; 4 had PD at ILD diagnosis, 1 was not RC due to persistent CT findings. Med time to RC was 42d (r: 20-236); 2 pts received intervening therapy before RC. After RC, 15 pts remained on T-DXd without recurrent ILD for a med of 95d (r: 20-365); 1 pt remained on T-DXd at data lock (6/26/25); 7 pts (32%) had recurrent ILD at a med of 47d after RC (r: 20-331; 5-G1, 2-G2). Four pts with recurrent G1 ILD were RC a 2nd time (2 dose reduced) and remained on T-DXd for a med of 152d (r: 92-370); 1 pt with recurrent G1 ILD, 3 with PD, 1 remained on T-DXd at data lock. Imaging review showed complete resolution of ILD in 4 of 27 pts with G1 ILD, partial response in 15, stable disease in 2, worsening ILD in 1, and 4 were unevaluable due to missing follow-up imaging. Med time from initial ILD diagnosis to complete or partial response was 32d (r: 9-209). All 7 pts with recurrent ILD after RC had partial imaging responses before RC. 22 pts had imaging findings consistent with possible ILD but were not diagnosed with or treated for T-DXd associated ILD. Per clinician assessment, imaging findings were attributed to PD (n=7), viral infection (n=5), aspiration/infection (n=4), radiation changes (n=2), and in 4 cases, ILD findings were not documented in the official report. These 22 pts remained on T-DXd for a med of 152d (r: 20-949) and were never clinically diagnosed or treated for ILD; 21pts stopped T-DXd for PD and 1 remained on T-DXd at data lock. Conclusions: In this real-world, single institution cohort, high rates of T-DXd RC after G1 ILD were reported with long duration of clinical benefit. After RC, low rates of low grade recurrent ILD were seen with no G5 events. Four pts with recurrent G1 ILD were RC a 2nd time with only 1 case of recurrent G1 ILD. Most pts with G1 ILD did not have complete resolution of ILD findings on CT. By imaging review, some patients had CT findings possibly concerning for ILD; however, providers identified alternative causes—underscoring the challenge of diagnosing T-DXd related ILD. This cohort provides clinically important information on RC safety and imaging characteristics associated with T-DXd related ILD.
Presentation numberPS1-01-16
Effectiveness of post-discharge management program of breast cancer patients based on Smart Cancer Care 2.0 using patient reported outcome, a preliminary analysis of a randomized clinical trial
Su-Jin Koh, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, Korea, Republic of
S. Koh1, J. Kwon2, Y. Noh3, J. Na1, J. Kim2, C. Kim4, D. Kang1, J. Seo5, M. Ock5; 1Department of Hematology and Oncology, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, KOREA, REPUBLIC OF, 2Department of Surgery, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, KOREA, REPUBLIC OF, 3Department of Radiation Oncology, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, KOREA, REPUBLIC OF, 4Department of Rehabilitation, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, KOREA, REPUBLIC OF, 5Department of Preventive Medicine, University of Ulsan, College of Medicine, Ulsan University Hospital, Ulsan, KOREA, REPUBLIC OF.
Title: Evaluating the effectiveness of post-discharge management program of breast cancer patients based on Smart Cancer Care 2.0 using patient reported outcome: a preliminary analysis of a randomized clinical trial
Authors: Su-Jin Koh, MD, PhD1*, Jin Ah Kwon2*, Young Ju Noh3, Jihyun Na1, Jin sung Kim2, Chung Reen Kim4, Dong Yoon Kang, MD, PhD5, Jeong-Wook Seo5, Minsu Ock, MD, PhD5† Affiliations:1Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea2Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea3Department of Radiation Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea4Department of Rehabilitation, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea5Department of Preventive Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Republic of Korea
†Corresponding authorsMinsu Ock, MD, PhDUlsan University Hospital, University of Ulsan College of MedicineDaehagbyeongwon-ro 25, Dong-gu, Ulsan 44033, Republic of KoreaEmail: ohohoms@naver.com
Funding: This study was supported by a grant from the National R&D Program for Cancer Control, Ministry of Health & Welfare, Republic of Korea (HA21C0107 and HA23C0478).Evaluating the effectiveness of post-discharge management program of breast cancer patients based on Smart Cancer Care 2.0 using patient reported outcome: a preliminary analysis of a randomized clinical trial
Introduction: In Korea, a “Smart Cancer Care” platform that utilizes patient reported outcomes has been developed to manage treatment side effects of cancer patients and resolve various unmet needs. In this study, we applied a cancer patient post-discharge management program based on Smart Cancer Care 2.0 to breast cancer patients and evaluated its effectiveness.Methods: Breast cancer patients receiving various treatments such as surgery, radiation therapy, and chemotherapy were randomly assigned to a group that received a cancer patient post-discharge management program (group A) and a group that did not (group B). The cancer patient post-discharge management program monitored and managed various treatment side effects that cancer patients may experience, and even provided information on cancer rehabilitation and health behavior improvement. After applying the program in group A for 3 months, breast cancer patients in both groups were followed up for up to 6 months to evaluate and compare health-related quality of life, unexpected visits to medical institutions, etc.Results: As of December 2023, a total of 190 breast cancer patients were registered, and 169 breast cancer patients were preliminary analyzed in this study. As a result of comparing health-related quality of life measured by the EORTC QLQ-C30, no difference in average scores between groups A and B was confirmed at 1 month (Group A: 65.4, Group B: 65.2) (Figure 1). However, the difference in scores at 3 months was statistically significant (Group A: 73.8 points, Group B: 64.7 points, p-value: 0.017). Furthermore, even at 6 months, the difference in scores was statistically significant (Group A: 75.7 points, Group B: 64.6 points, p-value: 0.027).Conclusions: The cancer patient post-discharge management program based on Smart Cancer Care 2.0 was estimated to be effective in improving the health-related quality of life of breast cancer patients.
Figure 1. Evaluation of the effectiveness of a cancer patient post-discharge management program through QLQ-C30 summary score
Keywords: Patient Reported Outcome Measures; Mobile Applications; Breast Neoplasms; Patient Discharge
Presentation numberPS1-01-17
Defining a Neuroinflammatory Signature of Chemotherapy-Induced Peripheral Neuropathy
Elaine P. Kuhn, Dartmouth-Hitchcock Medical Center, Lebanon, NH
I. S. Marchal1, E. P. Kuhn2, J. M. Mikecz3, P. A. Pioli4, L. T. Vahdat2; 1Medicine, Geisel School of Medicine, Hanover, NH, 2Medical Oncology, Dartmouth-Hitchcock Medical Center, Lebanon, NH, 3Research Data Services, Dartmouth College, Hanover, NH, 4Microbiology and Immunology, Geisel School of Medicine, Hanover, NH.
Background: Chemotherapy-induced peripheral neuropathy (CIPN) affects at least 50% of patients (pts) who undergo systemic therapy. CIPN not only impacts quality of life but also adversely affects survival by frequently causing dose reductions, delays, or premature cessation of therapy. While the best strategy for preventing CIPN symptoms is currently cryotherapy, the exact mechanism remains elusive. It is hypothesized that cryotherapy prevents toxicity by modulating the neuroinflammatory process. Therefore, we posited that by systematically profiling the neuroinflammatory microenvironment in sera of patients enrolled in a completed prospective CIPN trial, the mechanisms of CIPN and those patients who are at highest risk for its development could be identified. Methods: This cohort consists of 14 pts with metastatic breast cancer enrolled in a phase 2 study of ixabepilone in which the primary endpoint of the original trial was to define the effect of ixabepilone on the development of CIPN and on the ultrastructure of neurons evaluated by electron microscopy of serial skin biopsies. Pts enrolled in this trial underwent an extensive clinical neurologic evaluation every 3 weeks using the Total Neuropathy Score-clinical (TNSc), a validated tool that utilizes both physical exam and patient report, to assess CIPN. In this study, severe CIPN was defined as the presence of a TNSc score of 10 or greater, a score increase of 4 or more points from baseline, or peripheral neuropathy that led to dose reduction, delay, or cessation of therapy. Using these criteria, 5 patients with severe CIPN and 3 patients without severe CIPN had sufficient serum sample available for analysis from the completed trial. Serum samples were collected at baseline and over time throughout the study, 18 timepoints among the patients with severe CIPN and 10 timepoints among the patients without severe CIPN were evaluated. Next, neuroinflammatory biomarker concentrations of the banked patient serum samples were measured with the Luminex 48-plex assay and custom discovery panels, allowing for the levels of 80 cytokines, chemokines, and growth factors to be measured in duplicate. Diluent and serum were combined in a well, incubated with mixed micro-beads, and then re-incubated with detection antibody and later streptavidin-phycoerythrin. The labeled micro-beads were then re-suspended in sheath fluid and analyzed by the Luminex 100 system. The instrument readout of mean fluorescent intensity (MFI) was converted to pg/ml based on MFI values of standards run alongside the samples in the assay. Results: Seven cytokines were significantly differentially expressed between the two groups. The MFI of the chemokine CCL24 was decreased over 3.2-fold in the group with severe CIPN compared to the group without severe CIPN (p<0.0001). CXCL2 (p<0.01) and IL-12 (p<0.05) were also significantly decreased in the severe CIPN group. Four biomarkers were elevated in the group with severe CIPN compared to those without severe CIPN: CCL8 (p<0.05), CCL18 (p<0.05), CCL23 (p<0.05), and PDGF-AA (p<0.05). Conclusions: Together, these seven biomarkers reveal a novel neuroinflammatory signature of CIPN. Elevated serum levels of CCL18 and PDGF-AA in patients with severe CIPN are suggestive of chemotherapy-induced pro-fibrotic macrophage activation, which has been shown to exacerbate neuropathy. While CCL8 and CCL23 have been reported in the literature in relation to peripheral neuropathy, this study marks the first report identifying CCL24 in this context. Decreased expression of CCL24 may be associated with a negative feedback regulatory mechanism, in which increased occupancy of CCR3, the receptor for CCL24, by additional endogenous ligands leads to downregulation of CCL24 over time. These results will be validated in a forthcoming prospective trial.
Presentation numberPS1-01-18
Temperature-controlled hand-foot cooling prevents chemotherapy-induced polyneuropathy (cipn): a real-world data collection in 500 patients
Athina Kostara, MVZ Medical Center Duesseldorf, Duesseldorf, Germany
A. Kostara, T. Schaper, O. Opra; Gynecologic Oncology, MVZ Medical Center Duesseldorf, Duesseldorf, GERMANY.
Introduction: Advancements in oncology have significantly improved overall survival rates for cancer patients, particularly those with breast cancer. Consequently, the long-term side effects of chemotherapy—still a cornerstone of cancer treatment—on patients’ quality of life (QoL) are gaining increasing attention.Chemotherapy-induced peripheral neuropathy (CIPN) is a common adverse effect of taxanes (paclitaxel, nab-paclitaxel, docetaxel). CIPN can significantly reduce QoL and may lead to dose delays, reductions, or even treatment discontinuation. Reported incidence rates of grade 2-3 CIPN reach up to 50% with three-weekly and up to 30% with weekly taxane-based regimens, which are frequently used in breast cancer treatment.Temperature-controlled hand-foot cooling (Hilotherapy®) has shown potential in preventing CIPN. At our oncology department, Hilotherapy® is an established component of supportive care. We present here the results of our real-world data collection on 500 breast cancer patients who received prophylactic Hilotherapy® during chemotherapy. Methods: Temperature-controlled hand-foot cooling (Hilotherapy®) was used as a standard supportive care measure during each neurotoxic chemotherapy session. Hilotherapy® is a processor-controlled cooling system (Hilotherm ChemoCare CIPN) equipped with hand and foot cuffs that deliver continuous and precise cooling. The device uses distilled water and electricity to maintain the temperature.The standard procedure involved cooling the hands and feet for 30 minutes before, during, and 30 minutes after chemotherapy infusion. The device temperature was set to 15-17°C, resulting in a consistent skin surface temperature of 18-20°C.CIPN symptoms were assessed at each treatment cycle according to CTCAE v5.0 criteria. Patients were stratified by taxane dosing schedule (weekly vs. three-weekly). Follow-up data are currently being collected to evaluate long-term outcomes. Results: To date, 489 of the planned 500 patients have completed chemotherapy. Among them, 90.4% (n = 442) experienced only mild or no symptoms (CIPN grade 0-1), 9.4% (n = 46) developed CIPN grade 2, and only one patient (0.2%) experienced grade 3 CIPN.Key endpoints under evaluation include: Effectiveness of Hilotherapy® in preventing ≥ grade 2 CIPN; Time of first onset of CIPN; Dose reductions and/or treatment discontinuations due to CIPN; One-year follow-up data. Conclusion: Temperature-controlled hand-foot cooling (Hilotherapy®) effectively prevented ≥ grade 2 CIPN in over 90% of breast cancer patients. This method is a simple, well-tolerated, and easily implementable supportive care strategy. Patient satisfaction data will be presented in future analyses. Follow-up is ongoing to assess the long-term sustainability of the preventive effect.
Presentation numberPS1-01-19
Exploratory Study on the Efficacy and Safety of Combined Scalp Cooling and HairRepro MEDIα for Chemotherapy-Induced Alopecia in Japanese Breast Cancer Patients
Emi Kanaya, University of Toyama, Toyama city, Japan
E. Kanaya, K. Matsui, A. Urasaki, M. Furukawa, S. Nagasawa, M. Araki, K. Sukegawa, S. Sekine, T. Fujii; Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama city, JAPAN.
Chemotherapy-induced alopecia (CIA) is one of the most distressing side effects experienced by breast cancer patients undergoing adjuvant chemotherapy. In some cases, hair loss is incomplete or permanent (permanent CIA), significantly impairing quality of life, psychological well-being, and social reintegration. Although scalp cooling systems such as PAXMAN have shown effectiveness in reducing CIA, complete hair preservation is not achieved in all patients, and cases of permanent CIA still occur.HairRepro MEDI α is a novel scalp lotion containing a newly developed α-lipoic acid derivative, DHLH (histidine dithioloctamide with Na/Zn), which has been shown in preclinical studies to reduce inflammatory infiltration in the scalp induced by cytotoxic agents. A prior multi-institutional trial in Japan suggested its potential benefit in promoting hair regrowth among CIA-affected patients.This study is the first exploratory clinical trial in Japan to evaluate the combined use of the PAXMAN cooling system and HairRepro MEDI α, with the aim of improving not only CIA prevention but also post-chemotherapy hair regrowth and overall patient satisfaction. This retrospective exploratory study enrolled 22 breast cancer patients receiving adjuvant chemotherapy at our institution between 2022 and 2024. Chemotherapy regimens included HP+DTX→ddAC (n=8), HP+DTX (n=4), TC (n=2), ddAC→ddPTX (n=6), and others (n=2). HairRepro MEDI α was applied topically to the scalp and eyebrows at a dose of 4 mL twice daily, starting 6 days prior to the initiation of chemotherapy and continuing until day 90 post-final chemotherapy. PAXMAN scalp cooling was performed during each chemotherapy cycle. Photographs were taken at baseline (day −6), prior to each chemotherapy course, and on days 30, 60, and 90 after the final chemotherapy to assess hair loss percentage. Adverse events were recorded throughout. Among the 22 patients, the median hair loss at the final chemotherapy session was 45%, with 11 patients (50%) exhibiting Grade 1 or lower alopecia, indicating successful prevention. No adverse events related to the use of HairRepro MEDI α were observed.Hair loss typically began 4 to 6 weeks after the first chemotherapy dose. Compared to historical data from PAXMAN-only users, the onset of alopecia appeared delayed. Notably, all patients showed full scalp hair regrowth by 3 months post-chemotherapy, with hair loss scores returning to 0%, suggesting effective recovery.These findings indicate a potentially additive benefit of HairRepro MEDI α in both prevention and recovery from CIA when used alongside scalp cooling. The combination of PAXMAN and HairRepro MEDI α achieved favorable outcomes in the prevention and recovery of CIA among Japanese breast cancer patients receiving adjuvant chemotherapy. A 50% success rate for Grade ≤1 alopecia, along with complete hair regrowth by 3 months, suggests enhanced efficacy compared to previous studies involving scalp cooling alone. Notably, this is the first clinical report from Japan to evaluate this combination therapy, providing new insights into supportive care strategies tailored for Asian populations. Further randomized trials are warranted to confirm these preliminary findings and refine CIA management protocols.
Presentation numberPS1-01-20
Alopecia Associated with Antibody-Drug Conjugates and Impact of Cold Caps For Patients with Breast Cancer
Samer Alkassis, UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA
S. Alkassis, L. Zhang, E. Yang, M. Lipsyc-Sharf, K. McCann, N. McAndrew, A. Master, J. LaBarbera, M. Sedrak, N. Palaskas, K. Greene, M. Kuiper, J. Glaspy, D. Elashoff, R. Callahan, A. Bardia; Hematology/ Oncology, UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA.
Background: Antibody-drug conjugates (ADCs) such as trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG) have transformed the treatment of metastatic breast cancer (MBC). However, ADC-related alopecia remains poorly characterized. The effectiveness of scalp cooling in this setting is also unclear. Here, we evaluated the incidence and patterns of alopecia in patients (pts) with MBC treated with T-Dx and SG explored the potential protective effect of cold cap use. Methods: We identified pts with MBC treated with ADCs between June 2014 to June 2024 at UCLA using institutional databases. Pts were included based on ICD-9/10 codes for MBC, documentation of ADC use, and keywords related to scalp cooling discussion/ use in provider notes. Data were extracted on alopecia prior to ADC initiation (baseline alopecia), cold cap use during treatment, and alopecia development during ADC therapy based on clinical notes, patient self-report, and photographic documentation. Pts were stratified by pre-treatment alopecia status and ADC regimen (T-DXd, SG, or both sequentially). Those with alopecia attributed to neo/adjuvant chemotherapy administered at least one year prior to diagnosis of MBC were considered to have no baseline alopecia. Descriptive analyses were conducted to explore associations. Results: A total of 103 pts with MBC received ADC; T-DXd (n = 39), SG (n = 38), or both sequentially (n = 26). Disease subtypes included HR+/HER2− (36.8%), HR+/HER2+ (13.6%), HR−/HER2+ (12.6%), and TNBC (36.8%). Median age at MBC diagnosis was 54 years (range 26-80), and median duration of ADC treatment was 6.3 months (range 0-52.5). In the T-DXd group, 10/39 (25.6%) pts had baseline alopecia. Of the 29 pts without baseline hair loss, 10 (34.5%) developed alopecia while others did not have clear documentation of hair loss. Overall, 7 (of 29) pts without baseline alopecia used cold caps (4 [57.1%] developed alopecia) and 22 did not (6 [27.3%] had hair loss). In the SG cohort, 12/38 (31.6%) pts had baseline alopecia. Among the other 26 pts, 12 (46.2%) developed hair loss. Cold caps were used in 3 pts (2 [66.6%] had alopecia); of the 23 who did not use cooling, 10 (43.5%) experienced hair loss. Among the 26 pts who received both ADCs sequentially, 11 (42.3%) had baseline alopecia. Of the remaining 15 pts, 6 (40%) developed hair loss (5 during SG, 1 during T-DXd). One patient used a cold cap and did not have alopecia; among the 14 pts who did not use cooling, 6 (42.9%) developed alopecia. Conclusions: Alopecia is a common treatment-related toxicity among pts with MBC treated with T-DXd and/or SG. In this study, scalp cooling during ADC therapy was infrequent and appeared to offer limited protection against ADC-related alopecia. These findings underscore the need to develop innovative strategies to reduce hair loss risk with currently approved ADCs, as well as need to develop novel ADCs that do not confer alopecia risk for pts with breast cancer.
| Group | Patients Without Baseline Alopecia | Cold Cap Used | Alopecia w/ Cold Cap | No Cold Cap | Alopecia w/o Cold Cap | Total Alopecia |
| T-DXd Only | 29 | 7 | 4 (57.1%) | 22 | 6 (27.3%) | 10 (34.5%) |
| SG Only | 26 | 3 | 2 (66.6%) | 23 | 10 (43.5%) | 12 (46.2%) |
| Both Agents | 15 | 1 | 0 (0%) | 14 | 6 (42.9%) | 6 (40%) |
| All Groups Combined | 70 | 11 | 6 (54.5%) | 59 | 22 (37.3%) | 28 (40%) |
Presentation numberPS1-01-21
Smaller implant volume compared to native breast volume increases capsular contracture risk
Hyoung Won Koh, Seoul National University Bundang Hospital, Seongnam, Korea, Republic of
H. Koh, K. Yoon, H. Shin, E. Kim; Surgery, Seoul National University Bundang Hospital, Seongnam, KOREA, REPUBLIC OF.
Purpose: This study aimed to identify clinical, surgical, and treatment-related predictors of capsular contracture (CC) following immediate implant-based breast reconstruction after mastectomy, with particular emphasis on the influence of implant size relative to resected breast volume. Methods: We conducted a retrospective review of 465 patients who underwent nipple- or skin-sparing mastectomy and immediate reconstruction (direct-to-implant or two-stage) between 2004 and 2020. CC was defined as Baker grade II or higher. Capsular contracture-free interval (CCFI) was measured from final implant placement to first documented CC. Univariable and multivariable logistic regression identified CC risk factors. CCFI was assessed by Kaplan-Meier analysis and Cox proportional-hazards modeling. The optimal implant-to-breast weight ratio cut-point was determined using maximally selected rank statistics (surv_cutpoint). Results: Over a median follow-up of 63.7 months, 50 of 465 patients (10.8%) developed CC. In multivariable logistic regression, subpectoral placement (OR 3.28; 95% CI 1.05-10.21; P = 0.041) and postmastectomy radiotherapy (PMRT) (OR 4.94; 95% CI 2.27-10.74; P < 0.001) remained independent predictors of CC, whereas implant undersizing < 80% did not reach significance (OR 2.14; 95% CI 0.90-5.10; P = 0.085). In time-to-event analysis, PMRT alone independently shortened the capsular contracture-free interval (CCFI) (HR 4.04; 95% CI 2.11-7.74; P < 0.001). A maximally selected cut-point of 69.1% implant-to-breast ratio optimally stratified CC risk: patients with ratios ≤ 69.1% had significantly lower 1- and 5-year CCFI (84.2% and 75.8% vs. 96.5% and 90.3%; log-rank P < 0.001) and a threefold increased CC hazard in both unadjusted (HR 3.25; 95% CI 1.63-6.50; P < 0.001) and adjusted Cox models (HR 3.02; 95% CI 1.47-6.21; P = 0.003). Conclusion: Implant undersizing, particularly implant volumes ≤ 69.1% of resected breast volume, together with subpectoral placement and PMRT, are independent risk factors for capsular contracture. Careful matching of implant size to native breast volume may reduce CC risk in immediate reconstruction.
Presentation numberPS1-01-22
Genomic Insights into Anthracycline Cardiotoxicity: Enhancing Survivorship Care through Early Risk Identification
Pooja P Advani, Mayo Clinic, Jacksonville, FL
P. P. Advani, A. McPherson, J. Reddy, J. Schneider, S. Baheti, T. Nguyen, J. C. Ray, N. Norton; Hematology and Oncology, Mayo Clinic, Jacksonville, FL.
BACKGROUND: Anthracycline chemotherapy-related cardiomyopathy (CCM) is a serious adverse event that can occur several years after completion of breast cancer (BC) therapy. Demographic and clinical risk factors have failed to predict which patients will experience CCM. Genetic variants can account for a significant proportion of inter-individual variation. Knowledge of genetic risk variants prior to chemotherapy will be informative for risk stratification and early intervention. The goal of our study is to identify genetic variants that predispose patients to CCM.METHODS: We developed a cardiotoxicity registry at Mayo Clinic, Florida that enrolls patients who are to undergo standard of care chemotherapy in the early BC setting. Patients were (and continue to be) enrolled and consented for chart review and DNA sequencing. We sequenced whole exomes of the first 136 patients (anthracycline N=55, anti-HER2 (no anthracycline) N=71, other chemotherapy, N=10), primarily focusing on Titin (TTN) truncating variants, known to be present in ~25% of patients with primary dilated cardiomyopathy, and previously reported in 7.5% of patients with CCM, followed by exploration of increased burden of rare non-synonymous variants in 60 established genes for primary cardiomyopathy. Filtering of rare non-synonymous variants included minor allele frequency <0.5% (in GnomAD database European, African and Asian ancestries) and CADDv1.7 PHRED score ≥20.RESULTS: 18/55 (33%) patients treated with anthracycline experienced CCM. TTN truncating variants in were identified in 2/18 (11%) CCM patients and absent in 37 patients who did not experience CCM. Both patients with TTN variants had severe HF and required cardiac transplant. We identified a pathogenic variant in LMNA (p.Arg190Gln) in 1/18 (5.5%) patients and the same rare (p.Glu1127Gly) variant in RYR2 occurred in 2/18 (11%) of patients (Table 1). We noted enrichment of rare missense variants in anthracycline CCM patients compared to HER2 treated without anthracycline, CCM, p=0.0001.CONCLUSIONS: Pathogenic variants in TTN, LMNA, are enriched in BC patients (17%) who develop anthracycline-related cardiomyopathy. Early genetic screening may improve risk stratification and guide personalized cardiac monitoring to prevent CCM. Further research is needed to clarify the role of additional rare variants like RYR2 and to develop a CCM risk score based in idiopathic CM genes. Incorporating genetic risk assessments into treatment planning has the potential to enhance oncologic outcomes as well as long-term survivorship by preserving cardiac health.
| (Likely) pathogenic variant | Anthracycline CCM, N=18 | Anthracycline No CCM, N=37 | Anti-HER2 (no anthracycline) CCM, N=7 | Anti-HER2 (no anthracycline) No CCM, N=64 | Other chemotherapy, No CCM, N=10 |
| TTN frameshift (A-band) | Asn29967Metfs27, Thr29949Serfs11 (11%) | None | None | None | None |
| LMNA missense | Arg190GIn (5.5%) | None | None | None | None |
Presentation numberPS1-01-23
Dose Adjustments and Adherence to Adjuvant Abemaciclib in a Brazilian Real-World Scenario
Renata Colombo Bonadio, Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
R. Colombo Bonadio1, L. Holland1, C. A. Cavalcanti1, J. Bessa1, K. Cayres2, P. do Amor Divino1, R. Naves1, J. Bines3, L. Testa1; 1Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 2Oncology, Instituto D’Or de Pesquisa e Ensino, Recife, BRAZIL, 3Oncology, Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, BRAZIL.
Background: The use of abemaciclib in combination with endocrine therapy (ET) is an established adjuvant option for patients with hormone receptor-positive (HR+), HER2-negative early-stage breast cancer at high risk of recurrence. However, the prolonged treatment duration (up to two years) in the curative setting presents unique challenges related to tolerability and adherence, particularly given the known toxicity profile of CDK4/6 inhibitors. Diarrhea, fatigue, and hematologic adverse events are common and may lead to dose interruptions or permanent discontinuation. Real-world data are essential to better understand patient tolerance, frequency of dose adjustments, and their impact on treatment persistence and outcomes. We aimed to evaluate adjuvant abemaciclib use in a Brazilian real-world setting, focusing on adherence and the need for dose modifications. Methods: We retrospectively reviewed records of patients with high-risk, early-stage HR+ breast cancer who received adjuvant abemaciclib between 2019 and 2025. The endpoints evaluated included: initial dose, treatment interruptions, dose reductions, final maintenance dose, permanent drug discontinuation, and reasons for discontinuation. Results: A total of 189 patients were included, with a median age of 47 years (range 24-79); 58.7% were premenopausal and 37.0% postmenopausal. Most tumors were ductal (70.4%) or lobular (15.9%) in histology, stage II (45.0%) or III (38.1%), grade 2 (51.8%) or grade 3 (33.3%), and had a Ki-67 index ≥ 20% (77.2%). Nearly half of the patients (52.9%) had received neoadjuvant chemotherapy. For adjuvant ET, most patients (86.8%) initiated treatment with an aromatase inhibitor, and 53.9% received ovarian suppression.The majority (93.3%) initiated abemaciclib at the label dose of 300 mg daily (150 mg twice daily). At the time of analysis, 35 patients (18.5%) had completed the planned two years of treatment. In the overall cohort, dose interruptions occurred in 27.6% of cases and dose reductions in 43.1%. Maintenance dosing after adjustment was: 300 mg daily in 58.2%, 200-250 mg in 26.8%, 150 mg in 3.9%, and <150 mg in 11.1%. Permanent discontinuation due to adverse events was required in only 6.3% of patients, and all but one had undergone prior dose reductions. Grade 3 or higher diarrhea and neutropenia were registered in 6.3% and 7.4% of the cases, respectively. Among 152 patients with available adherence data, 91.5% reported good adherence (defined as taking ≥70% of prescribed doses). Conclusions: Our findings align with other real-world studies showing frequent need for dose adjustments during adjuvant abemaciclib therapy. Nevertheless, permanent discontinuation due to toxicity was infrequent, suggesting that proactive management through dose reductions can effectively support treatment persistence. Given that reduced doses do not appear to compromise efficacy, close monitoring of adverse events and timely dose adjustments are essential to maintain adherence and maximize benefit in the adjuvant setting.
Presentation numberPS1-01-24
Association Between Cumulative Symptom Burden and Cognitive Difficulties Among Breast Cancer Survivors: A Cross-Sectional Study
Xiaotong Li, Memorial Sloan Kettering Cancer Center, New York, NY
X. Li1, K. Lampson1, K. Liou1, Y. Li2, Q. Li1, T. Ahles3, J. Root3, J. Mao1; 1Integrative medicine service, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Epidemiology and Biostatistics; Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, 3Neurocognitive Research Lab, Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: Cancer-related cognitive difficulties (CRCD) is a multifaceted condition associated with various comorbid symptoms, making its management challenging. This study aims to evaluate cumulative symptom burden and its association with subjective and objective CRCD in breast cancer survivors. Method: We analyzed baseline data from a randomized clinical trial for addressing CRCD. The study included women with a history of stage 0-III breast cancer, currently free of oncologic disease, who reported moderate or greater CRCD and insomnia, as indicated by an Insomnia Severity Index score≥8. Subjective CRCD was assessed using the perceived cognitive impairment subscale from the Functional Assessment of Cancer Therapy-Cognitive Function (FACT-Cog PCI). Objective CRCD was measured using the normed delayed T score from the Hopkins Verbal Learning Test—Revised (HVLT-R). Comorbid insomnia, fatigue, pain, anxiety, and depression were measured using Insomnia Severity Index, Brief Fatigue Inventory, PROMIS-Global pain item, and Hospital Anxiety and Depression Scale. We calculated a Cumulative Symptom Score based on co-morbid symptom severity to evaluate the cumulative symptom burden. Multivariable linear regression models were used to assess the associations between cumulative symptom burden and CRCD, adjusted for clinical and sociodemographic variables. Results: Among the 260 participants, the mean age was 56.6 years (SD 10.4), with 54 (21.0%) identifying as non-white and 26 (10.0%) as Hispanic survivors. The mean FACT-Cog PCI score and HVLT-R delayed T-score was 36.0 (SD 14.0) points and 46.0 (SD 12.7) points, respectively. A high prevalence of moderate to severe comorbid symptoms was observed in this population: fatigue (90%) being the most common, followed by insomnia (70.0%), pain (45.2%), anxiety (35.4%), and depression (11.5%). In multivariate analyses adjusted for age and chemotherapy, a higher Cumulative Symptoms Score was significantly associated with greater subjective CRCD (Coef. = -3.0, 95% CI = -3.6 to -2.4, p<0.001). Each comorbid symptom was also correlated with subjective CRCD (all p<0.001). However, the Cumulative Symptoms Score was not associated with objective CRCD (Coef. = -0.2, 95% CI = -0.9 to 0.5, p = 0.58); only insomnia had a significant correlation (r = -0.20, p<0.001). Conclusion: Higher cumulative symptom burden was associated with subjective perception of cognitive difficulties but not objective cognitive function among breast cancer survivors; however, insomnia was associated with worsened objective functions. Comprehensive symptom management has the potential to improve subjective appraisal of cognitive difficulties; targeted treatment of insomnia has the potential to improve objective cognitive functions in women with breast cancer.
Presentation numberPS1-01-25
Let’S talk about sexuality in cancer care: a cross-sectional analysis of a multicenter survivorship program.
Camila Chiodi, Oncoclínicas&Co, Salvador, Brazil
C. Chiodi1, F. Santos Dumont Sorice2, T. Santana3, F. Oliveira4, M. Mathias Machado5, R. Brant Costa2, M. de Azevedo Kalile1, C. Pavei6, M. Borges Bittencourt6, D. Strassburger Nunes2, H. Pantoja7, T. de Melo Passarini2, M. Couto8, M. Behrends Pinto9, C. Perini10, C. Cerqueira Mathias1, M. Laloni6, M. Cruz Rangel Silva2, C. Alves Costa e Silva1, L. Landeiro1; 1Oncoclínicas Bahia, Oncoclínicas&Co, Salvador, BRAZIL, 2Oncoclínicas Minas Gerais, Oncoclínicas&Co, Belo Horizonte, BRAZIL, 3Oncoclínicas Sergipe, Oncoclínicas&Co, Aracaju, BRAZIL, 4Oncoclínicas Espirito Santo, Oncoclínicas&Co, Vitória, BRAZIL, 5EBMSP, Escola Bahiana de Medicina e Saúde Pública, Salvador, BRAZIL, 6Oncoclínicas São Paulo, Oncoclínicas&Co, São Paulo, BRAZIL, 7Oncoclínicas Rio de Janeiro, Oncoclínicas&Co, Rio de Janeiro, BRAZIL, 8Oncoclínicas DF, Oncoclínicas&Co, Brasília, BRAZIL, 9Oncoclínicas Rio Grande do Sul, Oncoclínicas&Co, Porto Alegre, BRAZIL, 10Oncoclínicas Paraná, Oncoclínicas&Co, Curitiba, BRAZIL.
Background: Sexual dysfunction is a highly prevalent yet frequently underrecognized and undertreated issue among breast cancer survivors (BCS). Despite its impact on quality of life, discussions and management strategies are often lacking in routine care. This study examines post-treatment sexual symptoms, their association with emotional well-being, and partner relationships in BCS. Methods: We analyzed data from breast cancer survivors enrolled in “OC sobre VIVER”, a multicenter survivorship program coordinated by the Oncoclínicas Group (Brazil), representing one of the few structured survivorship care models in Latin America. Participants were 3-18 months post-active treatment. Structured interviews assessed sexual symptoms (vaginal dryness, dyspareunia, low libido), emotional status (self-esteem, depression, anxiety), and partner communication. Data was managed via REDCap. Descriptive and bivariate analyses were performed using GraphPad Prism, version 10. Results: From July 2023-March 2025, 391 women (median age 50 years [range: 30-86]) were enrolled. Of 376 women with staging data, 360 (95%) had invasive breast cancer (stage I: 55%, II: 33%, III: 12%). Notably, 92% (349/381) expressed a desire to address sexual health concerns during clinical encounters. Sexual dysfunction was prevalent: low libido in 67% (253/376), vaginal dryness in 65% (236/363), and dyspareunia in 31% (116/374). Psychosocial findings included low self-esteem in 31% (118/383) of participants, anxiety in 58% (225/387), and depression in 27% (104/387). Poor partner communication (among partnered) was reported in 10% (27/264). Dyspareunia was significantly associated with anxiety symptoms (p = 0.0003), but not with cancer stage, depression, or communication quality (p > 0.05). Low libido was more frequent with low self-esteem (75% vs. 64%, p = 0.043), higher stage (stage III: 15% vs. 4%, p = 0.008), hormonotherapy use (71% vs. 56%, p = 0.012) and showed a trend toward higher prevalence with poor partner communication (78% vs. 63%, p = 0.143). No associations with age, relationship status, or chemotherapy use (p > 0.05). Conclusions: Sexual symptoms were highly prevalent in this BCS cohort and encompassed both physical and psychosocial factors, including emotional distress and partner communication. These findings highlight critical opportunities for intervention. Routine assessment of sexual health and the integration of tailored, multidisciplinary strategies should be prioritized in survivorship care.
Presentation numberPS1-01-26
Associations between Financial Toxicity and Illness-related Communication among Advanced Breast Cancer Patients and their Spouses
William Nicholas Riley, MD Anderson, Houston, TX
W. N. Riley1, A. Ahn1, J. Kroll2, M. Chavez Mac Gregor3, M. R. Jones1, G. Smith1, K. Milbury1; 1Behavioral Science, MD Anderson, Houston, TX, 2Palliative Care medicine, MD Anderson, Houston, TX, 3Health Services Research, MD Anderson, Houston, TX.
Background: Patients with advanced-stage breast cancer and their spouse/romantic partners experience various challenges throughout their cancer journeys, including economic hardships sustained from treatment known as financial toxicity. Financial toxicity has been associated with poor physical and mental health, with some findings suggesting a bidirectional effect of financial toxicity on the dyad’s wellbeing. Previous literature indicates couples’ open and supportive communication behavior is associated with improved cancer-related adjustment. However, these associations have not yet been studied in the context of financial toxicity. This study aims to identify prevalent financial concerns, examine convergence and divergence in the reported financial concerns between patients and their spouse, and explore the associations between the couples’ communication behaviors and psychosocial wellbeing. Methods: The current cross-sectional data are part of a longitudinal study examining the psychological and behavioral impact of financial stress on patient-caregiver dyads. Eligible participants included women with stage III-IV breast cancer and their spouse. Upon enrollment, dyads completed separate baseline assessments for financial toxicity (COST), depressive (CES-D) and anxiety (GAD-7) symptoms, relational wellbeing (DAS-7), and positive and negative affect (PANAS). Afterwards, participants engaged in a standardized 30-minute spousal interaction task, where each dyad member identified and discussed a finance-related concern with a focus on problem solving for the selected issues. Results: 85 dyads (n=2 same-sex dyads; > $100,000 income: 64%; DAS-7 < 21: P=15%, S=12%; ≥Grade 1 COST: P=52%, S=47%) completed the spousal interaction task. Concerns regarding treatment side effects (P. = 19%, S. = 28%), future financial stability (P = 13%, S=20%) and discussing financial stressors with each other (P = 12%, S = 19%) were rated as most frequent concerns for both members of the dyad. Yet participants tended to avoid selecting some of the most pressing financial concerns as a discussion topic for the spousal interaction task. For instance, although spouses reported maintaining a steady income as a frequent concern (20%) but rarely (5%) discussed this topic during the interaction task. Changes in both positive and negative affect after participating in discussion were compared using paired t-tests. A significant reduction in negative affect (P < 0.001) was found for spouses, but not for patients. Significant correlations were found between COST to DAS-7, GAD-7 and CES-D for patients and spouses. For patients only, significant correlations were found between COST to Cancer Communication Assessment Tool (CCAT) and discrepancy and Social Provisions Scale (SPS) total score. Discussion: The results of this study support the notion that even well-adjusted, affluent couples may benefit from professional assistance in discussing and solving financial concerns. For the issues listed as most frequently concerning that remained unaddressed, there may be several underlying explanations (e.g. a perceived lack of control, a desire to avoid confrontation, etc.) for why dyad avoid discussion for certain topics. However, our results demonstrated that couples’ problem-solving discussions regarding financial toxicity decreased – rather than increased – negative affect in caregivers, indicating that dyads should embrace discussion of financial topics they may find distressing. We recommend that healthcare providers consider encouraging this behavior in breast cancer dyads to reduce stress and negative affect overall.
Presentation numberPS1-01-27
Guided cannabis care and improvements in PROMIS metrics among cancer patients: a prospective assessment
Brooke Worster, Jefferson Health, Philadelphia, PA
B. Worster1, K. Parton2; 1Supportive Oncology, Jefferson Health, Philadelphia, PA, 2Senior Data Modeler, EO Care, Boston, MA.
Guided cannabis care and improvements in PROMIS metrics among cancer patients: a prospective assessmentBackground:Cancer-related symptoms such as pain, sleep disturbances, anxiety, and gastrointestinal (GI) complaints often negatively impact patients’ quality of life. Patient-Reported Outcomes Measurement Information System (PROMIS) instruments were developed to quantify these domains and provide standardized, validated measures for clinical interventions. Given the growing interest in cannabis as an adjunctive therapy, we sought to determine whether guided cannabis care could enhance PROMIS-defined symptom outcomes among individuals with active cancer diagnoses.Methods:A total of 52 patients were enrolled from multiple oncology centers, dedicated marketing efforts, and various cancer advocacy organizations. At baseline, each patient completed PROMIS assessments for Pain Interference, Sleep Disturbance, Anxiety, and GI Nausea & Vomiting, as well as an intake survey covering health history and other personal information. Using a Bayesian model built from existing research and real-world data, a cannabis care plan was developed by integrating patient profiling inputs into individualized recommendations for products, dosages, and times of use. A clinician reviewed the plan before sending it to the patient. Patients then had scheduled follow-up surveys every 1–2 weeks to monitor efficacy, tolerability, and potential side effects. Regimens were adjusted as needed, based on patient feedback, to optimize therapeutic benefit and minimize side effects. Outcomes were evaluated at 4 weeks and again at 12 weeks. This approach allowed investigators to track changes in PROMIS metrics and assess any correlation between cannabis interventions and improved symptom management.Statistical Analysis:The primary endpoint looked at the proportion of patients achieving clinically meaningful improvement – defined as at least a 3-point reduction in T-score from baseline in PROMIS domains. At 4 and 12 weeks, the percentage of patients seeing meaningful improvement was calculated.Results:After 4 weeks of guided cannabis care (n=52), patients reported notable improvements across all measured PROMIS domains:Pain Interference: 69% of patients achieved clinically meaningful improvement.Sleep Disturbance: 75% saw improvements in sleep-related symptoms. Anxiety: 81% experienced reductions in anxiety.GI Nausea & Vomiting: 92% reported meaningful improvement.For those who continued treatment through 12 weeks (n=21), the proportion of patients demonstrating meaningful improvement increased further:Pain Interference: 88%Sleep Disturbance: 80%Anxiety: 100%GI Nausea & Vomiting: 100%Only 15% of patients experienced side effects, which included dizziness, anxiety, feeling overly intoxicated, or elevated heart rate. Qualitative feedback indicated that patients appreciated having structured guidance on which cannabis products to use, how to dose, and how to adapt their regimens to optimize their outcomes. This close monitoring and individualized adjustment appear to have supported sustained engagement and symptom relief.Conclusions:These findings suggest that guided cannabis care may significantly improve cancer-related symptoms as measured by PROMIS, a validated patient-centered metric. The high rates of clinically meaningful improvement in domains such as pain, sleep, anxiety, and GI symptoms underscore the potential role of guided cannabis use as an adjunct therapy.
Presentation numberPS1-01-28
Navigating a path back to work: a cross-sectional analysis of a multicenter survivorship program.
Maria Luisa Mathias Machado, Escola Bahiana de Medicina e Saúde Pública, Salvador, Brazil
M. Mathias Machado1, T. Santana2, F. Santos Dumont Sorice3, F. Oliveira4, C. Chiodi5, R. Brant Costa6, M. de Azevedo Kalile7, C. Pavei8, M. Borges Bittencourt8, D. Strassburger Nunes6, H. Pantoja9, T. de Melo Passarini6, M. Behrends Pinto10, M. Couto11, C. Perini12, C. Cerqueira Mathias5, M. Laloni8, M. Cruz Rangel Silva6, C. Alves Costa e Silva5, L. Landeiro5; 1EBMSP, Escola Bahiana de Medicina e Saúde Pública, Salvador, BRAZIL, 2NOS, Oncoclínicas Sergipe, Aracaju, BRAZIL, 3Cancer Center Oncoclinicas, Oncoclínicas Minas Gerais, Belo Horizonte, BRAZIL, 4Cancer Center Oncoclinicas, Oncoclínicas Espirito Santo, Vitória, BRAZIL, 5NOB Ondina, Oncoclínicas Bahia, Salvador, BRAZIL, 6Oncoclínicas Minas Gerais, Oncoclínicas&Co, Belo Horizonte, BRAZIL, 7Oncoclínicas Bahia, Oncoclínicas&Co, Salvador, BRAZIL, 8Oncoclínicas São Paulo, Oncoclínicas&Co, São Paulo, BRAZIL, 9Oncoclínicas Rio de Janeiro, Oncoclínicas&Co, Rio de Janeiro, BRAZIL, 10Oncoclínicas Rio Grande do Sul, Oncoclínicas&Co, Porto Alegre, BRAZIL, 11Oncoclínicas DF, Oncoclínicas&Co, Brasília, BRAZIL, 12Oncoclínicas Paraná, Oncoclínicas&Co, Curitiba, BRAZIL.
Introduction: With the growing number of cancer survivors, new challenges are emerging for this population. Among them, returning to work often represents a significant hurdle due to persistent physical and psychological toxicities resulting from both the disease and its treatment. Understanding the factors associated with work reintegration is key to promoting long-term quality of life and socioeconomic stability for breast cancer survivors (BCS). Methods: We analyzed data from breast cancer survivors enrolled in “OC sobre VIVER”, a multicenter survivorship program coordinated by the Oncoclínicas Group (Brazil), one of the few structured survivorship care initiatives in Latin America. Participants were 3-18 months post-active treatment. Structured interviews assessed employment status (work interruption during treatment, and return to work afterward), psychosocial aspects (supportive psychosocial network, self-esteem, emotional distress, anxiety, and depression), and treatment-related toxicities (cognitive deficits, arthralgia, pain, motor limitations, and neuropathy). Data was managed via REDCap. Descriptive and bivariate statistical analyses were performed using GraphPad Prism, version 10.Results: From July 2023-March 2025, 391 women were enrolled with a median age of 50 years (range: 30-86). Among 376 participants with available staging data, 360 (95%) had invasive breast cancer, distributed as follows: stage I (55%), stage II (33%), and stage III (12%). Employment status analysis revealed that 40.9% (158/386) of participants discontinued work during treatment. Among these, 56% (n=88) successfully returned to work, and 28% (n=43) required workplace accommodations to resume employment. Overall, 55% (197/356) were working at the time of data collection. BCS who received chemotherapy (CT) tended to have lower return-to-work rates compared to those who did not (48% vs. 62%, p = 0.093). In contrast, patients treated exclusively with hormonotherapy (HT) had a trend toward higher return-to-work rates compared to those who received both CT and HT (66% vs. 52%, p = 0.144). Similarly, BCS reporting symptoms of depression showed lower return-to-work rates than those without depression (44% vs. 56%, p = 0.117). Significantly lower return-to-work rates were also observed among participants who experienced treatment-related toxicities, including motor limitation (39% vs. 57%, p = 0.016), neuropathy (41% vs. 59%, p = 0.015), arthralgia (43% vs. 66%, p = 0.0004), and cognitive impairment (42% vs. 58%, p = 0.049). No statistically significant associations were found with tumor stage (p = 0.572), type of breast surgery (quadrantectomy vs. unilateral or bilateral mastectomy; p = 0.702), HER2-targeted therapy (p = 0.454), immunotherapy (p = 0.710), pain (p = 0.112), supportive psychosocial network (p = 0.844), emotional distress (p = 0.887), anxiety symptoms (p = 0.507), or low self-esteem (p = 0.202). Conclusion: The return to work after breast cancer treatment remains a significant challenge for many survivors, often contributing to psychosocial distress and financial toxicity. The development of structured survivorship care programs that proactively address physical, cognitive, and psychosocial sequelae is essential to support and facilitate a successful and sustainable return to the workforce.
Presentation numberPS1-01-29
Real-world immune toxicity and survival outcomes by age in triple-negative breast cancer patients receiving pembrolizumab
Grace Gorecki, Allegheny Health Network, Pittsburgh, PA
G. Gorecki1, O. Abioye1, K. Babu1, R. Srinishant1, N. Gupta1, C. Hilton2; 1Internal Medicine, Allegheny Health Network, Pittsburgh, PA, 2AHN Cancer Institute, Allegheny Health Network, Pittsburgh, PA.
In triple-negative breast cancer (TNBC), pembrolizumab combined with chemotherapy has become a standard of care in both neoadjuvant and adjuvant settings, as established by the KEYNOTE-522 trial. Despite these clinical advances, immune checkpoint inhibitors (ICIs) are associated with immune-related adverse events (irAEs) that can affect multiple organ systems and result in substantial morbidity. In other cancer types, older patients have been shown to experience higher rates of irAEs, potentially due to age-related immune dysregulation and a higher burden of comorbidities, while younger patients often face distinct survivorship challenges. However, real-world data on age-specific irAE patterns and outcomes in TNBC patients receiving pembrolizumab are lacking. To date, no studies have specifically evaluated the differential impact of pembrolizumab-based therapy on irAE incidence, severity, and outcomes across age groups in TNBC populations.Methods This is a retrospective cohort study using TriNetX, TNBC patients treated with pembrolizumab between 2016 and 2024. Two age-stratified cohorts were created: younger (≤54 years) and older (≥55 years) of 1,661 patients per group after propensity score match. irAEs covering endocrine, hepatic, pulmonary, hematologic, and gastrointestinal toxicities were identified. Corticosteroid therapy and hospitalization were assessed as markers of toxicity severity. Kaplan-Meier survival curves were used.Results Older patients had a higher incidence of irAEs at 1 month (OR 1.43, 95% CI 1.01-2.04, p=0.048) and 3 months (OR1.43, 95% CI 1.11-1.79, p=0.0049), mainly due to thyroiditis (OR 1.67, 95% CI 1.08-2.13 p=0.017). Differences in irAE rates were no longer significant at 6 or 12 months. Despite lower irAE rates, younger patients had higher dexamethasone use at 3 months (OR 1.64, 95% CI 1.1-2.3, p=0.0047) and significantly worse survival (log-rank p=0.0071; HR 1.4, 95% CI 1.1-1.9). A composite marker of corticosteroid use and hospitalization showed younger patients consistently experienced more severe toxicities and poorer survival at all timepoints. At 1 month: OR 1.45, 95% CI 1.03-2.17, p=0.03; HR 1.3, 95% CI 1.01-1.9, log-rank p=0.04. At 3 months: OR 1.57, 95% CI 1.13-2.1, p=0.0087; HR 1.4, 95% CI 1.08-1.88, log-rank p=0.0105. At 6 months: OR 1.38, 95% CI 1.0-1.9, p=0.048; HR 1.3, 95% CI 1.03-1.69, log-rank p=0.027. At 12 months: OR 1.57, 95% CI 1.13-2.1, p=0.0063; median survival 171 days for younger vs. not reached in older group (log-rank p=0.0023; HR 1.49, 95% CI 1.1-1.8). At 3 months, total corticosteroid use and hospitalization remained higher in younger patients (OR 1.4, 95% CI 1.02-2.1, p=0.0359; HR 1.36, 95% CI 1.01-1.83; log-rank p=0.046). At 12 months, younger patients still had lower median survival (212 days vs. not reached; log-rank p=0.11; HR 1.2, 95% CI 0.95-1.59). While all-cause mortality was higher in older patients (p<0.0001), hospice referrals were similar between groups.ConclusionsIn this real-world cohort of TNBC patients treated with pembrolizumab, older patients experienced a higher incidence of early irAEs—particularly thyroiditis—but demonstrated superior survival. In contrast, younger patients had fewer documented irAEs but showed greater corticosteroid use, more frequent hospitalizations, and significantly worse survival across all timepoints when toxicity severity was considered. These findings suggest that irAE incidence alone may not fully capture clinical burden or risk and that age-related differences in immune response, toxicity manifestation, and management practices may contribute to divergent outcomes. Further investigation is warranted to elucidate the immunologic mechanisms of aging in the context of ICI use and safety to guide toxicity monitoring and supportive care strategies.
Presentation numberPS1-01-30
Protein Intake, Exercise, and Chemotherapy-Related Fatigue in Women with Breast Cancer
Isabella Victoria Critchfield-Jain, Saint Louis University, St Louis, MO
I. V. Critchfield-Jain1, M. A. Fiala2, Y. Park2, E. Salerno3, L. Peterson2; 1School of Medicine, Saint Louis University, St Louis, MO, 2Division of Oncology, Washington University School of Medicine, St. Louis, MO, 3Institute of Public Health, Washington University School of Medicine, St Louis, MO.
Background: For individuals with breast cancer undergoing chemotherapy, a nutritious, protein-rich diet may reduce side effects and improve outcomes. While emerging evidence supports the role of protein in managing cancer-related fatigue, few studies have focused on non-metastatic breast cancer or considered physical activity. This study examined the relationship between protein intake, physical activity, and fatigue in women with Stage I-III breast cancer receiving chemotherapy. Methods: This is a secondary analysis of the Genetics, Molecular Mechanisms, and Symptom Science (GEMMS) study, a longitudinal cohort of women undergoing curative-intent chemotherapy with follow up for 5 years. Protein and total caloric intake were assessed at baseline using the Automated Self-Administered Dietary Assessment (ASA24). Suboptimal protein intake was defined as 55 indicating clinically significant fatigue. Linear and logistic regression models evaluated associations between protein intake, exercise, and fatigue post-chemotherapy completion while adjusting for age, stage of cancer, chemotherapy intensity, and baseline fatigue. Results: A total of 95 women, median age 53 years [IQR 42-63] were included. Of these, 39% (n = 37) had suboptimal protein intake, with median protein intake of 0.88g/kg [IQR 0.60-1.12]. The characteristics of women with optimal and suboptimal protein intake are compared in Table 1. At chemotherapy competition, 41% (n = 39) reported clinically significant fatigue. This was numerically higher among women with suboptimal protein intake (43.2% compared to 39.7%) but did not reach statistical significance. In multivariable models, neither protein intake nor exercise were associated with fatigue. Baseline fatigue was the only consistent predictor: for each 1-unit increase in baseline fatigue T-score, the odds of post-treatment fatigue increased by 10% (aOR 1.10; 95% CI 1.03-1.17; p = 0.003). Conclusion: Protein intake and exercise were not significantly associated with fatigue during chemotherapy; however, baseline fatigue was a strong predictor of post-treatment fatigue. Future studies may benefit from exploring the micronutrient composition of dietary intake to discover other contributing factors to chemotherapy fatigue.
Presentation numberPS1-03-01
Secondary prevention of cancer therapy-induced thrombocytopenia with hetrombopag in breast cancer: a prospective,multi-center,self-controlled exploratory trial
Min Yan, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
H. Sun1, H. Lv1, W. Chen2, Y. Zhao3, M. Zhang1, L. Niu1, Z. Liu1, X. Guo1, X. Chen1, Y. Feng1, L. Wang1, H. Zeng1, J. Wang1, Y. Cui1, M. Yan1; 1Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, CHINA, 2Breast Cancer Centre, The Third Hospital of Nanchang, Nanchang, CHINA, 3Breast Oncology, Union Hospital, Tongji Medical College, Wuhan, CHINA.
Background: Cancer therapy-induced thrombocytopenia (CTIT) is a common complication of anti-tumor therapy and causes treatment delays or interruptions, increased bleeding risk and compromised outcomes. We aimed to evaluate hetrombopag, an oral thrombopoietin receptor agonist that prevented CTIT in breast cancer patients.Methods: In this multi-center, prospective exploratory trial (NCT05394285), breast cancer patients with platelet counts (PLT) 100×10⁹/L, which was defined as the thrombocytopenia treatment phase (TTP). Eligible patients were scheduled to continue the same anti-tumor regimen in their subsequent treatment cycle (Cycle 2). The secondary prevention phase (SPP) was initiated using a self-controlled design, with prophylactic hetrombopag (7.5 mg/day for 14 days) administration beginning one day after the start of Cycle 2. The primary endpoint was the response rate during the SPP, defined as the proportion of patients meeting all predefined criteria before the next treatment cycle (Cycle 3): 1. no platelet transfusion requirement; 2. no anti-tumor treatment modification (≥20% dose cut, ≥5 days delay, or discontinuation) ; 3. absence of severe thrombocytopenia (PLT <25×10⁹/L, or PLT <50×10⁹/L for ≥7 days).Results: Between Sep 2022 and May 2025, 67 breast cancer patients were enrolled in the study. After excluding 1 patient who withdrew informed consent, 66 patients comprised the full analysis set (FAS). During the trial, 6 patients discontinued participation in the SPP, the remaining 60 patients completed both TTP and SPP, forming the per-protocol (PP) population. In the FAS, 51 (77.3%) patients received antibody-drug conjugates (ADCs) monotherapy, 14 (21.2%) received regimens containing chemotherapy (RCC), and 1 (1.5%) with targeted therapy plus ADC. In the PP population, the response rate in the SPP was 85.0% (51/60). During the TTP, 86.7% (26/30) of patients in the hetrombopag group achieved response, compared to 80% (24/30) in the rhTPO group. No treatment-emergent severe adverse events occurred.Conclusions: As the first prospective trial to evaluate hetrombopag for preventing CTIT in breast cancer patients, this self-controlled exploratory study demonstrated hetrombopag may be a promising option for the secondary prevention of CTIT. The promising response rate observed during the TPP, coupled with a favorable safety profile, supports further large-scale investigation of hetrombopag for the prevention of CTIT in multi-cycle anticancer regimens.
Presentation numberPS1-03-02
Modulation of Inflammatory Gene Expression by Mind-Body Transformations Therapy (MBT-T) in Breast Cancer Patients
Stefania Cocco, INT IRCSS Fondazione G. Pascale, naples, Italy
S. Cocco1, M. Cozzolino2, M. Piezzo1, R. Caputo1, G. Celia3, D. Barberio4, V. Abate4, M. De Filippo5, V. Vaira6, A. Calabrese1, C. Della Bella1, A. Leone7, P. Mussnich De Freitas1, E. Di Gennaro7, A. Budillon8, M. De Laurentiis1; 1Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, INT IRCSS Fondazione G. Pascale, naples, ITALY, 2Department of Human, Philosophical and Educational Sciences, University of Salerno, salerno, ITALY, 3Department of Psychology and Health Sciences, University of Pegaso, naples, ITALY, 4Departmental Structure of Clinical Psycho-oncology, INT IRCSS Fondazione G. Pascale, naples, ITALY, 5Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, ITALY, 6Department of Pathophysiology and Transplantation, University of Milan, Milano, ITALY, 7Experimental Pharmacology Unit, INT IRCSS Fondazione G. Pascale, naples, ITALY, 8Scientific Directorate, INT IRCSS Fondazione G. Pascale, naples, ITALY.
Psychotherapeutic interventions, particularly those based on mind-body approaches, have demonstrated the ability to modulate inflammatory responses by downregulating the expression of inflammation-related genes and proteins. This has particular clinical relevance in breast cancer, where chronic inflammation is a well-established contributor to disease progression and resistance to therapy. Mind-Body Transformations Therapy (MBT-T) is an evidence-based therapeutic protocol designed to harness natural biological rhythms and biological plasticity in order to modulate gene expression. By reducing stress-induced dysfunctions, MBT-T facilitates the activation of endogenous healing mechanisms through the establishment of an optimal mind-body state. In our previous study (MBT-T Protocol 11/17) we demonstrated that MBT-T significantly reduced the serum levels of multiple cytokines and chemokines in breast cancer patients who had completed loco-regional treatment and adjuvant chemotherapy, compared to a matched control group not exposed to MBT-T. To further elucidate the molecular basis of these effects, we conducted a NanoString gene expression analysis on RNA extracted from blood samples collected from participants enrolled in the MBT-T study. The nCounter® Human Inflammation Panel, comprising 249 genes implicated in inflammatory and immune-related pathways, was used to evaluate gene expression changes at the end of the treatment (Tt) compared to baseline (T0). Matched timepoints were also analyzed in control patients. Consistent with our previous findings, MBT-T-treated patients exhibited a generalized downregulation of inflammatory gene expression at Tt compared to T0. Notably, we observed a significant reduction in the expression of genes associated with the interferon (IFN) signaling pathway, including OASL, OAS2, STAT2, IFIT1, IFIT3, and MX1. In contrast, the control group did not display significant modulation of inflammatory or IFN-related gene expression across timepoints. These findings are particularly meaningful given the complex role of the IFN pathway within the tumor microenvironment, where it influences immune cell dynamics, tumor cell behavior, and response to therapy. Elevated expression of IFN-related genes—such as OASL, OAS2, STAT2, and the IFIT family members—has been associated with immune evasion, angiogenesis, and tumor aggressiveness in several malignancies, including breast cancer. Collectively, our data suggest that MBT-T, through the reduction of psychophysiological stress, can downregulate key inflammatory signaling pathways, particularly those mediated by type I interferons. This modulation may influence tumor progression, reduce the risk of relapse, and enhance therapeutic responsiveness in breast cancer patients. These findings support the integration of mind-body interventions as adjunctive strategies in oncological care, with potential biological impact at the molecular level.
Presentation numberPS1-03-03
Patient experience with intramuscular vs oral endocrine therapy in metastatic breast cancer
Erica Fortune, Cancer Support Community, Washington DC, DC
E. Fortune1, A. Newell1, R. Speck2, A. Gilligan2, M. Gonzalo1, A. Searles Vitko3; 1Research, Cancer Support Community, Washington DC, DC, 2HEOR Oncology, Global Medical Affairs, Eli Lilly and Co, Indianapolis, IN, 3Oncology, Medical Affairs, Eli Lilly and Co, Indianapolis, IN.
Background: Endocrine therapy (ET) is commonly used in the treatment of breast cancer, including both oral and intramuscular (IM) options. While ET is an effective treatment for breast cancer, the two administration modes may generate disparate burdens and benefits on patients’ everyday lives. This study aims to explore metastatic breast cancer (MBC) patients’ experiences with ET by describing the burdens (including challenges to administration and adherence), benefits, and preference of oral and IM ET. Method: Adults in the US with a history of taking both oral and IM ET consecutively for their MBC for at least 3 months completed an online survey. Participants were recruited via email and in-person at Cancer Support Community’s national network of 190 locations and 14 advocacy groups. Participants provided sociodemographic information and clinical history. Survey content included assessment of treatment-related considerations for oral and IM ET administration (i.e., convenience, pain, time, transportation, remembering, cost, access, distress, impact on eating) on a 5-point scale of burden vs. benefit (major or minor burden, neutral, major or minor benefit), and preference for oral vs IM ET. Participants also rated the importance, on a 5-point scale (not at all important to very important), of 11 factors when considering their MBC treatment options. Data collection is ongoing; results and conclusions are based upon interim data. Results: Interim data include responses from 100 women with MBC with a mean age of 54 years (range: 29-78), 69% non-Hispanic White, 24% urban. The average time since metastatic diagnosis was 6.6 years (median = 6). Approximately 58% of participants lived more than 30 minutes away from a treatment center. Duration on ET was longer for oral (median = 28 months) than for IM (median = 17 months); 76% indicated they started oral prior to IM ET. More than 80% of participants responded that oral ET was less painful to take, required less transportation and time, and was more convenient compared to IM ET. Most (65%) reported that oral ET interfered less with daily life compared to IM (17%). Top benefits of oral ET included ease of access (71% [major or minor benefit]), minimal time commitment (64%), and convenience (60%), while the stress or anxiety associated with having to take daily medication was the most common burden (37% [major or minor burden]). For IM ET, the main benefit reported was ease of remembering (60%), while the most frequently reported burden was pain or discomfort from the injection (84%). When comparing treatment modalities, 63% preferred oral, 18% preferred IM, 12% expressed no preference, and 7% were unsure. Self-reported adherence was better for IM vs oral ET, with 84% and 57% reporting they never missed a single dose, respectively. Among the 41% that missed at least one dose for oral ET, 88% missed ≤3 times a month. When asked about treatment priorities, nearly all participants (98%) found the ability to slow disease progression when considering treatment options an important priority (quite important or very important) followed by the ability to cure the disease (87%) and impact of treatment on daily life (85%). Conclusions: Findings suggest a clear preference for oral over IM ET, with most participants citing ease of access, convenience, and minimal disruption to daily life as benefits to oral ET. However, having to take oral medication daily was associated with stress and anxiety for some. Although self-reported adherence was better for IM ET, missing a single dose has different implications for the IM ET, which is given every 28 days, rather than daily like oral ET. While benefits and burdens are observed for both modes of administration, oral therapy is clearly preferred. These findings support the need for shared decision-making to help ensure treatment decisions are optimized with patient preferences.
Presentation numberPS1-03-04
Predictive Factors of Thyroid Adverse Events in Neoadjuvant Immunotherapy for Triple-Negative Breast Cancer
Yumeng Li, First Hospital of China Medical University, Shenyang, China
Y. Li, Y. Cheng, L. Ji, Y. Wang, L. Yao, H. Zhang, H. Dong, M. Wang, X. Yingying; Department of Breast Surgery, First Hospital of China Medical University, Shenyang, CHINA.
Background:Thyroid dysfunction is among the most common immune-related events (irAEs) of immune checkpoint inhibitors (ICIs). This study aimed to investigate the prevalence, risk factors, and prognostic implications of thyroid irAEs in triple-negative breast cancer (TNBC) patients receiving neoadjuvant anti-PD-1 therapy combined with chemotherapy, based on data from the TREND phase II prospective clinical trial.Methods: The TREND study is a phase II prospective clinical trial involving TNBC patients treated with tislelizumab combined with nab-paclitaxel, followed by epirubicin-cyclophosphamide before surgery. Data on demographics, thyroid function, anti-thyroid autoantibodies were collected, and univariate and multivariate logistic regression models were used to identify risk factors. Peripheral immune analysis and whole-exome sequencing were conducted to explore underlying mechanisms associated with thyroid irAEs. Results: Among the 50 patients analyzed, 14 (28%) developed thyroid irAEs. Multivariate analyses revealed that baseline positivity of thyroglobulin antibody (OR 9.311, p=0.011) and lower levels of free triiodothyronine levels (OR 0.056, p=0.034) are independent predictors of thyroid irAEs. Peripheral immune analysis revealed that patients with thyroid irAEs exhibited elevated levels of CD4+ T cells, CD8+ T cells and CD16+CD56+ NK cells, along with higher levels of CD4+T cells before the onset of thyroid irAEs. Additionally, increased tumor mutational burden, and mutations in KLF17 and LRIG1 were observed in patients with thyroid irAEs.Conclusions: This study highlights the risk factors and underlying pathogenesis associated with thyroid irAEs in TNBC patients treated with neoadjuvant anti-PD-1 therapy combined with chemotherapy. These findings may assist clinicians in identifying patients at elevated risk for irAEs during ICI therapy, thereby improving patient management in clinical practice.Keywords:Thyroid immune-related adverse events; neoadjuvant immunotherapy; triple-negative breast cancer; predictive biomarkers
Presentation numberPS1-03-06
Radiation therapy for breast cancer in patients with Li-Fraumeni syndrome (LFS): a single institution cohort study
Adnan Antoine Shrebati, Institut Curie, Paris, France
A. A. Shrebati1, P. Loap1, E. Mouret-Fourme2, K. Cao1, M. Belotti2, D. Stoppa-Lyonnet2, Y. Kirova1; 1Department of Radiation Oncology, Institut Curie, Paris, FRANCE, 2Department of Genetics, Institut Curie, Paris, FRANCE.
Background: Li-Fraumeni syndrome (LFS), caused by monoallelic germline pathogenic variant (PV) in TP53 gene, causes an elevated risk of developing certain cancers, mainly early onset breast cancer, sarcoma and brain tumors. While LFS patients are believed to be at an increased risk for radiation-induced sarcomas, only a few retrospective studies have been published studying the impact of radiationtherapy in the treatment of breast cancer among these patients. This study reviews the data concerning LFS patients treated at the Institut Curie with adjuvant radiation therapy for localized breast cancer. Methods: A retrospective study was conducted on female breast cancer patients with confirmed TP53 PV treated at the Institut Curie. Data concerning patients’ characteristics, treatments received, radiotherapy if received, rates of recurrences, rates of radiation induced sarcomas (RIS) and patient outcomes were collected. Results: From 1989 to 2024, we identified 47 female LFS breast cancer patients who met our inclusion criteria. Median age at diagnosis was 31 years (range 18-72). Of them, 76.6% had a family history of cancer suggestive of LFS. 31.9% had tumoral HER2 amplification. 51.1% received a mastectomy and 31.9% received a lumpectomy. 23 patients (48.9%) received radiotherapy as a part of their treatment strategy. The median follow up was 111 months (range 7-413). Among patients who received radiotherapy, 3 (13%) had a known diagnosis of LFS prior to treatment. No case of radiation-induced sarcomas was observed during the follow-up period. In the radiotherapy group, 5 patients (21.7%) later developed a malignancy within the irradiated field. At 10 years post-diagnosis, overall survival did not differ significantly between patients who received radiotherapy and those who did not (58.2% [95%CI: 36.8-91.9] vs. 80.1% [95%CI: 64.2-100]). Similarly, there was no significant difference in loco regional recurrence-free survival (95.2% [95%CI: 86.6-100] vs. 74.0% [95%CI: 54.8-99.9]) or in survival free from subsequent non-breast cancers (59.6% [95%CI: 36.5-97.2] vs. 67.2% [95%CI: 49.8-90.7]). Univariate analysis did not find any correlation between hormonal status, grade, surgical technique, stage, use or non-use of radiotherapy and overall survival, cancer-specific survival, loco regional recurrence-free survival, metastasis-free survival. Conclusion: The incidence of radiation-induced sarcomas and local recurrences in LFS breast cancer patients was lower than anticipated. Further studies with larger cohorts and extended follow-up are required to validate these findings.
Presentation numberPS1-03-07
The association between adverse childhood events and pain, subjective cognitive decline and psychosocial distress over time in patients treated for early breast cancer.
Ayelet Shai, RAMBAM Health care Campus, Haifa, Israel
A. Alpert1, E. Sher-Cenzor2, M. Makarov1, A. Shai1; 1Oncology, RAMBAM Health care Campus, Haifa, ISRAEL, 2School of Psychology, University of Haifa, Haifa, ISRAEL.
Background: Chronic pain, subjective cognitive decline (SCD) and psychosocial distress (PD) are reported by > 40-60 % of patients recovering from breast cancer (BC). We previously reported that adverse childhood experiences (ACEs) are associated with increased pain and SCD before treatment. The effect of ACEs on pain and SCD during follow up has not been explored. Methods: We are conducting a prospective study to assess if ACEs increase chronic adverse effects in patients with localized BC and identify mediating mechanisms. Patients with stage I-III BC, age 18-75 without a history of prior cancer are recruited before starting (neo)adjuvant oncological treatment and assessed every 6 months by validated questionnaires, including ACEs, the brief pain inventory, FACT-COG and FACT-ES (measuring PD). Blood samples for inflammatory biomarkers are stored. This sub-study tested if ACEs are associated with pain, measured as a composite of pain intensity and interference, SCD and PD 6 months after treatment initiation, and with changes in these symptoms over time. Results: This sub-study included 116 patients with data collected 6 months from treatment initiation. Missing data ranged from 0.9% (n = 1) in ACEs to 9.5% (n = 11) in PD at 6 months and were imputed using the expectation maximization algorithm, as data were missing completely at random, Little’s MCAR test (229) = 242.51, p = .258. Of study participants, 68.1% were treated with chemotherapy, 62.9% had an academic degree, and 97.4% were employed prior to diagnosis. Univariate analysis did not detect an association between demographic or treatment factors and outcomes at 6 months (p’s > .14). Correlation analyses indicated that a higher number of ACEs was associated with more pain, SCD, and PD across time points (r range = .19 – .31, ps = < .048). A repeated-measures MANCOVA followed by Univariate analyses was conducted to examine changes over time in pain, SCD, and PD. Analyses included the number of ACEs (as a covariate), chemotherapy status (between-subjects variable), and time (within-subjects variable). The multivariate effect of time was significant, indicating that patients’ functioning declined over the 6 months, [Wilk’s lambda = .95, F (3,111) = 6.80, p < .001, partial η² = .16]. Specifically, pain [F (1,113) = 4.59, p = .034, partial η² = .04] and SCD [F (1,113) = 16.14, p < .001, partial η² = .13], but not PD [F (1,113) = 3.24, p = .074, partial η² = .03], increased between baseline and 6 months. ACEs had a significant main effect, F (3,111) = 4.81, p = .003, partial η² = .12. , and a higher number of ACEs was associated with worse pain, higher SCD, and higher PD on both timepoints [Fpain (1,113) = 13.43, p = .009, partial η² = .11; FSCD (1,113) = 6.97, p = .009, partial η² = .06; and FPD (1,113) = 7.96, p = .006, partial η² = .07]. The interaction between time and ACEs was not significant (p = .210) As expected, the interaction between time and chemotherapy was significant [F (3,111) = 3.50, p = .018, partial η² = .09; Univarite F’s (1, 113) > 4.14, p’s .03].Conclusion: Increasing numbers of ACEs predicts poor functioning of BC patients in terms of pain, SCD, and PD across the first 6 months post-diagnosis.
Presentation numberPS1-03-08
Breast Edema Following Breast Cancer Surgery: A Systematic Review of Risk Factors, Diagnosis and Management
Jasmine El-Taraboulsi, Imperial College Healthcare NHS Trust, London, United Kingdom
J. El-Taraboulsi1, C. Rossou2, M. Boland3, D. Leff1, S. Cleator4, V. Harding4, P. Thiruchelvam1; 1Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, UNITED KINGDOM, 2Department of Surgery and Cancer, United Lincolnshire Hospitals NHS Trust, Boston, UNITED KINGDOM, 3Department of Surgery and Cancer, St Vincent’s University Hospital, Dublin, IRELAND, 4Department of Oncology, Imperial College Healthcare NHS Trust, London, UNITED KINGDOM.
Introduction: Breast edema is a frequently under-reported and increasingly prevalent complication of breast cancer surgery. Currently, no unified definition or standardised diagnostic criteria exists, and there is limited evidence regarding its effective management and preventative strategies. The current literature suggests that breast edema is a long-term sequela of treatment, with a significant negative impact on patients’ quality of life. Methods: A systematic review of the literature was conducted according to PRISMA guidelines, examining the risk factors, diagnostic criteria, and management of breast edema following breast cancer surgery. Two independent reviewers conducted a comprehensive search of the Cochrane Library, PubMed, EMBASE, and additional sources from January 2000 to June 2025. Results: An initial keyword search identified 269 studies; following the application of eligibility criteria, 23 studies were included, encompassing 3,806 female patients with a mean age of 58.5 years-old, and an average BMI of 27.9. Of the included patients, 93.4% (n = 3,555) had undergone breast-conserving surgery, 4.4% (n = 168) have had mastectomies with free-flap or expander prosthesis reconstructions, and 2.2% (n = 83) have had therapeutic or prophylactic mastectomies without reconstruction. One study investigated breast edema in oncoplastic surgery, reporting this as a risk factor. Of the studies included, 21 reported on breast edema following adjuvant radiotherapy, despite inconsistencies in reporting, the most commonly cited radiotherapy schedule was 50Gy/25#. 39.1% (n = 9) of the articles investigated risk factors of breast edema, patient-related factors such as increased BMI and breast cup size were consistently reported as positive predictors of breast edema. There were conflicting findings with regards to tumour size and location, nodal involvement, and the extent of axillary surgery in relation to breast edema. Adjuvant radiotherapy and tumour bed boosts were associated with increased breast-edema, peaking at three to six months post-radiotherapy. Further potentially modifiable risk factors such as wound infection, and segmental post-operative scar-tissue burden were also proposed across studies. The reported incidence of breast edema following adjuvant radiotherapy was extremely variable, ranging from 7.1% to 75.5%, with high discordance based on the variability of definition and diagnostic methods. 43.5% (n = 10) of included studies focussed on the diagnosis of breast edema, including patient-reported outcomes measured with breast-edema related questionnaires, clinical examination, and imaging. Emerging investigations include dermal thickness measurement using High Resolution Ultrasound (HRUS) and tissue dielectric constant (TDC) as an accurate, non-invasive, and efficient quantitative measures of breast edema. Various studies have reported an inter-breast TDC ratio >1.40 as diagnostic for breast edema, while consensus has yet to be reached for the dermal thickness diagnostic threshold. The management and prevention of breast edema remains largely under-reported, with 17.4% (n =4) of included studies reporting this. Management options investigated in this review include compression garments, structured exercise programs and manual lymphatic drainage, though evidence supporting their efficacy remains inconsistent. Conclusion: Breast edema is a commonly overlooked but profoundly debilitating complication of breast cancer surgery, its aetiology is multifactorial, complex and distinct to breast-cancer related lymphedema of the arms. Improved prevention strategies, more timely and accurate diagnosis, and further assessment of more treatments is essential to optimise patient outcomes and quality of life.
Presentation numberPS1-03-09
Symptoms, survivorship concerns, and alleviation methods among young adults newly diagnosed with early-stage breast cancer
Kate E. Dibble, Dana-Farber Cancer Institute, Boston, MA
J. Stal1, K. E. Dibble1, S. M. Rosenberg2, C. Snow1, A. H. Partridge1; 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Department of Population Health Sciences, Weill Cornell Medicine, New York, NY.
Background: Many adolescents and young adults (AYAs) with early-stage breast cancer experience acute and chronic symptoms and survivorship concerns. Approaches used by AYAs to alleviate unmanaged symptoms and concerns and their perceived effectiveness remain poorly understood. This study qualitatively explored patient-reported symptoms, survivorship concerns, and alleviation methods reported by female AYAs newly diagnosed with early-stage breast cancer enrolled in a supportive care intervention study. Methods: Women (aged 18-39) newly diagnosed (<3 months) with early-stage (0-III) breast cancer at Dana-Farber Cancer Institute who were enrolled in Young, Empowered, and Strong (NCT04379414), a study testing a web and app-based supportive care intervention, were invited to participate in a virtual semi-structured qualitative interview. The interview guide was developed to explore experiences with management of symptoms and survivorship concerns. Areas of inquiry included burdensome symptoms and concerns, and challenges managing them. Transcripts were thematically analyzed. Results: AYAs (N=20) were, on average, 35 years at diagnosis (range: 33-39) and 1 year from diagnosis at interview (range: 0.65-1.77); most were non-Hispanic white (75%), partnered (80%), had stage I (40%) or II (40%) disease. Symptoms: Physical symptoms were the most frequently reported symptoms (e.g., menopausal symptoms, fatigue, nausea, pain, concentration/memory) by AYAs. For example, regarding pain, one AYA stated, “The mastectomy, the dissection, combined with the radiation side effects. I still experience a lot of pain in that area.” Methods to alleviate symptoms: Various methods were effective at alleviating (e.g., oncologist support, time) or somewhat alleviating (e.g., cold/heat therapy, medical marijuana) symptoms. Several methods, including diet, exercise, medication, and physical therapy, were effective for some but ineffective for others. For example, regarding exercise, one AYA shared, “Walking helps with the fatigue, and it helps with your sleep during the night,” while another shared, “For the exhaustion and fatigue, I haven’t found anything. I try everything they recommend…I tried riding a bike to exercise and that made my cording worse.” Survivorship concerns: Concerns spanned psychosocial (e.g., stress, anxiety, depression, isolation, fear of long-term treatment or recurrence, financial toxicity, fertility, sexual health), and clinical (e.g., transition from treatment to survivorship) domains. Methods to alleviate survivorship concerns: Various methods were effective at alleviating (e.g., religion/spirituality, medical marijuana) or somewhat alleviating (e.g., journaling, online research) concerns. Other methods were identified as ineffective (e.g., staying busy). Several methods, including therapy, medication, positivity/advocacy, and social support, were effective for some but ineffective for others. For example, regarding attending support groups for social support, one AYA shared, “I’m going to a seminar…I don’t have anyone to talk about [sex and relationships with], so I don’t think anyone would understand except cancer patients,” while another shared, “I joined [a] program thinking it would help to talk to other people, and I haven’t gone because I feel like I would actually feel worse if I were to hear other stories right now.” Conclusion: Female AYAs newly diagnosed with early-stage breast cancer who are receiving a supportive care intervention continue to report symptoms and survivorship concerns. AYAs use multiple methods to manage symptoms and concerns that vary in effectiveness, highlighting the need for diverse methods tailored to evolving patient needs alongside personalized survivorship care delivery to improve patient quality of life.
Presentation numberPS1-03-10
Women’s Insights on Sexual Health After Breast Cancer (WISH-BREAST) Online Survey: Metastatic Breast Cancer Cohort
Laila S Agrawal, Norton Cancer Institute, Louisville, KY
L. S. Agrawal1, T. Kluthe2, C. Menn3, E. Teplinsky4; 1Medical Oncology, Norton Cancer Institute, Louisville, KY, 2Department of Pediatrics, University of Louisville and Norton Children Medical Group, Louisville, KY, Louisville, KY, 3Women’s Health, Alloy Women’s Health, New York, NY, 4Medical Oncology, Valley-Mount Sinai Comprehensive Cancer Center, Paramus, NJ.
Background: Sexual health symptoms are common and distressing in breast cancer survivors. There is limited data regarding the experience with sexual health among patients with metastatic breast cancer (MBC). Given the chronic nature of MBC and ongoing treatments, sexual health concerns may be especially complex and underrecognized in this population. We previously reported data from the WISH-BREAST study evaluating sexual health in breast cancer survivors. Here, we present findings from the WISH-BREAST MBC cohort. Methods: An anonymous online survey on sexual health in breast cancer survivors was distributed through a social media platform (Instagram) and e-mail. Questions included demographics, breast cancer history and treatment, sexual health symptoms and experience with medical care for sexual health symptoms. Results: There were a total of 1775 responses to the survey. 42 reported having MBC and 1420 reported Stage 0-III; these respondents were included in this analysis. The median age of MBC respondents was 44 years (range 39-53 years). Median age at diagnosis was 39 years (range 33-45 years) – younger than the non-MBC respondents. MBC respondents were more likely to report early menopause (57.1% vs 28.2%). 100% of respondents with MBC reported changes to sexual health. 95.2% reported a moderate to great deal of distress related to sexual health changes. The sexual health concerns reported are summarized in Table 1. Respondents with MBC were more likely to report painful sex (78.6% vs 58.8%), no sexual pleasure (52.4% vs 33.4%), and that sexual health concerns impacted relationships (85.7% vs 71.4%) than non-MBC respondents. The majority of respondents with MBC obtained information about sexual health from social media, although a lower percentage compared to those with non-MBC (66.7% vs 80.5%). While 95.2% of respondents with MBC reported vaginal dryness, 64.3% reported that treatment with vaginal hormones was not discussed. A similar proportion of respondents with MBC reported being prescribed vaginal estrogen compared to respondents with non-MBC (23.8% vs 22.7%). Conclusions: In this online survey, respondents with MBC reported high rates of sexual health concerns and distress. Despite the prevalence of symptoms such as vaginal dryness, painful sex and loss of sexual pleasure, conversations around management options, including vaginal hormones, remain limited. These findings underscore critical gaps in clinical care and patient-provider communication regarding sexual health in the metastatic setting and highlight the potential role of social media an information source. Integrating sexual health discussions into routine MBC care, improving access to evidence-based treatments and sexual health programs, and including patients with MBC in clinical trials focused on sexual health are essential steps toward enhancing quality of life for this population.
| Sexual health concern | Percentage |
| Decreased interest in sex or libido | 95.2% |
| Vaginal dryness | 95.2% |
| Decreased lubrication | 81% |
| Painful sex | 78.6% |
| Decreased arousal | 76.2% |
| Fatigue | 57.1% |
| Body image concerns | 54.8% |
| Sex is not pleasurable | 52.4% |
| Difficulty with orgasm | 38.1% |
| I am unable to have penetrative sex | 19% |
| Loss of orgasm | 19% |
| Frequent urinary tract infections | 19% |
| Changes to bladder health (frequent urination, urinary urgency) | 11.9% |
| I have no concerns | 0% |
Presentation numberPS1-03-11
Relationship between the Incidence of Osteonecrosis of the Jaw due to Bone-Modifying Drugs Administered for Bone Metastases of Breast Cancer, Risk Factors, and Overall Survival
Akari Murakami, Ehime University Hospital, Toon,Ehime, Japan
A. Murakami1, M. Manae1, U. Kaho1, T. Kana1, N. Megumi1, K. Erina1, N. Haruna1, T. Kana1, K. Yoshiaki1, U. Yuzo2; 1Breast center, Ehime University Hospital, Toon,Ehime, JAPAN, 2Department of Hepato-Biliary-Pancreatic Surgery and Breast Surgery, Ehime University, Toon,Ehime, JAPAN.
Background:Bone-modifying agents (BMAs), such as zoledronic acid and denosumab, are standard treatments for reducing skeletal-related events in breast cancer patients with bone metastases. Medication-related osteonecrosis of the jaw (MRONJ) is a well-recognized adverse event, particularly after prolonged treatment. Despite this risk, little data exist on the long-term incidence and prognostic implications in real-world practice.Methods:We retrospectively reviewed breast cancer patients with bone metastases who received BMAs for ≥3 months between January 2015 and December 2024 at our institution. Patients who received radiotherapy to the oral cavity or jaw were excluded from the study. Data were collected from the electronic medical records. MRONJ was diagnosed according to established clinical criteria. Overall survival (OS) was compared between patients with and without MRONJ using the Kaplan-Meier method.Results:A total of 46 female patients (median age, 58 years; range, 35-90 years) were included. Thirty-six patients (78.2%) underwent dental assessment before BMA initiation, and 19 (41.3%) underwent tooth extraction. The median treatment duration was 33 months (range 6-85). MRONJ occurred in 16 patients (34.8%) with a median onset time of 23.5 months. Six cases were triggered by tooth extraction during the treatment. Fourteen patients were managed conservatively, and two required surgical debridement.The median OS was significantly longer in the MRONJ group (2048 vs. 1092 days; p=0.0332), with no difference in BMA duration between the groups.Discussion and Conclusion:Our cohort showed a high incidence of MRONJ, which is consistent with previous long-term data. Notably, MRONJ was associated with a longer OS. Although its causality remains unclear, MRONJ may reflect prolonged disease control or more intensive care. These findings support the continued use of BMAs with proper dental monitoring and aggressive management of MRONJ. Prospective studies are warranted to explore prognostic implications and optimize supportive care strategies.These findings highlight a rare but clinically significant adverse event associated with prolonged use of BMAs in patients with breast cancer. Although the sample size was limited, the long-term follow-up and detailed analysis of dental intervention strategies provided valuable insights for real-world clinical management.As this analysis was conducted exclusively in an Asian population, the findings may offer unique insights into MRONJ risk and outcomes in non-Western breast cancer cohorts, where data remain limited.
Presentation numberPS1-03-12
Digni-t-uy: efficacy of dignicap® scalp cooling in uruguayan breast cancer patients – a public vs. private care comparison
Dahiana Amarillo, Universidad de La Republica (UdeLaR), Facultad de Medicina, Hospital de Clínicas, Montevideo, Uruguay
D. Amarillo, J. Manzanares, B. Insagaray, M. García, N. Camejo, C. Castillo, G. Krygier; Unidad Academica Oncología Clínica, Universidad de La Republica (UdeLaR), Facultad de Medicina, Hospital de Clínicas, Montevideo, URUGUAY.
Background:Chemotherapy-induced alopecia (CIA) is one of the most distressing side effects of systemic treatment in early-stage breast cancer (ESBC), negatively impacting quality of life (QoL) and sometimes leading patients to reject curative regimens. DigniCap® is an FDA-approved scalp cooling system recommended by NCCN and ESMO guidelines. However, regional evidence for Latin America is scarce, and differences in hair type, access, and health system structure may affect outcomes. This study examines the real-world efficacy and tolerability of DigniCap in Uruguayan patients, comparing results between the public university hospital (Hospital de Clínicas) and private clinics. Methods:A prospective single-arm cohort included Uruguayan women with ESBC receiving adjuvant or neoadjuvant anthracycline/taxane-based chemotherapy (March 2022-June 2025). Hair loss was evaluated using Dean’s scale (success: Dean 0-2). The primary analysis focused on CIA prevention rates and device discontinuation by care setting. Results:Among 122 patients, overall success was 83.6% (≤50% hair loss). Device discontinuation due to chemotherapy toxicity or DigniCap intolerance occurred in 17%. By care setting: Public (HC, n=49): Success 75%; discontinuation 24.5%. Private patients (n=73): Success 84.9%; discontinuation 4,1%. The highest success rates were seen with the TCHP (85,7%) and TC (85.4%) regimens. Reported adverse events included headaches, scalp discomfort, and cold intolerance, which were mostly mild. 83.1% of participants completed all planned cycles with DigniCap. Conclusions: This first Uruguayan cohort demonstrates that DigniCap is effective and well-tolerated for CIA prevention, achieving outcomes comparable to those of international trials. The success gap and higher discontinuation rate among public hospital patients underscore the need to address logistical barriers, provide hair care counseling, and offer supportive resources to ensure equitable benefits across all settings. Regional real-world data support the advocacy for broader insurance coverage and tailored protocols to enhance patient acceptance and treatment adherence. Local evidence on DigniCap® strengthens the case for implementing scalp cooling programs in Latin America, considering specific hair characteristics, health system inequities, and patient-centered QoL goals.
Presentation numberPS1-03-13
Comparative Outcomes in Breast Cancer Patients Treated with Alpelisib With and Without Type 2 Diabetes Mellitus: A Real-World Analysis from the TriNetX Network
Maha Zafar, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY
F. Khan1, M. Zafar1, B. Singeltary2; 1Hospice and Palliative Medicine, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Hematology and Oncology, TriHealth Good Samaritan Hospital, Cincinnati, OH.
Background: Alpelisib, a PI3Kα-specific inhibitor, is approved for use in hormone receptor-positive (HR+), HER2-negative advanced breast cancer with PIK3CA mutations. However, its known metabolic side effects, particularly hyperglycemia, raise concerns for patients with pre-existing Type 2 Diabetes Mellitus (T2DM). This study aimed to evaluate real-world treatment patterns and clinical outcomes in breast cancer patients treated with Alpelisib, stratified by diabetes status. Methods: We conducted a retrospective analysis using the TriNetX US Collaborative Network. Two cohorts were identified: (1) patients with breast cancer and T2DM treated with Alpelisib (n=655), and (2) patients without diabetes treated with Alpelisib (n=1,102). Treatment pathways were analyzed separately for each cohort. A comparative outcomes analysis was performed using 1:1 propensity score matching (503 patients per cohort) to balance baseline characteristics. Kaplan-Meier survival analysis was used to evaluate time-to-event outcomes. Patients with prior outcomes were excluded from each respective analysis. Results: After matching, each cohort included 503 patients. Mean follow-up was 562.2 days (SD 484.1) for the T2DM group and 525.1 days (SD 459.7) for the non-diabetic group. Among those without prior death records, 268 of 502 (53.4%) T2DM patients and 274 of 502 (54.6%) non-diabetic patients died during follow-up (median survival: 651 vs. 575 days; HR 0.948, 95% CI: 0.801-1.122, p=0.747). For hospitalization, 67 of 201 (33.3%) T2DM patients and 83 of 233 (35.6%) non-diabetics were hospitalized (median time: 1,178 vs. 1,009 days; HR 0.808, 95% CI: 0.584-1.116, p=0.631). Cancer progression occurred in 24 of 79 (30.4%) T2DM patients and 27 of 75 (36.0%) non-diabetics (median time: 799 vs. 765 days; HR 0.769, 95% CI: 0.443-1.337, p=0.381). Regarding adverse events, T2DM patients had a higher risk of hyperglycemia (95 of 294 [32.3%] vs. 116 of 463 [25.1%]; HR 1.336, 95% CI: 1.018-1.752, p=0.036). For hypokalemia, rates were similar (88 of 360 [24.4%] T2DM vs. 93 of 406 [22.9%] non-diabetics; HR 0.985, 95% CI: 0.735-1.318, p=0.444). Dermatitis was seen in 35 of 453 (7.7%) T2DM patients vs. 42 of 455 (9.2%) non-diabetics (HR 0.791, 95% CI: 0.505-1.239, p=0.249). Heart failure occurred in 37 of 441 (8.4%) T2DM vs. 34 of 474 (7.2%) non-diabetics (HR 1.120, 95% CI: 0.703-1.785, p=0.282). Myocardial infarction rates were 19 of 494 (3.8%) T2DM and 17 of 484 (3.5%) non-diabetics (HR 1.055, 95% CI: 0.548-2.031, p=0.593). Diarrhea occurred in 62 of 324 (19.1%) T2DM and 87 of 386 (22.5%) non-diabetics (HR 0.831, 95% CI: 0.600-1.152, p=0.283). Pneumonitis developed in 20 of 468 (4.3%) T2DM vs. 15 of 467 (3.2%) non-diabetics (HR 1.284, 95% CI: 0.657-2.509, p=0.806). Treatment pathway analysis revealed that breast cancer patients with T2DM were seven times more likely to discontinue or not proceed to subsequent therapy after Alpelisib compared to non-diabetics, despite similar follow-up durations. Conclusions: In this large real-world cohort, breast cancer patients with T2DM treated with Alpelisib experienced comparable survival and cancer progression outcomes to those without diabetes. However, they were at significantly higher risk for hyperglycemia and had less complete treatment documentation, potentially reflecting clinical complexity or data capture limitations. These findings underscore the importance of proactive metabolic monitoring and individualized care strategies for patients with comorbid diabetes receiving Alpelisib. Further research is warranted to optimize treatment sequencing and mitigate toxicity in this high-risk population.
Presentation numberPS1-03-14
Spire-bc: a clinically-integrated prognostic tool to identify breast cancer patients in need of early supportive care
Assia Konsoulova, University Specialized Hospital for Active Treatment in Oncology Prof. Ivan Chernozemski, Sofia, Bulgaria
T. Yordanov, P. Spasov, M. Musin, I. Pandzharova, S. Tuncheva, T. Karanikolova, S. Velchova, A. Konsoulova; Medical oncology, University Specialized Hospital for Active Treatment in Oncology Prof. Ivan Chernozemski, Sofia, BULGARIA.
Background:Early supportive care integration improves quality of life and may enhance survival in patients with advanced breast cancer. However, identifying the right timing and patient subset for such interventions remains a challenge in routine clinical settings. SPIRE (Supportive care & Prognostic Insights in Risk Evaluation) is a pragmatic screening tool that incorporates clinician judgment and key clinical variables to stratify oncology patients into risk groups for targeted early supportive care intervention. Methods: We performed a subgroup analysis of breast cancer patients (ICD code 50) within a prospective departmental implementation of SPIRE from April to June 2025 at the Medical oncology clinic, University Hospital of Oncology Prof. Ivan Chernozemski, Sofia. The SPIRE score integrates the surprise question (12-month mortality), ECOG status, metastatic burden, brain metastases, and comorbidities. Patients were stratified into low (0-1), medium (2-3), or high (≥4) risk categories. Referral to supportive care and preliminary follow-up status were tracked. A prospective validation strategy was pre-planned with three time-based cutoffs (month 4, 8, and 12) to evaluate the prognostic value of SPIRE in predicting 12-month mortality. Due to the prospective, pragmatic design, data reflects visit-level documentation; patient-level analyses are planned as the dataset matures. Results:Across 1,711 recorded visits between April and June 2025 (representing 737 unique breast cancer patients), 1,279 visits (74.7%) were categorized as low risk, 342 (20.0%) as medium risk, and 85 (5.0%) as high risk. Metastatic disease was present in 946 visits (55.3%), brain metastases in 86 (5.0%), and clinically significant comorbidities in 313 (18.3%). ECOG ≥2 was recorded in 89 visits. Referral to supportive care was documented in 190 visits (11.1%). Follow-up SPIRE score transitions were analyzed in 969 instances. Preliminary trend analysis showed worsening in 131 cases (13.5%) and rapid deterioration (defined as ≥2-point score increase within ≤7 days) in 8 patients. The mean normalized score change was +0.03 (SD ±2.74). These findings support the tool’s dynamic tracking potential, pending formal prospective validation. Conclusions:SPIRE is feasible to implement in routine breast cancer care and effectively stratifies patients based on prognosis and complexity. Its prospective validation will assess real-world prognostic performance, with the goal of enhancing timely and personalized supportive care delivery. Early findings suggest that SPIRE may serve as a valuable clinical trigger for palliative care referrals in breast cancer patients.
Presentation numberPS1-03-15
A Randomized Double-Blind Controlled Study on La Roche-Posay New B5 Multi-Effect Soothing and Repairing Cream in the Treatment of Skin Adverse Reactions Related to EGFR Inhibitors and Capecitabine
Xueqi Yan, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
X. Yan, Y. Liang, X. Huang, Y. Yin; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, CHINA.
Objective: To evaluate the efficacy and safety of topical La Roche-Posay New B5 Multi-Effect Soothing and Repairing Cream in the treatment of skin adverse reactions related to EGFR inhibitors and capecitabine.Methods: A randomized, double-blind, controlled clinical study was conducted. From October 2024 to June 2025, 40 patients with skin adverse reactions (such as rash, hand-foot syndrome, pruritus, dryness, discoloration, and skin chapping) during anti-tumor therapy (EGFR inhibitors and capecitabine) were recruited from the First Affiliated Hospital of Nanjing Medical University. They were divided into two groups with 20 cases each. The experimental group was treated with topical La Roche-Posay New B5 Multi-Effect Soothing and Repairing Cream, while the control group was treated with topical silicone oil cream, twice daily for 8 weeks. Follow-ups were conducted before treatment and at 2, 4, 6, and 8 weeks after treatment. Efficacy was evaluated based on the improvement rate of rash grade, the improvement time of rash grade, the improvement rate of hand-foot syndrome grade, the improvement time of hand-foot syndrome grade, Visual Analogue Scale (VAS), and Dermatology Life Quality Index (DLQI). Adverse events were recorded. Repeated measures analysis of variance and chi-square test were mainly used to compare efficacy and safety.Results: A total of 40 patients participated in this clinical study, including 22 males and 18 females, with age of (54.21 ± 14.47) years. Before treatment, there were no statistically significant differences between the two groups in gender, age, rash grade, hand-foot syndrome grade, VAS score, or DLQI score (all p > 0.05). After 2 weeks of treatment, the improvement rates of rash grade and hand-foot syndrome grade in the experimental group were 20.00% (2/10) and 20.00% (2/10), respectively, while those in the control group were 10.00% (1/10) and 20.00% (2/10), respectively. There were no statistically significant differences between the two groups (p > 0.05). After 4 weeks of treatment, the effective rate (rash grade improvement rate + hand-foot syndrome grade improvement rate) in the experimental group was 65.00% (13/20), which was significantly higher than that in the control group (40.00%, 8/20, p < 0.05). After 8 weeks of treatment, the effective rate in the experimental group was 70.00% (14/20), which was significantly higher than that in the control group (45.00%, 9/20, p < 0.05). After 4, 6, and 8 weeks of treatment, the VAS and DLQI scores in the experimental group were significantly lower than those in the control group (all p < 0.05).Conclusion: La Roche-Posay New B5 Multi-Effect Soothing and Repairing Cream is safe and effective in the treatment of skin adverse reactions related to EGFR inhibitors and capecitabine. It can significantly improve rash and hand-foot syndrome after 4 weeks of use and can be applied clinically.
Presentation numberPS1-03-16
Analysis of the frequency and associated factors of skin toxicity in patients receiving ribociclib based therapy for metastatic breast cancer
Kyoung Eun Lee, Ewha Womans University Mokdong Hospital, SEOUL, Korea, Republic of
K. Lee1, E. Kim1, L. Youra1, A. Ham1, J. Jo1, S. Lee1, H. Kim2, J. Lee2, J. Woo2, W. Lim2, B. Moon2, S. Ahn2, H. Lee3; 1Medical Oncology, Ewha Womans University Mokdong Hospital, SEOUL, KOREA, REPUBLIC OF, 2Surgery, Ewha Womans University Mokdong Hospital, SEOUL, KOREA, REPUBLIC OF, 3Statistics, Ewha Womans University Mokdong Hospital, SEOUL, KOREA, REPUBLIC OF.
Introduction: In the treatment of hormonal receptor positive (HR+), HER2 negative (HER2-) metastatic breast cancer (MBC), most guidelines recommend endocrine therapy including CDK4/6 inhibitors as the first-line drug for quality of life. Ribociclib is one of the CDK4/6 inhibitor and has been used with aromatase inhibitors or fulvestrant in HR+, HER2- metastatic breast cancer patient. Various drug reactions related with ribociclib has been reported including skin reaction, liver toxicity and hematologic toxicity. In this study, we aimed to evaluate clinical manifestations and risk factors of dermatologic toxicities in metastatic breast cancer patients who used ribociclib. Method: This study included the patients with metastatic/recurrent breast cancer who were prescribed ribociclib from April 2021 to December 2024 in our single institution. We retrospectively reviewed the medical records of the patients, identified the frequency of recorded skin lesions, the time of occurrence and clinical characteristics of skin reactions. Logistic regression analysis was performed with several clinical factors including BSA (body surface area) and concomitant medications to analyze risk factors related to the occurrence of skin lesions. Results: A total of 110 MBC patients were enrolled during the period. The median was 53 years-old (28-82), 110 patients (100.0%) were female, median BSA was 1.56 m2(range, 1.29~2.07) and 33 patients (30.0%) showed pre-menopausal status. There were 44 de novo metastatic patients (40.0%) and 66 recurrent patients (60.0%). Ribociclib+letrozole was used in 81 patients (73.6%) and ribociclib+fulvestrant was used in 29 patients (26.4%). Skin toxicity occurred in 29 patients (26.4%), and the median time to skin toxicity onset was 84 days (range, 3-498). Skin toxicity patterns included pruritus, erythematous patches, vitiligo, bullous patches, and desquamation. Grade 1 or 2 skin toxicity occurred in 93.1% of patients, grade 3 skin toxicity occurred in 1 patient and grade 4 toxicity, toxic epidermal necrolysis (TEN), was reported in 1 patient. Dose reduction was done in 7 patients (24.1%) and permanent discontinuation of ribociclib occurred in 1 patient (3.4%). Clinical improvement was achieved in 86.2% of patients with skin reactions following supportive care. Logistic regression analysis results for dermatologic toxicity risk showed that age, ECOG PS, BSA, treatment regimen, hyperlipidemia, and use of statins (HMG-CoA reductase inhibitors) were not risk factors for skin toxicity development. Conclusions: CDK4/6 inhibitors are one of the important treatments for HR+, HER2- MBC. Regardless of clinical efficacy, skin toxicity is a common cause of patient discomfort. Therefore, detailed clinical attention and supportive cares can improve the patient’s quality of life.
Presentation numberPS1-03-17
Clinical and Economic Impact of Preemptive UGT1A1*28 Genotyping in Metastatic Breast Cancer Patients Treated with Sacituzumab Govitecan: Real-World Experience
ISABEL BLANCAS, HOSPITAL, GRANADA, Spain
I. BLANCAS1, M. MARTINEZ PEREZ2, X. DIAZ VILLAMARIN2, F. RODRIGUEZ SERRANO3, R. MORON4; 1MEDICAL ONCOLOGY, HOSPITAL, GRANADA, SPAIN, 2PHARMACY, HOSPITAL, GRANADA, SPAIN, 3Associate Professor Department of Human Anatomy and Embryology, UNIVERSITY, GRANADA, SPAIN, 4MEDICAL ONCOLOGY, PHARMACY, GRANADA, SPAIN.
The UGT1A1*28/*28 genotype is linked to reduced UGT1A1 enzyme activity and increased toxicity risk. This study assesses the potential clinical and economic impact of preemptive UGT1A1*28 genotyping in SG-treated patients. Methods:We conducted a retrospective study (Aug 2024-Jul 2025) involving 15 metastatic breast cancer patients treated with SG. UGT1A1*28 genotyping (via KASP probes; €10.51/patient) was performed in those experiencing grade ≥3 toxicities (n=8). Clinical, hospitalization, and cost data were retrieved using official Andalusian Health Service pricing (May 2024), including DNA extraction and lab expenses.Results: Three patients required hospitalization:
- One UGT1A1*28/*28 patient was admitted to the ICU (5 days) and internal medicine ward (6 days) due to septic shock with febrile neutropenia (grade 4) and other toxicities (total cost: €14,553.08)One UGT1A1*1/*28 patient was hospitalized for 11 days due to febrile neutropenia (grade 4) and gastrointestinal toxicity (€8,485.73)Another UGT1A1*28/*28 patient was admitted to the oncology ward for 3 days due to grade 4 neutropenia (€2,314.29)
The total cost of preemptively genotyping all 15 patients would have been only €157.65.
The median PFS among evaluable patients was 6.5 months. Two patients remained on treatment without progression at data cutoff. Although no formal comparison by genotype was performed due to sample size, patients with the UGT1A1*28/*28 genotype showed a trend toward shorter treatment duration due to early-onset toxicity and treatment discontinuation.According to RECIST 1.1 criteria, there were 2 partial responses (PR), 4 cases of stable disease (SD), 1 progressive disease (PD), and 1 unevaluable case due to early discontinuation caused by persistent toxicity (UGT1A1*28/*28 carrier).Regarding clinical management, all UGT1A1*28/*28 patients required dose reductions, treatment discontinuation, or G-CSF support. Two underwent progressive dose reductions (up to 70%), and one discontinued SG after two reductions due to persistent toxicity. Both UGT1A11/28 patients required dose adjustments: one with a 25% reduction and filgrastim prophylaxis, and another with a 75% dose. In contrast, neither of the UGT1A1*1/*1 patients required dose modification.Conclusions:This real-world series suggests that the UGT1A1*28 genotype is associated with a higher risk of severe toxicity, hospitalization, and dose modification in SG-treated patients. Although one UGT1A1*1/*1 patient also experienced severe toxicity, most serious events occurred in UGT1A1*28 carriers. Given the low cost of genotyping, implementing preemptive UGT1A1*28 testing prior to SG initiation may be a cost-effective strategy to anticipate and reduce serious adverse events. Prospective studies with larger cohorts are needed to confirm these findings.
Presentation numberPS1-03-18
Effects of cancer treatment on bone mineral density in premenopausal patients with early-stage breast cancer
Aya Nishikawa, Yokohama City Univ. Medical Center, Yokohama, Japan
A. Nishikawa1, K. Narui1, Y. Fujiwara2, Y. Shibata1, S. Adachi1, K. Kawashima2, M. Oshi2, A. Yamada2, H. Yoshikata3, T. Tsuburai3, I. Endo2; 1Surgical Oncology, Yokohama City Univ. Medical Center, Yokohama, JAPAN, 2Surgical Oncology, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 3Gynecology, Yoshikata Clinic, Yokohama, JAPAN.
Background: Systemic therapies for breast cancer (BC) can lead to cancer treatment-induced bone loss (CTIBL), increase fracture risk, impair the quality of life, and potentially worsen prognosis. Although CTIBL prevention has been established in postmenopausal patients with BC, CTIBL in premenopausal women remains unclear owing to the limited number of reports. Chemotherapy often induces ovarian suppression and estrogen deficiency, thereby accelerating bone loss. Furthermore, the effects of tamoxifen on the bone are yet to be established in premenopausal patients with BC. In this study, we prospectively evaluated CTIBL in premenopausal patients with BC by monitoring bone mineral density (BMD), hormones, and bone turnover markers. Methods: We recruited 64 premenopausal females diagnosed with early-stage BC. We compared patients receiving tamoxifen monotherapy (T group, n=19; median age, 45 [range, 37-55] years) with those receiving chemotherapy followed by tamoxifen therapy (C group, n=31; median age, 45 years [range, 31-50] years). BMD at the lumbar spine and femoral neck was measured at baseline, immediately after chemotherapy (C group), and at 6, 12, and 18 months after tamoxifen initiation. Serum levels of estradiol (E2), follicle-stimulating hormone (FSH), bone turnover markers, procollagen type 1 N-terminal propeptide (P1NP), tartrate-resistant acid phosphatase 5b (TRACP-5b), and undercarboxylated osteocalcin (ucOC) were concurrently assessed to evaluate hormonal and metabolic effects on bone health. Results: 1. BMD Changes: T group: The changes(±SD) in BMD were minimal, with a 2.2±5.1% decrease (p=0.13) at the lumbar spine and a 1.6±4.4% increase(p=0.19) at the femoral neck at 24 months.C group: Lumbar spine BMD decreased by 2.4±3.9% post-chemotherapy (p=0.0057) and by 3.4±3.1% at 24 months (p=0.0060). Femoral neck BMD decreased by 1.3±4.6% post-chemotherapy (p=0.24) and 4.1±4.1% at 24 months (p=0.0047). 2. Hormonal Changes: T group: E2 increased by 328.1±385.6% at 6 months (p=0.003) but returned to baseline by 24 months. FSH levels showed no significant changes.C group: E2 decreased by 76.2±49.6% post-chemotherapy (p<0.001), with no significant differences observed at 24 months. FSH increased by 1061.1±890.7% post-chemotherapy (p<0.001), gradually decreased, and remained 53.6±89.8% above baseline at 24 months (p=0.038). 3. Bone Turnover Markers: T group: P1NP and TRACP-5b levels showed no significant changes over 24 months (all p > 0.05). ucOC decreased by 22.6±36.5% at 6 months (p=0.036), stabilizing thereafter.C group: P1NP increased by 98.9±90.0% post-chemotherapy (p<0.001) and gradually returned to baseline levels by 24 months (-5.4±45.0%, p=0.90). TRACP-5b increased by 89.9±106.6% post-chemotherapy (p<0.001) and gradually returned to baseline levels by 24 months (5.7±44.3%, p=0.38). ucOC decreased by 82.6±11.5% post-chemotherapy (p<0.001) and remained suppressed, with no significant recovery observed at 24 months (-89.1±9.2%, p<0.001). Conclusions: The C group showed significant and sustained reductions in BMD. The observed bone loss could be attributed to chemotherapy-induced ovarian suppression and estrogen deficiency. Increased bone turnover markers were consistent with a high bone resorption state and bone loss. Conversely, BMD was preserved in the T group, consistent with stable hormone levels and bone turnover markers. These findings indicate the need for proactive bone health management, including regular BMD monitoring and consideration of bone-protective treatments, especially in premenopausal patients with BC receiving chemotherapy, to improve long-term quality of life without fracture.
Presentation numberPS1-03-19
Interventions acceptable to women with metastatic breast cancer for their sexual problems
Nusrat Jahan, University of Alabama at Birmingham, Birmingham, AL
N. Jahan1, E. Sringer-Reasor1, H. Sarfraz1, K. Khoury1, M. Escobar1, C. P. Williams2, I. Starks1, T. Padalkar1, S. Olisakwe1, N. Henderson1, S. A. Ingram1, A. Azuero3, K. Keene4, M. E. Melisko5, E. H. Shinn6, G. B. Rocque1; 1Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 2Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 3UAB School of Nursing, University of Alabama at Birmingham, Birmingham, AL, 4Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 5Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, 6Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Metastatic breast cancer (MBC) and its therapies can cause sexual problems, leading to distress and relationship problems. Unfortunately, limited information is available on acceptable interventions to address sexual problems in women with MBC. Methods: This qualitative study focuses on acceptable interventions for sexual problems of women with MBC. A medical anthropologist conducted semi-structured interviews with women diagnosed with MBC at the University of Alabama at Birmingham. Interviews were transcribed verbatim and inductively coded to identify recurring themes and exemplary quotes. Results: Between September 2024 and May 2025, 20 women aged 30-77 with MBC participated in interviews. Of 20, 11 (55%) were White and 9 (45%) were African American; 95% were heterosexual and 1 (5%) was bisexual; 10 (50%) were married or partnered and 10 (50%) were widowed, single, or divorced; and 90% were in menopause (natural or treatment-related). Participants expressed key intervention preferences reflecting both favorable views and concerns (Table). The most common interventions discussed were lubricants during sexual intercourse. Most patients expressed positive experiences with using lubricants, which provided a degree of symptom relief. However, some women expressed concerns about a burning sensation associated with lubricants. The second category was sex toys, including mostly different types of vibrators. Although the majority of the women expressed positive experiences about using sex toys, some expressed hesitancy. A third intervention discussed was hormonal vaginal preparations. Many women expressed concerns about the long-term side effects of using estrogen- or testosterone-containing vaginal preparations resulting in a limited willingness to use. Another intervention discussed was non-hormonal vaginal moisturizers, which they perceived as having limited benefits. Although a few women were open to using medications to improve sexual desire, the majority are hesitant due to potential side effects and an additional pill burden. The majority of women in relationships adopt or are willing to adopt changes beyond sex to improve communication, relationships, and intimacy. However, women were reluctant to use formal therapy, including couple and behavioral therapy, due to lack of time, other priorities, and skepticism about their effectiveness. Conclusions: This study identifies acceptable interventions to address the sexual problems of women with MBC. Over-the-counter interventions such as lubricants, sex toys, and behavioral techniques were preferred. They were more reluctant to use prescription medications such as estrogen- or testosterone-containing vaginal preparations, medications to enhance sexual desire, and formal couple and behavioral therapies due to concerns about side effects, skepticism about their effectiveness, and additional therapeutic burdens.
| Interventions | Exemplary quotes | ||
| Lubricants | Yeah, my number one go-to, I always use *** gel and that helped me out a lot. | ||
| Sex toys | I have a rose, if you ever heard of those. Those are pretty good! | ||
| Vaginal estrogen or testosterone | I know estrogen not as risky as it once was, thought it was for hormone treatment. I think in my case, I’d probably prefer not to do hormones if I could. | ||
| Vaginal non-hormonal moisturizer | It didn’t really help. | ||
| Libido enhancing medications | I don’t want to take a pill that’s like six pills a day. I got enough of those. | ||
| Intimacy beyond sex | Find a shared interest that you can do together, even if it’s not sex. | ||
| Behavioral and couple therapy | To a therapist. No, I’ve done that. |
Presentation numberPS1-03-20
Impact of early vs delayed referral to physical therapy and occupational therapy after mastectomy and the risk of postmastectomy Lymphedema: A Real-World Propensity-Matched Study
Krishna Doshi, UT San Antonio Health Center/Mays Cancer Center, San Antonio, TX
K. Doshi1, S. Afridi2, N. Iyer3, S. A. Haddad4; 1Department of Hospice and Palliative Medicine, UT San Antonio Health Center/Mays Cancer Center, San Antonio, TX, 2Department of Internal Medicine, SUNY Upstate Medical Center, Syracuse, NY, 3Department of Internal Medicine, Loyola University Health Center- MacNeal Hospital, Berwyn, IL, 4Department of Hematology and Oncology, UT San Antonio Health Center/Mays Cancer Center, San Antonio, TX.
Background:Postmastectomy lymphedema remains a significant source of morbidity in breast cancer survivors. Physical therapy (PT) and occupational therapy (OT) are commonly employed for prevention and management, but optimal referral timing remains unclear. We evaluated whether early PT and OT referral after mastectomy reduces two-year lymphedema. Methods: We conducted a retrospective cohort study using the TriNetX Global Collaborative Network, which includes de-identified electronic health records from 151 healthcare organizations worldwide. We identified adult women (≥18 years) with a diagnosis of breast cancer (ICD-10: C50) who underwent mastectomy between January 1, 2000, and December 31, 2020. Patients with pre-existing lymphedema or hereditary lymphedema were excluded. Two cohorts were defined based on the timing of PT and OT initiation relative to the mastectomy date: Early cohort: PT/OT initiated within 14 days post-mastectomy (n = 5,197) and delayed cohort: PT/OT initiated more than 14 days post-mastectomy (n = 18,817). PT/OT procedures were identified using relevant CPT codes for therapy evaluations, manual lymphatic drainage, gait training, and self-care training. The primary outcome was the incidence of lymphedema occurring between 90 and 1095 days following the index event. A Cox proportional hazards model was used to assess the association between timing of PT/OT and lymphedema risk, adjusting for age, extent of breast resection, axillary dissection, and sentinel lymph node biopsy. Results In the multivariable Cox model, early initiation of PT/OT within 14 days post-mastectomy was significantly associated with reduced lymphedema risk (HR 0.737, 95% CI 0.608-0.893, p=0.002). Sentinel lymph node biopsy was associated with a protective effect (HR 0.721, 95% CI 0.546-0.953, p=0.021). In contrast, axillary lymph node dissection was associated with more than a twofold increased risk of lymphedema (right: HR 2.217, p=0.001; left: HR 2.101, p=0.002). Bilateral mastectomy was also protective (HR 0.597, p<0.001), and increasing age was modestly protective (HR 0.981 per year, p<0.001). Unilateral breast resections were not significantly associated with lymphedema risk. Conclusion Early initiation of PT/OT and the use of sentinel lymph node biopsy were independently associated with reduced lymphedema risk following mastectomy. In contrast, axillary lymph node dissection was strongly predictive of increased lymphedema risk. These real-world findings support current American Society of Clinical Oncology (ASCO) and National Comprehensive Cancer Network (NCCN) guidelines advocating early rehabilitation and limited axillary surgery to minimize long-term morbidity in breast cancer survivors.
Presentation numberPS1-03-22
Predictors of cardiotoxicity with HER2-directed antibody therapy in a real-world setting across a large, Western US hospital system
Cameron Smith, Providence Portland Medical Center, Portland, OR
C. Smith1, M. Layoun2, H. Li3, E. Koltner3, M. T. Imboden3, Z. Taylor2, K. J. Spinelli3, D. Page1; 1Providence Cancer Institute, Providence Portland Medical Center, Portland, OR, 2Providence Heart Institute, Providence St Vincent Medical Center, Portland, OR, 3Center for Cardiovascular Analytics, Research + Data Science (CARDS), Providence St Vincent Medical Center, Portland, OR.
Background: Incidences of cardiotoxicity related to HER2-directed antibody therapy (anti-HER2) vary considerably from 0.5% to 30% depending on comorbidities and chemotherapy regimen. Recent studies have posited whether low-risk cohorts could decrease or eliminate surveillance echocardiography given low incidences of treatment-related heart failure (HF). Objectives: The aim of this study was to characterize the incidence of cardiotoxicity in patients receiving HER2-therapies, evaluate predictors of cardiotoxicity, and examine the incidence of LVEF decline and new HF among a low-risk subgroup. Methods: We performed a retrospective cohort study of early stage (I-III) breast cancer patients who received anti-HER2 between 1/1/2016-3/20/2024 and completed baseline and surveillance echocardiography in the 7-state Providence healthcare network. The primary outcome was development of LVEF ≤50% during anti-HER2. Secondary outcomes included new HF diagnosis (based on ICD-10 codes I50.x), any hospitalization, and all-cause mortality. Incidences of LVEF ≤50% and new HF were also obtained in a low-risk subgroup. Logistic regression models accounting for demographics, comorbidities, HER2 treatment regimen, and preceding cardiac medications were performed.Results: The study included 1,215 patients (Table). Fifty-two patients (4%) developed LVEF ≤50% during anti-HER2. Among this subset, 52% were hospitalized and 42% had a new HF diagnosis during the treatment period. No patients died while receiving anti-HER2 or during 1-year follow up. Univariate modeling revealed that CAD (aOR [95% CI] = 3.22 [1.21, 8.61]) and prior HF (aOR [95% CI] = 3.67 [1.23-10.99]) were associated with treatment-related LVEF ≤50%, though no comorbidities were predictive on multivariate analysis. After risk adjustment, HER2 treatment regimen was the only predictor of LVEF ≤50% during therapy (taxanes + platinum vs. anthracyclines: aOR [95% CI] = 0.22 [0.10, 0.48]; taxanes vs. anthracyclines: aOR [95% CI] = 0.29 [0.13, 0.65]. New HF was observed in 11% of patients receiving anthracyclines compared to 4-5% in other treatment arms (p = 0.024). Patients deemed low-risk (non-anthracycline regimen, no prior CAD or HF) developed an LVEF ≤50% in 3.3% of patients and new HF in 2.6% of patients. Conclusions: In a real-world clinical setting, anti-HER2-associated LVEF declines were uncommon but were associated with increased hospitalizations and HF diagnoses. Exposure to anthracyclines was the only predictive risk factor for LVEF decline. Interim analysis of a low-risk cohort (non-anthracycline regimen, no prior CAD or HF) still demonstrated a modest incidence of LVEF decline and HF, suggesting that enhanced risk stratification should be considered before deferring echocardiographic surveillance in these subgroups.
| Variable | All Patients (N=1,215) | LVEF ≤50% subgroup (N=52) | |||
| Baseline Characteristics | |||||
| Age* | 56.8 (12.8) | 56.7 (12.3) | |||
| Female, N (%) | 1211 (99%) | 50 (96%) | |||
| Race*, White, N (%) | 900 (76%) | 42 (84%) | |||
| Ethnicity*, Hispanic, N (%) | 119 (10%) | 4 (8%) | |||
| BMI*, ≥40 | 103 (9%) | 7 (13%) | |||
| Baseline comorbidities*, N (%) Hypertension Hypercholesterolemia Diabetes Smoking status (% current or passive smokers) Coronary artery disease Heart Failure Atrial fibrillation Cerebrovascular disease | 360 (30%) 423 (35%) 146 (12%) 76 (6%) 54 (4%) 38 (3%) 43 (4%) 59 (5%) | 15 (29%) 18 (35%) 8 (15%) 4 (8%) 6 (12%) 4 (8%) 3 (6%) 3 (6%) | |||
| Referral to cardiology prior to treatment | 44 (4%) | 3 (6%) | |||
| Cardiac meds prior to HER2* | 292 (24%) | 15 (29%) | |||
| HER2 treatment group* Anthracycline Taxane Taxane plus Other | 97 (8%) 465 (38%) 596 (49%) 52 (4%) | 12 (23%) 22 (42%) 17 (33%) 1 (2%) | |||
| Outcomes While on HER2 Therapy | |||||
| Decline in LVEF, average | -5.4 (3.2) | -8.7 (3.8) | |||
| Decline in max LVEF, range | (-40, -1) | (-20, -1) | |||
| Referral to cardiology while on treatment | 34 (3%) | 8 (15%) | |||
| Cardioprotective meds after start of HER2 | 119 (10%) | 19 (37%) | |||
| New HF diagnosis | 70 (6%) | 22 (42%) | |||
| Patients with ED and hospital encounters | 476 (39%) | 27 (52%) | |||
| Total number of ED and hospital encounters** Cancer related Cardiac related Other | 955 67 (7%) 10 (1%) 878 (92%) | 73 1 (1%) 4 (5%) 68 (93%) | |||
| Mortality | 0 (0%) | 0 (0%) | |||
| Data presented as n (%), mean (SD), or range *Variables included in the multivariate model **Patients could have more than 1 hospital encounter. Abbreviations: BMI = body mass index, ED = emergency department, HER2 = human epidermal growth factor receptor 2, HF = heart failure, LVEF = left ventricular ejection fraction |
Presentation numberPS1-03-23
Digital Tool to Examine the Characteristics and Disparities Associated with Immune-Related Adverse Events (IrAEs) among Breast Cancer Outcomes.
Parikshit Padhi, University at Buffalo, Williamsville, NY
P. S. Satheeshkumar1, S. Panginikkode2, V. Gopalakrishnan3, D. Kewlani4, N. Alam1, T. Odemuyiwa5, R. Pili1, P. Padhi5; 1Internal Medicine-Division of hematology and oncology, University at Buffalo, Buffalo, NY, 2Department of Rheumatology, Tufts Medical Center, Boston, MA, 3Internal Medicine, University of Massachusetts, Worcester, MA, 4Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 5Internal Medicine-Division of hematology and oncology, University at Buffalo, Williamsville, NY.
AIM AND OBJECTIVES: Analyzing IrAEs data provides an opportunity to investigate the impacts of targeted therapy, especially as the implementation of immunotherapy in breast cancer has enhanced survival rates. We utilized digital tools to illustrate that IrAEs, including characteristics, are essential to manage for cancer patients receiving treatment, and identifying outcomes and disparities is significantly more critical. METHODS: We examined breast cancer patients admitted to the hospital in the year 2021 (National Inpatient Sample database) and further examined IrAEs among them. IrAEs examined included dermatologic, endocrine, gastrointestinal, hepatic, neurologic, pancreatic, pulmonary, and renal. We utilized approaches—propensity score matching (PSM) and difference-in-difference (DID), and additional PSM DID combined and further multivariable generalized linear models accounting for weights—to examine the outcomes—mortality, infectious complications (mainly septicemias and healthcare-associated complications), and complicated diabetes (diabc), complicated hypertension (hypc), fluid and electrolyte disorders (fed), and depression (depre) were examined. And additionally, we examined the disparities in the outcomes. RESULTS: We examined 108,170 breast cancer patients admitted to the hospital in the year 2021, and we further observed 28,390 (26%) IrAEs among them. The mean [SD] age of the patients admitted to the hospital with IrAEs was 66 [13.25] years. The breast cancer patients who reported with IrAEs were associated with higher odds of mortality (aOR: 1.18, 95% CI: 1.02-1.38). Compared to the high-income group, the low-income group was associated with higher mortality (1.26; 1.02-1.56). And further, those with IrAEs were associated with higher septicemias (1.33; 1.21-1.46). Compared to Whites, Blacks (1.06; 1.05-1.45), Hispanics (1.26; 1.05-1.45), and Native Americans and Others (1.26; 1.07-1.48) were associated with higher septicemias. And compared to high-income neighborhoods, low-income neighborhoods were associated with higher septicemias (aOR: 1.16; 95% CI 1.03-1.32). Similarly, the trend of IrAEs outcomes and disparities was examined among fluid and electrolyte disorders(1.66; 1.07-1.27), hypertension (1.16; 1.07-1.27), and depression (1.48; 1.36-1.6). Disparities observed (Blacks were having higher hypertension, and depression was higher among Whites compared to all other racial groups, and fed was higher among low-income groups) were examined. We additionally evaluated the impact of IRAES on individual outcomes using PSM DID. The treatment effect (TE) and the average treatment effect (ATE) were examined as specific metrics that measure the average causal influence of a treatment across breast cancer cohorts. CONCLUSION: Investigating the IrAEs in breast cancer patients is crucial, since the five-year survival rate for these patients is key for patient treatment. Consequently, the competing risks within this population are particularly significant to identify, as they fall under this framework. This marks the inaugural application of digital health techniques to investigate inequities. Black individuals, Native Americans, and low-income communities are disproportionately impacted by the new regimes.
Presentation numberPS1-03-24
Real-world evaluation of hepatotoxicity during treatment with ribociclib
Brooke Kielkowski, West Virginia University Cancer Institute, Morgantown, WV
J. Hill1, B. Kielkowski1, S. Nolfi2, C. Marino2, L. Thomas2; 1Pharmacy, West Virginia University Cancer Institute, Morgantown, WV, 2Hematology/Oncology, West Virginia University Cancer Institute, Morgantown, WV.
Background: Ribociclib, a CDK4/6 inhibitor, is widely used in hormone receptor-positive, HER2-negative breast cancer. While hepatotoxicity is a known adverse event associated with ribociclib, its real-world incidence and characteristics may differ from those observed in clinical trials. This study aimed to evaluate the incidence, severity, and clinical course of hepatotoxicity in patients receiving ribociclib in routine clinical practice. Methods: We conducted a retrospective chart review of 90 patients treated with ribociclib at our institution. Patients who received fewer than 14 days of treatment (n=25) were excluded, leaving 65 patients for analysis. We assessed liver function test abnormalities during treatment and graded hepatotoxicity according to CTCAE v5.0 criteria. Data on baseline liver disease, liver metastases, concurrent hepatotoxic medications, and CYP3A4 inhibitors were collected. Results: Among the 65 patients included, 10 (15.4%) developed grade 3-4 hepatotoxicity: 5 patients (7.7%) with grade 3 and 5 patients (7.7%) with grade 4. The observed incidence of grade 4 hepatotoxicity was approximately three-fold higher than reported in pivotal ribociclib clinical trials, including MONALEESA-2, MONALEESA-3, MONALEESA-7, and NATALEE, where incidence of grade 4 hepatotoxicity ranged from 0.9-2.5%.1-4 Notably, only 2 of the 10 patients had baseline liver metastases, and just 1 patient had known baseline liver disease (hepatic steatosis). None of the patients who experienced grade 3-4 hepatotoxicity were receiving strong CYP3A4 inhibitors or other hepatotoxic medications at the time of onset. The median time to onset of hepatotoxicity was 15 weeks (range: 2-29 weeks), and the median time to recovery was 11 weeks (range: 4-30 weeks). Among the 5 patients who developed grade 3 hepatotoxicity, 3 were rechallenged with a reduced dose of ribociclib. All three patients experienced recurrent hepatotoxicity following rechallenge. Conclusions: In this real-world cohort, the incidence of grade 4 hepatotoxicity with ribociclib was significantly higher than reported in clinical trials, underscoring the importance of vigilant hepatic monitoring beyond trial conditions. Hepatotoxicity occurred independently of baseline liver metastases, pre-existing liver disease, or concurrent use of known hepatotoxic agents. Rechallenge following grade 3 hepatotoxicity was associated with recurrence, suggesting cautious consideration before re-initiation. These findings highlight the need for personalized risk assessment and further investigation into predictors of severe hepatotoxicity in patients treated with ribociclib. References Hortobagyi GN, Stemmer SM, Burris HA, et al. Overall Survival with Ribociclib plus Letrozole in Advanced Breast Cancer. N Engl J Med 2022;386(10):942-950. Slamon DJ, Neven P, Chia S, et al. Phase III randomized study of ribociclib and fulvestrant in hormone receptor-positive, human epidermal growth factor receptor 2-negative advanced breast cancer: MONALEESA-3. J Clin Oncol 2018;36:2465-2472. Lu YS, Im SA, Colleoni M, et al. Updated Overall Survival of Ribociclib plus Endocrine Therapy versus Endocrine Therapy Alone in Pre- and Perimenopausal Patients with HR+/HER2- Advanced Breast Cancer in MONALEESA-7: A Phase III Randomized Clinical Trial. Clin Canc Res 2022;28(5):851-859. Slamon DJ, Lipatov O, Nowecki Z, et al. Ribociclib plus Endocrine Therapy in Early Breast Cancer. N Engl J Med 2024;390(12):1080-1091.
Presentation numberPS1-03-25
Incidence of hypersensitivity reactions in patients receiving trastuzumab biosimilars
Alicia C McGhie, MD Anderson Cancer Center, Houston, TX
E. M. Grannan1, A. C. McGhie1, A. Zheng1, S. Cherian1, A. B. Newman2, R. M. Layman3, S. Damodaran3; 1Division of Pharmacy, MD Anderson Cancer Center, Houston, TX, 2Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 3Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX.
Background: Currently multiple biosimilars of trastuzumab (Herceptin®) are approved for use in HER2-positive breast cancers. Accordingly, MD Anderson Cancer Center updated its formulary from trastuzumab (Herceptin®) to trastuzumab biosimilars: trastuzumab-qyyp (Trazimera®) and trastuzumab-anns (Kanjinti®), with trastuzumab-qyyp designated as the preferred agent. While these biosimilars have been approved based on extensive structural and functional characterization demonstrating similar clinical activity, differences in hypersensitivity reactions (HSRs) have been reported. We evaluated the incidence of HSRs in patients receiving trastuzumab biosimilars compared to the reference product. Methods: A retrospective chart review was conducted on patients who received trastuzumab products from January 1, 2019, to January 1, 2025. Patients with documented trastuzumab allergies in EMR were included in this analysis. Incidence of HSR between patients who received trastuzumab (Group A, n=612) and trastuzumab biosimilars (Group B, n=503) was compared. Results: HSRs were significantly more common in patients in patients who received trastuzumab biosimilars (Group B; n=86, 17.1%) compared to trastuzumab (Group A; n=22, 3.6%) (p<0.001). Majority of the patients in Group B received Trastuzumab-qyyp (n=79, 92%). Chills and/or rigors were the most common HSR symptom across both groups (Group A: n=18, 75%); Group B: n=75, 87.2%). Severe symptoms, such as dyspnea (Group A: n=2 [9.1%] vs Group B: n=10 [11.6%], p=0.74) and hypotension (Group A: n=1, [4.5%] vs Group B: n=5 [5.8%], p=0.82), were not significantly different between the groups. Two patients in Group B required epinephrine, while no patients in Group A required epinephrine. Seven patients in Group B required a change in trastuzumab product. Conclusion: The incidence and severity of HSRs were higher in patients receiving biosimilar trastuzumab products compared to the reference product. Data collection for patients without documented trastuzumab allergies is ongoing. Based on this analysis, a prospective study utilizing prophylactic medications is being planned.
Presentation numberPS1-03-26
Changing patterns of attention network in Breast Cancer Survivors
Jingjing Li, The Second Affiliated Hospital of Anhui Medical University, hefei, China
F. li1, J. Li1, X. Chen2; 1Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, hefei, CHINA, 2Department of Neurology, The First Affiliated Hospital of Anhui Medical University, hefei, CHINA.
Background: Cognitive impairments, particularly in attention, are frequently reported among breast cancer survivors undergoing systemic anticancer therapies. However, the changes across distinct attention functions remain poorly characterized. This study aims to investigate alterations in the attention networks among breast cancer survivors treated with chemotherapy and tamoxifen and to explore their association with treatment duration. Methods: Between September 2018 and December 2024, a total of 435 breast cancer survivors who underwent chemotherapy followed by tamoxifen treatment, along with 242 age-matched healthy female controls were recruited. The Attention Network Test was utilized to assess the efficiency of attentional networks. Linear regression and restricted cubic spline analyses were employed to evaluate changes in the three attention networks among breast cancer survivors. Results: Compared with healthy controls, breast cancer survivors exhibited significantly reduced efficiency in the alerting (P < .001) and executive (P < .001) networks, while the orienting network efficiency remained intact. Notably, the executive network demonstrated a nonlinear (V-shaped) trajectory over treatment duration, characterized by an initial improvement peaking around at approximately 15 months, followed by a subsequent decline (nonlinear P < .001). This pattern was consistent across different chemotherapy subtypes. The alerting network showed persistent impairment throughout the five-year observation period (linear P = .247), potentially indicative of underlying neural alterations. Conclusions: Systemic anticancer treatments in breast cancer survivors are associated with selective impairments in attention networks, characterized by deficits in alerting efficiency and a V-shaped trajectory of executive function changes over a five-year period. These findings suggest that early chemotherapy-related cognitive effects may be transient, whereas prolonged tamoxifen use could contribute to persistent executive dysfunction. Longitudinal studies are essential to elucidate the underlying causal mechanisms and inform strategies for cognitive rehabilitation. Keywords: Breast cancer; Chemotherapy; Tamoxifen; Cognitive function; Attention network
Presentation numberPS1-03-27
Impact of Trastuzumab Emtansine with DoseEscalation on Thrombocytopenia inChinesePatientswithHER2-Positive Breast Cancer: A Multicenter Retrospective Study
Meng Wang, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliate Cancer Hospital Of University Of Electronic Science and Technology of China (UESTC), ChengDu, China
M. Wang1, M. Huang2, X. Zhao3, H. Che4, J. Li1, M. Cheng5, S. Aili6, X. Zhao7, X. Wang8, Y. Zou9, H. Wang1; 1Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliate Cancer Hospital Of University Of Electronic Science and Technology of China (UESTC), ChengDu, CHINA, 2Breast Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, CHINA, 3Thyroid Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, CHINA, 4Thyroid Breast Surgery, Zigong First People’s Hospital, Zigong, CHINA, 5Breast and thyroid surgery, Suining Central Hospital, Suining, CHINA, 6Thyroid Breast Surgery, Guangyuan Central Hospital, Guangyuan, CHINA, 7Breast and thyroid surgery, Leshan People’s Hospital, Leshan, CHINA, 8Oncology, Neijiang Second People’s Hospital, Neijiang, CHINA, 9Breast and thyroid surgery, Dazhou Central Hospital, Dazhou, CHINA.
Background: Trastuzumab Emtansine (T-DM1), an antibody-drug conjugate of trastuzumab and emtansine, is an established adjuvant or second-line therapy for HER2-positive breast cancer (BC), widely used in China for both early-stage and advanced HER2-positive BC. Thrombocytopenia represents a common adverse event associated with T-DM1 in Chinese patients, with the incidence of grade ≥3 thrombocytopenia reported as 40.4% in the ELAINA study. This study was designed to investigate the impact of T-DM1 with dose escalation on thrombocytopenia incidence. Method: This multicenter, retrospective study enrolled HER2-positive BC patients treated with T-DM1 between May 2024 and March 2025, all of whom had received at least one prior systemic therapy. Patients were stratified into two cohorts: (1) early-stage disease with residual disease (non-pCR) post-neoadjuvant therapy; (2) advanced-stage disease with progression following prior systemic treatments. T-DM1 dose was escalated in a stepwise manner: 2.4 mg/kg administered in Cycle 1, followed by 3.0 mg/kg in Cycle 2, and culminating in 3.6 mg/kg in Cycle 3. Safety was closely monitored at each dose level, especially for thrombocytopenia. Non-pCR patients received up to 14 cycles of T-DM1, while those with advanced-stage continued treatment until disease progression or unacceptable toxicity. The primary outcome was thrombocytopenia incidence. Data on treatment patterns, adverse events, and clinical efficacy were also collected and analyzed. Result: A total of 46 patients were included (37 non-pCR; 9 advanced). The mean age was 52.8±8.9 years. Five patients (10.9%) with visceral crisis were all in advanced-stage. Prior systemic therapy was reported in all patients. All 37 non-pCR patients received neoadjuvant chemotherapy: 78.4% platinum-based, 13.5% anthracycline-based, and 8.1% other regimens. For advanced breast cancer, Seven and 2 patients received T-DM1 as first and later-line therapy, respectively. The initial dose of all the patients was 2.4 mg/kg. Maximum dose levels were 2.4 mg/kg in 39.1%, 3.0 mg/kg in 17.4%, and 3.6 mg/kg in 43.5% of patients, respectively. The median treatment duration was 75 days (range 7-377). Overall, 4 patients (8.7%) discontinued T-DM1: 3 patients (6.5%) due to disease progression and 1 patient (2.2%) as per physician’s advice (unrelated to thrombocytopenia). Thrombocytopenia of any grade occurred in 58.7% of patients, while grade ≥3 thrombocytopenia was observed in 30.4% of patients. Thrombocytopenia of any grade occurred with a median onset of 8 days, while grade ≥3 thrombocytopenia occurred with a median onset of 280 days. Conclusion: T-DM1 dose escalation was well tolerated and effective with a relatively low incidence of grade ≥3 thrombocytopenia. Further prospective studies are warranted to confirm the benefit of initial low-dose T-DM1.
|
Variables |
Total(N=46) |
|
Age, years, Mean ± SD |
52.81±8.92 |
|
Treatment setting, n (%) |
|
|
Advanced |
9 (19.6) |
|
Non-pCR (non-pathologic complete response) |
37 (80.4) |
|
Prior systemic therapy, n (%) |
46 (100.0) |
|
T-DM1 Treatment Pattern |
|
|
Initial dose, mg, Mean ± SD |
139.05±21.20 |
|
Maximum dose, mg, Mean ± SD |
171.33±38.38 |
|
Maximum dose level, n (%) |
|
|
2.4 mg/kg |
18 (39.1) |
|
3.0 mg/kg |
8 (17.4) |
|
3.6 mg/kg |
20 (43.5) |
|
Treatment exposure duration, days, median (range) |
75.00 (7.00, 377.00) |
|
Treatment discontinuation rate, n (%) |
4 (8.7) |
|
Reason for discontinuation, n (%) |
|
|
Permanent discontinuation due to physician’s advice |
1 (2.2) |
|
Permanent discontinuation due to disease progression |
3 (6.5) |
|
Thrombocytopenia, n (%) |
|
|
Grade 1 |
8 (17.4) |
|
Grade 2 |
5 (10.9) |
|
Grade 3 |
13 (28.3) |
|
Grade 4 |
1 (2.2) |
|
None |
19 (41.3) |
Presentation numberPS1-03-29
Development of an evidence-based patient guide to support symptom management in individuals with metastatic breast cancer
Victoria Barghout, Viver Health, Morristown, NJ
L. Medina1, V. Barghout1, R. L. Best1, J. Patten-Coble2, T. Buckingham3, H. Hammond3, C. Hogea3; 1n/a, Viver Health, Morristown, NJ, 2n/a, Little Pink Houses of Hope, Burlington, NC, 3Patient-Focused Implementation Sciences, Gilead Sciences, Inc., Foster City, CA.
Background: Metastatic breast cancer (MBC) is breast cancer that has spread beyond the breast and regional lymph nodes. For MBC, treatment goals are typically controlling disease progression and maintaining quality of life (QoL). However, disease-related symptoms and treatment-related side effects often impair QoL and may result in decreased treatment adherence. Treatment-related side effects are among the leading reasons patients with MBC discontinue recommended therapy. There is an urgent need for evidence-based tools that inform and empower patients to effectively manage symptoms and treatment-related side effects, optimize their QoL, and facilitate informed patient-provider communication. Methods: We used a qualitative stakeholder-informed approach with literature review and iterative resource creation to identify gaps in patient education, symptom management, and shared decision-making in MBC. This review informed the IRB-approved protocol and discussion guide for semi-structured interviews held with US individuals living with MBC (n=9; from rural and urban areas and diverse racial and ethnic backgrounds (Asian, Black, White—Hispanic and non-Hispanic))and the healthcare providers (HCPs) who treat them (n=4). Interview findings guided the co-creation of a printed symptom management resource, Your Guide to Managing Symptoms from Metastatic Breast Cancer Treatment, focused on the topics participants identified as most needed. The same participants reviewed the Guide for understandability, clarity, and usability. The Guide underwent medical review for accuracy and alignment with clinical guidelines and received endorsement by the Academy of Oncology Nurse & Patient Navigators (AONN+). An online survey was created to track usage and assess satisfaction, behavior change, and reach. Results: Individuals reported struggling with the mental and emotional symptoms of MBC, navigating available support resources, and managing therapy side effects—including fatigue (100%), pain (67%), neutropenia, and digestive issues (100%). Our results underscored the unmet need for a patient-focused educational resource to support symptom management and provider-patient communication. The resulting resource, Your Guide to Managing Symptoms from Metastatic Breast Cancer Treatment, is an accordion-style four-fold printed handout backed by 142 references. Based on interview findings, it focuses on evidence-based whole-person interventions to help manage symptoms and side effects and enhance patients’ QoL. It includes integrative strategies and resources to manage physical, emotional, nutritional, and financial concerns, including managing fatigue, treatment-related side effects, and cost-of-care resources. HCPs described it as “timesaving” and engaging, with one noting, “The fatigue panel should be given to everyone.” Patients appreciated its practical actionable tools, stating, “[it’s] very empowering, allows people to do something.” Preliminary feedback indicated greater confidence in managing symptoms, increased resource awareness, and greater participation in shared decision-making with HCPs. Patients rated the use likelihood as extremely likely (10/10). Conclusions: Your Guide to Managing Symptoms from Metastatic Breast Cancer Treatment fills key gaps in patient disease state and financial support education, symptom management, and shared decision-making for those living with MBC. By integrating evidence-based co-created content with stakeholder insights, the Guide offers a practical accessible tool for whole-person care. Early feedback suggests it enhances patient confidence, promotes meaningful dialogue with HCPs, and may contribute to improved QoL. Ongoing usage and impact evaluation will inform future resource development and dissemination.
Presentation numberPS1-03-30
Assessment of risk factors, monitoring strategies, incidence, and management of trastuzumab deruxtecan (T-DXd)-related interstitial lung disease (ILD) in metastatic breast cancer with brain metastases
Heather Moore, Duke Cancer Institute, Durham, NC
H. Moore1, A. Kupper1, R. Bansal2, C. Anders2, J. Burrows3, P. Alice3, A. Erkanli3; 1Pharmacy, Duke Cancer Institute, Durham, NC, 2Medicine, Duke Cancer Institute, Durham, NC, 3Biostatistics and Informtics, Duke Cancer Institute, Durham, NC.
BackgroundTrastuzumab deruxtecan (T-DXd) is a highly effective antibody-drug conjugate (ADC) indicated for the treatment for metastatic, HER2-positive or HER2-low/ultra-low, breast cancer (MBC) with objective response rates between 50-80% as reported within DESTINY-BREAST01, -03, -04, and -06. Despite its efficacy, T-DXd carries a black box warning for interstitial lung disease (ILD)/pneumonitis with approximately 10-15% risk of ILD development including some grade (G) 5 events. Given G2-4 ILD result in permanent discontinuation of T-DXd, identifying ILD earlier at G1 is essential to maintain therapy; however, monitoring guidance remains limited and variable amongst individual practice. The tumor microenvironment (TME) plays an important role in CNS tumor growth as cancerous cells create complex relationships that reorganize the local TME and reprogram CNS cells, including lymphocytes. It is not yet known if an association exists between patients (pts) with brain metastases (BrM) and decreased total lymphocyte counts and an increased risk of developing opportunistic infections (OI) and/or T-DXd-related ILD. However, the DESTINY-Breast12 study suggested potentially higher rates of ILD in pts with BrM than in pts without and 5 pts in the BrM group concurrently developed an OI. As T-DXd use continues to extend across other tumor types, identifying risk factors (RF) that may increase ILD risk as well as improved monitoring strategies is essential. This retrospective study aims to assess the incidence of ILD and OI, associated RF, monitoring strategies, and management of T-DXd-related ILD in MBC with BrM or leptomeningeal diseases (LMD) as well as assess any correlation with lymphocyte depletion. MethodsThis is a single-center, retrospective, cohort study in pts with MBC with BrM and/or LMD at the Duke Cancer Institute (DCI) Center from 12/2019 to 9/2024. Eligible pts were ≥18 years of age with a diagnosis of HER2 ultra-low/low/positive MBC with BrM and/or LMD receiving T-DXd, with or without additional ILD monitoring to include CT-ILD or standard restaging (PET or CT). ResultsA total of 36 pts with HER2-ultra low, low or positive MBC who had received T-DXd were included. A total of 23 pts had HR+ MBC, and 6 pts with TNBC. Approximately 36 pts and 6 pts had BrM and LMD, respectively. In the study cohort, 7 pts (19.4%; 95% CI 8.2%, 36%) developed ILD. One pt had G1 ILD, 5 pts had G2 ILD, and 1 pt had G3 ILD. Among pts who developed ILD, the average time from start of T-DXd treatment to ILD onset was 7.3 (range: 0.3-15.4) months. All 7 pts with confirmed ILD were treated with corticosteroids. Of pts who had a decreased lymphocyte count at the time of cycle 1 day 1 of T-DXd treatment, 4/15 (26.7%) developed ILD. Among pts with a history of smoking tobacco and pts with no history of smoking tobacco, 3/12 (25.0%) and 4/24 (16.7%) developed ILD, respectively. Among pts < 60 yo and pts ≥60 yo, 4/20 (20.0%) and 3/16 (18.8%) developed ILD, respectively. No pts in the study cohort had a history of COPD, ILD, or CKD. Two pts developed an OI while on treatment with T-DXd (5.6%). One pt who developed an OI did not develop ILD while one pt did develop ILD. Additional results to be presented with increased sample size and ILD monitoring strategy. ConclusionA numerically higher rate of ILD was observed in pts with BrM and/or LMD compared to rates previously reported. The results of this study do not suggest an association between the RF assessed and T-DXd-related ILD, however, this may be due to the small sample size. Further investigation is needed to determine if pts with BrM and/or LMD may be at an increased risk of OI or ILD, however, 26.7% of pts with a decreased lymphocyte count at T-DXd initiation developed ILD, suggesting a potential correlation.
Presentation numberPS1-05-01
Predictors of medication-related osteonecrosis of the jaw in HR+/HER2- metastatic breast cancer patients treated with CDK4/6 inhibitors
Tajana Silovski, University Hospital Center Zagreb, Zagreb, Croatia
S. Jaman1, J. Jaman2, T. Tokic3, I. Todoric3, L. Trkmic3, M. Krizic1, K. Cular1, T. Silovski1; 1Department of Oncology, University Hospital Center Zagreb, Zagreb, CROATIA, 2Department of Plastic, Reconstructive and Aesthetic Surgery, Dubrava University Hospital, Zagreb, CROATIA, 3School of Medicine, School of Medicine, University of Zagreb, Zagreb, CROATIA.
Predictors of Medication-related Osteonecrosis of the Jaw in HR+/HER2- Metastatic Breast Cancer Patients Treated with CDK4/6 Inhibitors Background: Hormone receptor positive, human epidermal growth factor receptor 2 negative (HR+/HER2-) metastatic breast cancer (mBC) represents 60-70% of all mBC, with bones being the most common site of metastases in ~60% of cases. Combination of endocrine therapy (ET) and cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) is the standard treatment in HR+/HER2- mBC with addition of antiresorptive therapy (ART) in cases of bone metastases. ART implies application of parenteral or peroral bisphosphonates as well as denosumab. Medication-related osteonecrosis of the jaw (MRONJ) is a known side effect of all ART. This study aimed to assess the incidence and predictors of MRONJ in HR+/HER2− mBC patients receiving CDK4/6 inhibitors and ART, with emphasis on treatment type, duration, and cumulative dose. Methods: A retrospective cohort of 193 female patients with HR+/HER2− mBC treated with CDK4/6i (ribociclib, palbociclib, abemaciclib) and ART (zoledronate, ibandronate, denosumab, clodronate and pamidronate) in the University Hospital Center Zagreb was analyzed with prior Ethics Committee approval. Descriptive statistics, group comparisons, and logistic regression models were used to identify clinical and treatment-related predictors of MRONJ. Results: MRONJ was developed in 14 out of 193 patients (7.3%), who were significantly older (71.6 ± 8.0 vs. 63.8 ± 13.8 years; p = 0.0039) and had longer CDK4/6i exposure (median 48.0 vs. 22.0 months; p = 0.047). MRONJ incidence was higher in patients treated with abemaciclib (13.0%) and ribociclib (9.9%) than with palbociclib (2.5%), though this difference did not reach statistical significance (p = 0.0952). Among 193 patients receiving ART, most commonly used ART agents were zoledronic acid in 104 patients (53.9%), followed by ibandronate in 71 patients (36.8%) and denosumab in 18 patients (9.3%). Among the 14 patients who developed MRONJ, 8 (57.1%) had received zoledronic acid, 4 (28.6%) ibandronate, and 2 (14.3%) denosumab. Adjusted cumulative ART doses were calculated, and higher doses were observed in MRONJ compared to the non-MRONJ group (6266,85 mg vs 3848,8 mg respectively), without reaching statistical significance (p = 0.47). Total ART duration was significantly longer in the MRONJ group (52.71 vs. 33.98 months), with a p-value of 0.033. Conclusion: Older age and longer CDK4/6i exposure was associated with an increased risk of MRONJ in patients with HR+/HER2− mBC. Although MRONJ incidence appeared higher with abemaciclib and ribociclib compared to palbociclib, it did not reach statistical significance, possibly due to small numbers. These findings highlight the need for careful monitoring and risk assessment in patients undergoing long-term CDK4/6i therapy in combination with ART.
Presentation numberPS1-05-02
Efbemalenograstim alfa significantly reduces incidence of incidence of severe chemotherapy-induced neutropenia in later cycles: Results of a meta analysis
John A. Glasby, David Geffen School of Medicine at UCLA, Los Angeles, CA
J. A. Glasby; Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA.
Introduction: Efbemalenograstim alfa, a non-PEGylated, long-acting, human granulocyte-colony stimulating factor was recently evaluated for the management of chemotherapy-induced neutropenia in a phase III trial of 393 patients with stage I-III invasive breast cancer. The study met the primary end point, demonstrating that a single dose of efbemalenograstim alfa was noninferior to pegfilgrastim, decreasing the duration of severe neutropenia in cycle 1. An additional endpoint of the phase III trial was the incidence of severe neutropenia (ISN; grade 4, ANC <0.5 × 109/L) across all chemotherapy cycles, which demonstrated significant reductions in ISN at later cycles. Here, we aimed to investigate is these reductions in ISN in later chemotherapy cycles occurred in previous trials of efbemalenograstim alfa and with different chemotherapy regimens. Methods: ISN rates from four cycles from phase III trials of efbemalenograstim alfa and three different chemotherapy regimens, docetaxel + cyclophosphamide (TC) (Trial 1), epirubicin + cyclophosphamide (EC) (Trial 2), and high myelotoxic docetaxel + doxorubicin (TA) (Trial 3 + historic pegfilgrastim and filgrastim comparative data) were evaluated. Results: Trial 1 included 197 patients randomized to efbemalenograstim alfa, compared to 196 randomized to pegfilgrastim that received TC chemotherapy. During cycle 1, ISN for both groups was 11.7%, but for subsequent cycles, ISN was lower for patients that received efbemalenograstim alfa compared to pegfilgrastim, 4.6% vs 5.1% for cycle 2, 2.6% vs 6.3% for cycle 3 (p=0.08) and significantly lower, 1.6% vs 5.3% for cycle 4 (p=0.05), respectively. Trial 2 included 122 patients randomized to efbemalenograstim alfa, compared to 120 randomized to filgrastim that received EC chemotherapy. During cycle 1, ISN was 14.2% for the efbemalenograstim alfa, compared to 16.0% for the filgrastim groups, with subsequent cycles at 1.7% vs 0.9%, respectively, for cycle 2, significantly lower, 0% vs 3.9% for cycle 3 (p=0.048) and 1.8% vs 4.0% for cycle 4. Trial 3 included 83 patients randomized to efbemalenograstim alfa and 39 randomized to placebo, therefore ISN for the TA chemotherapy regimen was compared to a historic pegfilgrastim + TA and filgrastim + TA phase III trial data, which showed an ISN at cycle 1 of 69.9% for efbemalenograstim alfa and 77-84% for pegfilgrastim, and 79-83% for filgrastim. ISN at cycle 2 was 15% for efbemalenograstim alfa and much higher for pegfilgrastim 45-57% and filgrastim 54-55%, and similarly for cycle 3, 20%, 37-56% and 53-60% and cycle 4, 10%, 45-51% and 49-55%, respectively. Conclusion: Across studies, non-PEGylated efbemalenograstim alfa has demonstrated equivalent or lower incidence rates of severe neutropenia (grade 4, ANC <0.5 × 109/L) during the first cycle of three types of chemotherapy (TC, EC and TA) and importantly, significantly lower rates in later cycles when compared to pegfilgrastim and filgrastim. These findings demonstrate that efbemalenograstim alfa may be more effective at preventing severe, life-threatening neutropenia at later chemotherapy cycles, which has meaningful implications for real-world breast cancer patients that are likely to receive chemotherapy for longer durations.
Presentation numberPS1-05-03
Vitiligo-like Lesions Associated with CDK4/6 Inhibitors in Metastatic HR+/HER2− Breast Cancer: Multiple case reports from a Brazilian experience.
Rafaela Melo Campos Borges, Rede Américas, Hospital 9 de Julho, São Paulo, Brazil
R. M. Borges1, M. C. Gouveia1, C. Giro1, M. Aisen1, L. C. Oliveira1, N. J. Gomes2, A. C. Ferrari3, T. G. Rivelli3, M. S. Neto3, M. L. Brito4, R. B. Sousa5, K. P. Sacardo6, B. M. Zucchetti1; 1Oncologia Clínica, Rede Américas, Hospital 9 de Julho, São Paulo, BRAZIL, 2Oncologia Clínica, Hospital São Domingos, São Luís-MA, BRAZIL, 3Oncologia Clínica, Rede Américas, Hospital Santa Paula, São Paulo, BRAZIL, 4Oncologia Clínica, DASA Oncology – Clínica AMO, Salvador-BA, BRAZIL, 5Oncologia Clínica, Brasília Hospital, Oncologia Américas, Brasilia, BRAZIL, 6Oncologia Clínica, Rede Américas, Hospital Christóvão da Gama, Santo André – SP, BRAZIL.
Introduction CDK4/6 inhibitors have significantly changed altered the natural history of HR+/HER2− metastatic breast cancer, leading to substantial improvements in progression-free and overall survival. These drugs are generally well tolerated, with manageable toxicity profiles. However, rare dermatologic adverse events may be underreported and poorly understood. In this study, we describe case series of patients who developed vitiligo-like lesions during treatment with ribociclib or abemaciclib. Case Series Description A total of eight women with HR+/HER2− metastatic breast cancer receiving ribociclib (n=7) or abemaciclib (n=1), all in combination with endocrine therapy (aromatase inhibitor or fulvestrant), developed well-demarcated depigmented lesions consistent with vitiligo-like lesions. None had a prior history of autoimmune or dermatologic disease. Dermatologic evaluations and lesion photographs were obtained. In two cases, skin biopsies revealed pigmentary incontinence and chronic perivascular mononuclear infiltrate.Cutaneous changes developed within a median of 8 months (minimum 6 and maximum 24 months) after treatment initiation. The lesions were asymptomatic, hypopigmented, and primarily involved the face, neck, and upper limbs. No systemic symptoms were reported. Dermatological evaluation ruled out other differential diagnoses and suggested an immune-mediated mechanism. In one patient, treatment was permanently discontinued after two years due to cutaneous toxicity from vitiligo-like lesions, with sustained oncologic response; another patient had a temporary 3-month drug interruption due to lesion worsening, after which the skin changes stabilized. All patients were treated with topical corticosteroids, resulting in partial improvement. Complete lesion regression was observed in the patient who discontinued therapy; three patients had stable lesions while continuing medication; others showed partial regression after treatment change due to disease progression. Discussion The most common skin toxicities associated with ribociclib and abemaciclib include rashes and dermatitis (10-20%). Vitiligo-like reactions are rare. CDK4/6 inhibitors may affect the proliferation of keratinocyte precursors, compromising the secretion of melanocyte-supporting cytokines. Treatment-induced melanocyte apoptosis may contribute to depigmentation. Additionally, ultraviolet radiation-induced melanocyte damage may trigger immune responses, highlighting the importance of sun protection. However, the underlying pathophysiology remains unclear. Conclusion This case series highlights a potentially underrecognized cutaneous adverse event associated with CDK4/6 inhibitors (ribociclib and abemaciclib). Oncologists should be aware of vitiligo-like reactions as part of the toxicity spectrum of these agents. It is important to point out that this is not a life-threatening condition but it can affect appearance, specially of the face, causing burden for patients. Early recognition and collaborative management with dermatology can allow continuation of cancer therapy without compromising treatment outcomes or quality of life. Further studies are needed to better understand this phenomenon.
Presentation numberPS1-05-04
A multi-practice quality initiative evaluating antiemetic use in metastatic breast cancer patients treated with trastuzumab deruxtecan-nxki (T-DXd)
Stephan Rosenfeld, Highlands Oncology Group (HOG), Fayetteville, AR
S. Rosenfeld1, O. Ajayi1, R. Choksi2, S. W. Champaloux3, D. Parris3, A. Rui3, A. Lamb3, M. Gart3, C. Wall3, B. Wang3, P. Varughese3, J. Donegan3, L. Morere3, R. Geller3, J. Scott3, V. Gorantla2; 1Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 2Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3PrecisionQ, IntegraConnect, West Palm Beach, FL.
Background: As of January 2023, the National Comprehensive Cancer Network (NCCN) guidelines recommend using combinations of 5HT3, NK1, olanzapine (O), and dexamethasone (DEX) drugs as prophylaxis with highly emetic therapies, including fam-trastuzumab deruxtecan-nxki (T-DXd). An IntegraConnect PrecisionQ multi-practice quality initiative was conducted with Highlands Oncology Group (HOG), University of Pittsburgh Medical Center (UPMC), and Regional Cancer Care Associates (RCCA) to assess adherence to recommended antiemetic use in HER2-positive (HER2+) and HER2-low metastatic breast cancer (mBC) patients (pts) treated with T-DXd. This initiative compared current practices to NCCN guidelines, identified potential gaps in care, and implemented strategies to address these gaps. Our goal was to demonstrate improvements in antiemetic utilization for mBC pts treated with T-DXd as a result of this quality initiative. Methods: Using the IntegraConnect PrecisionQ de-identified database, HER2+ and HER2-low pts treated with T-DXd were evaluated for specific time periods at each practice related to antiemetic utilization. HOG (baseline: 1/1/2022-12/31/2023 and follow-up: 01/01/2024-06/04/2025), UPMC (baseline: 1/1/2022-04/30/2024 and follow-up: 05/01/2024-06/04/2025), RCCA (baseline: 1/1/2022-01/01/2024 and follow-up: 02/01/2024-06/04/2025). For the baseline period, we completed a retrospective analysis of the antiemetic utilization within the practices’ HER2+ and HER2-low patient cohorts. The results of the initial baseline analysis were shared with practice leaders and among oncologists at the practices. The practices updated antiemetic order-sets, and we evaluated the order-set changes during the follow-up period to account for any improvement in antiemetic utilization. Data are presented using descriptive statistics. Results: The total pts evaluated in the initiative were: HOG baseline (49 pts) and follow-up (27 pts); UPMC baseline (212 pts) and follow-up (110 pts); and RCCA baseline (132 pts) and follow-up (99 pts). In the follow-up analysis, utilization of the recommended combinations of antiemetics (5HT3+NK1+O±DEX or 5HT3+NK1±DEX) increased from 47% to 85% at RCCA, from 29% to 100% at UPMC, and from 49% to 100% at HOG, with the overall rate increasing from 37% to 94%. Conclusion: This multi-practice quality initiative led to improved antiemetic utilization that is in line with NCCN guidelines in HER2+ and HER2-low mBC pts treated with T-DXd. These findings highlight that with established guidelines, oncology practices can benefit from regularly evaluating their own data to assess their treatment compared to the latest clinical standards.
Presentation numberPS1-05-06
Breast Calciphylaxis in a Patient with ESRD and Prior Radiation – A Diagnostic and Therapeutic Challenge
Nikki S Verma, UTHSCSA, San Antonio, TX
N. S. Verma1, R. Kannaiyan2, K. Lathrop2; 1Internal Medicine, UTHSCSA, San Antonio, TX, 2Hematology and Oncology, UTHSCSA, San Antonio, TX.
Intro Calciphylaxis is commonly associated with CKD and results from calcium deposition in dermal and adipose vasculature1. Diagnosis is often challenging with lesions typically presenting on the lower extremities or abdomen; involvement of the breast is rare2. This case describes a patient with breast calciphylaxis requiring multi-specialty management. Case Presentation A 69-year-old female with a history of grade 3 ER+/PR+/HER2− right breast intraductal carcinoma, status post partial mastectomy, axillary dissection and right breast radiation, presented with progressive skin changes of the right breast. Prior systemic therapy included two cycles of cyclophosphamide and docetaxel (discontinued due to rectal bleeding requiring exploratory laparotomy and partial colectomy), hormonal therapy, and radiation. Medical history is also significant for ESRD on HD, CHF, and pulmonary hypertension. Patient was diagnosed with breast cancer in 2012 and had been under routine oncologic surveillance. In 2022, she noticed skin changes involving the right breast. Examination revealed skin thickening beneath the right breast, with satellite lesions and areas of hyperpigmentation and hypopigmentation. A biopsy of the right inframammary fold demonstrated dermal sclerosis and minute calcifications. These findings raised suspicion for radiation dermatitis versus calcium deposition. However, malignancy could not be definitively excluded, as the initial punch biopsy failed to penetrate the dense, plate-like nodules adequately. Screening mammograms remained negative since her initial diagnosis. The patient’s symptoms progressed with skin breakdown and drainage in areas of calcification. Laboratory studies remained consistent with ESRD but showed no acute uremic exacerbation. A recent breast ultrasound revealed multiple calcified areas, most prominent in the inferior breast, consistent with calcinosis cutis. The patient’s pain and tenderness became severe enough to preclude further mammography. Treatment attempts included antifungal and barrier creams, calcium binders, and a trial of sodium thiosulfate, none of which provided significant relief. The patient was not a suitable candidate for calcium channel blockers or bisphosphonates. Surgical excision was deemed high-risk due to the extensive resection area required, with concerns regarding poor wound healing, dehiscence, and recurrence. The patient declined surgical intervention and opted for continued follow-up with dermatology and the wound care. A repeat biopsy, two years after the initial one, demonstrated dermal fibrosis, perivascular plasmocytic inflammation, and vascular ectasia, without evidence of malignancy. Discussion Calciphylaxis is a rare but often fatal condition, with wound-related infections contributing to mortality rates of up to 60% within one year3. This case highlights the need for a high index of suspicion, particularly in patients with ESRD and lesions in uncommon locations such as the breast. Diagnosis can be difficult due to overlapping features with conditions like radiation dermatitis or recurrent malignancy and may require multiple biopsies. In this patient, differentials included calcinosis cutis, scleroderma, inflammatory breast cancer, and chronic radiation dermatitis. Classic calciphylaxis pathology findings include medial calcification of dermal arterioles, fibrointimal hyperplasia, microthrombi, and necrosis, demonstrating some overlap with pathology seen in this case. Although calciphylaxis is strongly associated with ESRD, most dialysis patients do not develop it. In this patient, the coexistence of malignancy, prior radiation, and ESRD may have played a synergistic role in disease development.
Presentation numberPS1-05-07
Guided cannabis care and its impact on personally important outcomes among cancer patients: a prospective assessment
Brooke Worster, Jefferson Health, Philadelphia, PA
B. Worster1, K. Parton2; 1Supportive Oncology, Jefferson Health, Philadelphia, PA, 2Senior Data Modeler, EO Care, Boston, MA.
Guided cannabis care and its impact on personally important outcomes among cancer patients: a prospective assessmentBackground:Increasingly, patients with cancer explore cannabis as an adjunct therapy to help manage symptoms and improve quality of life. However, rigorous evaluations of its effectiveness in supporting meaningful functional gains—particularly those prioritized by patients themselves—remain limited. Personally Important Outcomes (PIOs) provide a patient-centered metric that targets specific daily activities each individual deems critical to their quality of life. We set out to determine whether a structured, guided cannabis care program could meaningfully improve patient-designated PIOs.Methods:Patients were recruited from multiple oncology centers, a dedicated marketing site, and various cancer advocacy organizations. Each patient completed an intake survey covering their health history, personal information, and a baseline PIO survey, identifying up to three activities they wished to track (e.g., walking, cooking). Each activity was scored on a 10-point scale, with a total of 119 activities tracked across 49 patients. Using a Bayesian model built from available research and real-world outcomes data, a personalized care plan was constructed by integrating patient profiling inputs into individualized recommendations for products, dosages, and times of use. The plan was then reviewed by a clinician before being sent to the patient. Regular check-ins were conducted to monitor efficacy and side effects, and the regimen was adjusted as needed based on patient feedback. Throughout treatment, periodic reassessments of PIO scores allowed investigators to track functional changes over time and determine whether cannabis interventions correlated with improvements.Statistical Analyses:To quantify the magnitude of changes in PIO scores, we conducted a paired-sample Cohen’s d analysis comparing baseline and an average of follow-up assessment scores.Results:Analysis of changes in PIO scores revealed a large effect size (Cohen’s d=1.01), indicating a substantial improvement in patients’ ability to engage in their activities. This effect was statistically significant (p=0.000). At baseline, participants frequently cited mobility challenges, fatigue during daily tasks, and difficulty with daily activities as key concerns. Following guided cannabis care, many reported higher levels of daily functioning and reduced perceived difficulty. At 4 weeks, only 15% of patients experienced side effects, which included dizziness, anxiety, feeling overly intoxicated, or an elevated heart rate. Qualitative feedback suggested that patients valued having a structured approach to cannabis, including guidance on product selection and dosing strategies tailored to their symptoms. Such individualized support may have enhanced treatment adherence and optimized outcomes. Conclusions:These data underscore the potential of guided cannabis care to deliver meaningful functional improvements for both survivors of cancer and for patients in active cancer treatment. Because PIOs reflect personal priorities—rather than generic benchmarks—this method captures patient-specific progress in a more nuanced manner than many existing patient-reported outcome measures. An effect size of 1.01 demonstrates that the improvement is both clinically relevant and statistically robust, reinforcing the importance of incorporating guided cannabis care into oncology care when appropriate.
Presentation numberPS1-05-08
Incidence and predictors of interstitial lung disease associated with antibody-drug conjugates in breast cancer: a systematic review and meta-analysis
Heather S. Wenning, Oncology Hematology Care, Inc, Cincinnati, OH
H. S. Wenning1, T. Takahashi2, A. Argulian3, R. Bardhan4, K. Chida5, R. Paredes6, Y. Fujiwara7, A. M. Roy8; 1Hematology-Medical Oncology, Oncology Hematology Care, Inc, Cincinnati, OH, 2Department of Medicine, University of Hawaii, John A. Burns School of Medicine, Honolulu, HI, 3Icahn School of Medicine, The Mount Sinai Hospital, New York, NY, 4Department of Medicine, The Jewish Hospital – Mercy Health, Cincinnati, OH, 5Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 6Department of Medicine, Icahn School of Medicine at Mount Sinai Beth Israel, New York, NY, 7Hematology-Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 8Department of Medicine, The Ohio State University, Columbus, OH.
Introduction: Antibody-drug conjugates (ADCs) have emerged as a transformative therapeutic class in breast cancer (BC), offering targeted cytotoxic delivery with improved efficacy; however, interstitial lung disease (ILD) and pneumonitis have been increasingly recognized as serious, and sometimes fatal, toxicities. Their incidence, severity, and clinical predictors remain incompletely characterized across trials. This meta-analysis quantifies and explores the potential predictors of ILD and pneumonitis in BC patients treated with ADCs. Methods: We conducted a systematic review and meta-analysis of phase I-III clinical trials evaluating ADCs in BC to estimate the pooled incidence of ILD, including drug-related pneumonitis, and to assess potential contributing factors. ILD rates were stratified by Common Terminology Criteria for Adverse Events (CTCAE) grade. The incidence was pooled using a random-effects proportional meta-analysis. Subgroup and meta-regression analyses evaluated the impact of ADC type, payload, dose, BC subtype, drug-antibody ratio (DAR), and line of therapy. Study and subgroup heterogeneity were evaluated using the I² statistic, with I² greater than or equal to 50% considered high. Results: Across the 48 included trials (N = 10607 patients), 664 developed ILD/pneumonitis, yielding a pooled incidence of ILD/pneumonitis of 7.5% (95%CI: 4.5%-11.2%; I² = 96.2%, τ² = 0.0321, p < 0.0001). Most events were low-grade (grade 1-2: 6.4%), with fewer high-grade (grade 3-4: 0.4%) and fatalities (grade 5: 0.2%) events.The ADC, ARX788 had the highest overall ILD incidence (47.6%), largely driven by grade 1-2 events (42.0%). Trastuzumab deruxtecan (T-DXd) also demonstrated notable ILD incidence at 12.5%, including a fatality rate of 0.93% (grade 5).In a mixed-effects meta-regression model, payload type significantly influenced ILD incidence (R² = 90.3%, p 0.05). ILD incidence by payload was highest with DXd (10.7%), and lower with microtubule inhibitors (3.3%). Notably, studies that excluded patients with pre-existing lung disease reported a slightly higher pooled ILD rate (10.0%).Treatment modifications due to ILD/pneumonitis (dose reduction, delay, or discontinuation) occurred most frequently with T-DXd, particularly in DESTINY-Breast(DB)03 (11.9%), DB-02 (9.4%), and DB-01 (9.2%). Across studies reporting these outcomes, discontinuation rates ranged from 0.0% to 11.9% (median: 2.8%, IQR: 0.3%-4.8%). Conclusion: ILD, including pneumonitis, is a clinically relevant toxicity of ADC therapy in BC. Incidence and severity vary across agents. Payload type emerged as the strongest predictor of ILD risk, underscoring its importance in ADC development and patient selection. ARX788 was associated with the highest ILD risks, while T-DXd had the highest rate of treatment interruptions.
Presentation numberPS1-05-09
Utilization and safety of concurrent antibody drug conjugates alongside extracranial radiation therapy in the real-world setting
Ameer Basta, University of South Florida, Tampa, FL
A. Basta1, A. Klug1, G. Ni1, L. Wajahat1, W. Alhassani1, M. Awad1, V. Khatri2, R. Costa3, M. Mills2, I. Washington2, R. Diaz2, K. Ahmed2; 1Morsani College of medicine, University of South Florida, Tampa, FL, 2Radiation Oncology, Moffitt Cancer Center, Tampa, FL, 3Breast Oncology, Moffitt Cancer Center, Tampa, FL.
Utilization and safety of concurrent antibody drug conjugates alongside extracranial radiation therapy in the real-world setting Ameer Basta, Alexander Klug, Gabris Ni, Leena Wajahat, Wid Alhassani, Mark Awad, Vaseem Khatri, Ricardo Costa, Matthew Mills, Iman Washington, Roberto Diaz, Kamran Ahmed Background Antibody-drug conjugates (ADCs) have become an integral part of systemic therapy for breast cancer. As radiation therapy (RT) continues to play a central role in the management of breast cancer, the safety of combining ADCs with RT has become an emerging area of clinical interest. However, there remains limited data regarding the safety of ADCs administered concurrently with extracranial RT. This study aims to assess real-world data on the safety of combining ADCs with extracranial RT. Method A retrospective chart review of patients who received concurrent ADC therapy (trastuzumab emtansine (T-DM1), sacituzumab govitecan (SG), and trastuzumab deruxtecan (T-DXd)) and extracranial RT at a single academic institution was conducted. RT was delivered to the breast and/or chest wall with or without regional nodal irradiation (RNI). Demographic, clinical, and treatment data as well as adverse events were extracted from the electronic medical record. Concurrent therapy was defined as ADC administration within 4 weeks before or after the start of RT. Results A total of 49 patients who received extracranial RT in conjunction with ADCs were included in this retrospective analysis. Average months of follow up for the patients was 22 months after receiving RT. The median age of patients was 54 (range 30-76) at the time of RT. A majority patients were stage 0-2 (n=32, 65%), HR+ (n=35, 71%), HER2+ (n=40, 82%), post-menopausal (n=31, 63%), and received T-DM1 (n=43, 88%). Four patients received SG and two patients received T-DXd. The most common site treated was the chest wall with regional nodal irradiation (n=26, 53%) with 50 Gy in 25 fractions (n=34, 69%). The most frequently reported side effects were grade 1-2 skin dermatitis which was reported in 36 patients (73%), and fatigue in 22 patients (45%). Three patients had grade 3 dermatitis and one had grade 1 esophagitis. Importantly, no radiation courses were discontinued due to adverse effects. Conclusion In our study of concurrent use of ADCs with RT to breast and/or chest wall +/- RNI, no unexpected toxicities occurred. This study adds to real-world evidence that extracranial radiation therapy can be safely administered alongside ADCs.
Presentation numberPS1-05-10
Clonal Hematopoiesis in Breast Cancer: Underrecognized Predictor of Anthracycline Cardiotoxicity Reviewing Prevalence, Mechanisms, and Prognostic Implications
Chinemerem M Emeasoba, UAM, fayettville, AR
C. M. Emeasoba1, C. okoye2; 1Internal medicine, UAM, fayettville, AR, 2Internal medicine, North east Georgia, Gainesville. GA, AR.
Background: Clonal hematopoiesis of indeterminate potential (CHIP), defined by somatic mutations in hematopoietic stem or progenitor cells without cytopenias, dysplasia, or overt hematologic malignancy, is a common age-related phenomenon. Its prevalence increases with age, affecting 10-20% of individuals over age 70 and up to 40% of older women with breast cancer. CHIP is enriched in patients exposed to cytotoxic therapies—particularly anthracyclines and alkylating agents—which may drive the selection and expansion of clones harboring high-risk mutations, notably in TP53 and PPM1D, genes involved in the DNA damage response. While initially described in hematologic contexts, CHIP has emerged as a potent risk factor for cardiovascular disease (CVD), including coronary artery disease, myocardial infarction (MI), and heart failure (HF). Independent of traditional cardiovascular risk factors like hypertension, diabetes, and hyperlipidemia, CHIP confers a two- to four-fold increase in CVD risk. Despite these associations, CHIP remains unaccounted for in current cardio-oncology guidelines from the American Heart Association (AHA) and American College of Cardiology (ACC). Methods: We conducted a comprehensive literature review using PubMed, Scopus, and Embase through June 2025. Peer-reviewed studies were included if they assessed CHIP prevalence, gene mutation profiles, CVD outcomes, or anthracycline-induced cardiotoxicity (AIC) in breast cancer patients. Key data extracted included mutation frequencies, clonal evolution post-chemotherapy, hazard ratios (HRs) for cardiovascular events, and left ventricular ejection fraction (LVEF) changes. Results: CHIP mutations—particularly in DNMT3A, TET2, ASXL1, TP53, and PPM1D—are found in 15-40% of breast cancer patients, often detectable prior to chemotherapy. TP53 and PPM1D mutations frequently expand post-anthracycline exposure. Population studies report a 25-100% increase in risk for HF and MI in individuals with CHIP, with TET2 and ASXL1 mutations showing the strongest associations. Mechanistically, CHIP promotes chronic inflammation via IL-1β and NLRP3 inflammasome activation, leading to myocardial injury and fibrotic remodeling. One prospective cohort study found CHIP to be independently associated with an eight-fold increased risk of symptomatic HF or LVEF decline after anthracycline therapy, even after adjusting for age, cumulative dose, and baseline risk factors. However, data remain limited by small sample sizes, heterogeneous cancer types, and inconsistent CHIP definitions. Conclusion: CHIP is a common, age-related genomic alteration with significant cardiovascular implications in breast cancer patients, especially those treated with anthracyclines. Despite emerging evidence linking CHIP to AIC, it remains absent from current risk stratification models. Prospective, breast cancer-specific studies are needed to validate CHIP as a predictive biomarker and inform its integration into personalized cardio-oncology management strategies.
Presentation numberPS1-05-11
Exploratory Analysis of Nausea and Vomiting Probability over Multiple Cycles of NEPA (netupitant/palonosetron) Prophylaxis in Metastatic Breast Cancer Patients Treated withTrastuzumab Deruxtecan
Serena Capici, Fondazione IRCCS San Gerardo, Monza, Italy
M. E. Cazzaniga1, S. Capici2, G. Passarella3, F. F. Pepe2, T. Clementi1, F. Cicchiello3, F. Riva3, P. Cafaro4; 1Medicine and Surgery, University of Milano Bicocca, Monza, ITALY, 2Phase 1 Research Centre, Fondazione IRCCS San Gerardo, Monza, ITALY, 3Oncology Unit, Fondazione IRCCS San Gerardo, Monza, ITALY, 4Oncology Unit, Fondaizone IRCCS San Gerardo, Monza, ITALY.
Background:Nausea and vomiting (NV) are the most frequent side effects in patients with metastatic breast cancer (MBC) treated with trastuzumab deruxtecan (T-DXd). Since MBC patients may receive T-DXd for an extended period, effective management of NV could help maintain dose intensity, reduce the risk of treatment discontinuation, and improve quality-of-life.Preventing NV can also contribute to reduce other adverse events, like fatigue Effective NV control from the first cycle with a NK1 receptor antagonist (RA)-based regimen is crucial for optimal management throughout subsequent cycles.This exploratory retrospective analysis examined the probability of patients experiencing NV over multiple cycles of T-DXd following antiemetic prophylaxis with NEPA, a fixed-combination of an NK1 RA (netupitant/fosnetupitant) and 5-HT3 RA (palonosetron).Methods:Occurrence and severity of NV in patients with HER2-positive or HER2-low MBC over the first 5 cycles of T-DXd for each patient were extracted and analyzed from clinical records at a single-center in Italy.Probability of NV occurring in each of 5 cycles of T-DXd was estimated using a logistic regression with NV as a dependent variable, cycle as an independent variable and with Generalized Estimating Equation to account for repeated measures. Results:Twenty-five patients were included for a total of 118 cycles. NEPA with dexamethasone was administered as prophylaxis in all but 6 cycles. The median age at T-DXd initiation was 64 years (range, 41-78). The estimated probability of developing NV was low and diminished over each cycle (Table), suggesting that NV was well-controlled with NEPA from the first cycle onward. Table: Probability of NV Occurrence During Each Cycle (Estimates Obtained from a Logistic Regression) Conclusion:These exploratory findings indicate that NEPA is likely to be highly effective in preventing NV over multiple cycles in MBC patients receiving T-DXd. It also underscores the value of using an appropriate upfront antiemetic regimen from the first cycle of T-DXd.
| Cycle | Chances of NV occurrence | Lower 95% CI | Upper 95% CI | ||||
| 1 | 0.2800 | 0.1397 | 0.4822 | ||||
| 2 | 0.1200 | 0.0392 | 0.3130 | ||||
| 3 | 0.0812 | 0.0192 | 0.2856 | ||||
| 4 | 0.0390 | 0.0042 | 0.2812 | ||||
| 5 | 0.0408 | 0.0026 | 0.4101 |
Presentation numberPS1-05-12
Primary G-CSF Prophylaxis in Sacituzumab Govitecan-Treated mTNBC: Real-World Evidence from a Multinational Cohort
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
Z. Bielčiková1, M. Pieniążek2, A. Polakiewicz-Gilowska3, J. Żubrowska4, M. Holánek5, R. Soumarová6, H. Študentová7, A. Konieczna8, M. Malejčíková9, A. Młodzińska8, K. Winsko-Szczęsnowicz10, M. Lisik-Habib11, A. Pękala11, D. Krejčí12, J. Šustr13, I. Kolářová14, I. Danielewicz15, M. Szymanik-Resko15, T. Ciszewski16, L. Rusinova17, B. Czartoryska-Arłukowicz10, A. Łacko18, R. Pacholczak-Madej19, M. Jarzab3, M. Püsküllüoğlu20, M. Kubeczko3; 1Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 2Department of Oncology; Breast Unit Clinical Oncology Day Care Department, rocław Medical University; Lower Silesian Comprehensive Cancer Center, Wrocław, POLAND, 3Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 4Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, POLAND, 5Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, and Masaryk Memorial Cancer Institute, Brno, CZECH REPUBLIC, 6Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 7Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 8Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, POLAND, 9Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 10Department of Clinical Oncology, M. Skłodowska-Curie Bialystok Oncology Center, Białystok, POLAND, 11Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 12Breast Cancer Center, First Faculty of Medicine, Charles University in Prague; Bulovka University Hospital, Prague, CZECH REPUBLIC, 13Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University; University Hospital Pilsen, Plzen, CZECH REPUBLIC, 14Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Králové, CZECH REPUBLIC, 15Oddział Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 16Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 17Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 18Department of Oncology; Breast Unit Clinical Oncology Day Care Department, Wrocław Medical University; Lower Silesian Comprehensive Cancer Center, Wrocław, POLAND, 19Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND, 20Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND.
Background. Sacituzumab govitecan (SG) has emerged as a key agent in the treatment of metastatic triple-negative breast cancer (mTNBC), with use increasingly shifting toward earlier treatment lines. Consequently, the number of patients exposed to SG will continue to grow. Although recent updates to the SG summary of product characteristics list granulocyte colony-stimulating factor (G-CSF) primary prophylaxis as an option, the need for its routine use in all patients remains uncertain. We aimed to evaluate the impact of G-CSF primary prophylaxis on clinical outcomes and treatment-related toxicities in a multinational real-world cohort of patients with mTNBC treated with SG. Methods. Baseline characteristics were balanced with respect to prior systemic treatment for early-stage disease, the number of previous treatment lines in the metastatic setting, comorbidities, metastatic sites, and prior episodes of febrile neutropenia. However, ECOG performance status of 0 was more frequent in the prophylaxis group (50% vs. 37.1%, p=0.034). Efficacy endpoints included progression-free survival (PFS) and overall survival (OS), analyzed using the Kaplan-Meier method. Adverse events were assessed according to CTCAE criteria. All analyses were conducted using Stata Statistical Software version 19.0. Results. G-CSF primary prophylaxis was not associated with improved clinical outcomes. Median PFS was 4.2 months in the primary prophylaxis group versus 5.1 months in the no-primary prophylaxis group (p=0.20), with 6-month PFS rates of 38.5% vs. 42.1%, respectively. Median OS was 10.9 vs. 11.6 months (p=0.95), and 12-month OS rates were 44.9% vs. 47.1%, respectively. There were no statistically significant differences in the rates of all-grade toxicities between groups. Grade ≥3 neutropenia occurred less frequently in patients receiving G-CSF primary prophylaxis (33.0% vs. 50.7%, p=0.005), but this was not accompanied by a reduction in febrile neutropenia rates (4.0% vs. 4.4%, p=1.00). Rates of other all grade adverse events—including anemia, diarrhea, nausea, vomiting, and alopecia—were similar between groups. Dose reductions due to toxicity occurred in 36.0% of the prophylaxis group and 38.4% of the non-prophylaxis group (p=0.71). Among those who did not receive G-CSF primary prophylaxis, 74.4% eventually required secondary G-CSF support. Ultimately, only 17.2% of patients were treated with SG without any G-CSF support. Notably, SG dose reductions due to toxicity were not associated with inferior outcomes. In contrast, patients who required dose reduction had significantly longer PFS (median 6.6 vs. 3.3 months, p<0.001) and numerically longer OS (median 13.0 vs. 10.7 months, p=0.12) compared to those without dose modification. Conclusions. In this large real-world cohort, primary G-CSF prophylaxis was not associated with improved survival or reduced febrile neutropenia, though it significantly reduced the incidence of grade ≥3 neutropenia. Given that the majority of patients ultimately received G-CSF as secondary prophylaxis, and considering the relatively short treatment duration in later lines, primary G-CSF prophylaxis remains a valid option in this setting. Consistent with prior reports, SG dose reductions due to adverse events were not associated with worse survival.
Presentation numberPS1-02-01
Knowledge, risk perception, and decision making in patients with a history of ductal carcinoma in situ (DCIS) or atypical breast lesions: a secondary analysis from the PORTAL Study
Carmine Valenza, Dana-Farber Cancer Institute, Boston, MA
C. Valenza1, S. M. Rosenberg2, K. Crowell3, K. L. Schreiber4, I. Bedrosian5, K. S. Hughes6, T. Lynch3, D. Basila7, D. Collyar8, E. S. Frank9, S. Darai1, C. Lanahan6, J. R. Marks3, J. K. Plichta3, A. M. Thompson10, T. Hyslop11, S. Hwang12, A. H. Partridge1; 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Department of Medical Oncology, Weill Cornell Medicine, New York, NY, 3Department of Medical Oncology, Duke University, Durham, NC, 4Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA, 5Department of Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 6Department of Surgery, Massachusetts General Hospital, Boston, MA, 7., Patient Leadership Team, Boston, MA, 8., Patient Advocates in Research, Danville, CA, 9., Patient Leadership Team, ., MA, 10Department of Medical Oncology, Baylor College of Medicine, Houston, TX, 11Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 12Department of Surgery, Duke University, Durham, NC.
Background: The COMET trial demonstrated that active monitoring (AM) is not inferior to guideline concordant care (GCC) in patients with low-risk ductal carcinoma in situ (DCIS), with comparable rates of ipsilateral invasive breast cancer (iBC) and quality of life outcomes. Similarly, patients with atypical breast lesions have an increased risk of iBC and are managed with an AM strategy which could serve as a proxy for future omission of surgery in low-risk DCIS. Acknowledging that acceptance of treatment deintensification is likely linked to patient preferences, understanding, and risk perception, this analysis assessed these patient-reported outcomes (PROs) among patients with atypical breast lesions and DCIS. Methods: In this secondary analysis of the Patient-reported Outcomes after Routine Treatment of Atypical Lesions (PORTAL) Study – a multicenter cross-sectional survey study – we compared PROs from women with DCIS who received GCC, to women with high-risk breast lesions (e.g., atypical ductal hyperplasia [ADH], lobular intraepithelial neoplasia [LIN]) who underwent AM. We report on participants’ fear of invasive breast cancer (using Quality of Life in Adult Cancer Survivors [QLACS]), and disease understanding and decision quality (using Breast Cancer Surgery Decision Quality Instrument-Sections 1 and 3 [BCS-DQI-S1/3], SURE scale, Decision Regret Scale [DRS]; and Control Preferences Scale [CPS]). Descriptive statistics were provided, with means and standard deviations (SD), or proportions and 95% confidence intervals (CI). Results: 903 patients were included: 538 underwent GCC for DCIS; 365 underwent AM for ADH/LIN (Table). The mean QLACS score was 1.9/4 in both groups, indicating low to moderate fear of developing iBC, with minimal impact on quality of life. The mean knowledge of the natural history of breast lesions was 77% of patients in the DCIS group and 67% in the ADH/LIN group. In the DCIS group, 84% of patients received treatment aligned with their personal goals (concordance score, BCS-DQI-S1). Mean Decision Process Scores (BCS-DQI-S3) were 77% and 63%, respectively, indicating a high level of shared decision-making. According to the CPS, 61% of patients reported a collaborative role in decision-making. On average, there was low or no regret about treatment decision (DRSs were 12/100 and 10/100). Among patients with DCIS, 81% reported no decisional conflict (SURE scale). Conclusions: These findings suggest that women with ADH/LIN and DCIS report high levels of knowledge, goal-concordant care, and involvement in shared decision-making, potentially suggesting that AM is acceptable and presents a future opportunity to de-intensify treatment for low-risk DCIS, pending results of clinical trials.
|
DCIS group (n=538) |
ADH/LIN group (n=365) |
|
| Age, mean (range) | 61 (34-88) | 58 (26-94) |
| QLACS score, mean/4 (SD) | 1.9/4 (1.1) | 1.9/4 (1.1) |
| Knowledge of natural history, mean (SD) | 77% (16%) | 67% (21%) |
| Concordance score (BCS-DQI, Section 1), % (95% CI) | 84% (81-87%) | NA |
| Decision Process Score (BCS-DQI, Section 3), mean (SD) | 77% (21%) | 63% (30%) |
| Control Preferences Scale (CPS) – collaborative role, % | 58% | 66% |
| Decision Regret Scale (DRS), mean/100 (SD) | 12/100 (20) | 10/100 (17) |
| SURE Score, mean (SD) | 3.7/4 (0.7) | NA |
Presentation numberPS1-02-02
Long-term worry after early-stage breast cancer diagnosis: longitudinal study of a population-based cohort
Reshma Jagsi, Emory University School of Medicine, Atlanta, GA
R. Jagsi1, A. Furgal2, S. Pottow3, S. Hawley4, L. Wallner4; 1Radiation Oncology, Emory University School of Medicine, Atlanta, GA, 2Biostatistics, University of Michigan, Ann Arbor, MI, 3Psychology, University of Michigan, Ann Arbor, MI, 4Medicine, University of Michigan, Ann Arbor, MI.
Background/Purpose: Worry influences treatment decision-making, surveillance behaviors, and quality of life of people with cancer. The long-term experience of worry among patients with early-stage breast cancer requires better understanding and validated instruments for efficient measurement. Methods: ICanCare enrolled 2502 women with early-stage breast cancer, diagnosed in 2013-15 and reported to Georgia and Los Angeles SEER registries, who responded to surveys in the year of diagnosis (T1). After excluding those deceased, too ill, or otherwise unable to participate, 2361 were sent follow-up surveys in 2021-22 (T2), of whom 1412 responded. We asked at both times, “During the past month, how often has worrying about your cancer coming back”: 1) “made you feel upset”; 2) “made it difficult for you to carry out your usual daily activities at home or at work”; 3) “made you feel distant from family and friends.” Response options were “almost never,” “rarely,” “sometimes,” “often,” or “almost always.” A scaled measure across the 3 items was developed using factor analysis and evaluated for internal consistency and validity in bivariate analyses with tumor and patient characteristics, using Wilcoxon/Kruskal-Wallis tests for all variables (categorical) except age (continuous), which was compared using Spearman’s correlation. The 3-item scale was also compared using Spearman’s correlation to a single item asking, “In the past month, how often have you worried about your breast cancer coming back?” Results: Table 1 shows responses among the 1165 without recurrence or second cancer at T2. Over 1 in 5 respondents (27.6% at T1 and 21.1% at T2) endorsed that worry about their cancer coming back made them feel upset at least sometimes. Factor analysis retained a one-factor solution. Cronbach’s alpha for the scale was 0.87 at T1 & 0.82 at T2. Scaled scores had mean (SD) of 1.64 (.83) at T1 and 1.43 (.67) at T2; median (IQR) 1.33 (1.00-2.00) at T1 and 1.00 (1.00-1.67) at T2. Scaled scores were higher among patients with higher stage (p=.009 at T1 and .007 at T2) and those who were younger (p<0.001 at T1 and T2), Asian and Latina (p<.0001 at T1 and T2), with lower education (p=.003 at T1, p<0.0001 at T2), divorced/separated or never married (p=.0007 at T2 only), and with lowest income (p=.002 at T2 only). At T1, the mean score on the single item was 2.16 (SD 1.13), with 37.8% worrying sometimes or more; at T2, the mean was 2.1 (SD 1.08), and 38.3% worried sometimes or more. The 3-item scale correlated with the single item (correlation coefficients T1: .71, p<.001; T2: .67, p<.001). Conclusions: Three-item and single-item measures of worry appear valid for assessing patients with early-stage breast cancer, suggesting that >1 in 5 patients has worries that extend well beyond 5 years after diagnosis. Recognizing patient and tumor characteristics associated with long-term worry may help target resources and supportive interventions.
| Made upset | Made upset | Impaired daily activitie | Impaired daily activities | Affected relationships | Affected relationships | |
| T1 | T2 | T1 | T2 | T1 | T2 | |
| Almost never | 561 (49.4%) | 670 (59.0%) | 758 (66.8%) | 892 (79.4%) | 767 (67.8%) | 905 (80.2% |
| Rarely | 260 (22.9%) | 226 (19.9%) | 222 (19.6%) | 151 (13.4%) | 196 (17.3%) | 132 (11.7%) |
| Sometimes | 230 (20.3%) | 185 (16.3%) | 110 (9.7%) | 63 (5.6%) | 122 (10.8%) | 74 (6.6%) |
| Often | 64 (5.6%) | 40 (3.5%) | 28 (2.5%) | 11 (1.0%) | 29 (2.6%) | 13 (1.2%) |
| Almost always | 20 (1.8%) | 14 (1.2%) | 16 (1.4%) | 7 (0.6%) | 17 (1.5%) | 4 (0.4%) |
| Missing | 30 | 30 | 31 | 41 | 34 | 37 |
Presentation numberPS1-02-03
Talking to Employers and Medical Staff about Work: An Intervention to Increase Access to Work Accommodations
Yashasvini Sampathkumar, Memorial Sloan Kettering Cancer Center, New York, NY
Y. Sampathkumar1, E. Kasuga2, S. Patil3, J. Suarez2, C. Lopez2, Y. De Los Santos2, B. Narang2, P. Enriquez4, D. F. Makower5, W. G. Lichtenthal6, P. A. Parker7, T. Bao8, S. Rosenberg9, F. M. Gany2, V. S. Blinder10; 1Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 2Immigrant Health & Cancer Disparities Service, Memorial Sloan Kettering Cancer Center, New York, NY, 3Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, 4School of Medicine, City University of New York, New York, NY, 5Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, 6Public Health Sciences, Sylvester Comprehensive Cancer Center, Miami, FL, 7Counseling Center, Memorial Sloan Kettering Cancer Center, New York, NY, 8Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 9Epidemiology, Weill Cornell Medicine, New York, NY, 10Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: Minimizing employment disruption during cancer treatment improves long-term job retention and may reduce financial hardship and its consequences, including treatment nonadherence, disease progression, and death. Patients who report receiving work accommodations (e.g., a flexible schedule) are more likely to remain employed during and after treatment. Our prior qualitative work showed that many cancer survivors were unaware of available accommodations and uncomfortable initiating requests. Symptom control during (neo)adjuvant therapy was also a barrier to continuing to work. To address these needs, we developed Talking to Employers and Medical Staff about Work (TEAMWork), a patient-informed intervention that provides education on work accommodations based on vocational rehabilitation principles, employer negotiation strategies, symptom self-management, and guidance on communicating with clinicians to enhance support (e.g., obtain work documentation). TEAMWork equips patients with strategies to effectively engage with employers and clinicians, thereby enhancing access to work accommodations and better symptom management during treatment. Methods: TEAMWork is available in English and Spanish, as a booklet and a mobile application (app). We have an ongoing randomized controlled trial comparing the TEAMWork app to the booklet. Here we present data regarding self-reported accommodations requested by study participants while enrolled on the trial. Eligible participants were women aged 18-65 years with stage I-III breast cancer who spoke English and/or Spanish, were employed pre-diagnosis, and planning to undergo systemic adjuvant chemotherapy. Patients were recruited between October 2021 – March 2025 from multiple New York City sites. Demographic and clinical data was collected through self-report and medical chart abstraction. Self-reported data on work accommodations was obtained at baseline and the midpoint of chemotherapy (mid-treatment). Results: Of the 420 participants at baseline, 89% spoke English and 11% spoke Spanish; 52.4% identified as White, 18.1% Black, and 18.1% Hispanic/Latino. The mean age was 48 years (standard deviation 9.2); 65.2% were college educated. All were employed, but only 78.5% were working at baseline. The baseline survey was completed prior to chemotherapy initiation for 47.9% of participants. Three hundred participants completed a mid-treatment survey. From baseline to mid-treatment, we observed an increased percentage of participants in both arms who reported new requests for the following accommodations: work from home (booklet: 30.1% to 41.3%; app: 28.4% to 34.4%), reduced hours (booklet: 33% to 53.8%; app: 28.4% to 45.9%), changes responsibilities or tasks (booklet: 7.7% to 29.4%; app: 10.4% to 22.9%), modified work environment (booklet: 6.2% to 18.2%; app: 8.5% to 12.1%), and additional breaks (booklet: 11% to 29.4%; app:11.8% to 22.9%). We observed a decline in new requests from baseline to mid-treatment for time off (overall: 50.7% to 45.3%) and time off to go to doctor’s appointments (overall: 76% to 60%). Conclusions: The TEAMWork intervention had promising early results in empowering patients with breast cancer to request work accommodations during chemotherapy. Declines in new requests for time off may reflect early awareness or consistent need for this type of accommodation from diagnosis through treatment, in contrast to growing needs for other accommodations throughout the treatment course. Both intervention formats, app and booklet, supported increased patient engagement in requesting accommodations, such as reduced hours and flexible work environments, which can help job retention during treatment. Future research will focus on identifying which intervention formats are most effective for different types of patients.
Presentation numberPS1-02-04
Loneliness among women with metastatic breast cancer and its relation to well-being: the moderating role of religiosity and spirituality
Michal Braun, Hadassah Medical Center, Jerusalem, Israel
M. Braun1, G. Goldzweig2, I. Hasson-Ohayon3, N. Gafni2, S. Paluch Shimon1; 1Oncology of Breast, Hadassah Medical Center, Jerusalem, ISRAEL, 2Behavioral Sciences, The Academic College of Tel Aviv Yaffo, Yaffo, ISRAEL, 3Psychology, Bar Ilan University, Ramat Gan, ISRAEL.
Objective: This study examined how Jewish and Arab women in Israel, with varying levels of religiosity, cope with metastatic breast cancer, specifically investigating the relationships between loneliness, depression, anxiety, and quality of life, and exploring the moderating roles of religiosity and spirituality. Methods: A mixed-method approach was employed, including a quantitative longitudinal component and a qualitative component consisting of in-depth interviews with a subset of the participants. The quantitative analysis involved 100 women who completed validated scales assessing loneliness (UCLA), religiosity and spirituality (SBI, FACIT-Sp), depression (PROMIS-D-SF), anxiety (PROMIS-A-SF), and quality of life (FACT-B). Results: Loneliness correlated positively with depression (r = 0.63, p < .01), anxiety (r = 0.50, p < .01), and negatively with quality of life (r = -0.55, p < .01). Cluster analysis identified four groups: (1) low loneliness-low religiosity, (2) high loneliness-low religiosity, (3) low loneliness-high religiosity, and (4) high loneliness-high religiosity, demonstrating significant differences in depression (F(3) = 18.96, p < .001), anxiety (F(3) = 10.61, p < .001), and quality of life (F(3) = 10.81, p < .001). Post-hoc analyses indicated lower depression levels in the highly lonely but highly religious group (cluster 4) compared to the highly lonely and less religious group (cluster 2). Qualitative interviews with Arab and ultra-Orthodox Jewish women highlighted belief in God as a critical resource alleviating distress and loneliness. Conclusions: High loneliness increases depression and anxiety and reduces quality of life among women with metastatic breast cancer. Religiosity and spirituality mitigate depression specifically under conditions of high loneliness. These quantitative findings, enriched by qualitative insights, underscore the importance of integrating religious and spiritual resources into psychosocial oncology care, particularly in culturally and religiously diverse populations.
Presentation numberPS1-02-05
Quality of life in women after breast biopsy with a benign result
Hans-Christian Kolberg, Marienhospital Bottrop, Bottrop, Germany
H. Kolberg1, E. Düster1, L. Akpolat-Basci1, H. Hoffmann2, I. Swienty1, K. Freienstein1, C. Kolberg-Liedtke3; 1OB/GYN, Marienhospital Bottrop, Bottrop, GERMANY, 2OB/GYN, Universitätsklinikum Essen, Essen, GERMANY, 3Applied Health Sciences, Hochschule Bochum, Bochum, GERMANY.
Introduction: Standard of care for the diagnosis of malign and benign breast tumors is a minimally invasive biopsy in cases of imaging classified BIRADS IVa and higher. This biopsy can be performed as a core cut biopsy or vacuum assisted biopsy and may be guided by sonography, mammography or MRI. The majority of biopsies are core cut biopsies guided by sonography. Whereas in cases of malign results patients in certified breast cancer centers are counseled by psychooncologists, patients with benign results are often informed in phone calls and without focus on their fears and psychological well-being. In this analysis we present data regarding the impact of breast biopsies with a benign result on patients‘ quality of life. Methods: Patients who had a core cut biopsy guided by ultrasound with a benign histological result between January 2021 and December 2023 were identified in our database. All patients received a written invitation to take part in the analysis, a questionnaire and an informed consent form. Patients who returned the questionnaire and the informed consent form were included in this analysis. The instrument we used for measuring quality of life is the short form health survey SF-36v2® (QualityMetric Inc., Johnston, USA) that contains 36 items used to measure eight domains of health-related quality of life. These parameters are compared with normative data from the QualityMetric 2009 General Population Sample. Results: 241 patients were identified who had undergone a core cut biopsy with a benign result between 2021 and 2023, 70 patients agreed to participate, submitted the questionnaire and were included in this analysis. In all eight domains of health related quality of life the results of the study population were in the category ,,same or better“ compared with the general population: physical functioning 84%, role limitations due to physical health 76%, bodily pain 73%, general health perceptions 74%, vitality 71%, social functioning 67%, role limitations due to emotional problems 67% and mental health 61%. Conclusion: We conducted a hypothesis generating analysis regarding the impact of a breast biopsy with a benign result on quality of life. We observed that all eight investigated domains of health-related quality of life were comparable or better than in the general population. Although this result may need validation in a prospective analysis it seems very improbable that the breast biopsy itself has a negative impact on quality of life. This result could help to increase the acceptance and reduce fears regarding the conduct of breast biopsy if indicated.
Presentation numberPS1-02-06
The Impact of Treatment-Induced Early Menopause on Body Image and Sexual Functioning Among Young Breast Cancer Survivors
Michal Braun, Hadassah Medical Center, Nes Harim, Israel
M. Braun1, G. Goldzweig2, O. Saar Sheffi1, S. Klein2, S. Paluch Shimon1; 1Oncology of Breast, Hadassah Medical Center, Nes Harim, ISRAEL, 2Behavioral Sciences, The Academic College of Tel Aviv Yaffo, Yaffo, ISRAEL.
Purpose: Young breast cancer survivors often face abrupt and premature menopause due to oncological treatments, resulting in sudden physical and psychological changes that profoundly affect their quality of life. This study aimed to examine the impact of treatment-induced menopausal symptoms on two critical domains: body image and sexual functioning, highlighting the mediating roles of menopausal symptoms and reproductive concerns, and exploring age as a potential moderator. Methods: Two complementary cross-sectional studies were conducted. In the first, 121 hormone receptor-positive breast cancer survivors and 114 healthy women completed validated measures: the Body Image Scale (BIS), Menopausal Rating Scale (MRS), and Reproductive Concerns Scale (RCS). In the second, 97 young breast cancer survivors and 75 healthy women completed the MRS and Fallowfield’s Sexual Activity Questionnaire (FSAQ). All cancer survivors also provided detailed medical history. Moderated mediation and structural equation modeling were used to examine direct and indirect pathways linking cancer survivorship, menopausal symptoms, body image, reproductive concerns, and sexual activity frequency. Results: Survivors reported significantly higher levels of negative body image, reproductive concerns, menopausal symptoms, reduced sexual pleasure, and lower frequency of sexual activity compared to controls. Menopausal symptoms and reproductive concerns mediated the relationship between survivorship and negative body image, with age moderating these pathways — indirect effects were stronger among younger women. In parallel, structural equation modeling indicated that the link between survivorship and reduced sexual frequency was explained by menopausal symptoms and diminished sexual pleasure. Somatic and urogenital menopausal symptoms emerged as key contributors to both outcomes. Conclusions: Treatment-induced early menopause plays a pivotal role in shaping young breast cancer survivors’ body image and sexual functioning. Findings underscore the importance of age-sensitive, tailored supportive care addressing both the physical side effects and the psychological and intimate dimensions of survivorship. Targeted interventions should aim to mitigate menopausal symptoms, support reproductive concerns, and enhance sexual well-being, ultimately promoting higher quality of life among this vulnerable group.
Presentation numberPS1-02-07
Follow-up in Early and Locally Advanced Breast Cancer Patients: An EORTC QLG-BCG-ROG Protocol-Results of EORTC QLQ-BR42 and EORTC QLQ-SHQ22
Vesna Bjelic-Radisic, Helios University Hospital, Wuppertal, Germany
V. Bjelic-Radisic1, K. Pogoda2, D. Cameron3, G. Velikova4, M. Beauvois5, C. Coens5, L. Lim5, T. Verbiest5, G. Sarlet5, N. Nevries1, S. Serpentini6, H. Abdel-Razeq7, J. Kaźmierska8, T. Kuhnt9, E. Fernández Lizarbe10, E. Cretella11, I. Meattini12, F. van Duijnhoven13, I. Rubio14, R. Popescu15, S. Hartup16, H. Roelstraete17, F. Cardoso18; 1Senology, Helios University Hospital, Wuppertal, GERMANY, 2Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, POLAND, 3Cancer Research Centre, University of Edinburgh, Edinburgh, UNITED KINGDOM, 4Institute of Medical Research, Leeds Cancer Centre, St. James’s, University of Leeds, Leeds (West Yorkshire), UNITED KINGDOM, 5Quality of Life Department, EORTC Headquarters, Brussels, BELGIUM, 6Psychoncology Unit/Breast Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, ITALY, 7King Hussein Cancer Centre, School of Medicine, The University of Jordan, Amman, JORDAN, 8Electroradiology Department, Greater Poland Cancer Centre, Poznan, Poland, University of Medical Sciences, Poznan, Poznan, POLAND, 9Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig, Leipzig, GERMANY, 10Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, SPAIN, 11Oncology, Ospedale Generale di Bolzano, Bolzano, ITALY, 12Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, Firenze, ITALY, 13Division of Surgical Oncology, The Netherlands Cancer Institute (NKI), Amsterdam, Amsterdam, NETHERLANDS, 14Breast Surgical Oncology, Clinica Universidad de Navarra, Madrid, SPAIN, 15Medical Oncology, Tumor Zentrum Aargau and Hirslanden Klinik, Aarau, SWITZERLAND, 16St. James University Hospitals, Leeds, UK, St. James University Hospitals, Leeds, UK, Leeds, UNITED KINGDOM, 17Medical Oncology, Onze-Lieve-Vrouwziekenhuis (OLVZ), Aalst, BELGIUM, 18Advanced Breast Cancer (ABC) Global Alliance, Advanced Breast Cancer (ABC) Global Alliance, Lisbon, PORTUGAL.
Background: Breast cancer (BC) is the most common cancer among women globally. However, evidence-based follow-up strategies that address long-term risks, patient needs, and quality of life (QoL) remain limited. This study investigates the prevalence and nature of physical, psychological, and social challenges faced by women after treatment for early (EBC) or locally advanced breast cancer (LABC). Initial findings presented at SABCS 2024 showed that EBC and LABC survivors reported better overall health/QoL than the general population, but worse outcomes in insomnia, cognitive, and social functioning (as assessed by EORTC QLQ-C30)—particularly among LABC patients. This abstract focuses on results from the breast cancer-specific (EORTC QLQ-BR42) and sexual health (EORTC QLQ-SHQ-22) modules, exploring patterns by demographic and clinical variables. Methods:EORTC 1617-QLG-BCG-ROG is an international, cross-sectional, non-interventional study involving disease-free patients 1–3 years post-primary treatment (excluding endocrine therapy) for EBC or LABC. Data on institutional, demographic and clinical characteristics were collected. Patients completed the EORTC QLQ-C30, QLQ-BR42, QLQ-SHQ-22, QLQ-OUT-PATSAT-7, a follow-up strategy question, and the Distress Thermometer. A sample size of 830 was required to detect a 10-point difference between cohorts with 90% power. Stratification by age, Nottingham Prognostic Index (NPI), and treatment ensured balanced accrual. Multivariable prognostic models were constructed for each PRO scale, adjusted for propensity with a relevant relationship defined by 5% significance level. Results:From November 2020 to September 2022, 833 patients ( 82 % EBC; 17% LABC) were enrolled at 25 centres in 10 countries. A total of 805 (97%) eligible patients either fully or partially completed their questionnaires. Regarding clinical and sociodemographic characteristics, better WHO status correlates with better hand/foot symptoms/polyneuropathy, greater sexual satisfaction and perceived importance of sexual activity. Older patients reported fewer endocrine, arm, and breast symptoms, better body image and future perspective compared to younger women, as well as fewer problems with decreased libido, fatigue, treatment-related sexual issues, partnership, body image, and pain. Older patients also reported lower sexual satisfaction and worse communication with professionals, despite attaching less importance on sexual activity. Younger women, women with higher education level and those living with partner reported better sexual functioning and greater importance of sexual activity. Women with longer follow up reported more systemic, endocrine, arm, hand/foot symptoms, more weight gain and more problems with incontinence, treatment-related sexual issues, and higher sexual satisfaction. High volume institutions were associated with fewer breast symptoms but worse breast satisfaction. Private setting was linked with more endocrine and arm symptoms. Patients in follow-up in breast units reported worse skeletal symptoms compared to institutions without a breast unit. Higher nodal status correlated with poorer body image, more arm symptoms, greater fatigue, and treatment-related sexual issues. Conclusions:Despite being disease-free, women with EBC and LABC continue to experience significant QoL and sexual health challenges. Younger patients are particularly vulnerable to post-treatment symptoms. Institutional factors, such as follow-up duration and center volume, influence outcomes. Sociodemographic factors like education and partnership status positively impact sexual health. These findings are essential for developing tailored, evidence-based follow-up strategies for breast cancer survivors.
Presentation numberPS1-02-08
Comparative Analysis of the Effectiveness of Scalp Cooling with PAXMAN Between Dose-Dense AC→PTX and Trastuzumab+Pertuzumab+Docetaxel Regimens in Japanese Breast Cancer Patients
Shiho Nagasawa, Toyama University, Toyama, Japan
S. Nagasawa, K. Matsui, M. Furukawa, E. Kanaya, M. Araki, S. Sekine, T. Fujii; Surgery and Science,Faculty of Medicine,Academic Assembly, Toyama University, Toyama, JAPAN.
Background:Chemotherapy-induced alopecia (CIA) remains one of the most distressing side effects of breast cancer treatment, especially for younger patients. Scalp cooling systems such as the PAXMAN Orbis have shown promising results in reducing the incidence and severity of CIA, particularly in regimens involving taxanes and anthracyclines. However, most existing studies focus on single regimens, and comparative evaluations across chemotherapy backbones remain limited. This study aimed to compare the efficacy of PAXMAN in two commonly used neoadjuvant/adjuvant regimens with differing first-line agents: a dose-dense anthracycline-first regimen (ddAC→ddPTX) and a taxane-based HER2-targeted regimen (trastuzumab + pertuzumab + docetaxel: HP+DTX). Methods:We retrospectively analyzed 62 patients with early-stage breast cancer who underwent scalp cooling using the PAXMAN system during chemotherapy at our institution between January 2021 and December 2024. Of these, 37 patients received the ddAC→ddPTX regimen, and 25 received the HP+DTX regimen. Scalp cooling was administered during each chemotherapy session. Hair loss was assessed at the end of each regimen using the CTCAE v5.0 grading system. Grade 1 or lower alopecia was defined as successful hair preservation. Results:The overall cohort had a mean age of 49 years. In the ddAC→ddPTX group, 59.5% of patients maintained Grade 1 or lower alopecia at the end of chemotherapy. In the HP+DTX group, 64% achieved Grade 1 or lower alopecia. No serious adverse events related to scalp cooling were reported. The treatment adherence and tolerance of the PAXMAN system were high in both groups. Discussion:Our study is one of the first to directly compare the effectiveness of scalp cooling between anthracycline-first and taxane-first chemotherapy regimens. The comparable success rates of hair preservation between the ddAC→ddPTX and HP+DTX groups suggest that PAXMAN is effective across different chemotherapy backbones, including those with known high alopecic potential. These findings provide reassurance that scalp cooling can be equally beneficial regardless of the sequencing of anthracyclines or taxanes. This is especially relevant in clinical settings where regimen choice is dictated by tumor subtype, as in HER2-positive disease. To our knowledge, this is the first comparative report from Japan on this topic. The results offer valuable evidence supporting the routine use of scalp cooling in Asian populations, who may differ in hair structure and density from Western cohorts studied previously. Further prospective, larger-scale studies are warranted to validate these findings and optimize personalized scalp cooling protocols.
Presentation numberPS1-02-09
Association between financial toxicity and mental health among metastatic breast cancer survivors
Haoting Shi, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
Z. Xu1, Y. Lu2, Y. Wu2, H. Shi1, C. Shangguan3, K. Shen1, Q. Qu3, N. Zhang1; 1Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA, 2Department of Nursing, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA, 3Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA.
Background: Metastatic breast cancer (MBC) survivors require long-term treatment, resulting substantial costs and financial toxicity. Thought the financial burden was observed to be associated with cancer survivors’ clinical outcome, limited evidence focused on the toxicity on mental health. Here, we aimed to determine the association between financial toxicity and mental health and its mediators among MBC survivors.Methods: Female MBC patients diagnosed in our institution during May 2023 to October 2023 were included. Financial toxicity was assessed using Comprehensive score for financial toxicity based on the patient-reported outcome measures (COST-PROM) instrument. Mental health status was profiled through Connor-Davidson Resilience Scale (CD-RISC). To further explore the modifiable psychological symptoms, the Cancer Loneliness Scale (CLS), Cancer-related Negative Social Expectations Scale (CRNSES), M. D. Anderson Symptom Inventory (MDASI) and Perceived Social Support Scale (PSSS) were adopted to characterize loneliness, social expectations, oncology symptom awareness, and social support, respectively. The association between financial toxicity and mental health status was determined using conditional linear regression, adjusted for demographic factors, disease characteristics, treatment, and socioeconomic status. Mediation analysis with structural equation modeling (SEM) was performed to explore the mediator of the association. Further, we conducted the network analysis to profile the correlation among identified mediators, including a binary variable that encoded whether survivor was of financial toxicity. A two-tailed P < 0.05 was considered as statistically significant.Results: A total of 226 MBC survivors were included, of whom the median age were 55 (interquartile range 45 to 64 years) years, and 173 (76.5%) were with financial toxicity. Financial toxicity was significantly associated with mental health after adjusted for demographic factors, disease characteristics, treatment, and socioeconomic status (coefficient, 0.197, 95% CI, 0.024 to 0.370). CLS, CRNSES, MDASI-part 2, and PSSS were identified as the mediators (indirect effect [95% CI], CLS 0.087 [0.04 to 0.134], CRNSES 0.045 [0.009 to 0.08], MDASI-part2 0.071 [0.027 to 0.115], and PSSS 0.043 [0.008 to 0.078],all P < 0.05), which corresponding to 72.7%, 37.1%, 59.2%, and 35.9% of the relative effect, respectively. The network analysis showed that “relations (MDASI-2_4)”, “social discomfort (CRNSES_1)”, and “felt empty (CLS_6)” were the mental health nodes with the top 3 largest strength (mean [SD] of bootstrapped standardized index value, 1.79 [0.69] for “relations”, 1.71 [0.85] for “social discomfort”, and 1.57 [0.99] for “felt empty”, respectively), while the “felt empty (CLS_6)” and “perceived avoidance (CRNSES_2)” were the node with largest betweenness and closeness (mean [SD] of bootstrapped standardized index value, 2.12 [3.4] for “felt empty and 1.36 [2.40] for “perceived avoidance”), respectively.Conclusion: Financial toxicity was significantly associated with worse mental health. Targeting certain dimensions regarding loneliness, social expectations, oncology symptom awareness, and social support might could reverse the toxicity.
Presentation numberPS1-02-10
Knowledge and adherence to exercise recommendations among oncologic patients.
Renata C Cangussu, Instituto D’Or de Pesquisa e Ensino, Salvador, Brazil
R. C. Cangussu1, L. Andrade1, L. Testa2, C. Lima3, A. S. Ferreira4, C. M. Moniz2, S. R. Chagas5, P. M. Hoff2, M. Estevez-Diz2, R. Colombo Bonadio2; 1Oncology, Instituto D’Or de Pesquisa e Ensino, Salvador, BRAZIL, 2Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 3Oncology, Instituto D’Or de Pesquisa e Ensino, Recife, BRAZIL, 4Oncology, Instituto D’Or de Pesquisa e Ensino, Brasília, BRAZIL, 5Oncology, Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, BRAZIL.
Knowledge and Adherence to Exercise Recommendations Among Oncologic PatientsBackground: Physical activity positively influences various aspects of cancer treatment. Among patients (pts) undergoing active therapy, it improves physical capacity, treatment tolerability, and reduces side effects. For cancer survivors, it lowers the risk of recurrence of tumors such as breast and colorectal. Despite its importance, the extent to which pts are informed about exercise recommendations remains unclear.Methods: We conducted a self-administered survey to evaluate exercise habits and knowledge among cancer pts treated between 2022 and 2024. Participants were recruited from a private oncology network (with units across Brazil) and two public cancer centers. Pts, irrespective of tumor type, disease stage, or treatment phase, completed the survey via RedCap.Results: Of the 394 pts who answered the survey, 76.4% were female. Median age was 56 years (range 23 – 92). The cohort included 52.8% self-identified white pts and 44.8% black/mixed race pts; 63.9% were treated in the private health system and 36.1% in the public. The most common primary tumors were breast (47.7%) and gastrointestinal (16.2%). While 85.9% reported receiving any exercise recommendations from physicians, only 62.5% described themselves as physically active. In addition, 30.7% stated they lacked knowledge about the type or duration of recommended exercises. Exercise guidance and adherence were lower among patients treated in public health centers, black/mixed race pts, and those with non-breast cancer (Table). Notably, 94.2% of pts without sufficient information expressed a desire to learn more about exercise.Conclusion: Although a majority of cancer pts receive general recommendations on physical activity, adherence and understanding of specific exercise guidelines remain suboptimal. Variability in exercise awareness based on health system type and race highlights disparities that must be addressed. Promoting physical activity awareness and accessibility, particularly in resource-limited settings, is crucial. Exercise should not remain a privilege for a few but rather a fundamental aspect of care accessible to all pts, regardless of their circumstances. Table
| Received recommendation | Adherence to physical activity | Knowledge of recommended type and duration | |
| Health system | – | – | – |
| Private | 96.3% | 69.4% | 77.8% |
| Public | 61.3% | 33.3% | 44.6% |
| P-value | <0.001 | <0.001 | <0.001 |
| Race | – | – | – |
| White | 92.7% | 67.9% | 76.5% |
| Black | 77.3% | 54.6% | 60.9% |
| P-value | <0.001 | 0.008 | 0.001 |
| Primary tumor | – | – | – |
| Breast | 92.5% | 73.3% | 76.9% |
| Other | 79.9% | 52.7% | 62.2% |
| P-value | <0.001 | <0.001 | 0.002 |
Presentation numberPS1-02-11
The Unbridged Gap in Sexual Health Counseling: A Global Oncology Trainee Perspective
Malak Alharbi, Roswell Park Comprehensive Cancer Center, Buffalo, NY
M. Alharbi1, K. Ahuja1, Z. Shah1, J. Mussel1, S. Gandhi2, A. AbuSanad3, V. Gupta1, L. S. Agrawal4, A. M. Roy5; 1Department of Medicine- Division of Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Department of Medicine- Division of Oncology, Emory Winship Cancer Institute, Atlanta, NY, 3Department of Internal Medicine- Devision of Oncology, King Abdulaziz University, Jeddah, SAUDI ARABIA, 4Department of Medicine- Division of Oncology, Norton Cancer Institute, Louisville, KY, 5Department of Medicine- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH.
Background Clinical guidelines recommend addressing sexual health (SH) as part of routine oncology practice, yet only 20-29% of patients with breast cancer (BC) report receiving SH counseling. SH remains mostly excluded from formal medical training. This study explored the current knowledge, attitudes, and barriers to SH counseling among oncology trainees working in breast oncology settings globally. Methods A cross-sectional study was conducted among medical oncology residents/fellows, breast and gynecology surgical fellows, and radiation oncology residents. Anonymous self-administered surveys were distributed via social media platforms, including X, LinkedIn, WhatsApp, and email. Surveys collected demographics, self-reported SH knowledge, practices, and barriers to SH counseling. Data were analyzed using descriptive statistics. Results Among 46 oncology trainees, 52% were male, and 48% female, with the majority aged 31-35 years (48%). Most were in advanced training levels (PGY4-6), and 91% were in medical oncology. Respondents were trained in the US (70%), Middle East (17%), Europe (9%), and Asia (4%). A majority (89%) reported limited knowledge of SH dysfunction during cancer treatment. Table 1 summarizes self-reported confidence in managing SH concerns. SH was infrequently assessed; 48% reported they rarely addressed it, and only 26% indicated they usually/always did. 65% indicated that SH counseling is their responsibility. Biases were common:70% were more likely to discuss SH with younger patients (<50), and 52% with those with early-stage BC. When asked to select all appropriate times to address SH, 72% chose active/post-treatment, 43% at diagnosis, 41% during survivorship, and only 15% throughout care. 46% of trainees were moderately concerned about prescribing vaginal estrogen to patients with BC on endocrine therapy; 33% had prescribed vaginal estrogen, and 37% had prescribed erectile dysfunction medication. Overall, 94% reported insufficient SH training, with around 60% receiving no training to counsel females, males, or transgender patients with BC. 81% supported individualizing SH discussions, 68% felt patients value them, 50% feared offending patients by initiating them. Awareness of institutional SH resources was low with many unsure of availability of pelvic floor therapy 50%, sex therapy 59%, and behavioral therapy 50%. Commonly reported barriers to SH counseling included lack of training 85% and time 65%, followed by cultural sensitivity, provider discomfort, and perceived lack of effective treatment. Importantly, 83% supported more SH education, with seminars 83%, shadowing SH experts 50%, online courses 50%, and simulation-based training 26%. Conclusion Oncology trainees worldwide treating BC report inadequate training in SH, low confidence, and limited awareness of available resources. Formal education and social media engagement may help bridge the counseling gap.
|
Sexual health concern |
Not at all confident |
Not very confident | Neutral | Somewhat confident | Very confident |
|
Low libido |
26% n=12 |
35% n=16 |
24% n=11 |
11% n=5 |
4% n=2 |
|
Dyspareunia |
17% N=8 |
48% n=22 |
20% n=9 |
13% n=6 |
2% n=1 |
| Erectile dysfunction |
15% n=7 |
22% n=10 |
39% n=18 |
22% n=10 |
2% n=1 |
| Genitourinary syndrome of menopause |
22% n=10 |
22% n=10 |
35% n=16 |
18% n=8 |
4% n=2 |
| Body image concerns |
9% n=4 |
41% n=19 |
28% n=13 |
15% n=7 |
7% n=3 |
| Impact on intimate relationships |
15% n=7 |
39% n=18 |
26% n=12 |
13% n=6 |
7% n=3 |
| Psychological contributors such as depression, and anxiety |
13% n=6 |
37% n=17 |
17% n=8 |
24% n=11 |
9% n=4 |
Presentation numberPS1-02-12
Comparison of health-related quality of life outcomes in breast cancer patients undergoing breast-conserving surgery, medically necessary mastectomy, and patient preference mastectomy: a prospective study
Claire Liu, University of British Columbia, Vancouver, BC, Canada
C. Liu1, M. Deban1, J. Newman-Bremang1, R. Warburton1, C. Dingee1, J. Pao1, A. Bazzarelli1, J. Sutherland2, E. McKevitt1; 1Department of Surgery, University of British Columbia, Vancouver, BC, CANADA, 2Centre for Health Services and Policy Research, University of British Columbia, Vancouver, BC, CANADA.
Background: Guidelines support both breast-conserving surgery (BCS) with radiation and mastectomy for treatment of early breast cancer, but emerging evidence favoring BCS challenges the practice of universally offering both options to patients. There is currently no comparative study assessing health-related quality of life outcomes (HRQoL) across BCS, medically necessary (Mast-MN), and patient preference mastectomy (Mast-PP). This study compared the preoperative and six-month postoperative change in HRQoL in aspects of mental, physical health, and satisfaction between these surgical approaches. Methods: This prospective cohort study enrolled breast cancer surgery patients between 2021 – 2024 at our provincial referral center. Participants completed surveys measuring mental and physical health preoperatively and six-months postoperatively. The BREAST-Q questionnaire was used to assess breast cancer specific outcomes including breast satisfaction, psychosocial, sexual, and chest physical well-being. General patient-reported outcomes were categorized in depression, anxiety, pain, and perceived health. Results: A total of 494 patients meeting inclusion/exclusion criteria completed both pre- and six-month postoperative surveys. Of these, 343 underwent BCS, 104 Mast-MN, and 47 Mast-PP. On average, BCS was associated with a clinically significant increase in breast satisfaction (+4.3) and physical-chest well-being (+7.5). Mast-PP showed a decline in physical-chest well-being (−5.6), while Mast-MN showed reductions in breast satisfaction (−6.2), psychosocial (−5.4), and sexual well-being (−12.2). In multivariable regression, both Mast-PP (β = −7.91, 95% CI: −15.2 to −0.63, p = 0.033) and Mast-MN (β = −12.3, 95% CI: −17.4 to −7.13, p < 0.001) were associated with significantly lower postoperative breast satisfaction compared to BCS. There was no significant difference in depression, anxiety, pain, or perceived health scores within and across the surgical modalities. Conclusions: BCS was associated with significantly greater breast satisfaction postoperatively, and trends toward improved chest physical well-being compared to both medically necessary and patient preference mastectomy. When deciding surgical approach, patients should be counseled on these quality-of-life considerations when BCS is feasible.
Presentation numberPS1-02-13
Effects of different radiation techniques and fields on breast cancer related Lymphedema after axillary dissection and Simplified Lymphatic Microsurgical Preventive Healing Approach
Hadas Kadar Sfarad, University of Miami, Miami, FL
H. Kadar Sfarad1, M. Feret2, C. Padilla3, A. Balari4, J. Janin5, E. Avisar1; 1Breast Surgical oncology, University of Miami, Miami, FL, 2Radiation Oncology, University of Miami, Miami, FL, 3School of Medicine, Universidad del Norte, Colombia, COLOMBIA, 4Surgery, University of Miami, Miami, FL, 5School of Medicine, University of Miami, Miami, FL.
Background: Patients with breast cancer undergo axillary lymph node dissection under certain indications. One of the main side effects of axillary dissection is upper limb lymphedema that occurs in 15-40% 0f cases and can be debilitating. An objective measurement of lymphedema is Bioimpedance spectroscopy, that produces an LDEX Score and can detect subclinical lymphedema. There are several risk factors for developing lymphedema after axillary dissection, among them obesity, radiation therapy, wound complication and higher age. SLYMPHA is a surgical prevention of breast cancer related lymphedema, in which the breast surgeon is performing connection of lymphatic ducts from the upper limb to venous source. This method has been proven to reduce incidence of lymphedema to 10%. Our aim is to explore the effect of different radiation techniques and fields on the development of breast cancer related lymphedema in patients after axillary dissection and and SLYMPHA. Method: We established a retrospective database of all patients who underwent axillary dissection and SLYMPHA for breast cancer from 2014-2021, and collected demographic, clinical, surgical and radiation data. For each patient we observed L-dex measurement at pre-operative setting and 6,9,12,24 months post-operative setting as well as clinical examination for LE development. Binomial logistic regression was used to identify associations between clinical data, radiation methods and field, and LE incidence. Results: Among 219 patients, 136 had LDEX assessments, with 30 patients (13.6%) developing BCRL. Factors found to be related to elevated risk of LE included older age, higher BMI, recurrent cancer, reconstructive surgery and having N2/N3 nodal disease. Radiation therapy was found to be correlated with higher levels of lymphedema, especially with radiation techniques such as Three-Dimensional Conformal Radiation Therapy and Intensity Modulated Radiation Therapy/Volumetric Modulated Arc Therapy. Radiation to chest wall field and Comprehensive radiation excluding dissected axilla fields had a higher association with increased levels of LE. Conclusions: Patients undergoing axillary dissection and SLYMPHA with higher BMI, recurrent cancer, reconstructive surgery, N2/N3 disease, or adjuvant radiation treatment, as well as those receiving Intensity Modulated Radiation Therapy/Volumetric Modulated Arc Therapy or Three-Dimensional Conformal radiation techniques, have an increased risk of developing breast cancer related LE. De-escalation of radiation and better selection of radiation techniques and fields could contribute to a better lymphedema free survivorship.
Presentation numberPS1-02-14
Bridging the psychosocial gap in young breast cancer survivors: a peer-led, trauma-informed framework for survivorship navigation
Whitney O’connor, The Boobie Queen Company, Evans, GA
W. O’connor, L. Colbert, K. Armstrong; mental health, The Boobie Queen Company, Evans, GA.
Purpose Young breast cancer survivors experience high rates of emotional distress, identity disruption, and trauma-related symptoms. One in three cancer survivors show signs of PTSD. Among breastcancer survivors, 50% report depression and 60% report anxiety post-treatment. Yet few survivorship care models address mental health in a trauma-informed or developmentally relevant way. In our 2025 survey of 46 young survivors, 93% reported distress when reminded of cancer-related events, 84% experienced intrusive thoughts, 73% had sleep disturbances, 69%reported flashbacks, and over 80% expressed discomfort with physical changes. These findings underscore the urgent need for structured, peer-informed mental health support. In response, we developed targeted mental health tools and the Phases of Cancer Survivorship (PoCS)Framework: a three-phase model (Grief, Repair, Celebration) that conceptualizes psychological healing as a progressive, survivor-centered process. Methods The PoCS Framework was developed through an iterative, survivor-informed process. From2022 to 2025, 90 young breast cancer survivors participated in semi-structured interviews and surveys following 12 immersive peer-led retreats. Thematic analysis of participant narratives was conducted by a licensed mental health counselor and a multidisciplinary team to identify common psychosocial challenges. Key themes included emotional overwhelm, fear of recurrence, identity disruption, body image distress, professional delays, loneliness, and a desire to feel “normal” again.The framework includes three nonlinear, cyclical phases reflecting the evolving emotional experience of young survivors. The Grief Phase survivors process fear, loss, and trauma through reflection and trauma-informed dialogue. The Repair Phase survivors build emotional regulation and self-compassion through psychoeducation and skill development. The Celebration Phase survivors reclaims joy and identity through rituals, empowerment, and community connection.Retreat programming integrated the framework through group therapy, psychosocial education, art therapy, and original workbook tools. Emotional impact and utility were assessed through post-retreat surveys and open-ended feedback. Results Retreat participants reported 92% increased understanding of their mental health, 90%reported improved body image, 92% felt less alone and more connected to community, and 88% felt more confident and empowered. Qualitative feedback described the framework as “validating,” “clarifying,” and “exactly what I needed but didn’t know how to ask for.” Participants highlighted symbolic and expressive activities as emotionally transformative. Emergent themes included reduced shame, increased hope, empowerment, and clearer emotional navigation. Feedback supported the framework’s resonance, relevance, and potential for broader use in trauma-informed survivorship care. Conclusion Findings highlight the urgent need for structured, developmentally relevant psychosocial support throughout survivorship. The PoCS Framework addresses critical, unmet mental health needs among young breast cancer survivors. This model offers an integrable, emotionally resonant structure that bridges gaps in care and provides a shared language for oncology and mental health providers and patients. Its alignment with survivors’ lived experiences positions it for broader use in clinical navigation programs, community-based services, and direct patient support. The PoCS Framework has the potential to become a universal language for the survivor experience, guiding emotional recovery, and enhancing quality of care across settings. Further study is warranted to validate its impact and guide implementation at scale.
Presentation numberPS1-02-15
The influence of non-medical drivers of health on quality of life among Hispanic/Latina breast and cervical cancer survivors
Derek Rodriguez, UT Health San Antonio, San Antonio, TX
D. Rodriguez1, P. Chalela1, Y. Lew1, N. Rodriguez1, J. Ramos1, J. Cardenas1, C. Carmona1, E. Munoz1, B. Choi1, C. Wang1, Y. Manresa2, M. Hernandez Krause2, D. Perdomo2, A. Natori2, S. Cole3, F. Penedo2, A. Ramirez1; 1Institute for Health Promotion Research, UT Health San Antonio, San Antonio, TX, 2Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 3Medicine, Hematology/Oncology, University of California, Los Angeles, CA.
Background: Hispanic/Latina (H/L) women in medically underrepresented regions, such as South Texas, face substantial disparities in cancer survivorship, including reduced quality of life (QOL) and inadequate supportive care access. Despite growing recognition of the role of non-medical determinants of health (NMDoH) – including financial strain, transportation barriers, and limited social support – few studies have quantitatively assessed their combined impact on survivorship outcomes among H/L breast and cervical cancer survivors. Objective: This study examined how various NMDoH factors predict QOL in breast and cervical cancer survivors participating in Avanzando Caminos, a large NCI-funded cohort study investigating social, cultural, behavioral, and medical influences on Hispanic/Latino cancer survivorship. Methods: Baseline data were analyzed from 170 H/L women who had completed treatment for breast or cervical cancer within the last decade. NMDoH were assessed by cross-referencing Avanzando Caminos data with items from the Avanzando Salud Center NMDoH Screener—an instrument developed by the Center to investigate NMDoH in STX—measuring financial hardship, transportation needs, physical activity, healthcare access, and social support. QOL was evaluated using cancer site-specific Functional Assessment of Cancer Therapy (FACT) scales: FACT-B for breast cancer and FACT-Cx for cervical cancer survivors. Multivariable linear regressions estimated associations between NMDoH factors and QOL, controlling for sociodemographic and clinical covariates. Results: Financial hardship emerged as a significant predictor of lower QOL across both cancer types (breast: β = -3.41; cervical: β = -8.65; p < 0.05). Among cervical cancer survivors, greater comorbidity burden also predicted reduced QOL (β = -4.49; p < 0.01). In breast cancer survivors, engagement in vigorous exercise was associated with higher QOL scores (β = 4.85; p < 0.05). Although similar trends were observed among cervical cancer survivors, smaller sample size limited statistical significance. Conclusions: Findings demonstrate that unmet NMDoH needs, particularly financial strain and comorbid conditions, adversely impact QOL among H/L cancer survivors. Tailored, culturally informed interventions addressing these social and economic challenges – such as patient navigation and community health worker models – are essential for improving survivorship experiences. Incorporating systematic NMDoH screening into survivorship care planning may enhance supportive service provision and optimize long-term health outcomes in this population.
Presentation numberPS1-02-16
Addressing the support needs of people with hereditary cancer risk through virtual peer-led meetings
Juanita D Rogers, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL
J. D. Rogers1, S. Cohen2, M. Rosas2, P. Welcsh1, K. N. Owens1, S. J. Friedman1, R. H. Pugh Yi3; 1Education, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 2Support, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 3President, Akeso Consulting, Vienna, VA.
Background:People with pathogenic variants (PV) associated with hereditary cancer risk face psychosocial challenges, including anxiety about risk-management, family communication, and feelings of isolation. While clinical guidelines are well-established for many genes linked to cancer risk, emotional support remains an unmet need. FORCE is a national nonprofit organization serving people affected by hereditary cancer risk. We provide virtual support meetings tailored for the hereditary cancer community. The meetings are organized by topic (e.g. mastectomy), situation (e.g. survivor, previvor, caregiver), gene (e.g. BRCA1/2, Lynch syndrome, others), and other factors. The meetings are co-led by genetics experts and trained FORCE peer volunteers. Past studies show that women with hereditary cancer risk experience significant emotional distress, and value referrals from their doctors to social support networks3. Perceptions of low social and emotional support are predictors of increased distress and poor mental health outcomes in this population. However, structured support groups, peer connections, and family-based emotional support improve coping, anxiety levels, and patient experience1-3.Integrating emotional support into hereditary cancer care meets a critical need and can improve patient outcomes.Methods:Between February and July 2025, 1000 people attended 40 FORCE virtual support meetings for people affected by hereditary cancer risk. Feedback was collected from 185 attendees via a post-meeting survey to assess satisfaction, emotional support, and perceived value of information gained. Participants could also provide open-ended feedback.Results:Participants expressed that the meetings provided a safe, inclusive space to connect with others facing similar challenges, helped reduce feelings of isolation, and validated participant’s emotions: •62.4% felt less anxious or worried. •87.5% felt more supported or less alone. •82.3% felt more confident in their knowledge.•65.9% felt more confident about their decisions.•94% were “very satisfied” (71.2%) or “satisfied” (22.8%). •Attendees received support on various topics including genetic counseling/testing (40%), breast reconstruction (34%) and communication with healthcare providers (33%).Qualitative Themes:•Greater sense of community: “The support group is the only outlet for me to express my hopes, frustrations and fears.”•Empowerment in medical decision-making: “I am definitely going through with my mastectomy and am so happy I met new friends to help me through my journey!”• Improved representation: Specific groups (e.g., LGBTQ+, young previvors) felt especially seen when breakout sessions included people who shared their lived experiences.Discussion:The emotional burden of navigating hereditary cancer risk can be substantial. Survey responses demonstrated that FORCE’s peer-and-expert-led virtual meetings create a needed space for people with hereditary cancer risk to process complex emotions, build resilience, and make informed decisions. Organizing meetings by topic highlights the diversity of attendees and the importance of tailored support meetings.Conclusion:Virtual support meetings represent a scalable, low-cost, and high-impact intervention to address the emotional needs of people with hereditary cancer risk. The positive reception underscores the value of integrating psychosocial support alongside clinical care.References:1.https://doi.org/10.1080/10410236.2016.1250187 2.FORCE. (2024, October 21). Results from FORCE’s 2024 Survey Highlighting the Needs of People at High Risk for Breast Cancer. FORCE Blogs. 3.https://doi.org/10.3389/fpsyt.2019.00208
Presentation numberPS1-02-17
Scrambler Therapy: a safe and effective way to treat chemo-induced neuropathy
Thomas J Smith, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD
Z. Wang1, C. L. Loprinzi2, S. Abdi3, T. B. Strouse4, T. J. Smith5; 1Medicine, Johns Hopkins, Baltimore, MD, 2Oncology, Mayo Clinic, Rochester, MN, 3Department of Pain Medicine, M D Anderson Cancer Center, Houston, TX, 4Psychiatry, UCLA, Baltimore, MD, 5Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD.
Background: Up to 68% of patients will experience CIPN. There are no FDA or ASCO recommended therapies. The pain (P), numbness (N), and tingling (T) can persist for month, years or even lifetime. Duloxetine reduces pain scores by 0.6-1.0 on the 0-10 scale but does not help N or T. Scrambler therapy (ST) is a novel form of neuromodulation that uses EKG electrodes on the dermatomes to capture the pain c-fiber receptors, and generate an action potential of “non-pain” information to replace the pain information. It appears to reset central sensitization by altering cerebral blood flow and re-establishing the balance of neuropeptides (e.g., NGF, GDNGF) in the blood (Smith et al NEJM 2023). Yet many professionals and patients are unaware of it. Methods: We reviewed all available studies. Results: Smith et al (2010, VCU) noted a 59% P reduction (5.8 to 2.4 at the end of 10 days (P<0.0001)) in 16 subjects, and 4 had 0 pain at day 10. Pachman et al (2015, Mayo) in 37 subjects showed a 53% P reduction by day 10; a 44% reduction in T; and a 37% reduction in N. In a randomized trial of 50 patients using ST versus TENS (transcutaneous electrical nerve stimulation), Loprinzi et al (2020, Mayo), in a randomized trial, showed more than twice as many ST patients reported a 50% improvement (36-56%) vs. TENS (16-28%). At 8 weeks the reductions were important: 33%P, 28%N, 41%T. In a subsequent crossover trial (Childs 2021) of the TENS patients to ST, 6 of 12 ST patients achieved a 50% reduction in P and T. Smith et al (2021 Hopkins) did a randomized sham-controlled trial with 35 patients but observed no lasting effect; in retrospect, the electrodes and electrode placement were incorrect in this trial. Abdi et al (2024, MDACC) did a Phase II trial of ST in 10 patients and found symptoms of N, T, trouble walking, and disturbed sleep had significant improvements at 6 months. Worst pain was reduced at 6 months by 3 points. Pain medication use decreased by 70% at the end of treatment and by 42% at 6 months. Patient satisfaction was high (82%) and no adverse events with ST treatment were reported. Wang Z. et al (Hopkins, unpublished data 2025) showed a 60% reduction in pain (6.2 to 2) in twelve subjects, with one achieving complete remission. Strouse et al (2025, UCLA) will soon report similar results in over 60 patients. Wang E. et al (2023, Hopkins) reviewed 1152 patients treated with ST and found just 3 adverse events: one hematoma and two red spots under the EKG electrodes. The only other therapy that reduces all the symptoms of CIPN is spinal cord stimulation (Gupta 2024) but that is invasive, expensive, and can cause a multitude of adverse events. Conclusions: Scrambler Therapy is a safe and effective treatment for the P, N, and T of CIPN with rare and minimal side effects in a syndrome with no other effective treatment. It is hindered by reimbursement ($32/30 minutes, the same as TENS even though the technology is completely different). Scrambler Therapy should become more available in the future.
Presentation numberPS1-02-18
Enhanced Scalp Cooling Efficacy in Breast Cancer Patients Through Team-Based Care and Regimen-Specific Protocol Optimization: A Single-Institution Study and Review.
Nobuko Tamura, Toranomon Hospital, Tokyo, Japan
N. Tamura1, Y. Nagaoka2, Y. Sano3, Y. Kobayashi1, K. Tanaka1, M. Kurikawa1, A. Shibata1, Y. Tanabe4, Y. Yamaguchi4, Y. Oda5, M. Ogura5, R. Okamoto6, H. Kawabata1; 1Department of Breast and endocrine surgery, Toranomon Hospital, Tokyo, JAPAN, 2Cancer Chemotherapy Nursing, Toranomon Hospital, Tokyo, JAPAN, 3In-hospital beauty salon、ANKS, Toranomon Hospital, Tokyo, JAPAN, 4Department of Medical Oncology, Toranomon Hospital, Tokyo, JAPAN, 5Department of Pharmacy, Toranomon Hospital, Tokyo, JAPAN, 6Department of Pathology Photo Gallery, Toranomon Hospital, Tokyo, JAPAN.
Background: Chemotherapy-induced alopecia (CIA) significantly impacts patients’ quality of life (QoL). While scalp cooling therapy (SCT) offers a proven method for preventing CIA, its widespread implementation faces various challenges. Objective: This study aimed to evaluate the impact of a refined team-based approach and optimized protocols on scalp cooling therapy (SCT) efficacy in breast cancer patients. Methods: A retrospective observational study compared SCT outcomes in breast cancer patients before and after implementing a refined team-based approach (Group A vs. Group B), supplemented by a concise literature review. Results: Our institutional experience demonstrated improved efficacy in preventing alopecia with enhanced team collaboration and the active involvement of appearance care professionals, including hairdressers. Importantly, chemotherapy regimen sequencing significantly influenced outcomes. In Group A, patients receiving weekly paclitaxel followed by dose-dense doxorubicin/cyclophosphamide (wPTX f/b ddAC) had a significantly shorter average number of Grade 2 or higher alopecia cycles (mean: 1.007, median: 0.5) compared to those receiving the reverse sequence (ddAC f/b wPTX, mean: 2.870, median: 3.0) (P = 0.0006). In Group B, while wPTX f/b ddAC also showed a shorter average (mean: 0.448, median: 0.0) compared to ddAC f/b wPTX (mean: 0.800, median: 0.0), this difference did not reach statistical significance (P = 0.0543), potentially reflecting the overall improved efficacy in this group. For patients receiving wPTX f/b ddAC, the percentage of Grade 2 alopecia at the end of treatment was 44.8% in Group B, significantly lower than 69.2% in Group A. The overall proportion of time spent in Grade 2 alopecia during the entire treatment period also demonstrated a shorter duration in Group B (6.5%) compared to Group A (17.3%). Furthermore, comparing the average number of cycles with Grade 2 or higher alopecia, Group B (0.448 cycles) demonstrated a significantly shorter duration compared to Group A (1.007 cycles; P=0.0055). For ddAC f/b wPTX, Group B (0.800 cycles) also showed a significantly shorter average number of cycles with Grade 2 or higher alopecia compared to Group A (2.870 cycles; P=0.0003). For TC therapy, the average number of cycles with Grade 2 or higher alopecia did not yield a statistically significant difference (Group A: 3.000 cycles; Group B: 2.550 cycles; P=0.1176). For the PEM/Cb/wPTX→PEM/AC regimen, Group B (0.000 cycles) demonstrated a significantly shorter average compared to Group A (3.000 cycles; P=0.0002).Conclusion: Maximizing SCT efficacy requires not only technical proficiency but also a comprehensive multidisciplinary team approach and personalized, high-quality care. Specifically, optimized chemotherapy regimen sequencing, alongside the active involvement of appearance care professionals like hairdressers, significantly enhances efficacy. This study highlights their crucial role in improving future SCT dissemination and quality.
Presentation numberPS1-02-19
Aging and inflammatory biomarkers in post-menopausal breast cancer survivors with and without treatment-induced menopause
Shuo Wang, University of Minnesota, Minneapolis, MN
S. Wang1, S. A. Everson-Rose2, A. Prizment1, A. H. Blaes2; 1Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 2Medicine, University of Minnesota, Minneapolis, MN.
Background: Menopause has been associated with increased aging biomarkers, such as epigenetic clocks and inflammatory biomarkers, including interleukin-6 (IL-6) and C-reactive protein (CRP). However, no studies have examined aging and inflammatory processes in breast cancer survivors who experienced treatment-induced menopause. This study compared aging and inflammatory biomarker levels in post-menopausal breast cancer survivors with and without treatment-induced menopause. Methods: This study included 25 women who participated in the Mindfulness-Based Stress Reduction (MBSR) study, which investigated the impact of an 8-week MBSR intervention on the aging process. All participants were diagnosed with locally advanced, estrogen receptor-positive breast cancer. They had undergone surgery and were taking an aromatase inhibitor. We divided participants into two groups: Group 1 included 8 females younger than 50 years who experienced treatment-induced menopause, and Group 2 included 17 females older than 50 years who had undergone natural menopause. Blood samples were collected at baseline and at the 8-week follow up. Using the baseline samples, we measured an aging biomarker, P16INK4a expression in T cells, which increases with age. We also measured two inflammatory biomarkers: IL-6 (pg/mL) and high-sensitivity CRP (hsCRP, mg/L). P16INK4a expression and hsCRP levels were log2 transformed to correct for skewness. We compared the levels of P16INK4a, IL-6, and hsCRP across groups in an unadjusted model and a model adjusted for age, chemotherapy, and radiation. Results: Participants’ characteristics for the two groups are presented in Table 1. Participants were predominantly White and highly educated. For Group 1 (treatment-induced menopause) and Group 2 (natural menopause), respectively, the mean age was 43.7 and 61.9 years, and the mean time since diagnosis was 3.8 and 4.2 years. All women in Group 1 had chemotherapy, whereas 38.9% in Group 2 had chemotherapy (p = 0.01). The prevalence of radiation was similar across groups [Table 1]. In both unadjusted and adjusted models, mean levels of P16INK4a (Group 1 vs. Group 2 in the model adjusted for age, chemotherapy, and radiation: 5.04 vs. 5.11, p = 0.19), IL-6 (1.03 vs. 2.30, p = 0.28), and hsCRP (1.04 vs. 0.92, p = 0.92) were similar across groups. Conclusions: Despite treatment-induced menopausal breast cancer survivors being, on average, 18 years younger than those with natural menopause, both groups showed similar levels of aging and inflammatory biomarkers. These findings suggest that treatment-induced menopause may accelerate the aging process among young female breast cancer survivors. Future studies should examine whether treatment-induced menopause accelerates aging more than natural menopause and investigate longitudinal aging trajectories in treatment-induced menopausal breast cancer survivors.
| Group 1 (age <50 years, treatment-induced menopause) | Group 2 (age >50 years, natural menopause) | p-value | |
| Number of participants | 8 | 17 | |
| Mean age, years (SD) | 43.7 (2.5) | 61.9 (7.8) | <0.01 |
| White, % | 85.7% | 100% | 0.11 |
| College degree, % | 100% | 88.9% | 0.65 |
| Mean time since diagnosis, years (SD) | 3.8 (2.3) | 4.2 (1.9) | 0.64 |
| Chemotherapy, % | 100% | 38.9% | 0.01 |
| Radiation, % | 85.7% | 83.3% | 0.88 |
| Comparison of biomarker levels across groups in an unadjusted model | |||
| P16INK4a expression (SD) | 5.08 (0.05) | 5.09 (0.07) | 0.69 |
| Interleukin-6 (IL-6), pg/mL (SD) | 1.99 (1.07) | 2.02 (1.74) | 0.97 |
| High-sensitivity C-reactive protein (hsCRP), mg/L (SD) | 0.96 (2.5) | 0.95 (2.3) | 0.99 |
| Comparison of biomarker levels across groups in the model adjusted for age, chemotherapy, and radiation | |||
| P16INK4a expression (SD) | 5.04 (0.05) | 5.11 (0.08) | 0.19 |
| IL-6, pg/mL (SD) | 1.03 (2.1) | 2.30 (1.7) | 0.28 |
| HsCRP, mg/L (SD) | 1.04 (2.3) | 0.92 (1.8) | 0.92 |
Presentation numberPS1-02-20
Feasibility of Implementing a Novel Dietary Assessment Platform for Breast Cancer Patients: A Kaiser Permanente Pilot Study
Isaac J Ergas, Kaiser Permanente Northern California, Pleasanton, CA
I. J. Ergas1, C. Mindemann1, J. Roh1, D. Lia1, L. Raymond2, V. Shim3, S. Siegel2, M. Kwan1, L. Kushi1, R. Zwerling Baltrushes3; 1Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, 2San Francisco Medical Center, Kaiser Permanente Northern California, San Francisco, CA, 3Oakland Medical Center, Kaiser Permanente Northern California, Oakland, CA.
Background: Breast cancer has one of the highest 5-year survival rates among cancers, with over 4 million survivors in the U.S. Studies suggest that adherence to a healthy diet can reduce morbidity and mortality risk in this population. However, dietary assessment is often overlooked in routine care due to limitations of existing tools. Traditional methods such as 24-hour recalls and food frequency questionnaires are time-consuming and burdensome, particularly for patients undergoing treatment. Diet ID™ is an online, image-based dietary assessment tool that uses Diet Quality Photo Navigation—a patented visual pattern recognition algorithm. Participants compare food photos representing different dietary patterns and select the one that best reflects their typical diet over the past month. The tool generates comprehensive nutritional profiles, and a Healthy Eating Index (HEI) score. This study aims to evaluate the feasibility of using the Diet ID™ platform to assess diet quality in breast cancer patients within a large integrated healthcare system. Methods: Eligible participants were women diagnosed with any stage of breast cancer within the past 24 months and receiving care at Kaiser Permanente San Francisco or Oakland. Recruitment is being conducted via email across three planned waves spaced three months apart, with the first wave launched in April 2025. Participants complete the Diet ID™ assessment online, with responses linked to electronic health records to capture additional demographic and clinical data. Feasibility metrics include recruitment yield, completion rates, and time to completion. Descriptive analyses summarize participant characteristics and HEI score distributions. Results: In the first wave, 1,349 potentially eligible patients were identified. After excluding 324 (24%) for reasons such as language preference, cognitive impairment, and lack of email, 1,021 were invited via email. Of those invited, 328 (32%) enrolled, with 93% completing the assessment in full. While enrollees were similar in age to invitees (mean = 58.9 vs. 60.7 years), White participants were overrepresented (62.8% enrolled vs. 47.0% invited) and Black/African American (5.8% vs. 13.7%), Asian/PI (18.6% vs. 23.4%), and Hispanic (6.4% vs. 8.0%) participants were underrepresented. Stage III/IV patients were also less likely to enroll (5.4% vs. 8.0%). Among 304 participants with complete dietary data, HEI scores ranged from 17 to 98 (mean ≈ 83, standard deviation ≈ 18) and were strongly right-skewed. Participants under age 50 were more common in the lowest HEI tertile (38%) than in the highest (27%). White participants were more likely to be in the top tertile (67% vs. 59% in the lowest), while Black/African American (5% vs. 9%) and Asian/PI (13% vs. 20%) participants were less represented. Higher HEI scores were linked to lower BMI: normal weight was more common in the top tertile (54% vs. 36%), while obesity was less prevalent (12% vs. 26%). Overall, about 15% of participants felt the tool did not accurately capture their diet, but dissatisfaction was more common in the lowest tertile (30%) compared to the highest (7%). Most participants found the tool quick (<5 minutes) and easy to use, with high acceptability (~85%). Results from the July and October waves will be included in the final analysis and presented at the conference. Conclusions: Preliminary findings suggest that Diet ID™ is a feasible and acceptable tool for assessing diet quality among breast cancer patients in clinical care. Early results show meaningful variation in diet quality and expected associations with BMI. However, the strong right skew in HEI scores highlights the need to assess the tool’s sensitivity to lower-quality diets. Ongoing data collection and follow-up will inform its broader use in dietary surveillance and survivorship care.
Presentation numberPS1-02-21
Personalizing breast cancer treatment for older adults: use of the geriatric assessment in chemotherapy decision making
Sacha A Roberts, NewYork-Presbyterian Weill Cornell, NEW YORK, NY
S. A. Roberts1, A. L. Tin2, J. Singh3, D. Lake3, E. Meditz4, A. Kulkarni5, M. Boparai6, B. Korc-Grodzicki6, A. Shahrokni6, K. Alexander6; 1Internal Medicine, NewYork-Presbyterian Weill Cornell, NEW YORK, NY, 2Department of Statistics, Memorial Sloan Kettering Cancer Center, NEW YORK, NY, 3Department of Breast Oncology, Memorial Sloan Kettering Cancer Center, NEW YORK, NY, 4Department of Nutrition, Memorial Sloan Kettering Cancer Center, NEW YORK, NY, 5Department of Hematology and Oncology, Columbia University Irving Medical Center, NEW YORK, NY, 6Department of Geriatrics, Memorial Sloan Kettering Cancer Center, NEW YORK, NY.
Purpose: To assess which patient characteristics and elements of the GeriatricAssessment (GA) are associated with not proceeding to chemotherapy in older womenwith breast cancer.Methods: Between April 2022 and February 2024, 50 women aged 65 and older with breastcancer were seen in the Cancer and Aging Interdisciplinary Team clinic at our institution forGA prior to initiating systemic treatment. We present characteristics among our cohort andevaluate differences in GA domains between patients who proceeded to chemotherapyand those who did not.Results: When comparing the cohort of patients who did not undergo chemotherapy vsthose who did, factors that were significantly associated with the decision to omitchemotherapy included older age (median 84 vs 75 years old, p<0.001), non-triple negativedisease (75% vs 42%, p=0.035), frailty (94% vs 55%, p=0.007), abnormal functionalperformance test (75% vs 19%, p<0.001), high risk CARG-TT score (50% vs 13%, p=0.007),chemotherapy not recommended by geriatrician (69% vs 0%, p<0.001), iADL dependency(88% vs 44%, p=0.047), KPS<80 (75% vs 19%, p=0.003), and limited social activity (100% vs52%, p=0.015). Of the women who proceeded to chemotherapy, 42% underwent upfrontchemotherapy modifications.Conclusion: Our study identified components of the GA associated with chemotherapydecisions and modifications in older women with breast cancer. Further research isneeded to determine the causal relationships between GA components and decision-making and evaluate how such decision-making could be improved.
Presentation numberPS1-02-22
Delayed Palliative Care in Metastatic Breast Cancer: Rationale for Early Integration
Daria Chelysheva, UPMC Harrisburg, Harrisburg, PA
D. Chelysheva, J. Fanizza, E. Nasrollahi, E. Aguilar Montoya; Internal Medicine, UPMC Harrisburg, Harrisburg, PA.
Background Metastatic breast cancer (MBC) is an advanced stage of disease with a poor prognosis and heavy symptom burden. Its five-year relative survival rate is about 30%, and the median survival time is roughly 2-3 years. Palliative care can alleviate symptoms and improve quality of life in advanced cancer, and major oncology guidelines (ASCO/ESMO) now recommend providing specialized palliative care early in the course of metastatic illness. However, in real-world practice palliative care is often introduced very late. This study examines how delayed palliative care referral affects outcomes in MBC and makes the case for earlier integration. Methods We conducted a retrospective cohort study using data from the TriNetX global health research network, which includes de-identified electronic health records from multiple healthcare organizations, predominantly located in the United States. Female patients who were diagnosed with stage IV (metastatic) breast cancer between 2011 and 2024 were included. The primary focus of the analysis was to assess the time interval between metastatic diagnosis and receipt of a palliative care consultation. Additional variables collected included demographics and documented clinical symptoms. Patients were grouped based on whether or not they received a palliative care consultation, and matched cohort analysis was performed to compare symptom burden between the groups. Results A total of 1036 women with stage IV breast cancer were included. The median age was 66 years (range 30-90; SD = 13). The median time from metastatic diagnosis to first palliative care consult was 408 days, indicating that referrals generally occurred late in the disease course. Notably, median referral delays varied by race: 425 days for White patients, 347 days for Black or African American patients, 699 days for Asian patients, 294 days for Hispanic or Latino patients and 388 days for American Indian/Alaska Native patients. When we analyzed matched cohorts of patients who did and did not receive palliative care consultations, it was discovered that symptom burden was significantly higher in the palliative care group. This included symptoms such as pain, fatigue, delirium, and dyspnea. Conclusions These results highlight that palliative care is frequently introduced late for patients with metastatic breast cancer. Earlier integration, ideally soon after stage IV diagnosis, is advocated in guidelines and has the potential to improve symptom control, enhance quality of life, and align care with patient goals. In our cohort, the median time to palliative care referral was over a year from diagnosis. These findings align with prior studies and underscore that late palliative care referral may limit its impact on meaningful outcomes. The observed racial differences in timing further suggest a need to address systemic and cultural barriers to equitable, timely access. Additionally, the significantly higher symptom burden observed in the palliative care group supports the concern that referrals are often made reactively, in response to advanced clinical decline. Overall, our findings reinforce that palliative care should be embedded earlier in the treatment trajectory – not as a last resort, but as a concurrent component of comprehensive cancer
Presentation numberPS1-02-23
The impact of optimism and coping on the experience of side effects of adjuvant chemotherapy in women with early-stage breast cancer
Keri Morgan Cronin, mt sinai west, New York, NY
K. M. Cronin1, J. Fasano2, C. Shapiro3, G. Montgomery4, s. julie4, a. tiersten4, a. bhardwaj4, h. irie4, p. klein4, a. goel4, t. sheng4, t. shao4; 1internal medicine, mt sinai west, New York, NY, 2Oncology, Mt Sinai Hospital, New York, NY, 3internal medicine, Mt sinai, New York, NY, 4internal medicine, mt sinai, New York, NY.
Background:Adjuvant chemotherapy for early-stage breast cancer is associated with numerous side effects, including nausea, fatigue, and neuropathy, which may lead to early discontinuation. Psychosocial variables may influence how patients experience and tolerate these side effects. Personality traits such as optimism and neuroticism at diagnosis may impact psychosocial outcomes and recurrence in long-term survivors. Optimism has been linked to better coping, reduced anxiety, and greater resilience, whereas neuroticism correlates with distress and poor coping. No prior studies have directly examined the relationship between optimism, coping, and chemotherapy side effects. This study aimed to evaluate whether psychosocial factors are associated with chemotherapy-related side effects.Methods:We conducted a prospective study of women with stage I-III breast cancer receiving standard neoadjuvant or adjuvant chemotherapy. At baseline (prior to treatment), participants completed four psychosocial questionnaires: the Revised Life Orientation Test (LOT-R), NEO-Five Factor Inventory Neuroticism Scale (NEO-N), Connor-Davidson Resilience Scale 10 (CD-RISC 10), and Brief COPE (B-COPE). Patients also completed the PRO-CTCAE v1 and PROMIS surveys at baseline, during chemotherapy, end of treatment, and one year post-therapy. The primary endpoint was to determine correlations between optimism, coping, and resilience with treatment-related toxicity (PRO-CTCAE). The secondary endpoint was to examine associations between psychosocial factors and physical, mental, and social domains of health-related quality of life (HRQOL). Spearman correlations assessed relationships between psychosocial and symptom scores. Linear mixed-effects models evaluated longitudinal associations, with time point and psychosocial variables as fixed effects and participant as a random intercept. Analyses were conducted in R (v4.4.0).Results:A total of 107 patients were enrolled (median age: 51 [range 42-58]). The cohort was diverse: 18.7% Hispanic, 37.3% White, 24.3% Black, 10.3% Asian, and 9.3% other. Symptom scores increased significantly by the end of chemotherapy compared to baseline (Estimate = 0.49, p < 0.01). Higher optimism was modestly associated with lower symptom burden (Estimate = -0.03, p = 0.05). Resilience was not significantly associated with symptom scores. Greater use of coping strategies was linked to higher symptom burden (Estimate = 0.02, p < 0.01), possibly reflecting increased efforts to manage symptoms. No specific coping strategy was significantly associated with symptom scores. Symptom burden increased during and shortly after chemotherapy but declined one year later. In the mixed-effects model, none of the psychosocial scores (optimism, coping, or resilience) remained significantly associated with symptom burden over time.Conclusions:Patients with higher optimism experienced fewer chemotherapy-related side effects, suggesting that optimism may serve as a potential psychological marker for reduced treatment burden. Greater coping effort was observed in those reporting higher symptom scores, possibly indicating a reactive coping response. These findings may inform future development of behavioral interventions aimed at reducing chemotherapy-related side effects in breast cancer patients.
Presentation numberPS1-02-24
Physical activity level differences among Non-Hispanic White and Chinese American breast cancer survivors
Can Cao, Mount Sinai Hospital, New York, NY
C. Cao1, T. Shao2, A. Tsang1, M. Kwa3, R. Brown4, L. Chen5, M. Schwartz6, D. Roblin7, M. H. Antoni8, L. Hilakivi-Clarke9, G. Kowk10, A. Lin9, K. Joseph3, J. X. Osorio6, J. H. Wang6, S. Cai1; 1Surgery, Mount Sinai Hospital, New York, NY, 2Hematology & Oncology, Mount Sinai Hospital, New York, NY, 3NYU Perlmutter Cancer Center, New York University Langone Health, New York, NY, 4Clinical Health Science, University of Wisconsin–Madison, Madison, WI, 5School of Public Health, Texas A&M University, College Station, TX, 6Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 7Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, 8Psychology, University of Miami, Miami, FL, 9Hormel Institute, University of Minnesota, Austin, MN, 10Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ.
INTRODUCTION: Increased physical activity (PA) in breast cancer patients has been associated with multiple benefits including lower fatigue, depression and anxiety levels, better tolerance of treatment related side effects, and lower cancer recurrence rates (1). The American Cancer Society recommends that cancer survivors undergo at least 150-300 minutes of moderate intensity activity or 75-150 minutes of vigorous activity per week and encourages meeting or exceeding these upper limits (2). Given limited data on PA in Chinese American breast cancer survivors, this study aimed to evaluate and better quantify PA in this population. METHODS: As a part of a broader prospective cohort study investigating survivorship in Chinese American breast cancer patients compared to Non-Hispanic white (NHW) patients over a 12-month period, participants underwent a baseline phone survey administered by trained staff. All participants were women aged over 18, diagnosed with stage 0-III breast cancer within 10 years, and had completed primary surgical or medical treatment. The survey included the Global Physical Activity Questionnaire developed by World Health Organization (WHO), which was used to evaluate time spent on moderate and vigorous activity during work, travel and leisure and to calculate weekly metabolic equivalent (MET) minutes per week for each group. Group differences were evaluated using T-tests for significance. RESULTS: A total of 133 NHW and 110 Chinese American patients completed the baseline survey. The average age was 56 for Chinese Americans and 59 for NHWs, and the difference was not statistically significant. The average BMI of Chinese Americans (23.6) was statistically significantly lower than that of NHWs (25.7). Chinese Americans spent an average of 71 minutes per week on vigorous activity and 424 minutes per week on moderate activity compared to NHWs who spent an average of 179 minutes per week and 627 minutes per week, respectively. Combined, Chinese Americans averaged 2266 MET-minutes per week, while NHWs averaged 3952 MET-minutes per week, this difference was statistically significant (p = 0.0053). Additionally, for patients reporting use of a step counter, the average daily step count for Chinese Americans was 6592 compared to 8432 for NHWs. The reported average daily sedentary time for Chinese Americans (337 minutes) was not statistically different from that for NHWs (348 minutes). CONCLUSION: While both groups exceeded recommended PA minimums, Chinese American women with breast cancer self-reported significantly lower levels of PA than their NHW counterparts. Further studies should focus on the impact of this discrepancy on survivorship and possible interventions to bridge the gap between the two groups to mitigate health disparity after breast cancer treatment. KEYWORDS: breast cancer; survivorship; exercise; disparity SOURCES:(1) Le Y, Gao Z, Gomez SL, Pope Z, Dong R, Allen L, Chang MW, Wang JH. Acculturation and Adherence to Physical Activity Recommendations Among Chinese American and Non-Hispanic White Breast Cancer Survivors. J Immigr Minor Health. 2019 Feb;21(1):80-88. doi: 10.1007/s10903-018-0721-x. PMID: 29569102; PMCID: PMC6151158. (2) Rock CL, Thomson C, Gansler T, Gapstur SM, McCullough ML, Patel AV, Andrews KS, Bandera EV, Spees CK, Robien K, Hartman S, Sullivan K, Grant BL, Hamilton KK, Kushi LH, Caan BJ, Kibbe D, Black JD, Wiedt TL, McMahon C, Sloan K, Doyle C. American Cancer Society guideline for diet and physical activity for cancer prevention. CA Cancer J Clin. 2020 Jul;70(4):245-271. doi: 10.3322/caac.21591. Epub 2020 Jun 9. PMID: 32515498.
Presentation numberPS1-02-25
Multimodal Exercise in Breast Cancer: Health Impact and Efficacy of Online Modalities. RCT
Soraya Casla, Universidad Pontificia Comillas, Ciempozuelos, Spain
S. Casla1, M. Castellanos-Montealegre2, L. Cantero3, M. Ayuso-Chico4, J. Haro5, Ejercicio y Cáncer; 1Escuela Enfermería y Fisioterapia, Universidad Pontificia Comillas, Ciempozuelos, SPAIN, 2Ciencias de la Actividad Física y el Deporte, Universidad de Castilla-La Mancha, Toledo, SPAIN, 3Centro Retiro, Centro Ejercicio y Cáncer, Madrid, SPAIN, 4Centro Cádiz, Centro Ejercicio y Cáncer, Madrid, SPAIN, 5Retiro, Centro Ejercicio y Cáncer, Madrid, SPAIN.
Multimodal Exercise in Breast Cancer: Health Impact and Efficacy of Online Modalities. RCTIntroductionExercise is widely recognized for its significant benefits in cancer survivors, helping improve cardiorespiratory fitness (CRF), reduce fatigue, and positively impact body composition. These advantages are crucial as cancer treatments often lead to reduced physical function and increased fatigue, negatively affecting quality of life (1,2). While traditional supervised exercise programs are effective, access remains a challenge due to logistical barriers. Therefore, exploring remote exercise options is essential for broader accessibility (3,4). This study evaluated the effectiveness and feasibility of a 16-week multimodal exercise intervention comparing online and onsite formats for breast cancer survivors at stages IA to IIIB.MethodologyThe study was conducted as a randomized controlled trial with 83 participants divided into intervention and control groups. The intervention group was further split into online and onsite subgroups, participating in aerobic, strength, and flexibility exercises. The primary outcomes measured were CRF (using the Bruce test) and body composition (via bioelectrical impedance). Secondary outcomes included functional capacity (sit-to-stand and 6-minute walk tests) and quality of life.Results demonstrated a 22.4% improvement in CRF in the intervention group versus a 4.96% decrease in the control group. Functional capacity also improved significantly in both the online and onsite groups, showing the potential of remote programs to match in-person results (5,6). Body composition changes included a 5.24% increase in lean mass and a 10% decrease in fat mass, supporting evidence that exercise contributes to improved metabolic profiles and lower inflammation, reducing long-term cancer risks (7,8).ConclusionsThe discussion highlighted a high adherence rate of 95.3% across both modalities, underscoring the effectiveness of flexible delivery for maintaining engagement. The perfect adherence in the online group (100%) emphasized the potential for virtual programs to fit into patients’ lives, especially for those balancing responsibilities or facing geographic limitations (9). Limitations included the use of the Borg scale for intensity monitoring instead of more objective measures, suggesting future studies should include precise tools and examine active treatment phases (10).In conclusion, both online and in-person multimodal exercise interventions were effective in enhancing CRF, body composition, and functional capacity in breast cancer survivors. The innovative aspect of demonstrating equivalent results between modalities highlights the potential of flexible, personalized programs to improve access and adherence, ultimately influencing long-term survival and quality of life.
Presentation numberPS1-02-26
Effectiveness of mindfulness on quality of life in women breast cancer survivors: a Systematic Review and Meta-Analysis
Paulo Gustavo Tenório do Amaral, FMB/UNESP, Botucatu/SP, Brazil
P. Amaral1, G. Tosello2, P. Silva Filho1, E. Nahas1, D. Buttros1; 1Pós-graduação em Tocoginecologia, FMB/UNESP, Botucatu/SP, BRAZIL, 2Mastologia, Intituto do Câncer Oeste Paulista, Presidente Prudente/SP, BRAZIL.
Introduction: The rising incidence of breast cancer, alongside decreased mortality rates and therapeutic advances, has led to an increasing population of breast cancer survivors. These individuals are susceptible to acute and chronic psychological distress. Mindfulness-based interventions have been proposed as integrative strategies to support mental health and well-being in this population. This study aimed to evaluate the effectiveness of mindfulness practices compared to standard care in improving the quality of life among women surviving breast cancer. Methods: A systematic review was conducted including randomized controlled trials (RCTs) involving women aged ≥18 years who had undergone treatment for breast cancer (surgery, radiotherapy, chemotherapy, or a combination), at any clinical stage, without psychiatric disorders, no prior mindfulness practice, and that assessed quality of life as a primary or secondary outcome. Studies not meeting these criteria were excluded. A comprehensive literature search was performed without language or date restrictions across PubMed, Embase, Scopus, Web of Science, CINAHL, SciELO, LILACS, Cochrane Library, WHO, and grey literature sources (Google Scholar, OpenGrey, ProQuest, and direct contact with field experts). Risk of bias was assessed using the RoB 2 tool. Standardized mean differences (SMD) were calculated for pooled effect estimates using R software. The certainty of evidence was evaluated using the GRADE framework. Results: Thirteen RCTs met inclusion criteria, comprising a total of 1,942 breast cancer survivors, of whom 1,016 received mindfulness interventions and 926 received standard care. Risk of bias was considered low across the included studies. Meta-analysis revealed that mindfulness practices resulted in a small but statistically significant improvement in quality of life (SMD = 0.33; 95%CI: 0.16 to 0.49). However, the certainty of evidence was rated as low due to moderate heterogeneity (I² = 65.9%), high risk of publication bias (funnel plot asymmetry and Egger’s test), and serious imprecision (confidence interval spanning trivial to small effect sizes). Discussion: Key limitations of this review include study heterogeneity, the predominance of Mindfulness-Based Stress Reduction (MBSR) as the primary intervention, limited representation of patients with metastatic disease, and possibly attenuated effects in those with early-stage tumors due to less intensive treatment regimens. Despite these limitations, mindfulness practices demonstrated potential benefits in enhancing quality of life and, due to their socioeconomic accessibility, may warrant clinical recommendation as part of survivorship care in breast cancer patients. Funding and Registration: This study did not receive external funding. The review protocol was registered with PROSPERO (CRD42024502665) on January 26, 2024.
Presentation numberPS1-02-27
Assessment of metabolic health in women with breast cancer: a prospective cohort study
Paulo Gustavo Tenorio do Amaral, FMB/UNESP, Botucatu/SP, Brazil
V. Prado1, D. Buttros1, P. Amaral1, G. Tosello2, S. Rizzi1, E. Nahas1; 1Pós-Graduação em Tocoginecologia, FMB/UNESP, Botucatu/SP, BRAZIL, 2Mastologia, Intituto do Câncer Oeste Paulista, Presidente Prudente/SP, BRAZIL.
Introduction: Women diagnosed with breast cancer exhibit an elevated risk of weight gain and metabolic dysfunctions, which are associated with a decline in overall and specific survival rates. Interdisciplinary assessment at the time of diagnosis can positively influence prognosis and survival. This study aimed to evaluate the impact of breast cancer diagnosis and treatment on the metabolic health and survival of women undergoing treatment for breast cancer. Methods: This was a single-center, prospective cohort study that included women with a recent diagnosis of breast cancer, aged ≥ 40 years, without metastatic disease or established cardiovascular disease, who were followed for the first two years after breast cancer diagnosis. The criteria used for the assessment of metabolic health/dysfunction were the presence of obesity (body mass index, BMI ≥ 30kg/m²), hypertension (blood pressure, BP ≥ 130/85mmHg), hypertriglyceridemia (triglycerides, TG ≥ 150mg/dL), low high-density lipoprotein cholesterol (HDL 100mg/dL), and the presence of metabolic syndrome (MetS, NCEP-ATPIII criteria). Clinical and anthropometric data (BP, BMI, and waist circumference/WC) were collected through interviews and physical examinations. Information on oncological treatments and tumor characteristics was extracted from medical records. Biochemical analyses included total cholesterol, HDL, LDL, TG, and glucose levels. Patients underwent interdisciplinary assessment (nutritional and psychological) at the time of diagnosis and continued with follow-up appointments with a breast specialist according to the service routine for 2 years. Assessments were conducted at five time points: initial consultation, and at six, 12, 18, and 24 months. Statistical analyses included non-parametric tests, logistic regression, and Kaplan-Meier survival analysis. Results: A total of 165 women were eligible for the study, of whom 39 dropped out during follow-up. The analysis included 126 women with a mean age of 59.0 ± 12.0 years and a mean BMI of 29.7 ± 5.8 kg/m²; 91% of tumors were in stages I and II, 71% had negative axillary lymph nodes, and 79% were luminal subtypes. Over the 2-year follow-up period, among the indicators of metabolic dysfunction, a significant reduction was observed in the occurrence of women with dysglycemia (p=0.04), elevated systolic blood pressure (p0.05). During follow-up, 15 patients died, and these patients had a higher prevalence of MS at initial assessment (73% versus 41% of survivors, p=0.020) and higher glucose levels (p=0.021) compared to survivors. The presence of MS was associated with lower survival (p=0.021), with a Hazard Ratio of 9.09 (95% CI: 1.73-47.9). Conclusions: In women with breast cancer, interdisciplinary follow-up and healthy lifestyle recommendations had a positive impact on metabolic health, specifically on glycemia, HDL levels, and blood pressure, without altering the occurrence of obesity and MetS. MetS presented a high prevalence and was associated with lower survival.
Presentation numberPS1-02-28
Quality of life of women after breast surgery: report from Mirebalais University Hospital in Haiti.
Temidayo Fadelu, Dana-Farber Cancer Institute, Boston, MA
F. Turenne1, J. Merci2, C. Mannley2, G. Benoit2, M. Guerrier2, W. Guerrier2, K. Dubique3, S. Estanis3, E. Durkin4, T. Fadelu5; 1Surgery, University Hospital Mirebalais/ Zanmi Lasante, Mirebalais, HAITI, 2Oncology, University Hospital Mirebalais/ Zanmi Lasante, Mirebalais, HAITI, 3Research, University Hospital Mirebalais/ Zanmi Lasante, Mirebalais, HAITI, 4Oncology, Partners In Health, Boston, MA, 5Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: Breast cancer remains the leading cause of female cancer mortality in low- human development index countries, including in Haiti. Treatment of breast cancer can impact patient’s quality of life (QOL). At the University Hospital of Mirebalais (HUM) in Haiti, due to the national absence of radiation therapy, patients treated with curative intent receive modified radical mastectomy (MRM) and systemic therapy with chemotherapy and hormonal therapy, as clinically indicated. This project aimed to measure QOL in women after breast cancer surgery. Methods: We conducted a cross-sectional study in adult women with pathologically-confirmed breast cancer and who underwent MRM at HUM between January 2018 to December 2022. Clinical and treatment data were collected from the electronic medical records and paper charts, while a comprehensive survey was administered assessing QOL and potentially associated factors. Functional Assessment of Cancer Therapy- Breast (FACT-B) 37-item questionnaire was used to assess the following domains: physical, emotional, social, and functional well-being, as well as breast cancer specific concerns; total score range is from 0-148, with higher scores representing better QOL. Hospital anxiety and depression scale (HADS) was also administered. These questionnaires were chosen due to broad use and prior validation in multiple settings; they underwent linguistic translation into Haitian Creole. Data were securely collected using Research Electronic Data Capture (REDCap) and Epi-Info version 7.2.5.0 was utilized for data analysis. QOL data were summarized descriptively in aggregate and by domain, groups were compared using non-parametric comparisons tests (Mann-Whitney and Kruskal-Wallis tests) and logistic regressions to explore potential predictors of anxiety and depression (cut-offs: HADS-A ≥8 for mild anxiety, HADS-D ≥15 for severe depression). Results: The study recruited 87 participants. The median age of the participants was 50 (interquartile range (IQR): 43-58). Educational attainment was modest, with 27 (31%) patients having only primary school level education and 41 (47%) patients reaching secondary school level. Most of the patients resided in the West department (60, 69%), most were post-menopausal (71, 82%), and 35 (40%) were married. Most patients (58, 67%) had locally advanced tumors, and majority (60, 69%) had an ECOG performance status of 1. Within this cohort, 86 (99%) received chemotherapy, and 78 (91%) received hormonal therapy. The total FACT-B score among all patients had a median of 111.5 (IQR: 97.2-122.7), reflecting generally good perceived quality of life. In summary, most demographic and clinical variables were not statistically significantly associated with QOL, except emotional well-being was notably lower among younger women (<50 years), and receipt of both neo- and adjuvant chemotherapy appeared to be negatively associated with well-being in aggregate and across multiple domains (physical, emotional and breast specific well-being). The total median HADS anxiety score was 8 (IQR: 6-11) and a depression score was 13 (IQR: 12-15), indicating at least mild anxiety and moderate to severe depression in most of the cohort. Again younger age (<50) was associated with higher rates of anxiety, while there was no clear association trend with depression among the factors that were explored. Conclusions: This study provides a summary of QOL and the prevalence of anxiety and depression among Haitian patients who underwent MRM. Younger patients had worse emotional well-being and more anxiety. The high prevalence of at least moderate depression in over 50% of patients indicates unmet psychosocial needs in this underserved population. The study highlights the need for improved mental health services for patients with breast cancer, especially for younger patients.
Presentation numberPS1-02-29
Sexual health communication preferences of women with metastatic breast cancer
Nusrat Jahan, University of Alabama at Birmingham, Birmingham, AL
N. Jahan1, E. Stringer-Reasor1, T. Padalkar1, K. Khoury1, M. Escobar1, C. P. Williams2, I. Starks1, H. Sarfraz1, A. Falcao1, N. Henderson1, S. Olisakwe1, K. Keene3, A. Azuero4, M. E. Melisko5, E. H. Shinn6, G. B. Rocque1; 1Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 2Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 3Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 4UAB School of Nursing, University of Alabama at Birmingham, Birmingham, AL, 5Division of Hematology and Oncology, University of California San Francisco, San Francisco, CA, 6Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Metastatic breast cancer (MBC) and its therapies can cause a myriad of sexual problems, which often go unaddressed. There is limited information on how women with MBC would like to communicate the sexual problems that may arise from their ongoing cancer therapy. Methods: This qualitative study explored the perspectives on sexual health communication preferences of women with MBC. A medical anthropologist conducted semi-structured interviews with women diagnosed with MBC at the University of Alabama at Birmingham from September 2024 to May 2025 via Zoom, phone, or in-person meetings. Interviews were transcribed verbatim and inductively coded using Dedoose software to identify recurring themes and exemplary quotes. Results: Of 20 interviewed women aged 30-77, 11 (55%) were White and 9 (45%) were African American; 95% were heterosexual and 1 (5%) was bisexual; 10 (50%) were married or partnered and 10 (50%) were widowed, single, or divorced; and 90% were in menopause (natural or treatment-related). Six key themes emerged from interviews (Table). First, women with MBC emphasized normalizing the conversation about sexual problems in the clinical setting and permitting women to bring sexual problems to providers. “Being comfortable with the conversation and making it not seem like it’s something that’s taboo, that then can help patients feel more comfortable talking about something that they may feel is taboo to talk about with.” Second, they recommended self-review materials, such as flyers, pamphlets, or web pages, that include expected sexual consequences of their cancer treatments to improve their knowledge and set realistic expectations. “Yeah and would be much more comfortable just reading about it rather than asking about it. And then once they read about it and knew that yes, there was a lot of people had this problem, I think that they could then they could be more open to talking about it to the doctor.” They preferred receiving information close to diagnosis and intermittent check-ups afterward. “If the doctor brought it up early on, and then the doctor could ask maybe on the second or third visit.” Women differed in their preferences for patient-initiated versus provider-initiated conversations, suggesting the use of a brief survey to gauge comfort levels and tailor the approach accordingly. “I think do a little survey or something to see if they are interested.” There was no consensus on the preferred provider to discuss sexual problems. Notably, most women preferred not to include their spouses in the initial conversation. “I also think that it should be brought up maybe the first time when the partner is not present.” Conclusions: Women with MBC recommended normalizing the conversation about sexual problems with the availability of evidence-based self-review materials to be delivered close to diagnosis and intermittently afterward. Further work is needed to apply these recommendations in clinical settings.
| Themes | Key recommendations |
| Normalization of conversation | Normalization of discussion of sexual problems in the clinical setting will permit women to discuss their sexual problems |
| Self-review materials | Receiving self-review materials to improve their knowledge and set up expectations (flyers, pamphlets, or web pages) |
| Timing of conversation | Close to the diagnosis and intermittently |
| Inclusion of spouse | Not including spouse in the initial conversation |
| Preferred providers | No consensus |
| Patient vs provider-initiated conversation | No consensus |
Presentation numberPS1-02-30
Beyond breast cancer survival: quality of life aspects in women undergoing endocrine therapy
Mariana Carolina Vilas Boas Carvalho, AC CAMARGO CANCER CENTER, São Paulo, Brazil
M. C. Carvalho, I. d. Oliveira, B. C. Figueroa, T. C. Lima, R. P. Souza, S. M. Sanches, V. C. Lima, M. G. Cesca; Clinical Oncology, AC CAMARGO CANCER CENTER, São Paulo, BRAZIL.
Background: Endocrine therapy (ET) is the cornerstone of hormone-receptor positive (HR+) breast cancer treatment. However, it is potentially associated with several adverse events, which can impact patients’ quality of life (QoL). Depicting different spheres of QoL affected by ET is essential to improve supportive care strategies. This study aimed to evaluate QoL among breast cancer patients on mid/long-term ET, exploring its diverse domains. Methods: Single-center, observational cohort study. Female patients with HR+ breast cancer undergoing systemic ET for at least 6 months were included. Participants fulfilled a unique online questionnaire, including the validated European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) + Breast Cancer-specific Module (BR23). QoL was assessed across several functioning and symptom domains. Each score domain was transformed into a score ranging 0 to 100, according to EORTC guidelines. Higher scores indicated a better quality of life for functioning domains/general health status, and a greater symptom burden. Descriptive statistics were used to characterize the domains (mean scores) and the population characteristics. Correlations between the patient’s characteristics and the scores were evaluated with Student’s T test. Results: 200 patients, with a median age of 44 years-old (range: 27-92), were included. The majority had at least higher education (87%), were self-declared as white (69%; brown/black: 24.5%), heterosexual (95.5%), were premenopausal (74.5%), overweight (68%) and had children (69.5%). 14% were under ET for metastatic disease (86% adjuvant ET), 94.5% had primary breast cancer surgery (50.5% conservative) and 49% breast reconstructive surgery. Chemotherapy was used in 62.5% and the ongoing ETs were: SERMs (22.5%), aromatase inhibitors (70.5%) and SERDs (7%); 58.5% associated with ovarian function suppression. Global health status (71.4) and physical and role functioning (76.9 and 71.8) were relatively well preserved in this cohort. Financial difficulties (25.7), and symptoms related to surgery, such as breast (23.3) and arm (31.9) pain were also not impactful. However, patients had significant impairment in sexual functioning (27.4), future perspective (27.5), body image (56.6) and in emotional (51.3) and cognitive (52.9) functioning. Insomnia (52.0) and fatigue (40.1) were the most impactful symptoms. Premenopausal status was correlated to worse body image (52.9 vs 67.1; p=0.014) and emotional functioning (49.4 vs 56.9; p=0.006). There was a trend for worse body image in patients who underwent breast reconstructive surgery (46.9 vs 66.5; p=0.055), worse sexual functioning for aromatase inhibitors versus SERMs (25.9 vs 32.6; p=0.059) and worse global health status in premenopausal women (70.1 vs 75.2; p=0.058). Conclusion: This study highlights the impact of ET and breast cancer diagnosis in the sexual, cognitive and emotional functioning, as well in the future perspective and body image of HR+ breast cancer patients. Premenopausal women appear to be the most affected. These insights underscore the importance of routine QoL assessment in clinical practice, and the urgent need for improvement in survivorship care for breast cancer patients.
Presentation numberPS1-04-01
Misguided Relief: Prevalence and Patterns of Symptom Management Misinformation for Breast Cancer on Instagram
Jasmin Hundal, Cleveland Clinic, Cleveland, OH
J. Hundal1, A. Loomba2, M. Mody3, N. Lopetegui-Lia4; 1Hematology and Oncology, Cleveland Clinic, Cleveland, OH, 2Hematology and Oncology, Maharaja Agrasen Medical College, Agroha, INDIA, 3Medicine, M. P. Shah Government Medical College, Bharuch, INDIA, 4Department of Internal Medicine, Division of Medical Oncology, Ohio State University, Columbus, OH.
Background: Patients with breast cancer (BC) often experience a range of treatment-related symptoms that significantly impact quality of life. Social media platforms such as Instagram have become popular sources of health information, including guidance on symptom management. However, unregulated nature of these platforms enables the rapid dissemination of health-related misinformation, which can influence patient behavior and decision-making. Despite this growing concern, limited data exist on the scope and nature of symptom-related misinformation in the context of BC. This study aimed to evaluate the prevalence, characteristics, and engagement patterns of misinformation related to BC symptom management on Instagram.Methods:We conducted a cross-sectional content analysis of publicly available Instagram posts to evaluate the prevalence and nature of misinformation related to symptom management in BC. Searches using breast cancer-related and symptom-specific hashtags (e.g., #cancerfatigue) were conducted between June 17-28, 2025. To reduce algorithmic bias, searches were done in incognito mode. Posts were eligible if in English, publicly accessible, referenced symptom management in the context of BC, and made a therapeutic claim. Posts unrelated to cancer or lacking health-related content were excluded. For each symptom, the first 30-50 eligible posts were included.For each post, data were extracted on symptom category, misinformation classification (accurate, misleading, false, or unproven), perceived risk level (none to high), creator type, emotional tone, follower count, engagement (likes/comments), and presence of promotional links. Two independent coders reviewed each post. Descriptive statistics summarized misinformation prevalence and engagement patterns. Thematic analysis of captions identified narratives conveying misinformation.Results:A total of 328 Instagram posts referencing BC symptom management were included, categorized by symptom: fatigue (n = 46), brain fog (n = 45), hair loss (n = 46), nausea (n = 48), neuropathy (n = 51), cancer pain (n = 44), and insomnia (n = 48). Misleading content was the most prevalent, comprising 55.2% posts (n=181), while accurate information was infrequent, with only 22.8% posts (n=75). Thematic analysis revealed that approximately 30% of posts referenced evidence-based interventions, while 50% promoted non-evidence-based therapies, such as “magic juices,” Ayurvedic cures, and commercial programs claiming to eliminate treatment side effects. Additionally, 20% of posts offered oversimplified advice, such as using ginger for nausea or turmeric for recurrence prevention.The predominant emotional tones were neutral (42%, n = 138) and hopeful (40%, n = 132). Commercial intent was noted in more than half of the posts (53%,n= 174).Despite 64% (n=210) accounts having >1,000 followers, overall engagement was low, with 82% (n=269) of posts receiving <100 likes and <30 comments. Additionally, 48.4% of the creators were influencers, compared to healthcare professionals, who comprised only 15%. Conclusion: Among Instagram posts addressing BC symptom management, misinformation was highly prevalent, with over half the content categorized as misleading and fewer than one-quarter considered accurate. Commercial intent was common, and non-medical influencers, rather than healthcare professionals, created a significant proportion of posts. Despite many accounts having large followings, overall user engagement remained low. These findings highlight the risk of widespread exposure to low-quality health information and underscore the need for greater engagement by oncology professionals and institutions to promote accurate, evidence-based content on social media platforms.
Presentation numberPS1-04-02
Prospective assessment of upper extremity range of motion changes following mastectomy and breast reconstruction using a novel smartphone app
David B Lipps, University of Michigan, Ann Arbor, MI
D. B. Lipps1, K. Russell-Bertucci1, S. M. Cain2, P. L. Myers3, A. O. Momoh3, D. L. Hertz4; 1School of Kineisology, University of Michigan, Ann Arbor, MI, 2Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV, 3Plastic Surgery, University of Michigan, Ann Arbor, MI, 4College of Pharmacy, University of Michigan, Ann Arbor, MI.
Background: Upper extremity range of motion (UEROM) deficits are common after mastectomy and breast reconstruction, limiting patient recovery and quality of life. Many breast cancer patients report shoulder pain and reduced arm motion months to years post-surgery, including significant declines in abduction and flexion. However, access to frequent UEROM assessment in clinics is limited by time, equipment, and specialized expertise. A smartphone app could offer an accessible solution for at-home UEROM evaluation. We hypothesized that a smartphone application could detect reductions in shoulder abduction and flexion following breast reconstruction, yielding results comparable to those from wearable inertial measurement units (IMUs).Methods: Thirteen female breast cancer patients (mean age 45.5 years, range 35-54) completed at-home UEROM assessments both before and 6-12 weeks after mastectomy with immediate breast reconstruction. Participants performed the assessments simultaneously using an IMU sensor system (placed on sternum, right/left upper arms, right forearm, left forearm) and an iPhone application (MotionDetect module, CareEvolution, Ann Arbor, MI). The app provided standardized audio and video instructions, ensuring consistent arm positioning and movement. UEROM was defined as the maximum angle of abduction and flexion from the torso, measured both by the IMU and iPhone’s onboard sensors. Pairwise changes in pre- and post-surgery mobility were compared within and across both measuring methods using paired t-tests. Agreement between IMU and iPhone app measures was determined using Pearson correlations.Results: Both IMU and iPhone app measures detected significant declines in the maximum shoulder abduction and flexion following breast reconstruction. 30% of enrolled mastectomy patients exhibited clinically meaningful UEROM limitations exceeding 20 degrees post-surgery, consistent with prior studies. Specifically, the IMU detected significant decreases in abduction (p = 0.007) and flexion (p = 0.007), while the iPhone app detected significant reductions in flexion (p = 0.004) and a trend toward decreased abduction (p = 0.08). The pre-post difference in UEROM between the elevation angles derived by the IMU sensors and the iPhone app were statistically similar (both p > 0.15), with strong correlations between the IMUs and iPhone app measures in abduction (r = 0.92) and flexion ranges of motion (r = 0.96). Discussion: This study demonstrates that a smartphone app is able to detect and quantify upper extremity range of motion deficits following breast reconstruction surgery with comparable results to a wearable IMU sensor system. The app consistently identified significant post-operative decreases in shoulder mobility, suggesting it can serve as a convenient, accessible means for remote UEROM monitoring. These app-based assessments are far more scalable for use in clinical trials and practice than sensor-based assessments. The smartphone app’s ease of use may facilitate telehealth-based monitoring, enabling clinicians to intervene earlier and address mobility restrictions, which could potentially improve patient outcomes and quality of life.
Presentation numberPS1-04-03
“We’Re both dealing with the same thing, but on different sides of it” – understanding metastatic breast cancer through a dyadic lens: a qualitative assessment of patient and caregiver perspectives and experiences
Nicole MG Fleege, University of Iowa Health Care, Iowa City, IA
N. M. Fleege1, E. E. Chasco2, L. Soweid2, S. Houghton2, A. T. Seaman3; 1Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa Health Care, Iowa City, IA, 2Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA, 3Department of Internal Medicine, Division of General Internal Medicine, University of Iowa Health Care, Iowa City, IA.
Background: The prognosis for patients with metastatic breast cancer (MBC) has substantially improved due to advancement in therapies. With prolonged survival, patients’ caregivers often experience an extended duration of caregiving. However, little is known about the impact of a MBC diagnosis and treatment on caregivers, or on the caregiver-patient relationship over time. Using a dyadic approach, this study explored how patients with MBC and their caregivers perceive a MBC diagnosis and subsequent caregiving needs over time on an individual and dyadic level. Methods: Patient-caregiver dyads participated in semi-structured qualitative interviews from July 2024-March 2025. Informal caregiver was defined as an unpaid individual who provides at least 50% of a patient’s care needs. Patients and caregivers were interviewed separately. The interview guide included questions about patient and caregiver perspectives on and experiences with MBC diagnosis and treatment, unmet needs, and facilitators and barriers associated with caregiving. Interviews were audio-recorded and transcribed; transcripts were imported into the qualitative data management program MAXQDA and analyzed using an adapted framework method. Results: Eleven patient-caregiver dyads [consisting of opposite sex-spouse pairs (n = 7), same-sex spouse pair (n = 1), opposite-sex engaged pair (n =1), and daughter-mother pairs (n=2)] were interviewed. In relating participants’ experiences, four themes were developed: 1) contrast of a prior cancer diagnosis to current MBC experience, 2) concerns related to individual health after MBC diagnosis and its role in caregiving needs, 3) MBC as a shared diagnosis and experience, and 4) impact of MBC diagnosis on the dyadic relationship. Participants contrasted MBC with patients’ previous cancer diagnoses, which typically included a shorter, intense period of treatment followed by a return to normalcy. Conversely, for MBC, participants highlighted the uncertainty of the survival period, prolonged symptoms, and the realization that there would be no return to normalcy. Both patients and caregivers shared concerns about the impact of MBC on their individual health and its impact on the dyad. While patients focused on feelings of uncertainty regarding their own functioning, mortality and potential increase in caregiving needs over time, caregivers shared concerns about their health, age, and ability to provide a prolonged duration of caregiving. Caregivers often viewed MBC diagnosis/treatment as a shared experience, frequently using “we” or “our” in responses. An important topic was the impact of a MBC diagnosis and treatment on their shared activities as a dyad (i.e. socializing, traveling, and exercising together). Patients emphasized that caregivers needed time for hobbies and self-care, but caregivers were frequently concerned about leaving patients alone or traveling far. In terms of the dyadic relationship, both patients and caregivers described monitoring and managing the others’ emotions. Most participants agreed that the MBC diagnosis either hadn’t affected their relationship or had brought them closer. Conclusions: Caregiving changes clinical interpersonal dynamics by bringing an additional person into the clinical relationship. Given this, dyadic approaches are particularly useful as they provide insight into the unique perspectives and experiences of each dyad member and their influence on interpersonal processes. Understanding patient and caregiver perspectives and experiences of MBC diagnosis, treatment, and caregiving needs from both an individual and a dyadic perspective is essential to improving clinical care and quality of life for patients with MBC and their caregivers.
Presentation numberPS1-04-04
Quality of Life and Patient-reported outcomes in HR+/HER2- metastatic breast cancer: a systematic literature review
Amin Haiderali, Merck, North Wales, PA
A. Haiderali1, J. R. Earla2, J. Nathani3, P. Singh4, S. Sharma4; 1Value & Implementation, Merck, North Wales, PA, 2Value & Implementation, Merck, Rahway, NJ, 3HEOR, Parexel International, Bengaluru, INDIA, 4HEOR, Parexel International, Chandigarh, INDIA.
Objectives: This study focused on summarizing the quality of life (QoL), health state utility values, and patient-reported outcomes (PRO) associated with hormone receptor-positive (HR+) /human epidermal growth factor receptor 2-negative (HER2-) locally recurrent inoperable or metastatic breast cancer (BC). Methods: A PRISMA-compliant systematic literature review (SLR) was conducted to identify English language studies published from database inception till July 2024. Embase and MEDLINE databases were searched, along with recent conference proceedings (2021-2024) and bibliographies of relevant SLRs, to comprehensively identify the evidence. Results: Thirteen studies (sample sizes: 15-2,352; mean/median ages: 46.1-66 years; data collection period: 2010-2023) assessed QoL in HR+/HER2- metastatic BC patients across the Netherlands (1), Canada (2), Germany (1) and the USA (3) and multiple countries (6). These studies (9 cross-sectional, 4 longitudinal) primarily reported evidence for chemotherapy, hormonal, and targeted therapies. Patients with progressive disease (PD) health state had worse QoL compared to those in progression-free (PF) health state across most QoL/PRO scales. Lambert-Obry et al (2018) reported that in first-line (1L) and second and/or subsequent line (≥2L) settings, EQ-VAS (66.2 vs 76.8; 66.1 vs 71.7), EORTC QLQ-C30 Global Health Status (GHS) (52.9 vs 68.2; 54.0 vs 66.0), and EQ-5D utility scores (0.64 vs 0.73; 0.65 vs 0.74) were lower and BPI scores (3.1 vs 2.0; 2.6 vs 2.0) were higher for PD vs. PF patients. Lower scores on functional scales and EQ-VAS, and higher scores on symptom scales and BPI indicating worse QoL. QoL generally declined as patients progressed through treatment lines (Table 1). Functional scales (EORTC QLQ-C30 and QLQ-BR23) worsened, while symptom scales in both instruments increased, with fatigue significantly impacting PD vs. PF disease in 1L (43.1 vs 36.5) and 2L settings (45.2 vs 37.1) (Lambert-Obry 2018). FACT-G scores significantly declined in patients with visceral + bone metastases at 3- and 6-months vs baseline (-6.6 and -3.6, respectively, p<0.05) (De Laurentiis 2018). Lower scores were seen in patients with visceral + bone metastases (FACT-G: 57.7; FACT-B: 79.7) compared to those with bone-only (63.7; 88.7) or visceral-only metastases (60.9; 84.8) (Wood 2016). This impacted Physical Well-Being (FACT-G: 17.1 vs 19.1 vs 18.7), Emotional Well-Being (FACT-G: 12.0 vs 13.7 vs 13.1), and Breast Cancer Subscale domains (FACT-G: 22.2 vs 25.0 vs 23.7), in visceral + bone vs. bone only and visceral only groups (Wood 2016). The EQUALS survey reported fatigue (74-78%), joint pain (64%), and vaginal atrophy (56%) as the most impacted symptoms. 12-20% of patients reported poor or very poor QoL (Sammons 2023 and Sammons 2024). Conclusion: The present SLR on HR+/HER2- locally recurrent inoperable or metastatic BC revealed significant humanistic burden with declining QoL and increasing symptom burden through progressive treatment lines. These findings highlight the need for new treatments balancing efficacy and QoL, particularly for patients with limited treatment options.
Presentation numberPS1-04-05
Incidence of relationship breakups during endocrine therapy for breast cancer: preliminary results of the ReBreCa trial
Mariana Carolina Vilas Boas Carvalho, AC CAMARGO CANCER CENTER, São Paulo, Brazil
M. C. Carvalho, M. G. Cesca, I. d. Oliveira, B. C. Figueroa, T. C. Lima, S. M. Sanches, V. C. Lima, R. P. Souza; Clinical Oncology, AC CAMARGO CANCER CENTER, São Paulo, BRAZIL.
Background: Breast cancer and its treatment significantly impact patients’ interpersonal relationships, with lasting effects. Adverse events of anticancer therapies, including sexual dysfunction and body image changes, frequently negatively affect patients’ quality of life and relationships. This study aimed to evaluate the incidence of relationship breakups (RBr) during endocrine therapy (ET) for breast cancer and its associated factors. Methods: This is a single-center, retrospective, observational cohort study. Participants were female ≥18 years-old, with hormone-receptor positive (HR+) breast cancer (metastatic or non-metastatic, including in situ neoplasia) undergoing ET for at least 6 months. Data were collected through an online questionnaire, which included demographic, relationship and quality of life aspects, and electronic medical records. The patients who reported being in a relationship at ET initiation were included in the primary outcome analysis. The primary endpoint was the incidence of RBr among those in a relationship at ET initiation. Secondary endpoints included factors associated with RBr and motivations for maintaining relationships. Descriptive statistics were used for describing the population characteristics and the endpoints. Chi-square or Fisher’s exact tests were used for determining categorical variables association. Results: Of 201 patients who completed the questionnaire, 162 were in a relationship at ET initiation, and were included in the primary analysis. The median age at diagnosis was 44 years-old (range: 26-71), and the majority worked outside home (74.1%), had at least higher education (86.4%), had children (72.8%), were premenopausal (75.9%) and overweight (67.9%). 13.6% had HER2+ tumors, 14.8% metastatic disease, and 6.8% in situ carcinomas. The ETs in use were: SERMs (23.5%), aromatase inhibitors (70.4%), and SERDs (6.2%). 58% received ovarian function suppression and 24.7% a target-therapy (24.1% iCDK4/6). 95.1% were heterosexual, 3.1% homosexual and 1.9% bisexual. 72.2% were married. Sixty-two (38.3%) reported a RBr after at least 6 months of ET. Among those, 35.4% (n=22) attributed the RBr to cancer treatment in general, and 27.4% (n=17) to ET. The RBr decision was made by the patient in 48.1% (n=13), by the partner in 37% (n=10), and by both in 14.8% (n=4). Some contributing factors to RBr were low libido (27.0%), sexual dissatisfaction (19.0%), prejudiced body image (22.2%), and difficulties in communication (20.6%). Only axillary node dissection was associated with RBr in univariate analysis (52.3% vs 33.3% for sentinel lymph node dissection; p=0.04). There was a trend of association between nulliparity and RBr (50% vs 33.9%; p=0.07). Despite 55.6% of responders referring to a conflictual RBr, 74.1% of patients who ended their relationships expected a lasting relationship in the future. Considering the 100 women who maintained their relationships, only 14% were dissatisfied with them. The main contributing factor for maintaining the relationship was affection (73%), with children (8%), financial aspects (2%), religion (2%), fear of loneliness (2%) and others (3%) being less relevant. Conclusion: Our findings reveal a considerable incidence of RBr among breast cancer patients undergoing ET, with more than one third of patients directly attributing the ending of the relationship to treatment. Factors such as libido, sexual satisfaction, body image, and communication were reported as crucial in this context of relationship vulnerability. This study underscores the urgent need to integrate psychosocial assessments and to offer targeted support to manage the impacts of ET/treatment on patients’ relational lives, aiming to preserve well-being and quality of life during and after breast cancer treatment.
Presentation numberPS1-04-06
Integrating Mental Health in Oncology: A Review of Psychiatric Symptom Monitoring in Recent Breast Cancer Trials
Fatma Nihan Akkoc Mustafayev, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
V. Andion Camargo, F. Akkoc Mustafayev, M. Jaramillo, Z. Sarfraz, K. A. Qidwai, L. S. Spiegelman, M. S. Ahluwalia, R. L. Mahtani; Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Breast cancer (BC) patients face a lifelong disease burden that includes significant psychological stress. While clinical trials often focus on treatment efficacy and physical toxicities, psychiatric side effects remain underrecognized despite their potential to impact quality of life, adherence, and outcomes. This study evaluated the extent to which psychiatric symptoms were monitored and reported in Phase III clinical trials of FDA-approved BC treatments over the past decade. Methods: We reviewed the FDA archives to identify BC treatments approved over the past decade and found 21 such treatments. We conducted a search on clinicaltrials.gov to identify Phase III trials evaluating these FDA-approved interventions initiated after January 1, 2015, with posted results. The search yielded 38 clinical trials, comprising a total of 26,298 participants. We then examined each trial cohort for the presence of reported psychiatric symptoms. Trial protocols were reviewed in detail to identify the specific assessment tools and criteria for monitoring psychiatric symptoms, including documentation during treatment and post-treatment follow-up. All symptoms were recorded as adverse events for each trial per the NCI CTCAE guidelines. Most trials reported symptoms only during the treatment period. Descriptive statistics were used for data analysis. Results: Among the 38 phase III trials, 37 (97%) reported at least one psychiatric symptom. The most frequently reported symptom was insomnia, affecting 2563 participants (9.7%), while nervousness was the least reported, documented in only 1 participant. Other reported symptoms included anxiety, stress, depression, suicide attempts or completions, and confusional states. The most used assessment tool was EORTC-QLQ C30, followed by EQ-5D-5L, NCI-PRO-CTCAE, and FACT-B. Additional psychiatric symptoms, such as disorientation, apathy, panic attacks, stress, mania, psychiatric decompensation, mental disorders and sleep disorders, were identified but inconsistently assessed across trials. Most trials recorded psychiatric symptoms only during the treatment period, with limited documentation in long-term follow-up. Conclusion: While most trials reported at least one psychiatric symptom, monitoring was largely limited to short-term effects, highlighting the narrow scope of assessment. Inconsistent use of tools and limited follow-up suggest psychiatric symptoms remain underrecognized. This study is limited by reliance on publicly available data and potential reporting bias. A more holistic and longitudinal approach to psychiatric symptom monitoring can significantly improve patient safety to ensure treatment adherence without compromising their well-being.
| Symptoms | Trials (n= 37) | Participants (n=26,298) |
| Insomnia | 36 | 2563 (9.75%) |
| Anxiety | 20 | 524 (1.99%) |
| Depression | 21 | 484 (1.84%) |
| Confusional state | 12 | 23 (0.09%) |
| Suicide Attempts/Completions | 8 | 12 (0.05%) |
| Delirium | 3 | 6 (0.02%) |
| Mental status change | 5 | 5 (0.02%) |
| Bipolar disorder | 2 | 3 (0.01%) |
| Agitation | 2 | 2 (0.01%) |
| Nervousness | 1 | 1 (0.00%) |
Presentation numberPS1-04-07
Sleep, depressive symptoms and quality of life among women with newly diagnosed breast cancer: findings from the AMBER cohort study
Lin Yang, University of Calgary, Calgary, AB, Canada
L. Yang1, T. Tayyab2, R. Kokts-Porietis2, Q. Wang2, J. McNeil3, M. Matthews4, L. Dickau2, J. Vallance5, M. McNeely6, N. Culos-Reed7, K. Kopciuk2, K. Courneya8, C. Friedenreich1; 1Oncology, University of Calgary, Calgary, AB, CANADA, 2Cancer epidemiology and prevention research, Alberta Health Services, Calgary, AB, CANADA, 3Kinesiology, University of North Carolina at Greensboro, Greensboro, NC, 4Metabolic Epidemiology Branch, National Cancer Institute, Rockville, MD, 5Faculty of Health Disciplines, Athabasca University, Athabasca, AB, CANADA, 6Physical therapy, University of Alberta, Edmonton, AB, CANADA, 7Kinesiology, University of Calgary, Calgary, AB, CANADA, 8Faculty of Kinesiology, Sport, and Recreation, University of Alberta, Edmonton, AB, CANADA.
Background: Sleep problems are common among breast cancer survivors and are associated with poor quality of life. Yet, few studies have accounted for depression. This study examines whether depressive symptoms attenuate the association between sleep and quality of life in newly diagnosed breast cancer survivors. Methods: Women with newly diagnosed early-stage breast cancer were recruited between 2012-2019 in Alberta, Canada, and completed the Pittsburgh Sleep Quality Index (PSQI) to assess global sleep quality. Quality of life was measured using the Short Form Survey (SF-36 version-2) to assess physical and mental well-being. Depressive symptoms were measured using the Patient Health Questionnaire (PHQ-9). Multivariable linear regressions were used to estimate the association of sleep measures with physical and mental well-being. Depressive symptoms were evaluated as a potential confounder and effect modifier. Results: Among 1454 breast cancer survivors with available data, 43% reported poor global sleep quality and 10.5% reported clinically meaningful depressive symptoms. Poor sleep quality was associated with lower physical (β=-3.0, 95%CI: -3.8 to -2.3) and mental well-being (β=-5.7, 95%CI: -6.7 to -4.7). These associations were attenuated to (β=-2.3, 95%CI: -3.1 to 1.5) and (β=-1.2, 95%CI: -2.1 to -0.3) after further adjusting for depression symptoms. Stratified analyses showed a slightly stronger association in women with non-minimal symptoms, though this association was not clinically meaningful. Conclusion: The association between sleep and quality of life was substantially attenuated after accounting for depressive symptoms. Longitudinal data are needed to clarify the temporal relationship and determine whether sleep can serve as an early indicator of emerging depression. Next steps: Based on these findings, we are currently investigating depressive symptoms as a mediator for the association between sleep health and quality of life, using longitudinal data collected from time of diagnosis to five-years follow-up. We will be able to complete the analyses and report findings from the mediation analyses at the time of conference.
Presentation numberPS1-04-08
Five-year longitudinal sociodemographic and lifestyle trajectories among Mexican young women with breast cancer in the Joven & Fuerte cohort
Cynthia Villarreal-Garza, Tecnologico de Monterrey, San Pedro Garza Garcia, Mexico
F. P. Pons-Faudoa1, F. Mesa-Chavez1, A. Platas2, B. F. Vaca-Cartagena1, A. S. Ferrigno3, A. Fonseca4, M. Miaja4, M. Cruz-Ramos4, J. E. Bargallo-Rocha5, P. Cabrera-Galeana6, A. Mohar7, C. Villarreal-Garza1; 1Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, MEXICO, 2Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, MEXICO, 3Department of Medicine, Yale University School of Medicine, New Haven, CT, 4Joven & Fuerte, Programa para la Atencion e Investigacion para Pacientes Jovenes con Cancer de Mama, MILC, Mexico City, MEXICO, 5Joven & Fuerte, Programa para la Atencion e Investigacion para Pacientes Jovenes con Cancer de Mama, MILC, San Pedro Garza Garcia, MEXICO, 6Breast Medical Oncology Unit, Instituto Nacional de Cancerologia, Mexico City, MEXICO, 7Epidemiology Unit, Instituto Nacional de Cancerologia, Mexico City, MEXICO.
Background: Breast cancer (BC) diagnosis and treatment can significantly disrupt employment, reproductive, and health behaviors, particularly in young women, with lasting implications for quality of life. However, longitudinal data in Latin American populations remain scarce. This study aimed to characterize five-year sociodemographic and lifestyle trajectories among Mexican young women with BC (YWBC). Methods: Females aged ≤40 years diagnosed with stage I–III BC between 2015 and 2020 were enrolled in the Joven & Fuerte prospective multicenter cohort. Sociodemographic and lifestyle variables including occupational status, income, relationship status, fertility preferences and outcomes, and behaviors such as exercise, alcohol use, smoking, and spirituality were assessed using generalized estimating equations and multinomial logistic regression. Risk ratios (RR), relative risk ratios (RRR), and 95% confidence intervals (CI) were calculated relative to baseline. Data were collected at diagnosis, 6 months, 1 year, 2–3 years, and 4–5 years post-diagnosis. Due to the longitudinal design, missing data varied by variable and timepoint. Each analysis was calculated based on the number of respondents with available. Results: Among 526 participants (median age: 36 years; IQR: 32–38), 52.9% had education up to high school. At diagnosis, ~40% were employed full- or part-time. Employment dropped at 6 months (RR = 0.46; 95% CI: 0.41–0.51) and 1 year (RR = 0.54; 95% CI: 0.49–0.60), both p < 0.001. Occupational changes, reported by 70% at diagnosis, increased at 2–3 years (RR = 1.15; 95% CI: 1.05-1.26; p = 0.003) and 4–5 years (RR = 1.23; 95% CI: 1.12-1.34; p < 0.001). Income reductions affected 67% at diagnosis and persisted, though likelihood declined at 4–5 years (RR = 0.74; 95% CI: 0.62–0.89; p = 0.001), suggesting partial financial recovery. Relationship status was stable: 78% had a partner at diagnosis; 75% stayed with the same partner at 6 months, and 68% at 4–5 years. Compared to this group, being with a different partner at 1 year (RRR = 0.24; 95% CI: 0.10–0.56; p < 0.001) or starting a new relationship at 2–3 years after being single (RRR = 0.31; 95% CI: 0.14–0.72; p = 0.006) was less likely. Fertility preferences declined: 39% desired biological children at diagnosis vs. 20% at 4–5 years (RR = 0.40; 95% CI: 0.17-0.95; p = 0.038). Fewer than 10 live births occurred, and most patients (95-99%) reported no attempts to conceive, with no significant change over time. Lifestyle behaviors improved during the study period. Physical inactivity dropped from 57% at diagnosis to 44% at 4–5 years. At follow-up participants were more likely to engage in <150 min/week (RRR = 1.80; 95% CI: 1.14-2.84; p = 0.01) or ≥150 min/week (RRR = 1.64; 95% CI: 1.07-2.51; p = 0.02). Alcohol intake declined: 2% consumed >20 drinks/week at diagnosis; nearly none by 4–5 years (p < 0.001). Less than 20 drinks/week intake also dropped at 6 months and 1 year (p < 0.001). Smoking (4-7%) and spirituality (87-93%) showed no significant changes over time. Conclusion: Employment and income disruptions were most pronounced during early post-diagnosis, with only partial recovery observed over time. These findings highlight the need for survivorship plans that incorporate financial support and job reintegration opportunities to help YWBC regain occupational stability. Fertility preferences and attempts to conceive declined during follow-up, while relationship status remained largely stable. Encouragingly, physical activity improved during survivorship. Collectively, these results emphasize the importance of longitudinal support strategies that address the socioeconomic, social, and reproductive health needs of YWBC.
Presentation numberPS1-04-09
Complexities of Weight Change in Breast Cancer Survivors: A Report from the Black Women’s Health Study
Vanessa B Sheppard, Virginia Commonwealth University, Richmond, VA
V. B. Sheppard1, D. Chiranjeev2, M. C. Edmonds1, A. M. Kumar1, L. L. Adams-Campbell2; 1Department of Social and Behavioral Sciences, Virginia Commonwealth University, Richmond, VA, 2Office of Minority Health & Health Disparities Research, Georgetown University, Washington DC, DC.
Background: Weight changes during treatment and survivorship impact health outcomes in breast cancer survivors. These treatment-related changes include both substantial weight gain and loss. Many patients gain 5-15 lbs during treatment due to reduced physical activity, chemotherapy, hormonal therapy, and corticosteroids. Conversely, some experience weight loss related to treatment side effects or cancer cachexia.Our prior work suggests that women undergoing chemotherapy may shift their weight to a higher weight category during active treatment. Up to 70% of Black breast cancer patients have reported weight gain during survivorship. Weight gain increases cardiovascular risk, which is concerning given the elevated cardiac risk Black BCS face from therapies like doxorubicin and radiation. Heart disease is a leading cause of death among BCS and is most common in Black women. Emerging data also suggest that post-diagnosis weight gain may increase recurrence and lower survival, particularly in hormone receptor-positive cancers—types in which Black women experience the highest recurrence rates. Up to 50-70% of Black breast cancer survivors (BCS) experience weight changes during treatment and survivorship.Weight fluctuations may worsen insulin resistance and increase diabetes risk, which could affect cancer outcomes. Aromatase inhibitors can intensify these metabolic effects. Previous studies on weight change during treatment often lacked sufficient power to focus on Black women specifically, excluded cancer-related endpoints, or had limited data on lifestyle factors. This study aims to fill these gaps using detailed pre-diagnosis data and long-term follow-up of weight, lifestyle, and psychosocial factors in Black BCS. Methods: We examined 1,665 incident breast cancer cases (1995-2015) from the Black Women’s Health Study (BWHS), a nationwide prospective cohort of 59,000 Black women. Mortality and cancer details were obtained through medical record review. Post-treatment weight change was defined as the percentage difference between weight reported near diagnosis and at the 2-year follow-up. A change exceeding 10% from diagnosis weight was considered clinically significant. Survival analysis (Kaplan-Meier, Cox proportional hazards) was used to evaluate the relationship between weight change and all-cause mortality, controlling for age at diagnosis, education, stage, ER status, comorbidities, menopausal status, physical activity, and BMI. Women with metastatic disease at diagnosis were excluded.Results: Among 1,248 eligible women, 82% had no significant weight change (±10%) by 2 years post-diagnosis. Ten percent had significant weight gain (≥10%) and 8% experienced significant loss. There were 248 deaths during follow-up. Risk of death was higher among women with >10% weight change. Significant weight loss was associated with higher mortality (adjusted hazard ratio [HR]: 1.65; p=0.02), but weight gain was not (HR: 1.24; p=0.26). However, among women receiving both chemotherapy and hormone therapy, weight gain was linked to higher mortality (HR: 3.44; p=0.02).Conclusion:Nearly half of Black breast cancer survivors experience weight changes that start during treatment and continue afterward. Monitoring weight fluctuations is crucial in survivorship care to improve health and cancer outcomes. Weight loss may indicate more advanced disease, while weight gain could increase cardiometabolic risks. Including dietary data in future research might help better understand modifiable factors behind these patterns. The link between higher weight gain and increased mortality among women undergoing both chemotherapy and hormonal therapy highlights the need for comprehensive interventions for this group.
Presentation numberPS1-04-10
Closer to Care, Closer to Help: Emotional Support Use in Breast Cancer Patients by Symptom Burden and Travel Time
Asiyah Allibhai, Holy Trinity School, Richmond Hill, ON, Canada
A. Allibhai, A. Allibhai; Student, Holy Trinity School, Richmond Hill, ON, CANADA.
Background: Breast cancer is a leading cause of cancer-related morbidity and mortality among women and is frequently associated with significant psychological distress, particularly around diagnosis, treatment, and survivorship transitions. Although up to 50% of patients experience clinically significant symptoms of anxiety or depression, mental health and emotional support services remain underutilized. This study aimed to evaluate the utilization of institutional psychosocial support services and identify factors associated with their use among breast cancer patients reporting moderate to severe psychological symptoms. Methods: We conducted a cross-sectional analysis using data from an electronic medical record-linked patient-reported outcomes registry at a single institution. Adult breast cancer patients who received care within the past year and reported moderate to severe psychological symptoms—defined as a score ≥4 on the Edmonton Symptom Assessment Scale (ESAS) for depression or anxiety—were included. Emotional support utilization was defined as having a documented referral to psychiatry, social work, or support groups. Patient demographic, clinical, and geographic factors were compared between users and non-users of psychosocial services using chi-square or two-proportion z-tests. Results: Among 340 eligible patients, only 118 (35.1%) utilized institutional emotional support services. Utilization was significantly higher among patients with greater symptom burden (ESAS 7-10: 63.3% vs. ESAS 4-6: 23.1%; p60 minutes (12.5%) (p<0.001). Trends toward higher utilization were observed among younger patients (<50 years: 41.3%) and those identifying as white (40.8%), although these differences did not reach statistical significance. No significant associations were found for gender or cancer stage. Conclusion: Despite strong evidence supporting the benefits of psychosocial interventions in breast cancer care, institutional support services remain underutilized, especially among patients with lower symptom burden or longer travel times. Accessibility and perceived need appear to be key drivers of engagement. Addressing geographic and systemic barriers, increasing provider referrals, and assessing pre-existing support systems may improve equitable access to mental health care for breast cancer patients.
| Yes | No | p-value | |||||
| Gender | |||||||
| Female | 118 (35.0%) | 222 (65.5%) | 0.51 | ||||
| Male | 0 (0%) | 4 (100%) | |||||
| Age | |||||||
| <50 | 31 (41.3%) | 44 (58.7%) | 0.068 | ||||
| 50-70 | 59 (37.3%) | 99 (62.7%) | |||||
| >70 | 28 (26.2%) | 79 (73.8%) | |||||
| Race | |||||||
| White | 75 (40.8%) | 117 (63.6%) | 0.229 | ||||
| East Asian | 16 (28.6%) | 39 (69.6%) | |||||
| South Asian | 13 (29.5%) | 28 (63.6%) | |||||
| Middle Eastern | 6 (25%) | 16 (66.7%) | |||||
| Afro-Caribbean | 5 (27.8%) | 12 (66.7%) | |||||
| Latin American | 3 (21.4%) | 10 (71%) | |||||
| Stage | |||||||
| 1-2 | 95 (36.0%) | 164 (62.1%) | 0.432 | ||||
| 3-4 | 23 (30.3%) | 58 (66.0%) | |||||
| Travel Time To Clinic | |||||||
| < 30 mins | 94 (43.5%) | 122 (56.5%) | <0.001 | ||||
| 30-60 mins | 20 (21.7%) | 72 (78.3%) | |||||
| >60 mins | 4 (12.5%) | 28 (87.5%) | |||||
| Highest ESAS for Anxiety or Depression | |||||||
| 4-6 | 56 (23.1%) | 186 (76.9%) | <0.001 | ||||
| 7-10 | 62 (63.3%) | 36 (26.7%) |
Presentation numberPS1-04-11
From Prediction to Prevention: The TIDE Score and a Navigator-Based Intervention to Reduce Delayed Breast Cancer Treatment in Southeast Texas
Nency Ganatra, Baptist Hospitals of Southeast Texas, Beaumont, TX
N. Ganatra, A. Jamal, J. Doshi, P. Jain, S. Patel; Internal Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX.
Background:Delays in breast cancer treatment initiation are associated with worse outcomes, particularly among underserved populations. While socioeconomic disparities are well documented, there is no standardized tool to identify patients at high risk for treatment delays. We aimed to (1) develop a predictive model to stratify delay risk and (2) design a scalable quality improvement (QI) intervention targeting high-risk patients in a community oncology setting. Methods:We analyzed data from 609 breast cancer patients treated between 2018 and 2023 across a multi-hospital network in Southeast Texas. Treatment delays were defined using established modality-specific thresholds. Logistic regression identified predictors of delay, and a point-based risk score—the TIDE Score (Treatment Initiation Delay Estimator)—was developed using significant socioeconomic and clinical variables. Based on this score, we proposed a QI model incorporating early navigator engagement, social work referral, and expedited care pathways. Results:The final predictive model demonstrated good performance (AUC = 0.81). The TIDE Score ranged from 0 to 6 and included Medicaid insurance (2 points), unmarried status (1 point), age under 50 years (1 point), AJCC stage ≥ II (1 point), and absence of Medicare coverage (1 point). Patients scoring ≥4 had 3.6-fold increased odds of treatment delay (p < 0.001). Using this risk stratification, we designed a navigator-led QI intervention including EMR-based alerts at diagnosis, navigator contact within 5 days, and support service linkage within 2 weeks. This dual approach bridges risk prediction with timely intervention. Conclusion:The TIDE Score is a simple, SES-informed tool to identify breast cancer patients at risk of treatment delay. Coupled with a navigator-driven workflow, it offers a low-cost, scalable solution applicable across Texas and other underserved oncology settings.
Presentation numberPS1-04-12
Access to Psychological Support and Psychotherapy for Women with Advanced Breast Cancer in Nigeria. A National Study.
Runcie CW Chidebe, Project PINK BLUE – Health & Psychological Trust Centre, Abuja, Nigeria
R. C. Chidebe1, S. Ipiankama1, M. C. Onyedibe2, I. I. Okoye3, S. Aruah4, O. Asogwa5, O. Ahmadu6, A. Adewumi7, L. Eriba8, D. Esiaka9, B. Olusegun10, M. Nandul11, I. Zubairu12, N. Mampak13, K. Omenukor14, D. Obi15, E. Ugwu1, H. Dogo16, C. Okwuonu17, J. Chukwuorji18; 1Patients Care and Research, Project PINK BLUE – Health & Psychological Trust Centre, Abuja, NIGERIA, 2Department of Psychology, University of Nigeria Nsukka, Enugu, NIGERIA, 3UNN Centre for Clinical Trials, University of Nigeria Teaching Hospital, Enugu, NIGERIA, 4Department of Radiotherapy and Oncology, National Hospital Abuja, FCT, NIGERIA, 5Medical Physics Department, Marcelle Ruth Cancer Centre, Lagos State, NIGERIA, 6Oncology Department, Federal Medical Centre, Abuja, NIGERIA, 7Oncology Department, NSIA-LUTH, Lagos, NIGERIA, 8Oncology and Radiotherapy Department, University of Benin Teaching Hospital, Edo State, NIGERIA, 9Center for Health Equity Transformation, University of Kentucky College of Medicine, Kentucky, KY, 10Oncology Department, University of Port-Harcourt Teaching Hospital, Rivers State, NIGERIA, 11Oncology Department, Jos University Teaching Hospital, Plateau State, NIGERIA, 12Oncology Department, Ahmadu Bello University Teaching Hospital, Kaduna State, NIGERIA, 13Oncology Nursing, National Hospital Abuja School of Post-Basic Oncology Nursing, FCT, NIGERIA, 14Patients Care and Research, David Omenukor Foundation, Dallas, Texas, TX, 15Department of Community Medicine and Primary Healthcare, Nnamdi Azikiwe University Teaching Hospital, Anambra State, NIGERIA, 16Urological Surgery, University of Maiduguri Teaching Hospital, Maiduguri, NIGERIA, 17Family Care, Federal Medical Centre, Abia State, NIGERIA, 18Department of Psychology, University of Nigeria Nsukka, Enugu State, NIGERIA.
Background: Advanced breast cancer (ABC) affects patients’ psychological well-being and quality of life, with a higher prevalence of psychiatric disorders such as anxiety, depression, sleep disorders, and post-traumatic stress disorder. Despite evidence underscoring the importance of psychological support and psychotherapy in improving patients’ well-being, access to these services remains unknown in this population. This study examined access to psychological support and psychotherapy for patients with ABC in Nigeria. Methods: Of the (N=1,389) cancer patients recruited in the national study, (n=208) patients with ABC (203 females and five males; mean age = 50.53 years, SD = 11.36) were included in this study. Participants completed a questionnaire evaluating their access to psychological support. The data were analyzed using descriptive statistics in SPSS. Results: The findings showed that 75.5% of participants received their cancer diagnosis in a comforting and humane manner, while 53.8% were offered psychological counseling immediately after diagnosis. However, only 13.5% received counseling from a qualified clinical psychologist on the day of diagnosis. Cancer diagnosis had a significant psychological impact on 83.7% of participants, yet only 44.2% received psychological support during treatment, leaving 55.8% without access. Notably, 65.9% expressed a need for psychological support and psychotherapy during their treatment, and 78.8% indicated a willingness to utilize such services if available. Conclusions: The study reveals critical gaps in the availability and accessibility of psychological support and psychotherapy for patients with ABC. Establishing psycho-oncology clinics and recruiting clinical psychologists to provide psychotherapy for patients could help bridge this gap and enhance mental well-being.
Presentation numberPS1-04-13
Expanding Access to the Advanced Breast Cancer Clinic: A Patient-Centered Screening Strategy and Characterization of Patients Served
Ginny Kirklin, The University of Texas MD Anderson Cancer Center, Houston, TX
G. Kirklin, A. Anderson, A. J. Kaler, M. Butaud, A. Singareeka, J. Harris, T. Jacobsen, B. Lim; Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Significance and Background: The diagnosis of advanced breast cancer (ABC) requires greater support and resource utilization compared to early-stage breast cancer. MD Anderson Cancer Center’s ABC Clinic is staffed with Advanced Practice Registered Nurses (APRNs) who provide personalized care through complex medical system, provide education about diagnosis/ treatment, assist with clinical trial process, explain genetic/genomic testing, discuss goals of care, and offer emotional support. In 2025, the ABC Clinic evaluated a new screening process for ABC patients seen in breast center to determine the fit for an ABC Clinic video visit, with the aim of increasing the number served. Purpose: To evaluate the impact of the screening process on access to the ABC Clinic by comparing the number and characteristics of patients served in 2025 versus 2024. Methods: Data were collected during visits between late March and June in 2024 and 2025. In 2024, patients were referred by a provider or self-referral. In 2025, ABC Clinic APRNs screened patients scheduled in breast oncology clinics. APRNs identified ABC patients who were Texas residents (due to licensing regulations), in active treatment, haven’t had a visit in the ABC Clinic in >365 days, and excluded patients who were going to hospice care. The oncology team would approve which patients to contact for an ABC Clinic video visit. Descriptive statistics were used for analysis. Findings: The new screening process increased provider referrals by 986% and patients with completed visits by 138% compared to 2024. In 2025, 188 unique patients were seen in 235 visits. Over half (54%) were new to the ABC Clinic, female (n= 186) with a diagnosis of ABC, and self-identified as White (68%) and non-Hispanic (79%). Most patients (86%) were diagnosed one to five years ago, married (62%), retired (32%), and had an ECOG status of 0 or 1 (89%). There were 56 total outgoing referrals to 15 different clinics or services with 30% of patients having at least one internal referral. Concerns identified by ABC patients: education (81%), coordination of care (46%), emotional concerns (42%), and physical concerns (40%). In 98% of visits, 40-60 minutes focused on care-related challenges. Patients seen in 2025 were more often be a new patient to the ABC Clinic (54% vs 22%), Hispanic (18% vs 12%), aged ≥ 70 years (20% vs 13%), have a ECOG score of 0 (34 vs 44%), and have a new ABC diagnosis (8% vs 6%). Patients seen in 2025 were more likely to present with knowledge deficit (81% vs 74%) and physical symptoms (40% vs 10%) compared to patients seen in 2024. Patients were more likely to discuss treatment related goals of care (38% vs 26%), have an internal referral (24% vs 20%), and the departments/services referred to more than doubled (15 vs 7) compared to referrals in 2024. Discussion and Implications: ABC patients presented with knowledge deficit and physical and emotional symptoms necessitating additional time at visits to assess and address concerns. An APRN-led visit allows for time to address education, physical symptoms, and goals of care. The screening process significantly increased referrals and new patients to the ABC Clinic and changed outgoing referral patterns and increased number of new patients. Future efforts will focus on outcomes for first time patients and long-term impact of the screening implementation.
Presentation numberPS1-04-14
A Service Evaluation Audit of a Holistic Health Follow Up Clinic for Early Stage Breast Cancer Patients
John R Benson, Addenbrooke’s Hospital, Cambridge, United Kingdom
R. Thomas1, C. Richardson2, C. Smith3, J. R. Benson2; 1School of Clinical Medicine, University of Cambridge, Cambridge, UNITED KINGDOM, 2Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge, UNITED KINGDOM, 3Patient Experience Team, Addenbrooke’s Hospital, Cambridge, UNITED KINGDOM.
Background: The Holistic Health Follow Up Breast Cancer Clinic provides a consultation service for early stage breast cancer patients that is available towards the end of their treatment pathway when all modalities of therapy have been completed. These clinics are run by specialist breast care nurses and assess a patient’s knowledge and understanding of treatments to-date. A plan for subsequent follow up and support is proposed and coordinated that aligns with current research suggesting the importance of a broad and multifaceted approach that encompasses various health domains including physical, emotional and social needs after treatment. Methods: A service evaluation audit was conducted to gauge the effectiveness of the holistic clinic in supporting patient knowledge and confidence in three key areas: self-examination, planning of follow up imaging and knowledge of treatments received and potential adverse effects that might impact on quality-of-life. Patients were further probed in terms of understanding different types of treatment and why these were recommended. A questionnaire was designed to assess the aforementioned areas using a combination of ordinal rating scales and free-text responses. Patients were allowed two weeks for completion and return of the questionnaire without any reminders being issued. Results were analyzed with graphic representation of numerical data and assessment of patient confidence in each designated area. Additional themes were extracted from the free-text responses to identify strengths and limitations of the holistic clinic assessment, thereby offering insight into further development of the service and improving patient care. Results: The questionnaire was issued to 206 patients with a response rate of 30% (61/206). More than 80% of patients reported improved understanding of follow up imaging whilst over 90% of patients considered their understanding of treatment and side-effects to be improved following a holistic consultation. By contrast, only 57% of responders either fully or partly declared any improved confidence in performing self-examination. Amongst the various treatment modalities, the proportion of patients who fully understood reasons for receiving surgery, radiotherapy and chemotherapy were 98%, 79% and 64% respectively. Moreover, only half (50%) understood the rationale for extended hormonal therapy and fewer than half (41%) fully appreciated the adverse impact of ovarian suppression. Free-text responses revealed some uncertainty about the exact purpose of the holistic clinic with requests for more emphasis on practical support in areas such as mental health and lymphedema and tailoring of support literature based on personal need. Conclusion: The holistic clinic achieves its overall aims with more than three-quarters of patients reporting improved understanding of treatments received, treatment-related side-effects and follow up imaging strategies. Areas that were less well addressed included hormonal therapies and breast self-examination where more thorough explanation, adjustments of information provision and better signposting of support services may be indicated.
Presentation numberPS1-04-15
A focused evaluation of young adults living with metastatic breast cancer: Patient-reported strategies to alleviate disease symptoms, treatment-related side effects, and survivorship concerns
Kate E Dibble, Dana-Farber Cancer Institute, Boston, MA
K. E. Dibble1, J. Stal1, S. M. Rosenberg2, C. Snow1, A. H. Partridge1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Population Health Sciences, Weill Cornell Medicine, New York, NY.
Background. Young women living with metastatic breast cancer (MBC) experience many physical and psychosocial difficulties heightened by a disrupted life trajectory and the burden of ongoing treatment. We conducted a focused evaluation of how survivors with MBC alleviate their symptoms/treatment-related side effects and survivorship concerns. Methods. Women diagnosed with MBC (18-39 years) participating in an ongoing prospective intervention study (Young, Empowered and Strong; NCT04379414) at Dana-Farber Cancer Institute were invited to participate in a semi-structured virtual interview. Descriptive statistics were used to describe the sample and quantify frequency of side effects, concerns, alleviation techniques, and perceived efficacy. We conducted a thematic analysis to further explore these topics. Results. The convenience sample comprised 20 women with a median age of 40 years at enrollment (SD=3.84, range: 30-46) and 34 years at MBC diagnosis (SD=3.03, range: 28-38). At time of interview, median time from initial breast cancer to MBC diagnosis was 2.14 years (SD=2.56, range: 0-9.51). Most were non-Hispanic (85%), white (80%); half had a graduate degree (50%). Symptoms/side effects: Overall, all women reported experiencing disease symptoms/treatment-related side effects. Fatigue (50%), gastrointestinal issues (35%), menopausal symptoms (25%), and nausea (25%) were most frequently reported. Of the 19 women who reported efforts to alleviate symptoms/side effects of MBC and its treatment, 18 (94.7%) tried more than two methods, and 7 (38.8%) tried more than four methods, including prescription medication, exercise, diet/hydration, over-the-counter medication, sleep, acupuncture/massage, and medical marijuana. Of these, participants reported utilizing several methods to alleviate symptoms/side effects in tandem with other methods. Concerns: All but one participant reported survivorship concerns including fear of disease progression (68.4%), mental health (52.6%), financial toxicity (36.8%), future of family/children (36.8%), feeling trapped (15.8%), fertility/childbearing (15.8%), and sexual health (15.8%). The most common methods to alleviate concerns were social support networks (53.3%), talk therapy (46.7%), staying busy (33.3%), and exercise (26.7%). Of the 15 women who tried more than one method to alleviate their concerns, all tried more than two methods and 10 more than four methods. Of these, most reported utilizing multiple methods because previous methods had not worked for them or updated treatments introduced new side effects. Conclusion. Young women with MBC have a high burden of symptoms/side effects and concerns and utilize numerous strategies to alleviate them. Multiple approaches were often employed, both because symptoms/side effects and concerns changed with new treatments and because methods were ineffective. Supporting women to streamline these efforts may address symptoms/side effects and concerns and may help to manage disease expectations and optimize overall well-being during MBC survivorship.
Presentation numberPS1-04-16
Reducing Stress and Improving Quality of Life for Adult Breast Cancer Patients
Danna Remen, Ellie Fund, Needham, MA
D. Remen, International Health Strategies; Strategic Initiatives, Ellie Fund, Needham, MA.
BackgroundÏEllie Fund provides essential support services for breast cancer patients to ease the stresses of everyday life, allowing the focus to be on family, recovery, and healing.ÏIn 2019, Ellie Fund launched an Outcomes Measurement Program to empirically measure the impact of its non-medical services on patients’ physical, emotional, and financial health and show improved quality of life (QOL) during treatment.ÏThis research is an essential continuation of the study conducted in 2024 and presented at SABCS, incorporating a broader dataset and deeper analysis of demographics (e.g., race, income), and examining how these factors correlate with stress reduction after receiving Ellie Fund services.ÏLonger term, this research aims to link support services to improved breast cancer outcomes. MethodsÏEach patient participant selected two services, such as grocery gift cards, delivered meals, transportation to medical appointments, childcare reimbursement, housekeeping and integrative therapies. Services were provided monthly for 3 months for non-metastatic patients (Stages 0-3) and 6 months for metastatic patients (Stage 4).ÏData were collected via voluntary surveys administered before and after service delivery, using the Perceived Stress Scale (PSS-14) which includes 14 standard questions scored numerically. Twelve additional questions assessed QOL changes using a 4-point Likert scale.ÏThis analysis focused on surveys from patients who started services 2019-2025 and completed both surveys. The McNemar test was used to measure the association between pre- and post-service stress improvements (primary outcome) and evaluate disease type in relation to perceived stress (secondary outcome). A Cochran-Mantel-Haenszel test was used to measure the association between stress and race, controlling for income (secondary outcome).ResultsA total of 1,047 patients enrolled in the program and completed at least one survey. As participation in survey items was not mandatory, sample sizes varied by question. Analyses were conducted only on cases with both pre- and post-survey responses.Primary outcomes highlighted the relationship between service use and patient-reported outcomes:ÏA statistically significant reduction in the proportion of participants reporting high perceived stress following receipt of Ellie Fund services (p < 0.05).ÏStatistically significant decreases were seen in emotional, physical, financial, and daily life stress levels (p < 0.05).ÏStatistically significant QOL improvements were reported in 6 key domains, namely (1) maintaining order in the home, (2) managing treatment side effects, (3) physical self-care, (4) food access, (5) focus on family, and (6) medication access.For secondary outcomes, among non-White patients, higher income is associated with 3.4x higher odds of stress reduction compared to lower income (statistically significant). For White patients, the association is weak (not statistically significant). Both patients with and without metastatic disease experienced significant improvements in their perceived stress pre- and post-services (p<0.05). In the non-metastatic group, stress was reduced about 67%, slightly higher compared with the metastatic group (60%). ConclusionEllie Fund’s services are associated with meaningful reductions in stress and improvements in various aspects of QOL among breast cancer patients, especially for higher-income non-White patients. Data suggest that below a certain income, more robust interventions may be needed. These findings underscore the value of supportive care to complement medical cancer treatment and also suggest a vital role for non-medical services in preventative breast care, and to shorten the time between diagnosis and initiating treatment. Further research is warranted to better understand the forces driving QOL improvements.
Presentation numberPS1-04-17
Real-world patient (pt) and caregiver experiences with breast cancer (BC) risk of recurrence (ROR) in the US: Results of an Online Survey and Social Media Analysis
Hope S. Rugo, City of Hope Medical Centre, Duarte, CA
H. S. Rugo1, M. Chase2, M. Rutt3, M. Smith4, S. Weldon5, E. Pain6, G. Vadnerkar7, T. Sen8, S. Padhi8, N. Nadimpalli8, L. Santarsiero7, C. Auld9, N. Harbeck10, A. B. Chagpar11; 1Duarte Cancer Center, City of Hope Medical Centre, Duarte, CA, 2n/a, Life on the Cancer Train, Atlanta, GA, 3n/a, Living Beyond Breast Cancer, Philadelphia, PA, 4n/a, Research Advocacy Network, Plano, TX, 5n/a, Unite for HER, Malvern, PA, 6Online Patient Community, Carenity, Paris, FRANCE, 7n/a, Novartis Pharmaceuticals Corporation, East Hanover, NJ, 8n/a, Novartis Healthcare Ltd, Hyderabad, INDIA, 9US Medical Affairs, Novartis Pharmaceuticals Corporation, East Hanover, NJ, 10Department of Obstetrics and Gynecology, LMU University Hospital, Breast Center, Munich, GERMANY, 11Department of Surgery, Yale University School of Medicine, New Haven, CT.
Background: While most new HR+/HER2− BC cases are diagnosed (dx) as early BC (EBC), recurrences are an urgent concern as approximately 50% of recurrences occur in ≤5 years despite standard-of-care endocrine therapy and this risk can persist for decades after dx. Little has been published on the awareness and understanding of BC ROR from the pt and caregiver perspective and how this may influence shared decision-making and quality of life (QoL). Here, we present the results of an online survey of pts and subsequent social media analysis (SMA) of pts and caregivers including important insights on ROR. Methods: Female pts from France, Germany, Italy, the UK, and the US were invited to participate in a survey developed with input from medical experts and pt advocates on the Carenity platform, an online pt community, between Dec 1, 2021 and Jan 24, 2022; results from the US are presented. Additionally, deidentified data from publicly available posts by US pts and caregivers on social media sites, forums, and blogs between Apr 1, 2023 and Mar 31, 2025 were analyzed using the Sprinklr social media aggregator. Results: Of 220 US pts who responded to the Carenity survey, 57 (26%) reported being dx with HR+/HER2− BC, 39% with HER2+, and 22% with triple negative BC (TNBC). Among pts with HR+/HER2− BC, 38 pts (67%) were initially dx with EBC (stage I-III); of those, 11 pts (29%) recurred as metastatic BC (MBC). In the SMA, a screen of 3440 posts found 1622 relevant to EBC and ROR; 82% of conversations were driven by pts. In the Carenity survey, many pts initially dx with HR+/HER2− EBC wished they had received more relevant information on chances/duration of survival (32%) or emotional support options (26%) from their healthcare provider (HCP) at the time of dx, some of whom indicated they received no information at dx (chances/duration of survival: 22%; emotional support options: 67%). Many pts who reflected back on initial dx and wanted but had not received information on chances/duration of survival (75%) or emotional support options (40%) experienced a metastatic recurrence. This was reinforced by the SMA which identified recurrence rate and peer support as topics pts were seeking more information on. Posts related to ROR highlight pt and caregiver concerns about late recurrences, limited long-term monitoring, and persistent worries about unexpected recurrences. Pts also expressed feelings of being overwhelmed by the fear of BC recurrence. Within the Carenity survey, limiting risk of cancer metastasis and living as long as possible ranked high among treatment (tx) expectations. Similar themes emerged from the SMA, in which ROR was identified as one of the top aspects leading to positive perceptions of available tx. Surveyed pts with HR+/HER2− BC felt they had less access to services and support programs compared to pts with HER2+ or TNBC. In the SMA, pts discussed challenges and stressors associated with the lack of ROR knowledge within their support systems. Conclusions: Pts prioritize reducing ROR when ranking their tx options yet lack sufficient information and understanding about ROR. This is especially important for those with HR+/HER2− EBC whose risk includes early and late recurrence, a majority of which are metastatic recurrences. Conversations between HCPs and pts can be complex, further complicated by the number of HCPs communicating ROR to a single pt, varying degrees of pt health literacy, and impact to pt QoL. These analyses shed light on the need for greater knowledge and education about ROR, while also ensuring pts are offered support services to help address the impact of ROR on QoL for both pts and loved ones, from initial dx, throughout tx, and beyond.
Presentation numberPS1-04-18
Identifying Smoking-Related Factors in Breast Cancer Patients: Mayo Clinic Registry Insight
Samina Hirani, Mayo Clinic Health System, Eau Claire, WI
S. Hirani1, R. A. Vierkant2, N. L. Larson3, S. D. D’Andre4, F. J. Couch5, J. E. Olson3, K. J. Ruddy4; 1Department of Hematology Oncology, Mayo Clinic Health System, Eau Claire, WI, 2Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 3Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 4Department of Medical Oncology, Mayo Clinic, Rochester, MN, 5Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
Background: Smoking, a known risk factor for numerous cancers, negatively impacts long-term survival in breast cancer (BC) survivors (Parada H Jr et al., 2017; Pierce et al., 2019). This study aims to characterize baseline smoking behaviors among breast cancer patients who self-reported smoking during an initial survey. Objectives: To assess attributes of patients self-reporting as current smokers near BC diagnosis, examining demographics, habits, quality of life, cancer treatments, comorbidities, and finances. Methods: Patients with newly diagnosed BC seen at Mayo Clinic Rochester between 2014-2024 were invited to enroll in the Mayo Clinic Breast Disease Registry. Consenting participants who completed self-reported questionnaires at baseline were included in this study. Smoking status and related variables at the time of survey completion were assessed via a baseline questionnaire, classifying participants as current, former, or never smokers. Of 3,959 participants completing baseline surveys, 3,729 were analyzed after excluding 103 with missing smoking data and 127 with their baseline survey returned >1-year post-diagnosis. Categorical variables were summarized with frequencies and percents, continuous variables with distributional statistics. Univariable associations with current smoking were assessed via two-sample t-tests (continuous) and chi-square or Fisher exact tests (categorical). Significant variables (p<0.05) were included in a multivariable logistic regression model. Results: Among 3,729 BC patients (mean age 58.3 years, SD 12.9), 99.1% were female, 97.1% White, and 98.7% non-Hispanic. Only 2.7% reported financial difficulty, 56.4% had a bachelor’s degree or higher, 81.7% were married or equivalent, and 21.7% consumed ≥5 alcoholic drinks/week. Tumor stage distribution was as follows: Stage 0 (13.9%), Stage 1 (43.9%), Stage 2 (28.9%), Stage 3 (8.9%), and Stage 4 (4.5%); 33.8% received chemotherapy, 56.5% radiotherapy, 63.6% endocrine therapy, and 42.2% underwent mastectomy. Current smokers (2.6%, n=95) had a median diagnosis age of 59.1 (IQR 31.8-87.1),with 32.6% consuming ≥5 drinks/week (vs. 21.5% in non-smokers, univariable p=0.012), 69.5% with less than a Bachelor’s degree (vs. 42.9%, p<0.001), 9.6% facing financial difficulty (vs. 2.5%, p<0.001), and 10.5% diagnosed within 270-365 days after survey return (vs. 4.2%, p=0.022). Tumor stage showed 47% with Stage 1 (vs. 43.7%, p=0.245); 33.7% received chemotherapy, 60.0% radiotherapy, and 58.9% endocrine therapy. Current smokers had a mean smoking initiation age of 19.9 (SD 6.2), smoked 12.5 cigarettes/day (SD 7.9) for 32.8 years (SD 14.1), with mean 22.3 pack-years (SD 17.2).Univariable associations of demographic and clinical characteristics with current smoking showed Compared to never or former smokers, current smokers at the time of survey took a longer time to return their baseline survey (p=0.022), were more likely to consume five or more alcoholic drinks per week (p=0.012), were more likely to have difficulty paying their bills (p<0.001) and were less likely to obtain a Bachelor’s degree or higher (p<0.001). These associations remained significant after multivariable adjustment. Conclusion: Current smoking (2.6%) soon after a BC diagnosis was independently associated with delayed survey return, higher alcohol use, financial difficulty, and lower education. Assessing smoking status and referring patients to smoking cessation programs are critical for addressing these risk factors and improving health outcomes. These findings highlight distinct socioeconomic and behavioral profiles among current smokers, suggesting potential targets for tailored interventions.
Presentation numberPS1-04-19
Sexual Function in Young Breast Cancer Survivors: A Subgroup Analysis of an Observational Study in Uruguay
Natalia Camejo, Hospital de Clinicas, Montevideo, Uruguay
N. Camejo1, C. Castillo1, R. Servetto1, G. Muñoz1, J. Manzanares1, D. Amarillo1, M. Guerrina1, G. Herrera2, C. Dörner3, G. Krygier1; 1Clinical Oncology, Hospital de Clinicas, Montevideo, URUGUAY, 2Department of Preventive and Social Medicine, School of Medicine, Montevideo, URUGUAY, 3Clinical Oncology, Hospital Departamental de Soriano “Zoilo A. Chelle”, Soriano, URUGUAY.
Introduction: Sexual dysfunction is a prevalent issue among breast cancer survivors and may have a particular impact on younger women, for whom long-term quality of life is crucial.Objectives: To explore the prevalence and predictors of sexual dysfunction in breast cancer survivors under 50 years old in Uruguay, and to assess the influence of different treatment modalities on their Female Sexual Function Index (FSFI) scores.Methods: This subgroup analysis included patients younger than 50 years from a multicenter observational ambispective study. Eligible women had completed treatment for stage I to III breast cancer (surgery, chemotherapy, and radiotherapy) at least 12 months prior. Sexual dysfunction was defined as FSFI less than or equal to 26.55. Logistic regression models were used to identify associations between treatment modalities and dysfunction.Results: Of the total cohort, 41 patients were under 50. In this group, 65.9 percent had FSFI scores indicating dysfunction. Mastectomy was associated with lower FSFI scores across all domains compared to breast-conserving surgery. The use of gonadotropin-releasing hormone agonists was significantly associated with higher odds of dysfunction (odds ratio 5.7, 95 percent confidence interval: 1.5 to 25.6). Chemotherapy showed no significant detrimental effect in this subgroup.Conclusions: Sexual dysfunction is highly prevalent among young breast cancer survivors in Uruguay and appears to be influenced by specific treatments. These findings support integrating sexual health counseling and fertility-preserving strategies into early survivorship care for this population.
Presentation numberPS1-04-20
A new comprehensive integrative care and navigation model for enhancing outcomes for Black breast cancer patients: evidence from the Care for HER program
Sue Weldon, Unite for HER, Malvern, PA
S. Weldon1, V. Worthy2, R. Fairley2, E. Powers2, G. Kelly1, J. Meakim1, E. Fortune3; 1Non Profit Organization, Unite for HER, Malvern, PA, 2Non Profit Organization, TOUCH, The Black Breast Cancer Alliance, Annapolis, MD, 3Research, Cancer Support Community, Washington, DC.
Background: Care for HER is a campaign that serves Black breast cancer patients nationwide with free access to: 1) integrative care therapies and services, and 2) 24/7 culturally tailored patient navigation by Black nurses and social workers who are also breast cancer survivors. This study aims to evaluate program outcomes and satisfaction, as reported by program participants. Method: After participating in the Care for HER program, participants provided feedback online for multiple program evaluation metrics using a combination of scaled response questions: satisfaction and resource use (5-point scales), perceived benefits (4-point agreement scale and yes/no items), pre- and post-program distress (0-10 scale), and likelihood of recommending to others (0-10 scale). Descriptive and bivariate results are presented.Results: Between April and May 2025, 57 Black women completed the survey, with an average age of 52 (SD=9.1; range 33-71) and average time since diagnosis of 14 months (SD=11.6; range 6-71 months). Results show 98% report being satisfied or very satisfied with the wellness passport program of integrative care therapies and services, with the average likelihood of recommending to others a 9.4 out of 10; 93% of program participants report using resources provided (47% often or very often and 46% sometimes), and 7% rarely or never. As a result of the program, participants reported being better able to find support to reduce treatment side effects (75%) and reducing at least one aspect of financial strain (83%). Nearly all participants reported improvements in their health behaviors, with 95% saying they felt more motivated to make healthier food choices and 93% indicating a better understanding of the benefits of physical activity. Many also experienced gains in navigating the healthcare system and advocating for themselves: 89% reported a better understanding of the resources and services available to them, 88% felt better equipped to follow their treatment plan, and 84% felt more confident advocating for themselves in healthcare settings. Additionally, 69% of participants agreed or strongly agreed that they were better able to understand their diagnosis and treatment options. Average distress score dropped from 6.6 before the program to 2.7 after, reflecting a 3.9-point decrease. Overall, 85% of participants reported reduced distress, 11% saw no change, and 4% reported an increase. High distress (scores of 8-10) declined from 39% to 0%, while those reporting no distress (score of 0) rose from 5% to 21%. Frequency of resource use was significantly associated with reduction in distress (F=4.252, p<.02), such that those who reported using available resources often or very often had the largest reduction in distress after program participation (M=-3.8), compared to those who only use resources sometimes (M=-3.0). Additionally, more frequent use was significantly correlated with satisfaction (r=.43, p<.001).Conclusions: Participation in the Care for HER program was associated with improvements in health behaviors, managing side effects, healthcare navigation, and understanding of diagnosis and treatment, as well as a notable reduction in distress, suggesting the program effectively supports practical and emotional well-being for women with breast cancer. Further, program satisfaction and perceived benefits were significantly correlated with frequency of resource use, indicating that greater engagement enhances program outcomes. Programs like Care for HER provide essential supportive services and empower patients to take a more active role in their cancer care. Expanding access is crucial, as patients—especially those from historically underserved communities—cannot obtain these services through traditional healthcare systems.
Presentation numberPS1-04-21
Correlations Between MOTS-c, Body Fat, and Muscle Strength in a Diverse Ethnic Cohort of Breast Cancer Survivors in Hawai’i
Lauren Imai, University of Hawaii Cancer Center, Honolulu, HI
J. Fukui1, L. Imai1, C. Shen1, K. Baron1, M. Toyama1, I. Pagano1, A. Oamil1, H. Kumagai2, J. Wan2, P. Cohen2, P. Yamada3, C. Teranishi-Hashimoto4; 1Cancer Biology, University of Hawaii Cancer Center, Honolulu, HI, 2Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 3Kinesiology and Rehabilitation Science, University of Hawaii at Manoa, Honolulu, HI, 4Women’s Health and Cancer Rehabilitation, Rehabilitation Hospital of the Pacific, Honolulu, HI.
Abstract: Correlations Between MOTS-c, Body Fat, and Muscle Strength in a Diverse Ethnic Cohort of Breast Cancer Survivors in Hawai’i Background:Mitochondria Open Reading Frame within the Twelve S rRNA c (MOTS-c) is known to respond to exercise, influencing metabolic, cardiovascular, and muscular systems. However, the connection between MOTS-c and markers of health and fitness in Asian cancer survivors is underexplored. Our study aimed to examine the impact of a 12-week concurrent exercise program on MOTS-c levels in Asian breast cancer survivors and determine if anthropometric and fitness-related factors correlate with pre- and post-exercise MOTS-c levels.Methods:Twenty-five breast cancer survivors participated in a 12-week concurrent exercise program. Metrics were assessed before and after the exercise program, including visceral/subcutaneous fat mass, volume, bone mass density (BMD), bone mineral content (BMC), body fat percentage, 1-repetition maximum (1-RM) strength, isokinetic peak torque and maximum work, as well as cardiorespiratory (VO2 peak) and muscular endurance. Plasma MOTS-c was measured using an in-house enzyme-linked immunosorbent assay. Statistical analyses, including t-tests and correlation analyses, were conducted.Results:There was a significant 8.3±13.0% increase in MOTS-c levels following exercise (pre: 254.6±39.9 pg/ml, post: 272.5±36.0 pg/ml, p=0.015). Based on varying responses, the participants were divided into two groups: low responders (n=11) and high responders (n=14). Eight low responders showed a decrease in MOTS-c after exercise, while high responders showed an 18% increase. Pre-exercise MOTS-c levels were positively correlated with baseline body mass, body fat percentage, muscular strength, isokinetic maximum work/torque, and changes in BMD and BMC (p0.05). When analyzed separately for low and high responders, these significant correlations were only present in the high responder group. Additionally, MOTS-c showed a weak negative correlation with muscular endurance as measured by the chair squat test, but for the high responders, a significant negative relationship was found between MOTS-c and core endurance, assessed by plank hold performance (p=0.0145).Summary:Our study showed significant improvement in MOTS-c levels following our exercise program, with high responders showing significant correlations between MOTS-c and body fat, skeletal muscular strength, and BMD and BMC. This exemplified that MOTS-c is associated with bone and metabolic health. Since MOTS-c is correlated with strength but not endurance, strength training should be emphasized in exercise programs aimed at improving cancer survivorship outcomes.
Presentation numberPS1-04-22
Impact of lifestyle coaching on physical activity, mental wellbeing, and cancer wellbeing scores
Zainab Wasim, Princess Alexandra Hospital, Harlow, United Kingdom
Z. Wasim, N. Goyal, A. Konstantis, S. Patel; Oncology, Princess Alexandra Hospital, Harlow, UNITED KINGDOM.
Background: Lifestyle interventions are known to impact physical activity, mental wellbeing, and cancer recovery outcomes. This project evaluated the effectiveness of a lifestyle medicine coaching program over 12 weeks for overweight and obese breast cancer survivors on follow up across three domains: physical activity (Exercise Vital Sign – EVS), mental wellbeing (Warwick-Edinburgh Mental Wellbeing Scale – WEMWBS), and cancer wellbeing (FACT-G7 scores). Methods: Fifteen participants were assessed pre- and post lifestyle medicine program of education and patient led goal-based coaching for healthy behaviour change led by a lifestyle medicine physician, nutritionist, cancer exercise specialist and yoga teacher. Exercise minutes per week, mental wellbeing scores, and cancer wellbeing scores were collected and analysed using paired t-tests and Wilcoxon Signed-Rank Tests. Results: Significant improvements were observed across all three outcome areas. Weekly exercise minutes increased from a mean of 75.2 to 201.0 (t (14) = -5.81, p < .001, Cohen’s d = 1.5). Mental wellbeing scores improved from a mean of 45.0 to 53.0 (p < .05), with 86% of participants showing a meaningful positive change. Cancer-related wellbeing scores improved from 14.3 to 20.5 (t (14) = -3.266, p = .006). The proportion of participants with low mental wellbeing reduced from 43% to 11% post-intervention, while those reporting high wellbeing increased from 7% to 16%. Conclusion: The lifestyle coaching program produced statistically significant and clinically meaningful improvements in physical activity, mental wellbeing, and cancer-related health outcomes. These findings support the integration of holistic, behaviour-based coaching interventions into health promotion and cancer survivorship care pathways.
| Outcome Area | Pre-Intervention Mean | Post-Intervention Mean | Statistical Test | Effect Size / % Change | Key Notes |
| Weekly Exercise Minutes | 75.2 | 201.0 | t(14) = -5.81, p < .001 | Cohen’s d = 1.5 | Very large effect size |
| Mental Wellbeing Score | 45.0 | 53.0 | Wilcoxon, p < .05 | 86% improved | Low wellbeing: 43% → 11%; High: 7% → 16% |
| Cancer-Related Wellbeing | 14.3 | 20.5 | t(14) = -3.266, p = .006 | Significant positive change |
Presentation numberPS1-04-23
Working status and financial security of breast cancer patients during chemotherapy: a prospective study of a Brazilian Cancer Center
Marina Sanches Montemor, Hospital das Clínicas da Faculdade de Medicina da USP, São Paulo, Brazil
M. S. Montemor1, S. S. Santos2, B. M. Figueroa2, K. G. Sousa2, F. B. Makdissi2, S. M. Sanches2; 1Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas da Faculdade de Medicina da USP, São Paulo, BRAZIL, 2Centro de Referência de Tumores de Mama, AC Camargo Cancer Center, São Paulo, BRAZIL.
Introduction: Breast cancer (BC) is a major burden, affecting many working age women. In Brazil, it is estimated an adjusted incidence rate of 41.9 new cases in 100 000 women per year, of these, around 40.0% are diagnosed in advanced stages (stages III and IV). An important fraction of recently diagnosed BC patients will undergo neoadjuvant or adjuvant chemotherapy (CT). The treatment impact in work and household routine is an important factor to be discussed by healthcare team and patients in each therapeutical plan. Objective: Evaluate working status of recently diagnosed BC patients and CT implications on social and economic stability. Methods: We conducted a prospective observational study among insurance-assured patients diagnosed with BC in a Brazilian Cancer Center with indication of neoadjuvant or adjuvant CT. Patients were evaluated with a self-related questionnaire at diagnosis and after CT finished. Results: 44 female patients diagnosed with BC from March to November of 2024 responded to the questionnaire. Median age of diagnosis was 50 years old, 61.4% of patients self-recognized as white, 72.7% were married, 27.3% had no children. About 11.4% patients were in use of antidepressants and 25.0% at psychological care prior to diagnosis. Clinical stages II (54.5%) and III (25.0%) were most prevalent, and all patients were non metastatic. Luminal subtype totalized 77.3% and triple negative 6.8%. Around 52.3% patients underwent neoadjuvant CT.Nearly 29.5% and 57.0% of patients had total or partial participation in family income, respectively. Additionally, 31.8% patients had no financial dependents. Patients worked 36.5 hours per week in average , and 70.5% were socially secured. The median satisfaction rate with work was 8.0/10.0. Almost 41.0% intended to maintain full time job, 31.8% planned to keep it partially and 27.3% desired to halt it during treatment. Approximately 43.2% of women didn’t follow the original labor plan during treatment: of these, 14/19 worked less than expected and 5/19 increased workload. The majority of patients (65.9%) kept fully or partial working, and all of them described adjustments to do so. Most common adaptations were home-office regimens and reduced working hours. Most of bosses were women (79.5%) and 60.0% were described as empathic to patient’s situation. Degree of work satisfaction was maintained after CT. The majority of the group (65.9%) didn’t experience income decrease during treatment.Finally, 59.0% of patients felt depressed or psychological altered during treatment and 47.7%received psychological support. Discussion: The group analyzed comprises mostly socially secured and economically active women, with decent social-family support. Most of the group managed to keep working and didn’t have income decrease during treatment. It is essential to notice that all of them needed to adjust to keep working. Therefore, flexibility and leadership empathy are key to secure working status for breast cancer patients. Conclusion: It is of utmost importance to approach working status and expectations with recently diagnosed breast cancer patients, as this population consists mostly of economically active people. Maintaining and returning to work enable women to be socially integrated. Moreover, returning to work has been shown to be associated with higher survival rates.
Presentation numberPS1-04-24
Factors associated with adherence to surveillance mammography among a racially and ethnically diverse cohort of breast cancer survivors
Angelica Antai, Columbia University, Midland, MI
A. Antai1, K. Crew2, P. Kakani3; 1Epidemiology, Columbia University, Midland, MI, 2Oncology/ Epidemiology, NewYork-Presbyterian/Columbia University Medical Center, New York, NY, 3Internal Medicine, NewYork-Presbyterian/Columbia University Medical Center, New York, NY.
Authors: Angelica Antai, Preeti Kakani, Katherine D. CrewCharacter count including title (no spaces): 2479Character limit including title (no spaces): 3400 Factors Associated with Adherence to Surveillance Mammography Among a Racially and Ethnically Diverse Cohort of Breast Cancer SurvivorsBackground: Annual mammography is a recommended component of post-treatment surveillance for breast cancer survivors and is associated with earlier detection of local recurrence and contralateral breast cancer. However, adherence to surveillance remains variable. Psychological distress, including depression and anxiety, has been proposed as a potential barrier. We investigated demographic, clinical, and mental health factors associated with adherence to surveillance mammography in a diverse cohort of breast cancer survivors.Methods: We conducted a retrospective cohort study of women diagnosed with stage 0-III breast cancer between 2018 and 2023 at Columbia University Irving Medical Center in New York, NY. We excluded women with stage IV disease, bilateral mastectomies, or less than 15 months of follow-up after breast cancer diagnosis. The primary outcome was receipt of at least one mammogram within 15 months of diagnosis. Psychological distress was defined using ICD-10 codes for depression and/or anxiety. Sociodemographic and clinical variables were extracted from the electronic medical record and tumor registry, including age at diagnosis, race and ethnicity, marital status, primary health insurance type, cancer stage, local and systemic treatments. Multivariable logistic regression was used to identify factors associated with adherence to surveillance mammography.Results: Among 1,970 eligible patients, the mean age was 59.2 years (SD, 11.9). The cohort included 33% Hispanic, 32% non-Hispanic White, 12% Black, and 5% Asian women. Overall, 94.6% (n=1,863) were adherent to surveillance mammography guidelines. On multivariable analysis, psychological distress was not significantly associated with adherence to surveillance mammography (odds ratio [OR]=0.89; 95% confidence interval [CI]=0.57-1.39). Compared to non-Hispanic White women, Hispanic ethnicity was associated with increased adherence (OR=1.95; 95% CI=1.15-3.28). Meanwhile, being married (OR=0.63; 95% CI=0.41-0.95) was associated with decreased adherence compared to being unmarried.Conclusions: In this large, racially and ethnically diverse urban breast cancer cohort, adherence to mammography surveillance was high. Psychological distress was not independently associated with non-adherence, suggesting that institutional supports may reduce its potential impact. Interestingly, Hispanic women had nearly 2-fold higher mammography adherence compared to their White counterparts, which may represent cultural differences or disparities in breast cancer worry or fear of recurrence. These findings highlight the importance of tailored survivorship care strategies to promote equitable adherence among breast cancer survivors.
Presentation numberPS1-04-25
Associations of Diet Quality with Symptom Severity and Functioning During Breast Cancer Treatment in American and Taiwanese Patients
Rosalind Tran, Lombardi Comprehensive Cancer Center, Washington, DC
R. Tran1, Y. Liu2, C. Huang3, K. Chang3, F. Hsiao2, T. Fung4, C. Lo3, W. Kuo3, G. Sander1, R. J. Wong1, K. N. Hitesman1, M. Thomas1, J. Chen1, S. Jeong1, M. Tan1, J. H. Wang1; 1Cancer Control and Prevention Program, Lombardi Comprehensive Cancer Center, Washington, DC, 2School of Nursing, National Taiwan University, Taipei City, TAIWAN, 3Department of Surgery, National Taiwan University Hospital, Taipei City, TAIWAN, 4Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA.
Background and Objective: Breast cancer (BC) patients undergoing curative treatment commonly experience appetite loss and subsequent poor dietary quality. The role of nutrition in symptom burden and functioning during treatment remains inadequately studied among BC patients. This study examined these associations among breast cancer patients and whether the associations varied across different cultural dietary practices. Methods and Analysis: Cross-sectional data from 81 American (non-Hispanic White, Black, Hispanic) and 81 Taiwanese female BC patients receiving primary treatment (e.g., surgery, chemotherapy, and/or radiation) were collected. Participants were diagnosed between 2021 and 2024 with stage 0-III primary BC and enrolled within six months of diagnosis. Dietary intake was assessed using an automated 24-hour recall tool (ASA24) on three randomly selected days. Diet quality was assessed by the Healthy Eating Index (HEI-2020) through four categories: ‘good’ (80-100), ‘needs improvement’ (70-79), ‘poor’ (60-69), or ‘very poor’ (0-59). A good quality diet refers to an adequate intake of healthy foods (e.g., fruits, vegetables, whole grains, dairy, protein foods, and fatty acids), and a limited intake of undesirable dietary components (e.g., refined grains, sodium, added sugars, and saturated fat). Poor-quality diets suggest an insufficient intake of healthy foods and an excess intake of undesirable components. Patient-reported symptoms (e.g., insomnia, fatigue, appetite loss, and diarrhea) and functional outcomes (e.g., physical, emotional, social, and role functioning) were measured via EORTC QLQ-BR30. Linear regression examined the associations of HEI and subcomponent scores with each study outcome, adjusting for nationality, age, cancer stage, treatment type, body mass index, calorie intake, physical activity, and financial difficulties. Appetite loss was included in all regression models as a confounder, except when it was a symptom outcome. Results: Only 11% of the sample (Americans: 7%, Taiwanese: 4%) had good diet quality. Over 68% had poor diet quality. A higher percentage of Americans (25%) than Taiwanese (13%) had very poor diets. Americans had higher intakes of added sugar and saturated fat, while Taiwanese had higher intakes of sodium (p < 0.001). However, nationality was not a predictor of the outcomes. The overall HEI scores were not associated with any study outcomes. Patients with more severe appetite loss reported higher total fruit intake (β = 3.21, 95% CI: 0.32, 6.09). Total vegetable intake was inversely associated with insomnia (β = -5.12, 95% CI: -10.1, -0.13). Higher seafood and plant protein intake was associated with a higher prevalence of diarrhea (β = 3.44, 95% CI: 0.37, 6.51), and higher saturated fat intake was associated with poorer physical functioning (β = -1.11, 95% CI: -2.16, -0.07). Conclusion: Most patients undergoing BC treatment had poor diet quality. These cross-sectional findings do not establish causation; however, we noted certain dietary characteristics with specific symptoms. Longitudinal investigations in a larger sample will be needed to further evaluate these observed associations to inform dietary recommendations for BC patients during treatment.
Presentation numberPS1-04-26
Use of Wearable Activity Trackers During Active Breast Cancer Treatment:A Systematic Review
Jasmin Hundal, Cleveland Clinic, Cleveland, OH
J. Hundal1, S. Peshin2, F. Bashir3, H. Bazroodi4, E. Takrori5, M. Kurian6; 1Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 2Internal Medicine, Norton Community Hospital, Norton, VA, 3Medical Sciences, Shiraz University of Medical Sciences, Shiraz, IRAN, ISLAMIC REPUBLIC OF, 4School of Medicine,, Shiraz University of Medical Sciences, Shiraz, IRAN, ISLAMIC REPUBLIC OF, 5College of Medicine,, Alfaisal University,, Riyadh, SAUDI ARABIA, 6Cancer Institute, St Elizabeth Healthcare, Edgewood, KY.
BACKGROUNDBreast cancer is the most diagnosed cancer in women and a leading cause of cancer-related morbidity. Active treatments, such as chemotherapy, radiation, and targeted therapies, are frequently associated with fatigue, reduced physical activity, and decreased quality of life. Wearable activity trackers, including fitness bands and smartwatches, are increasingly being integrated into oncology care to track physical activity, monitor symptoms, and enhance patient engagement. Their use during active treatment provides new opportunities for behavioral support and real-time, remote monitoring. This study systematically examines the use of wearable activity trackers during active breast cancer treatment to monitor activity, track symptoms, support adherence, and enhance patient engagement.METHODSA comprehensive search using relevant keywords and MeSH terms related to ‘breast cancer’, ‘breast cancer treatment’, and ‘activity trackers’ was conducted until March 2025 across PubMed/Medline, Scopus and Web of Science. Studies that used wearable activity trackers in adult breast cancer patients receiving active treatment as part of clinical care or research were included. Data were systematically extracted on how activity trackers were used during active treatment and their intended purposes. The findings were synthesized narratively.RESULTSThe final analysis comprised 40 studies involving over 8,000 participants. The included studies comprised of observational studies (prospective, longitudinal, and cohort) (n=25), randomized controlled trials (n=9), cross-sectional studies (n=4), and non-randomized interventional studies (n=3). Most studies (n = 18) were conducted in the United States. The most common wearable tracker was FitBit. Wearables were primarily used to track physical activity, assess symptoms such as fatigue and sleep issues, and enhance patient engagement and treatment adherence. 23 studies (57.5%) assessed physical activity, 18 (45%) studies assessed sleep, 10 (25%) studies evaluated fatigue, and 9 studies (22.5%) examined quality of life. 21 studies described adherence outcomes, with adherence rates ranging from roughly 16.9% to 96%, while 23 studies (57.5%) reported feasibility outcomes. The duration of time using wearables varied across studies. CONCLUSIONThis study demonstrated that wearable activity trackers are practical and can be effectively utilized during active breast cancer treatment to monitor physical activity, sleep, and fatigue, while also supporting patient engagement and adherence. Most studies demonstrated high feasibility and acceptable adherence rates, underscoring the potential of these devices to enhance supportive care. With technological advancements, wearable trackers offer the potential for real-time detection of treatment-related toxicities and delivery of personalized interventions. Future research is needed to validate wearable-derived metrics and support their integration into clinical decision-making pathways, thereby optimizing outcomes in breast cancer care. Other future directions include leveraging wearables for tracking lymphedema, monitoring long-term cardiac health following anthracycline exposure, and evaluating toxicity profiles associated with the rise of HER2-targeted antibody-drug conjugates. However, key challenges remain, including data overload, limited integration with electronic medical records, and persistent issues of equity and access. Collaborating with companies to incorporate wearable data collection in clinical trials may help address these barriers, while efforts to ensure rigorous validation and standardization of wearable technologies will be critical to their successful adoption in routine oncology care.
Presentation numberPS1-04-27
Advancing hormone therapy adherence among Latinas: A mobile app and navigation strategy to improve outcomes
Vivian Cortez, UT Health San Antonio, San Antonio, TX
P. Chalela1, V. Cortez1, S. Sivak1, A. Flores1, Z. Garcia1, M. Duran1, S. Sierra1, M. Sung-Cuadrado1, A. Trejo1, S. Basoor1, B. Choi2, E. Munoz1, C. Despres1, A. Gonzalez1, V. Kaklamani3, K. Lathrop3, M. Mazo Canola3, D. Inupakutika4, D. Akopian4, A. G. Ramirez1; 1Institute for Health Promotion Research, UT Health San Antonio, San Antonio, TX, 2Population Health Sciences, UT Health San Antonio, San Antonio, TX, 3Mays Cancer Center, UT Health San Antonio, San Antonio, TX, 4Software Communication and Navigation Systems Laboratory, University of Texas at San Antonio, San Antonio, TX.
Background: Hormone therapy (HT) significantly lowers the risk of recurrence and mortality for patients with hormone receptor-positive breast cancer, which accounts for roughly 80% of all breast cancer cases. Long-term adherence to HT can cut recurrence rates by nearly 50%. Yet, about one-third of women prescribed HT are non-adherent, taking less than 80% of the recommended daily dose. Latina patients are disproportionately affected by Non-Medical Drivers of Health (NMDoH), such as socioeconomic and access-related barriers, that hinder adherence and contribute to poorer outcomes. Objective: This three-arm randomized controlled trial evaluates the comparative effectiveness of a bilingual, culturally tailored mobile app (HT Helper) combined with Patient Navigation (PN), PN alone, and standard care on improving HT adherence. The study also examines how these interventions influence self-efficacy in recognizing and managing side effects and enhancing communication with healthcare providers. This work builds on earlier pilot findings that confirmed the HT Helper app’s feasibility and acceptability. We present key findings from the formative phase, including usability testing, focus group feedback, and final app modifications. Methods: Twelve Latina breast cancer patients receiving HT at the Mays Cancer Center and experiencing NMDoH-related barriers participated in the formative research activities. Participants used the HT Helper app for one week and then joined focus groups (conducted in their preferred language, English or Spanish) to provide in-depth feedback. Participants reviewed and commented on all app components, including navigation, educational videos, symptom-tracking tools, motivational messaging, the Chat with a Doctor feature, and the User Guide. Based on their recommendations, we updated the app and conducted a second round of testing. Findings: Most participants responded favorably to the HT Helper app, describing it as helpful, informative, and easy to use. Suggestions for improvement included expanding descriptions of specific HT-related symptoms, enhancing the app’s interactive features (e.g., Chat with a Doctor), updating patient testimonial videos, and refining the visual design of the home screen for better appeal and usability. Conclusion: A patient-driven, iterative refinement process was critical to ensuring the HT Helper app is user-friendly, relevant, and responsive to the needs of Latina patients. The resulting multi-component intervention has strong potential to improve breast cancer outcomes by enhancing medication adherence, promoting better self-management, and ultimately supporting longer-term survival. The intervention is designed to be scalable, evidence-based, and suitable for broader use among patients prescribed oral anticancer therapies.
Presentation numberPS1-04-28
<b>Treatment</b> <b> Cascade Disruption in Breast Cancer: Socioeconomic Predictors of Sequential Delays in a Southeast Texas Cohort</b>
Nency Ganatra, Baptist Hospitals of Southeast Texas, Beaumont, TX
N. Ganatra, A. Jamal, P. Jain, J. Doshi, S. Patel; Internal Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX.
Background:Sequential, timely delivery of multimodal treatment is essential to optimal outcomes in breast cancer. The majority of previous studies determine delays in treatment in isolation, without including disruptions across the entire treatment cascade (surgery → chemotherapy → radiation → hormone/immunotherapy). This study examines socioeconomic predictors of sequential and cumulative delays in the treatment of breast cancer. Methods:We conducted a retrospective cohort study on registry data from 609 breast cancer patients treated between 2018 and 2023 at a Southeast Texas multi-hospital system. Delay was classified as determined by modality-specific time thresholds as defined. Patients were classified as having 0, 1, 2, 3, or ≥4 modality delays. Logistic regression examined associations with demographic, insurance, and tumor characteristics. Results:53% of patients experienced one or more modality delays; 19% had ≥2 modality delays.Medicaid insurance (OR: 2.89, 95% CI: 1.70-4.92), age <50 (OR: 2.15, CI: 1.30-3.54), and unmarried status (OR: 1.76, CI: 1.22-2.54) were independent predictors of cumulative delay.Triple-negative patients (subset analysis, n = 98) with ≥2 delays had higher stage progression rates than on-time-treated counterparts (p = 0.03).Cascade analysis showed the greatest drop-off rate was after surgery (not initiating adjuvant chemotherapy or radiation). Conclusion:Delays in breast cancer care are not isolated events. Medicaid status, younger age, and social isolation are predictors of cumulative care disruption. Interventions must address continuity of all treatment cascade steps, especially among socioeconomically disadvantaged groups.
Presentation numberPS1-04-29
Longitudinal changes in health-related quality of life of early-stage breast cancer patients treated with adjuvant chemotherapy: Data from geicam/2003-02 study
Pilar Zamora-Auñón, Hospital Universitario La Paz; PhD Program in Medicine and Surgery. Doctoral School, Universidad Autónoma de Madrid; GEICAM Spanish Breast Cancer Group, Madrid, Spain
A. Blasco1, P. Zamora-Auñón2, R. Pastor3, M. Martín4, C. Reboredo5, A. Lahuerta-Martinez6, I. Álvarez6, S. Sonia7, M. Marin8, E. Martínez de Dueñas9, J. López-Vega10, A. Barnadas11, Á. Rodríguez-Lescure12, S. Salvador-Bofill13, C. Jose Ignacio14, V. Lope15; 1Statics, GEICAM Spanish Breast Cancer Group; PhD Program in Medicine and Surgery. Doctoral School, Universidad Autónoma de Madrid, San Sebastián de los Reyes, SPAIN, 2Medical Oncology, Hospital Universitario La Paz; PhD Program in Medicine and Surgery. Doctoral School, Universidad Autónoma de Madrid; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 3Epidemiology and cancer area, National Centre for Epidemiology, Instituto de Salud Carlos III; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, SPAIN, 4Medical Oncology, Hospital General Universitario Gregorio Marañón: Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 5Medical Onco, Complejo Hospitalario Universitario A Coruña (CHUAC); GEICAM Spanish Breast Cancer Group, A Coruña, SPAIN, 6Medical Oncology, Unidad de Gestión del Cáncer de Guipúzcoa (Osakidetza-OSI Donostialdea_Onkologikoa)-BioDonostia; GEICAM Spanish Breast Cancer Group, San Sebastián, SPAIN, 7Medical Oncology, Hospital del Mar; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 8Medical Oncology, Consorci Sanitari de Terrassa; GEICAM Spanish Breast Cancer Group, Terrasa, SPAIN, 9Medical Oncology, Consorcio Hospitalario Provincial de Castellón; GEICAM Spanish Breast Cancer Group, Castellón, SPAIN, 10Medical Oncology, Hospital Universitario Marques de Valdecilla; GEICAM Spanish Breast Cancer Group, Santander, SPAIN, 11Medical Oncology, Hospital de la Santa Creu i Sant Pau, IR Sant Pau, CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 12Medical Oncology, Hospital General Universitario de Elche; GEICAM Spanish Breast Cancer Group, Elche, SPAIN, 13Medical Oncology, Hospital Universitario Virgen del Rocio; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 14Medical Oncology, Hospital Universitario de Toledo; GEICAM Spanish Breast Cancer Group, Toledo, SPAIN, 15Cancer epidemiology and cancer area, National Centre for Epidemiology, Instituto de Salud Carlos III; Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP); GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: The multicenter, open-label, randomized phase III GEICAM/2003-02 study compared two regimens as adjuvant therapy for node-negative breast cancer (BC) patients: FAC (5-fluorouracil, doxorubicin, and cyclophosphamide) alone versus FAC plus weekly paclitaxel (FAC-wP). It demonstrated paclitaxel chemotherapy (CT) improved disease-free survival [1]. This study aims to compare health-related quality of life (HRQoL) in these arms of treatment. Methods: Early-BC patients from the GEICAM/2003-02 study were selected (157 FAC arm and 141 FAC-wP arm). HRQoL was assessed using the EORTC QLQ-C30 version 3.0 questionnaire. Random intercept lineal model with two nested levels (hospital and subject) for repeated measures adjusted by patient and tumor characteristics at diagnosis (body mass, age, histopathological grade, surgery type, stage, tumor subtype and menopausal status) were used to assess changes in the global, functional and symptom QLQ-C30 summary scores from baseline to end of treatment and 6 months later. Stratified analyses were also performed by menopausal status and endocrine-therapy (ET) post-CT according to tumor subtype. Results: QLQ-C30 summary score significantly decreased from baseline to end of treatment (FAC: [-2.08, 95% CI= -7.11, 2.94] and FAC-wP: [-2.82, 95% CI: -7.76, 2.13]) but slightly exceeded baseline scores 6 months post-treatment, with no significant differences between the two arms. Greater deterioration in post-treatment functional summary score was observed in the FAC-wP arm [-4.15, 95%CI: -10.40, 2.11], compared to FAC [-1.46, 95% CI= -7.78, 4.86]. In postmenopausal patients, post-treatment QLQ-C30 summary score deterioration was greater in the FAC-wP arm [-4.05, 95% CI= -9.96, 1.86]. Hormone receptor positive, human epidermal growth factor receptor 2 negative (HR+ HER2-) with ET post-CT patients showed more pronounced reductions in the QLQ-C30 summary score post-treatment (FAC: [-3.58, 95% CI= -8.42, 1.27] and FAC-wP: [-5.03, 95% CI: -9.77, -0.28]) than triple-negative patients (FAC: [-2.53, 95%CI= -7.43, 2.37] and FAC-wP: [1.05, 95% CI= -4.05, 6.16]). Further details are shown in Table 1. Conclusions: Taxane-based regimens do not have a great impact on HRQoL recovery after 6 months of treatment: While temporary deterioration in functional and symptom scales was observed during treatment, patients in both arms recovered their baseline HRQoL scores 6 months later. These results also emphasize the importance of considering patient characteristics variables when evaluating HRQoL outcomes in BC survivors. References: 1. M. Martín et al., “Fluorouracil, Doxorubicin, and Cyclophosphamide (FAC) Versus FAC Followed by Weekly Paclitaxel AsAdjuvant Therapy for High-Risk, Node-Negative Breast Cancer: Results From the GEICAM/2003-02 Study,” JCO, vol. 31, no.20, Jul. 2013.
Presentation numberPS1-04-30
Breast cancer chemotherapy nearly doubles the risk of treatment-related AML and MDS: A systematic review and meta-analysis
Sacha A Roberts, NewYork-Presbyterian Weill Cornell Medical Center, NEW YORK, NY
S. A. Roberts1, A. M. Kagerer1, N. J. Sareen1, T. Haque2, P. Razavi3; 1Internal Medicine, NewYork-Presbyterian Weill Cornell Medical Center, NEW YORK, NY, 2Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, NEW YORK, NY, 3Department of Breast Oncology, Memorial Sloan Kettering Cancer Center, NEW YORK, NY.
Introduction: Chemotherapy continues to be a pillar of treatment for breast cancer (BC), and while it improves survival and reduces recurrence, there are significant side effects to acknowledge. One of the most serious long-term complications is the development of myeloid neoplasms post cytotoxic therapy (MN-pCT) (previously referred to as therapy-related myelodysplastic syndromes or acute myeloid leukemia). Individuals that developMN-pCTs have significant morbidity and reduction of overall life expectancy, despite improved breast cancer outcomes. The risk of developing t-AML/MDS in breast cancer patients who undergo chemotherapy compared to those who have not had chemotherapy has not been previously reported. Methods: To address this gap, we conducted a systematic review and meta-analysis to quantify the risk of MN-pCT among breast cancer patients treated with chemotherapy compared to those who did not receive chemotherapy. PubMed, Embase, and Cochrane databases were searched in March 2025. The search strategy included three search strings: breast cancer, leukemia or myelodysplastic syndrome or hematologic malignancy, and chemotherapy. Included articles were experimental or observational studies. Full texts were independently reviewed by two reviewers, with discrepancies resolved via consensus. The primary outcome was the pooled odds ratio (OR) for the development ofMN-pCT following chemotherapy exposure in BC patients compared to those who did not receive chemotherapy. Random-effects meta-analysis was used to calculate pooled ORs and 95% confidence intervals (CIs). Publication bias was assessed using visual inspection of the funnel plot and Egger’s test for small-study effects. Heterogeneity was assessed with the I² statistic, and meta-regression explored the impact of study-level factors such as anthracycline use, radiation, follow-up duration, publication year, age, and sample size. Results: Nineteen studies from 1985 to 2023 met inclusion criteria and included a total of 1,215,260BC patients (527,761 chemotherapy; 687,499 no chemotherapy). The studies consisted of2 randomized controlled trials, 1 case control study, and 16 retrospective cohort studies.The median follow-up time was 5.8 years. The pooled incidence of MN-pCT was 0.31%(chemotherapy) and 0.23% (no-chemotherapy). Chemotherapy was associated with an early two-fold increase in risk of AML/MDS (pooled OR 1.91 (95% CI: 1.41-2.58), p <0.001), with substantial heterogeneity (I² = 82.9%). The estimated between-study variance(τ²) was 0.1810 (SE = 0.1167). Meta-regression showed that the proportion of patients receiving anthracyclines explained over half of the between-study heterogeneity (R² =56.3%) but was not a statistically significant predictor of risk (p = 0.21). Notably, non-anthracycline regimens were also associated with a significant increase in MN-pCT risk.No significant associations were found for publication year, sample size, follow-up duration, age, or radiation exposure. Conclusion: We present the first meta-analysis that directly compares the risk of MN-pCT in patients with breast cancer who received cytotoxic chemotherapy versus those who did not receive chemotherapy. This risk was not confined to a specific regimen and is likely multifactorial in nature. Our results highlight the importance of risk-benefit discussions when pursuing chemotherapy, especially taking into consideration co-morbidities, life expectancy, and breast cancer risk profile. Additionally, future studies are needed to optimize surveillance strategies and develop and validate biomarkers to identify patients at risk of MN-pCTs after chemotherapy exposure.
Presentation numberPS1-05-16
Patient understanding and impact on quality of life of serial minimal residual disease testing in patients with early-stage breast cancer
Shaili Tapiavala, Washington University in St. Louis, St. Louis, MO
S. Tapiavala1, I. Grigsby1, N. Bagegni1, F. Fa’ak1, F. Ademuyiwa1, K. Clifton1, E. L. Podany1, C. Cheng2, A. J. Medford3, C. Gianni4, L. Gerratana5, M. Lipsyc-Sharf2, A. Bardia2, M. Cristofanilli4, C. X. Ma1, A. A. Davis1; 1Division of Oncology, Washington University in St. Louis, St. Louis, MO, 2Division of Hematology/Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, 3Division of Oncology, Massachusetts General Hospital, Boston, MA, 4Department of Oncology and Hematology-Oncology, Weill Cornell Medicine, New York, NY, 5Department of Medical Oncology, IRCCS CRO Aviano National Cancer Institute, Aviano, ITALY.
Background: There are no standard guidelines regarding radiographic or laboratory monitoring for patients with early-stage breast cancer (EBC) in the adjuvant setting. Detecting minimal residual disease (MRD) through circulating tumor DNA (ctDNA) is associated with significantly higher probability of later distant recurrence, although clinical utility data are lacking. To date, patient understanding and emotional burden of serial MRD testing are understudied. Here we evaluate the impact of MRD surveillance on quality of life among patients with EBC through investigating anxiety, depression, fear of recurrence, and patient comprehension. Methods: This IRB-approved prospective study enrolled patients with EBC undergoing ctDNA surveillance testing at Washington University in St. Louis. All patients underwent blood collection for Signatera MRD testing (Natera Inc.) and plasma-biobanking at up to 8 serial timepoints over 4 years. Baseline testing was prior to neoadjuvant treatment (n=12) or after surgery prior to adjuvant therapy (n=29). Testing results were shared with the patient and the treating oncologist. Surveys were conducted to assess patient understanding of the rationale for ctDNA testing, Fear of Recurrence (FCR4), Hospital Anxiety and Depression Scale (HADS), and level of comfort with ctDNA testing, at baseline blood collection and again 12 months after the initial MRD testing. Qualitative data were obtained through free-response survey items. Descriptive statistics were used to summarize the data. Results: 41 patients with triple-negative (n=17) or HR+ HER2- (n=24) EBC completed the baseline surveys with 26 patients completing both the baseline and 12-month assessments as of July 1st, 2025. The cohort included 31 (75.6%) White, 8 (19.5%) Black, and 2 (4.9%) Asian patients with a median age of 50 years. Qualitative analysis demonstrated patients had a strong understanding of the use of ctDNA surveillance with 30 of 35 (85.7%) patients who responded to the free text question accurately describing it as a tool for early detection of cancer in the blood. At 12 months, 88.5% of patients (n=23 of 26) answered that they would consider starting treatment based on results of ctDNA testing if recommended by their oncologist; however, 42.3% of patients (n=11 of 26) expressed financial concern with ctDNA testing if not covered by insurance. Among patients with longitudinal survey data (n=26), 10 patients had at least one positive MRD test and one patient had serially positive MRD testing prior to 12-month surveys. For patients with negative MRD testing at all timepoints (n=16), the mean FCR4 score did not significantly change from baseline to 12 months. Similarly, for those with positive MRD testing (n=10), FCR4 score did not significantly change at 12 months. The average HADS-anxiety score decreased from 5.15 at baseline (n=40) to 4.54 at 12 months (n=26), while the average HADS-depression score increased from 1.89 to 2.52; however, this change was not significant, including in the subset of patients with positive MRD testing. On a 0-5 scale of evaluating future attitudes toward use of ctDNA testing, the average rating was 4.1, indicating a favorable opinion of undergoing MRD testing. Conclusion: This prospective study demonstrated that the rationale for MRD testing was well understood and viewed favorably by patients. Regardless of MRD testing results, fear of recurrence and levels of anxiety or depression remained relatively stable from baseline to 1 year, indicating that testing was not associated with significant psychological stress in our study. Nearly half of patients expressed concerns about costs associated with testing if not covered by insurance. Larger, confirmatory studies are needed to further evaluate the impact of MRD surveillance on quality of life of patients with EBC.
Presentation numberPS1-05-18
Association Between Sleep Continuity and Cognition in Breast Cancer Survivors: A Cross-Sectional Study
Kaitlin Lampson, Memorial Sloan Kettering Cancer Center, New York City, NY
K. Lampson1, X. Li1, A. Yang2, S. N. Garland3, J. C. Root2, T. A. Ahles2, J. J. Mao1; 1Integrative Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, 2Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York City, NY, 3Department of Psychology and Oncology, Memorial University of Newfoundland, St. John’s, NL, CANADA.
Background: Cancer-related cognitive difficulties (CRCD) is prevalent and distressing for breast cancer survivors and commonly co-occurs with insomnia. Although sleep tracking has become increasingly common in oncology settings, little is known about how sleep patterns influence cognition in cancer survivors. Therefore, we aim to characterize the associations between subjective and objective sleep continuity and cognition in breast cancer survivors with insomnia and CRCD. Methods: This analysis includes baseline data from a trial conducted among 260 breast cancer survivors with insomnia (Insomnia Severity Index score 8) and self-reported CRCD. This analysis is limited to trial participants who completed sleep tracking measures, including the Consensus Sleep Diary (CSD) and Actiwatch or FitBit smart watch (WA). Sleep continuity measures included wake after sleep onset (WASO), number of awakenings (NWAK), time spent in bed (TIB), total sleep time (TST), and sleep efficiency (SE). Subjective cognition was assessed by Functional Assessment of Cancer Therapy-Cognition Perceived Cognitive Impairment domain (FACT-Cog PCI), while objective cognition was assessed by Hopkins Verbal Learning Test-Revised (HVLT-R) Delayed Recall normed T-score. Multivariate linear regression modeling was used to explore associations between sleep continuity and cognition. All models included adjustments from covariates: chemotherapy use, cancer stage, race, and education. Results: Among 158 participants, the average age [range] was 57 [28-83] and 21.5% were nonwhite. None of the sleep metrics collected from either the CSD or WA were significantly associated with subjective cognition (all p > .05). In data collected by CSD, greater WASO was significantly associated with worse objective cognition (Coef. = -.243, p = .003, 95% CI [-0.135, -0.028]) and higher SE was significantly associated with better objective cognition (Coef. = .209, p = .011, 95% CI [0.054, 0.422]). In data collected by WA, greater NWAK was significantly associated with worse objective cognition (Coef. = -.298, p = .010, 95% CI [-0.294, -0.042]). Conclusion: Measured and reported sleep continuity was not significantly associated with subjective cognition, but sleep fragmentation, measured by both sleep diary and smart watch, was significantly associated with worse objective cognition in breast cancer survivors with comorbid insomnia and CRCD. Interventions targeting sleep could be used to promote cognitive health during breast cancer survivorship.
Presentation numberPS1-05-19
Prognostic Effect of Clinical Treatment Score Post-5 years (CTS5) in patients with Invasive Lobular Carcinoma and its relationship with Oncotype Dx
Jorge Avila, Montefiore Medical Center, Bronx, NY
J. Avila1, X. Xue2, A. Gyamfi3, J. D. Anampa3; 1Department of Hematology and Medical Oncology, Montefiore Medical Center, Bronx, NY, 2Department of Epidemiology, Albert Einstein College of Medicine, Bronx, NY, 3Department of Oncology, Montefiore Einstein Cancer Center, Bronx, NY.
Background:While patients with early-stage invasive lobular carcinoma (ILC) of the breast often present better survival rates initially, the risk of recurrence and mortality increases later in the disease course. The Clinical Treatment Score Post-5 years (CTS5) is a validated tool used to estimate risk of late distant recurrence in patients with Hormone Receptor positive breast cancer after 5 years of adjuvant treatment; however, its application in patients with ILC has not been fully analyzed. OncotypeDx provides prognostic information in breast cancer, but there is limited data about OncotypeDx to predict long-term survival outcomes in ILC. We aimed to assess whether CTS can predict long-term OS for patients with ILC, and to evaluate its prognostic effect beyond OncotypeDx recurrence score. Methods:We conducted a retrospective analysis using the National Cancer Database (NCDB) from 2006-2020. Inclusion criteria were female gender, age ≥18 years, diagnosis of ILC, breast cancer stage I-III, removal of breast tumor and axillary lymph nodes, and alive status at least 5 years after diagnosis. Patients were classified into CTS5 low, intermediate and high-risk categories. Our primary endpoint, long-term overall survival (OS) included survival analysis after 5 years from diagnosis. We used the Pearson X2 test to evaluate the relationship between categorical variables and the t test for continuous variables. Cox proportional models were conducted to identify factors associated with long-term OS. Results:67,927 patients were analyzed for our long-term survival analysis. Median age for our group was 63 years. Most of our patients were Non-Hispanic White (82%), with breast cancer stage I (50%), HR positive (ER+ [100%], PR+ [87%])/HER2 negative (73%) tumors. Patients were divided into 3 groups based on their CTS5 risk: low (n = 32,625 [48%]), intermediate (n = 21,007 [31%]) and high (n = 14,295 [21%]). 10-year OS rates differed significantly among these groups with OS rates of 91%, 84% and 64% respectively (p < 0.001 each). In multivariate Cox regression models adjusted for patient, tumor, and treatment characteristics, CTS5 remained an independent predictor of long-term OS. Patients with high CTS5 score had worse long-term OS when compared to those with low CTS5 (HR 3.63, 95% CI 3.42-3.86; p <0.001). We also analyzed 17,882 patients from the long-term cohort with available OncotypeDx results. Among patients with high OncotypeDx recurrence score, the percentage of patients with low, intermediate, and high CTS5 risk-group were 47.3%, 41.3%, 11.4% respectively, whereas the percentages were 50.9%, 38.6%, and 10.5% for those with low OncotypeDx recurrence score. We found that CTS5 provided significant prognostic information for long-term OS across all Oncotype Dx risks group. Patients with high CTS5 risk-group had worse long-term OS when compared to those with low CTS5 risk-group in patients with low (HR 4.6, 95% CI 2.90-7.30; p <0.001), intermediate (HR 4.33, 95% CI 3.43-5.46; p <0.001), and high (HR 3.99, 95% CI 2.19-7.28; p <0.001) OncotypeDx recurrence score. A multivariable model with CTS5 in addition to OncotypeDx recurrence score showed significant association with long-term OS (Likelihood ratio test, p <0.001). Conclusion:CTS5 is an independent predictor of long-term OS in patients with ILC and it can identify patients with substantially higher risk of late death beyond 5 years. CTS5 adds independent prognostic information that persists even after adjusting for genomic factors such as OncotypeDx recurrence score. Combining CTS5 score and OncotypeDx recurrence score into clinical practice may provide better information for treatment decisions such as extending endocrine therapy or intensifying surveillance in this population.
Presentation numberPS1-05-21
Socioeconomic disparities in opioid use and harm among Danish breast cancer survivors
Kirsten Marie Woolpert, Aarhus University and Aarhus University Hospital, Aarhus N, Denmark
K. M. Woolpert; Department of Clinical Epidemiology, Aarhus University and Aarhus University Hospital, Aarhus N, DENMARK.
Background: Opioids are frequently prescribed to manage pain after a breast cancer diagnosis, but prolonged use can lead to serious harm, including dependence and overdose. Socioeconomic position (SEP), often defined by income, education, and employment, may influence access to care, prescribing patterns, and the likelihood of long-term opioid use. Although Denmark provides universal healthcare, it is unclear whether SEP-related differences in opioid use persist after diagnosis. Methods: We identified all patients assigned female at birth who were diagnosed with non-metastatic breast cancer at age 18 or older between 1997 and 2020 using the Danish Cancer Registry. Patients with opioid use or a hospital history of opioid use disorder in the year before diagnosis were excluded. To be eligible, patients had to survive and reside in Denmark for at least one year after diagnosis. SEP was measured at diagnosis using household income, education level, and employment status. We defined three outcomes from the time of diagnosis: any opioid use (≥1 prescription within 6 months), prolonged use (≥1 prescription in the first 6 months and ≥2 in months 6-12), and opioid-related disorder or overdose within 5 years. Associations between SEP and each outcome were estimated using logistic regression, reporting crude odds ratios (OR) and 95% confidence intervals (CI). Results: Among 77,261 one-year breast cancer survivors, 52,321 (68%) redeemed an opioid prescription within six months of diagnosis, 992 (1.3%) became prolonged users, and 204 (0.3%) experienced an opioid-related disorder or a fatal/non-fatal overdose. Patients with low income were more likely to receive any opioid in the first six months after diagnosis compared with those with high income (OR: 1.40, 95% CI: 1.34, 1.46), with similar patterns observed across other measures of SEP. Among those who received opioids, there were no SEP differences in the type or strength prescribed. Unemployed patients were more likely to become prolonged users compared with those employed at diagnosis (OR: 1.68, 95% CI: 1.47, 1.93), and patients with a high-school education had higher odds of opioid-related disorder or overdose compared with those with a tertiary-level education (OR: 1.74, 95% CI: 1.21, 2.50). Discussion: In this nationwide study of breast cancer survivors, socioeconomic differences in opioid use and harms emerged despite universal healthcare. Although initial prescribing practices appeared equitable among users, patients with lower SEP were more likely to receive opioids, become prolonged users, and experience opioid-related harm. These findings highlight the need for equity-focused pain management and survivorship care in breast cancer. Because this was a descriptive study, we reported crude estimates to reflect overall patterns. Future work could build on these findings by exploring potential mechanisms or points for intervention.
Presentation numberPS1-05-22
Characteristics and weight loss outcomes in patients with breast cancer prescribed semaglutide
Janet Song, Kaiser Permanente Northern California, Santa Clara, CA
J. Song1, R. Liu2, S. K. Zhong3, P. Mishra3, S. Zhu3, E. Feliciano3; 1Internal Medicine, Kaiser Permanente Northern California, Santa Clara, CA, 2Hematology/Oncology, Kaiser Permanente Northern California, San Francisco, CA, 3Division of Research, Kaiser Permanente Northern California, Pleasanton, CA.
Background Obesity and diabetes are common comorbid conditions found in patients with breast cancer and these conditions have been linked to risk of recurrence and early mortality. GLP-1 receptor agonists have changed the paradigm for weight loss and diabetes management, but the prevalence, timing of initiation and duration of use among women with a breast cancer history is not well described. Further, efficacy for weight loss may vary by diabetes status and treatment factors such as the use of endocrine therapy. Methods To address these knowledge gaps, our retrospective analysis utilized tumor registry and electronic health record data to identify all patients with a breast cancer history who initiated semaglutide (and/or tirzepatide) therapy from 2020-2025 at Kaiser Permanente Northern California, a large integrated community oncology network in the US. Results 1,753 patients with breast cancer history initiated semaglutide in the years 2020-2025. The majority of patients were diagnosed with stage 0 or I/II breast cancer (344 [19.6%] and 1,146 [65.4%], respectively) and 1,456 (83.1%) of patients had estrogen receptor (ER) positive breast cancer. 1,245 (71.0%) had diabetes at initiation. The average age at initiation was 63.5 years, with a median (IQR) time from breast cancer diagnosis to semaglutide initiation of 7 (3-13) years. 1,296 (73.9%) had ever used endocrine therapy, with 685 (39.1%) on active endocrine therapy at semaglutide initiation. Most patients had >1 semaglutide dispense (1,718 [98.1%]), but more than one third continued use beyond 1 year. Mean (SD) BMI at semaglutide initiation was similar among the non-diabetic (37.4 [7.2] kg-m2) vs. the diabetic (36.4 [7.5] kg/m2) group, with a mean maximum achieved weight loss of 11.2% and 8.6%, respectively. When stratified by history of endocrine therapy, rates of continuation at one year and achieved weight loss were similar between groups. Conclusion In patients with breast cancer, semaglutide therapy was most commonly initiated for diabetes and was associated with weight loss, even among those receiving endocrine therapy. However, the maximum observed weight loss was less than that observed in clinical trials in non-cancer populations. Long-term adherence was limited, with only one-third continuing use beyond one year. Our study highlights the role of real-world data in understanding the effects of GLP1- receptor agonist drugs in patients with breast cancer. Future studies should investigate whether the use of these agents reduces risk of recurrence and mortality.
| Group | Overall (N = 1753) | Endocrine Therapy (N = 685) | Not on Endocrine Therapy (N = 1068) | ||||||||||||
| Age – Mean (SD) | 63.5 (10.3) | 60.5 (10.5) | 65.5 (9.7) | ||||||||||||
| Female sex – n(%) | 1737 (99.1%) | 679 (99.1%) | 1058 (99.1%) | ||||||||||||
| Race/Ethnicity – n(%) | |||||||||||||||
| Asian | 218 (12.4%) | 99 (14.5%) | 119 (11.1%) | ||||||||||||
| Black | 180 (10.3%) | 69 (10.1%) | 111 (10.4%) | ||||||||||||
| Hispanic | 353 (20.1%) | 146 (21.3%) | 207 (19.4%) | ||||||||||||
| White | 873 (49.8%) | 31 (48.3%) | 542 (50.8%) | ||||||||||||
| Other | 129 (7.4%) | 340 (5.8%) | 89 (8.4%) | ||||||||||||
| Diabetes – n(%) | 1245 (71.0%) | 463 (67.6%) | 782 (73.2%) | ||||||||||||
| ER Positive – n(%) | 1456 (83.1%) | 669 (97.7%) | 787 (73.7%) | ||||||||||||
| Stage – n(%) | |||||||||||||||
| 0 | 344 (19.6%) | 67 (9.8%) | 277 (25.9%) | ||||||||||||
| 1 | 836 (47.7%) | 445 (65.0%) | 391 (36.6%) | ||||||||||||
| 2 | 310 (17.7%) | 115 (16.8%) | 195 (18.3%) | ||||||||||||
| 3 | 73 (4.2%) | 29 (4.2%) | 44 (4.1%) | ||||||||||||
| 4 | 7 (0.4%) | 5 (0.7%) | 2 (0.2%) | ||||||||||||
| Years from Breast Cancer Dx to GLP1 Initiation – Mean (SD) | 8.9 (7.4) | 4.4 (4.5) | 11.8 (7.4) | ||||||||||||
| BMI – n(%) | |||||||||||||||
| Normal (18.5-24.9) | 44 (2.5%) | 11 (1.6%) | 33 (3.1%) | ||||||||||||
| Overweight (25-29.9) | 225 (12.8%) | 82 (12.0%) | 143 (13.4%) | ||||||||||||
| Obese Class 1 (30-34.9) | 486 (27.7%) | 189 (27.6%) | 297 (27.8%) | ||||||||||||
| Obese Class 2 (35-39.9) | 468 (26.7%) | 185 (27.0%) | 283 (26.5%) | ||||||||||||
| Obese Class 3 (40+) | 491 (28.0%) | 210 (30.7%) | 281 (26.3%) | ||||||||||||
| Maximum Weight Loss (lbs) – Mean (SD) | 19.0 (17.7) | 19.3 (17.3) | 18.8 (17.9) | ||||||||||||
| Maximum Weight Loss (%) – Mean (SD) | 8.7 (7.6) | 8.8 (7.5) | 8.6 (7.8) | ||||||||||||
| More than one dispense – n(%) | 1601 (91.3%) | 638 (93.1%) | 963 (90.2%) | ||||||||||||
| Dispenses greater than 1 year – n(%) | 618 (35.3%) | 250 (36.5%) | 368 (34.5%) |
Presentation numberPS1-05-23
Using Exercise to Improve Arthralgia and Aromatase Inhibitor Adherence in Older Breast Cancer Survivors: Preliminary Results from a Pilot Randomized Controlled Trial
Shirley Bluethmann, Wake Forest University School of Medicine, Winston-Salem, NC
S. Bluethmann1, E. Dressler2, M. Caru3, H. Klepin4, A. Willis2, C. Truica5, C. Peters1, N. Olsen5; 1Div. Public Health Sciences, Dept of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, 2Div. Public Health Sciences, Dept of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, 3Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, 4Division of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 5Department of Internal Medicine, Penn State College of Medicine, Hershey, PA.
Background: Aromatase inhibitors (AIs) reduce recurrence and mortality risk in estrogen receptor-positive female breast cancer survivors (BCS), the majority of whom are aged ≥60. However, up to 70% of users discontinue AIs before the recommended duration of therapy, often due to arthralgia, which affects more than 50% of AI users. Despite its prevalence, AI-associated joint pain remains poorly understood, particularly in older survivors. Physical activity (PA) and education may mitigate symptoms and support adherence, yet tailored interventions for this population are lacking. Methods: We tested REJOIN (Relieving Joint Pain and Improving AI Adherence in Older Breast Cancer Survivors), a remotely delivered, supervised exercise and education intervention. We recruited through two sites (Penn State Cancer Institute and Atrium Health Wake Forest Baptist Comprehensive Cancer Center). The pilot randomized controlled trial (RCT) assessed feasibility and early evidence of benefit. Participants (n=24 BCS) received a 16-week, program including resistance and aerobic training, education on AI-related side effects, and self-management strategies. Joint pain was measured via the Brief Pain Inventory (BPI), and AI adherence via the Morisky-Levine Medication Adherence Scale at baseline, 4, 6 and 12 months. Additional measures included physical activity (collected via Actigraph triaxial research-grade accelerometer), modified geriatric assessment battery and a fasting blood draw. Descriptive statistics were calculated for each outcome at baseline, 4 months and the change in-between the two time points to assess effects from the intervention. Between group differences in change were assessed using the Wilcoxon rank-sum test and within-group changes using the Wilcoxon signed-rank test (α=0.05). Effect sizes were calculated using Cohen’s d with corresponding 95% confidence intervals to estimate magnitude of changes within and between groups; values of d≥0.5 indicate moderate or greater practical significance. Results: Participants’ mean age was 72 years (range 60-88), included >33% from rural areas, and we had 80% retention over the 12 month pilot study. Our study revealed a moderate reduction in joint pain interference (d = -0.465, p=0.43) and a practically significant increase in AI adherence (d = 0.632, p=0.22) from baseline to 4-months post-intervention compared to standard care. From this pilot, we also determined that the exercise-based intervention was safe (no severe adverse event), feasible (we were able to recruit older BCS and complete the intervention) and acceptable (mean 9.7/10 likelihood of recommending to a friend). In accelerometry data, we observed increased physical activity from baseline to post-intervention at four months, and evidence that activity was more effectively maintained by the exercise group compared to standard care over four research assessments. Future Directions A future randomized efficacy trial will test REJOIN among BCS aged ≥60 within one year of initiating AI therapy. The primary outcome will be joint pain interference during daily activity and the secondary outcome will include AI adherence. As part of this future study, we aim to investigate the biological mechanisms driving AI-related joint pain in older breast cancer survivors, focusing on inflammatory biomarkers such as IL-6, TNFα, and CRP. This will provide evidence to clarify origins of AI-related joint pain and inform future therapeutic strategies. Conclusions: REJOIN offers a scalable approach to reduce AI-associated joint pain and support medication adherence in older BCS. This intervention addresses a critical survivorship gap in older adults and may improve quality of life and treatment outcomes, especially in rural and understudied populations.
Presentation numberPS1-05-24
Frequency, characterization, and clinical impact of unexpected cardiac and extracardiac imaging findings in premenopausal women on the CROWN study
Amanda Broderick, Duke University, Durham, NC
A. Broderick1, S. Hatcher1, R. B. D’Agostino, Jr.2, N. O’Connell2, R. Bansal1, C. Anders1, S. Telloni1, K. Westbrook1, A. Natarajan1, N. J. Pagidipati3, D. Wendell4, E. Douglas5, K. Ansley5, C. J. Park6, K. M. Richardson6, S. R. Sirkisoon7, M. Hackney8, H. Vachhani8, M. Ross8, L. N. Vélez-Torres9, J. H. Jordan10, A. Thomas1; 1Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, 2Department of Biostatistics and Data Science, Wake Forest University Health Sciences, Winston-Salem, NC, 3Division of Cardiology, Department of Medicine, Duke University, Durham, NC, 4Division of Cardiology, Cardiovascular Magnetic Resonance Center, Duke University, Durham, NC, 5Department of Cancer Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 6Department of Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 7Clinical Trials Office, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 8Division of Hematology, Oncology and Palliative Care, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, 9Division of Hematology, Oncology and Palliative Care, Massey Comprehensive Cancer Center, Virginia Commonwealth University and University of Puerto Rico Comprehensive Cancer Center, Richmond, VA, 10Pauley Heart Center, Department of Medicine and Department of Biomedical Engineering, Virginia Commonwealth University Health Sciences, Richmond, VA.
Background: Near complete estrogen deprivation (NCED) with ovarian function suppression and aromatase inhibition improves survival outcomes in premenopausal women with HR+ breast cancer (BC). Despite the association of premature menopause with cardiovascular (CV) morbidity in non-cancer populations, the CV health in premenopausal women undergoing BC treatment remains underexplored. As advanced CV imaging is uncommon and guidelines discourage routine cross-sectional imaging in early-stage BC, cardiac and extracardiac imaging findings in these young women are largely uncharacterized. Methods: The Cardiac Outcomes with Near-complete Estrogen Deprivation (CROWN) study is an NIH-funded 3-site study utilizing serial stress cardiac magnetic resonance (CMR) and coronary computed tomography angiography (CCTA) to characterize the natural history of CV health in premenopausal women with stage I-III BC receiving NCED for HR-positive disease and for a comparator cohort with HR-negative disease. Here we describe unexpected cardiac and extracardiac findings on baseline CMR and CCTA imaging of CROWN participants and assess whether these findings led to changes in clinical management. Findings noted were categorized as “unexpected” if they were not included in the study’s primary or secondary endpoints which are myocardial perfusion reserve and other surrogates of large and small vessel disease such as coronary calcium. We excluded clinically insignificant findings such as atelectasis, small hiatal hernias, trace valvular regurgitation, and trace effusions. Results: 72 patients were included (median age of 44 years). 61 patients had HR+ disease (3 HER2+) and 11 had HR- disease (3 HER2+). A total of 37.5% of patients had one or more unexpected finding (37 findings in 27 patients), of which 30 were extracardiac and 7 were cardiac [TABLE]. Of the cardiac findings, 3 were congenital heart disease, 3 were mild structural heart disease, and 1 was a cardiac (vascular) finding. Of all patients, 4.2% were referred to cardiology for management of congenital or vascular (cardiac) findings, and 1.4% were referred for cardiovascular procedures. Further, 9.7% of patients had serial imaging for monitoring of extracardiac findings. 2.8% had a biopsy resulting in diagnosis of BC recurrence. Conclusion: In this first prospective cohort of premenopausal women with BC undergoing serial advanced CV imaging, unexpected baseline cardiac and extracardiac findings leading to clinical action occurred with some frequency, with a handful of findings being of immediate clinical consequence. These results highlight the need to further understand CV health in premenopausal women with BC as we seek to evaluate the impact of both anti-neoplastic therapy and of cancer itself on women with decades of life to protect. [Full cohort data of 90 participants to be presented at the meeting.]
| Type of finding | Number of findings (% of all 37 findings in 27 patients) | Subsequent imaginging-single study (% of patients out of 72) | Subsequent imaging-serial imaging (% of patients out of 72) | Biopsy (% of patients out of 72) | Management and treatment for recurrence (% of patients out of 72) | Referral for cardiology (% of patients out of 72) | Referral for cardiovascular procedure (% of patients out of 72) |
| Congenital Heart Disease | 3 (8.1%) | – | – | – | – | 2 (2.8%) | 1 (1.4%) |
| Mild Structural Heart Disease | 3 (8.1%) | – | – | – | – | 0 | 0 |
| Vascular (cardiac) | 1 (2.7%) | – | – | – | – | 1 (1.4%) | 0 |
| Total Cardiac | 7 (18.9%) | – | – | – | – | 3 (4.2%) | 1 (1.4%) |
| Lung-post radiation | 11 (29.7%) | 0 | 0 | 0 | 0 | – | – |
| Lung- other | 10 (27.0%) | 1 (1.4%) | 7 (9.7%) | 1 (1.4%) | 1 (1.4%) | – | – |
| Liver | 2 (5.4%) | 0 | 0 | 0 | 0 | – | – |
| Vascular (non cardiac) | 3 (8.1%) | 0 | 0 | 0 | 0 | – | – |
| Lymph Node | 1 (2.7%) | 1 (1.4%) | 0 | 0 | 0 | – | – |
| Bone | 1 (2.7%) | 1 (1.4%) | 0 | 0 | 0 | – | – |
| Chest Wall | 1 (2.7%) | 0 | 0 | 1 (1.4%) | 1 (1.4%) | – | – |
| Spleen | 1 (2.7%) | 0 | 0 | 0 | 0 | – | – |
| Total Extracardiac | 30 (81.1%) | 3 (4.2%) | 7 (9.7%) | 2 (2.8%) | 2 (2.8%) | – | – |
| Total Cardiac and Extracardiac | 37 (100%) | 3 (4.2%) | 7 (9.7%) | 2 (2.8%) | 2 (2.8%) | 3 (4.2%) | 1 (1.4%) |
Presentation numberPS1-05-25
The Relationship between Post-Diagnosis Recreational Physical Activity and Breast Cancer Recurrence: A Systematic Review and Meta-Analysis
Jillian Weathington, University of Central Florida, Orlando, FL
J. Weathington, I. Lynch, B. Sukhu, T. Glatz, E. Lee; Health Sciences, University of Central Florida, Orlando, FL.
Background: Breast cancer (BC) recurrence risk is highest within the first 5-years following initial diagnosis, making survivorship care a critical window for intervention. As survivorship rates increase, identifying modifiable lifestyle factors that may reduce recurrence risk becomes increasingly important. While the protective factors of recreational physical activity (rPA) before BC diagnosis are well-documented, the role of post-diagnosis rPA, particularly its impact on recurrence remains less explored. Objectives: This study aimed to determine 1) if high levels of post-diagnosis rPA reduce BC recurrence risk in female survivors compared to minimal rPA and 2) if adhering to the Physical Activity Guidelines for Americans (i.e., 150 minutes of moderate-intensity physical activity weekly) reduces recurrence risk. Methods: We included observational cohort studies evaluating the association between post-diagnosis rPA and recurrence risk among adult female BC survivors published up to January 2025, in any language or geographic location. Excluded studies were interventional studies, hospital/clinical studies, male BC survivors, and those under 18 years. Databases (Medline, CINAHL, Web of Science) were searched in February 2025. Two reviewers independently screened studies at all stages. Data synthesis and analysis used pooled risk estimates (RR/HRs) and 95% confidence intervals. A meta-analysis was conducted on Stata using a random-effects model to examine the effects of post-diagnosis of rPA on BC recurrence, separately for minimal vs high rPA and meeting vs not meeting recommended guidelines. Subgroup analyses explored BC subtypes, study characteristics, menopausal status, and follow-up period as potential sources of heterogeneity. Heterogeneity was assessed using Cochran’s Q test and I2 statistics. Results: Out of 2,635 studies screened, 22 passed the initial screening for full-text review, and 9 were included in the meta-analysis (9 high versus low; 5 meeting vs not meeting guidelines). The analysis included 37,787 BC survivors and 4,524 recurrences were reported. Meta-analysis showed that female BC survivors who engaged in high levels of rPA after diagnosis had a 13% lower risk of BC recurrence compared to those with minimal activity (RR=0.87, CI=0.77-0.96, I2=0.00%, n=9). However, meeting the minimum recommendations of rPA alone was not associated with a reduced risk of recurrence (RR=0.93, 95% CI=0.85-1.02, I2=0.00%, n=5). Subgroup analysis showed stronger associations in studies conducted in the Netherlands and Germany, as well as in those with rPA assessment periods greater than 5 years after diagnosis. Using the Newcastle Ottawa Scale, study quality was classified as good (n=7) or fair (n=2). Conclusion: These findings suggest that high levels of rPA postdiagnosis reduces recurrence risk by 13%. Because the current minimum recommendations may not be sufficient to reduce recurrence risk, further research is needed to determine optimal rPA recommendations and guidelines for BC survivors. Additionally, these findings emphasize the value of healthcare professionals promoting higher levels of rPA within survivorship care plans for BC survivors.
Presentation numberPS1-05-26
Vaginal estrogen versus non-hormonal moisturizer for breast cancer survivors on aromatase inhibitor therapy: a randomized, controlled trial
Polly Niravath, Houston Methodist Hospital, Houston, TX
P. Niravath1, M. Rimawi2, A. Puri1, K. Sun1, H. Mai1, S. Mathur1, S. Hilsenbeck2, D. Antosh3, R. High3, S. Hoopes3, L. Langsjoen3, A. Brock1, J. Nangia2, J. Chang1, C. Osborne2; 1Internal Medicine, Houston Methodist Hospital, Houston, TX, 2Internal Medicine, Baylor College of Medicine, Houston, TX, 3Gynecology, Houston Methodist Hospital, Houston, TX.
Background: Among post-menopausal breast cancer survivors, more than 60% experience atrophic vaginitis, often resulting in impaired quality of life and sexual dysfunction. Nonetheless, vaginal dryness is commonly an unmet need among breast cancer survivors. While vaginal estrogen is a proven, effective treatment for atrophic vaginitis, it has not been adequately studied in Estrogen Receptor-positive (ER+) breast cancer survivors. Methods: In this randomized, controlled, unblinded clinical trial, we enrolled ER+ breast cancer survivors on adjuvant aromatase inhibitor (AI) therapy who were suffering from atrophic vaginitis. Women were assigned to receive either Replens® (a non-hormonal vaginal moisturizer) or vaginal estrogen. Based on patient preference, patients randomized to the estrogen arm chose between 6 months of Vagifem® vaginal tablets (10 mcg daily for the first 2 weeks, then twice a week thereafter) or Estring® vaginal ring (2 mg, placed every 3 months). We tested serum estradiol levels by tandem mass spectrometry (TMS) at several time points in the estrogen arm, and only at baseline & end of study in the Replens arm. Study duration was 24 weeks. The primary endpoint was vaginal dryness, as measured by a validated questionnaire. Secondary endpoints include sexual functioning, vaginal pH, and serum estradiol values Results: Twenty-three patients were enrolled on the study, with 12 patients assigned to the Replens arm. Of the 11 patients assigned to the vaginal estrogen arm, 8 chose Estring and 3 chose Vagifem. The Vaginal Dryness scores (assessed on a scale from 0 to 4) in the estrogen arm improved by an average of 1.6 points, compared to 0.9 points in the Replens arm, which was a statistically significant improvement (p = 0.044). Similarly, there was a more significant reduction in vaginal pH in the vaginal estrogen arm as compared to the non-hormonal treatment, indicating return to healthier vaginal environment in the estrogen arm (p = 0.0286). We observed an improvement in patient-assessed dyspareunia, which was assessed on a scale from 0 to 10. Women on the estrogen arm saw an improvement of 4.8 points, whereas the women receiving nonhormonal treatment improved by an average of 1.7 points (p = 0.048). However, other sexual functioning parameters (libido, ability to achieve orgasm, sexual satisfaction, and relationship with partner) were not significantly different between the 2 groups. Serum estradiol levels remined low through the first 12 weeks in the estrogen arm and exceeded safety threshold (27 pg/mL at 2 weeks, or 12 pg/mL for the remainder of the study) for two of the 11 patients in the estrogen arm at 24 week. One of these patients was taking Vagifem, and her estradiol level rose to 21.8 pg/mL at 6 months. The other patient was using Estring, and her estradiol level increased to 16 pg/mL. Conclusions: Vaginal estrogen is a more effective treatment for vaginal dryness and dyspareunia compared to nonhormonal vaginal moisturizer. Serum estradiol levels remained low for 12 weeks, suggesting this duration of treatment may be considered in select patients. Some patients had an increase in estradiol level at 24 weeks, though its cause and clinical significance are uncertain. Further study is warranted as to why the serum estradiol rise occurred with a longer period of vaginal estrogen treatment.
Presentation numberPS1-05-27
A Multicenter Randomized Controlled Trial to Verify the Preventive Effect of Denosumab against Cancer Treatment-Induced Bone Loss (CTIBL) in Breast Cancer Patients Receiving Postoperative Endocrine Therapy with Normal Bone Density.
Midori Morita, Kyoto Prefectural University of Medicine, Kyoto, Japan
M. Morita1, A. Watanabe1, K. Sakaguchi1, I. Takashi2, Y. Hasegawa3, A. Sato4, S. Takao5, K. Tane6, M. Takahashi7, K. Watanabe8, M. Kato9, Y. Ouchi10, N. Kono11, T. Kubota12, M. Suzuki13, J. Horiguchi14, K. Narui15, D. Miura16, K. Terata17, K. Imai18, R. Takekawa19, I. Yokota20, A. Naito21, S. Sakabayashi19, Y. Naoi1, T. Taguchi1; 1Department of Surgery, Division of Endocrine & Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, JAPAN, 2Department of Breast Oncology and Surgery, Tokyo Medical University Hospital, Tokyo, JAPAN, 3Department of Breast Surgery, Hachinohe City Hospital., Aomori, JAPAN, 4Department of Medical Oncology, Hirosaki University Graduate School of Medicine, Aomori, JAPAN, 5Department of Breast Surgery, Kohnan Medical Center, Hyogo, JAPAN, 6Department of Breast Surgery, Hyogo Cancer Center, Hyogo, JAPAN, 7Department of Breast Surgery, Hokkaido University Hospital, Hokkaido, JAPAN, 8Department of Breast Surgery, Hokkaido Cancer Center, Hokkaido, JAPAN, 9Breast Surgery, Kato Breast Clinic, Shiga, JAPAN, 10Breast Surgery, Saiseikai Shiga Hospital, Shiga, JAPAN, 11Breast Surgery, Kobe Kaisei Hospital, Hyogo, JAPAN, 12Department of Breast Surgery, Kamiiida Daiichi General Hospital, Aichi, JAPAN, 13Department of Breast Surgery, National Hospital Organization Chiba Medical Center, Chiba, JAPAN, 14Department of Breast Surgery, International University of Health and Welfare, Chiba, JAPAN, 15Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Kanagawa, JAPAN, 16Breast Surgery, Akasaka Miura Clinic, Tokyo, JAPAN, 17Department of Breast and Endocrine Surgery, Akita University Hospital, Akita, JAPAN, 18Department for Medical Innovation and Translational Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JAPAN, 19Division of Data Science, The Clinical and Translational Research Center, University Hospital, Kyoto Prefectural University of Medicine, Kyoto, JAPAN, 20Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Hokkaido, JAPAN, 21Department of Biostatistics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, JAPAN.
Background: Aromatase inhibitors (AIs) are widely used as postoperative endocrine therapy in patients with postmenopausal hormone receptor-positive breast cancer. The main side effect of AIs is bone loss, which is called cancer treatment-induced bone loss (CTIBL), and the use of bone resorption inhibitors are recommended when the risk of fracture is high. However, we do not yet have clear data on the preventive effects of bone resorption inhibitors and their safety in patients with normal bone density. In this study, we aimed to evaluate the preventive effects of denosumab on suppressing bone loss associated with postoperative endocrine therapy for Japanese breast cancer patients with normal bone mineral density.Methods: A multicenter, open-label, randomized controlled trial was conducted. Patients with postmenopausal hormone receptor-positive breast cancer of stage ≦IIIA with normal bone density meeting lumbar spine T-score ≧-1.0 and femoral neck T-score ≧-1.0 were included. Patients were assigned in a 1:1 ratio to receive the hormone alone (control group) or an additional 60 mg of denosumab every 6 months (denosumab group). The primary endpoint was the percentage change in lumbar spine (L1-L4) bone mineral density (BMD) at 12 months. Secondary endpoints included the percentage change in lumbar spine BMD (2-5 years later), femoral neck BMD change, pathological fracture incidence, adverse events, progression-free survival, and overall survival.Results: A total of 83 cases were registered, and 66 were included in the primary analysis. At 12 months, the mean percentage change in lumbar spine BMD was +4.56% in the denosumab group (N=32) and -3.66% in the control group (N=34), yielding a significant improvement of +8.1% (95%CI: 6.2%-10.1%, p<0.01) in analyses adjusting for several covariates in a linear model.Within three years, there were no fractures in the denosumab group and one in the control group (p=0.53). Adverse events of any grade occurred in 13 cases (33.3%) in the denosumab group and 12 (31.6%) in the control group; there were no cases of osteonecrosis of the jaw or atypical femoral fractures. No significant differences were observed in disease-free survival or overall survival.Conclusion: In Japanese breast cancer patients with normal bone density, the combination of denosumab and postoperative adjuvant endocrine therapy significantly prevented the decline in lumbar spine BMD. This intervention showed a good safety profile without severe adverse events and the potential to prevent the occurrence of fractures. These findings support the consideration of denosumab as part of a comprehensive strategy for preserving bone health in a broader range of patients, including those with initially normal BMD who are receiving endocrine therapy for breast cancer. Future research should aim to explore long-term fracture outcomes, cost-effectiveness, and the integration of bone health strategies into standard oncology care.
Presentation numberPS1-05-28
Evaluating all-cause mortality and endocrine therapy tolerability with sodium glucose cotransporter-2 inhibitors in hormone receptor-positive breast cancer: A real-world analysis
Colton Jones, University of Texas Health Science Center San Antonio, San Antonio, TX
C. Jones1, J. Michalek2, E. Obomanu3, Y. Arya4, A. Syal5, S. Haddad1, V. Kaklamani6, S. Shaikh6; 1Hematology/Oncology, University of Texas Health Science Center San Antonio, San Antonio, TX, 2Population Health Sciences, University of Texas Health Science Center San Antonio, San Antonio, TX, 3Hematology/Oncology, Jefferson Einstein Hospital, Philadelphia, PA, 4Hematology/Oncology, Mayo Clinic Jacksonville, Jacksonville, FL, 5Hematology/Oncology, Mayo Clinic Jacksonville, San Antonio, FL, 6Hematology/Oncology, Mays Cancer Center, MD Anderson, San Antonio, TX.
Introduction: Endocrine therapy (ET) is central to treating hormone receptor-positive breast cancer (BC), but its side effects impact adherence and quality of life, especially in patients with metabolic dysfunction. Sodium-glucose cotransporter-2 (SGLT-2) inhibitors offer cardiometabolic benefits, potentially improving inflammation, glycemic control, and physical well-being. However, data on SGLT-2 use in BC patients is scarce. This study is the first to evaluate the real-world effects of SGLT-2 inhibitors on all-cause mortality and ET tolerability in BC patients. Methodology: We conducted a real-world analysis using the TrinetX database, which includes data from 184 million patients in 156 healthcare organizations, to identify BC patients on ET (letrozole, anastrozole, exemestane, tamoxifen) who received SGLT-2 inhibitors vs. those who did not. Patients were grouped by age (≤50 and ≥51 years as a surrogate for menopausal status. Primary outcome: all-cause mortality from the start of the index event, defined as the first day meeting BC diagnosis and ET use, with or without SGLT-2. Secondary endpoint: ET tolerability. Propensity score matching completed by TriNetX adjusted for confounders. Multivariate analysis yielded odds ratios and risk differences with 95% CI. Results: Among 1,136 BC patients ≤50 years, 568 were assigned to each group (SGLT-2 vs non-SGLT-2). For patients ≥51 years (n=28,600), each group included 14,300. Demographics across cohorts were comparable. No mortality benefit was seen in premenopausal patients. In postmenopausal women, SGLT-2 use reduced all-cause mortality—647/13,676 events (SGLT-2) vs 953/13,517 (non-SGLT-2) [OR: 0.655 (0.591-0.726)]. In premenopausal patients, SGLT-2 reduced endometrial cancer risk [RD: -0.018 (-0.029, -0.007)] but increased diarrhea risk [OR: 2.305 (1.078, 4.930)]. No significant differences were found in hot flashes, PE, DVT, depression, osteoporosis, joint pain, fatigue, nausea/vomiting, abdominal pain, or dizziness. Among postmenopausal patients, SGLT-2 reduced PE [OR: 0.757 (0.605-0.948)] and osteoporosis [OR: 0.877 (0.784-0.981)], but increased risks of depression [OR: 1.153 (1.007-1.319)], nausea/vomiting [OR: 1.120 (1.001-1.253)], diarrhea [OR: 1.193 (1.058-1.345)], and dizziness [OR: 1.234 (1.103-1.380)]. No significant differences were found for hot flashes, DVT, EC, joint pain, fatigue, or abdominal pain. Conclusion: This analysis reveals novel evidence supporting the cardiometabolic potential of SGLT-2 inhibitors in hormone receptor-positive BC, particularly postmenopausal populations. These agents were linked to lower all-cause mortality and improved tolerance of specific ET-related effects. However, certain adverse risks require caution. Future prospective studies are essential to validate their role and safety in oncologic care.
Presentation numberPS1-06-01
Predictive Value of Breast-Specific Gamma Imaging for Pathologic Response and Prognosis in Early Breast Cancer After Neoadjuvant Therapy: A Prospective Trial
Tianyi Qian, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
T. Qian1, X. Ye1, Y. Wu2, L. Pang1, L. Li2, X. Yu1, Z. Wang1, J. Huang1; 1Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, CHINA, 2Department of Medicial Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, CHINA.
Background: Neoadjuvant chemotherapy (NAC) is the standard treatment for locally advanced breast cancer. Achieving a pathologic complete response (pCR) is strongly associated with favorable long-term outcomes. However, current imaging modalities, such as magnetic resonance imaging (MRI), have limitations in quantifying residual metabolic activity following NAC. Breast-specific gamma imaging (BSGI) provides functional assessment through the tumor-to-normal ratio (TNR), with established diagnostic value in prior studies. Prospective validation of TNR’s predictive value for pCR and survival outcomes is currently lacking. This study aims to evaluate whether post-NAC TNR can identify patients with chemo-sensitive tumors and a superior prognosis. Methods: This single-center prospective trial (NCT02556684) enrolled 137 patients with stage I-III breast cancer who received standard NAC followed by surgery between 2014 and 2023. Inclusion criteria required patients to have clinical stage T1-4 and N0-3, baseline biopsy-confirmed invasive carcinoma, and completion of the planned NAC regimen. Exclusion criteria were bilateral or metastatic disease or incomplete NAC. Breast-specific gamma imaging was performed after 2 cycles of NAC using 99mTc-sestamibi (MIBI) and dual-head gamma cameras. The tumor-to-normal ratio (TNR) was calculated as the maximum tumor uptake divided by the mean uptake in the contralateral breast parenchyma. The primary endpoints were pathological complete response (pCR) and 3-year disease-free survival (DFS). Statistical analyses included χ² tests for associations, Kaplan-Meier analysis with log-rank tests for survival comparisons, and multivariate Cox regression models adjusted for clinicopathologic covariates. Results: Baseline characteristics were balanced between groups for age, nodal status, grade, and Ki-67 (p>0.05), though TNR-low patients had significantly higher HER2+ subtype prevalence (65% vs. 46%, p=0.026). TNR-low was strongly associated with pathologic complete response (pCR), with 34.8% achieving pCR versus 14.7% in TNR-high group. Survival analysis demonstrated significantly improved disease-free survival (DFS) for TNR-low patients after median follow-up of 42.5 months. DFS benefits were consistently observed in the overall population and key clinical subgroups: overall cohort (hazard ratio [HR]=0.33, 95% confidence interval [CI]:0.14-0.81, p=0.002), HR+HER2- (HR=0.23; 95% CI:0.07-0.73; p=0.001), HER2+ (HR=0.23; 95% CI:0.04-1.48; p=0.009), pCR patients (HR=0, p=0.016), and non-pCR patients (HR=0.42; 95% CI:0.18-1.00; p=0.02). The triple-negative subgroup showed non-significant trends likely due to limited sample size (n=19). TNR demonstrated independent prognostic value in multivariate Cox regression. After adjusting for established factors (including pCR status, T stage, nodal stage, Grade and Ki-67 index), TNR-low status confirmed as a significant predictor of superior disease-free survival (DFS), with a hazard ratio of 0.43 (95% CI: 0.20-0.93; p=0.031). Notably, TNR demonstrated predictive capacity beyond pathologic complete response. No endpoint events occurred in TNR-low/pCR patients, whereas TNR-low/non-pCR patients had 58% lower risk of recurrence versus TNR-high/non-pCR patients. Conclusion: These results establish TNR as a robust imaging biomarker which serves as a valuable complement to current assessment methods. The prospective design and consistent survival benefit across multiple subgroups support clinical integration of BSGI-derived parameters to guide post-neoadjuvant risk stratification, potentially enabling therapy escalation for TNR-high/non-pCR patients and de-escalation strategies for TNR-low/pCR patients.
Presentation numberPS1-06-03
Lymph Node Evaluation in Breast Cancer: A Comparative Study of CNB and FNA
Marina Diogenes, Hospital da Mulher, São Paulo, Brazil
M. Diogenes1, M. Antonini2, A. Mattar1, F. P. Cavalcante3, F. Zerwes4, F. Brenelli5, A. Frasson6, M. Eduardo7, G. Facina8, H. L. Couto9, S. Rondelo1, R. Arakelian1, V. Rocha10, L. Chrispim11, M. Madeira12, L. Gebrim13, L. Okumura14, P. Baruel14, R. Lopes2; 1Mastology, Hospital da Mulher, São Paulo, BRAZIL, 2Mastology, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, BRAZIL, 3Mastology, Hospital Geral de Fortaleza, Fortaleza, BRAZIL, 4Mastology, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 5Mastology, Universidade Estadual de Campinas, Campinas, BRAZIL, 6Mastology, Hospital Albert Einstein, São Paulo, BRAZIL, 7Mastology, Americas Oncologia, Rio de Janeiro, BRAZIL, 8Mastology, Universidade Federal de São Paulo, São Paulo, BRAZIL, 9Mastology, Redimama – Redimasto, Belo Horizonte, BRAZIL, 10Mastology, Universidade Nove de Julho, São Paulo, BRAZIL, 11Mastology, Pérola Centro de Pesquisa em Oncologia, São Paulo, BRAZIL, 12Mastology, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, BRAZIL, 13Mastology, Hospital Beneficiencia Portuguesa de São Paulo, São Paulo, BRAZIL, 14Statistics, Verde Health Care Consultancy, Curitiba, BRAZIL.
Background: accurate lymph node evaluation is essential for breast cancer staging, treatment planning and diagnosis of recurrent disease. Fine-needle aspiration (FNA) and core needle biopsy (CNB) are the two main percutaneous techniques used, but they differ in diagnostic yield, complication rates and associated healthcare costs. Objectives: to compare the diagnostic performance and economic impact of FNA versus CNB for lymph node evaluation in breast cancer patients within the Brazilian public healthcare system, emphasizing the relevance of cost-effective strategies in low- and middle-income countries (LMICs). Methods: a retrospective cohort study was conducted, including women with suspicious lymph nodes who underwent FNA or CNB between 2015 and 2023. A 1-year budget impact model nested with a decision tree model was used as economic model. Probabilities from the cohort study were used as clinical inputs. Costs were obtained from Sigtap (Brazilian listing of procedures reimbursement). The economic analysis considered only the procedural cots, assuming that each patient could undergo either FNA or CNB as the initial approach. In cases of inconclusive results, the patient could then undergo CNB or surgery. All analyses were reported as American dollar (USD), using an exchange rate of 6 brazilian reais to 1 USD. Results: the 300 women included in the study had similar ages (FNA: 53 vs CNB: 54 years old, average). The overall rate of inconclusive results in the sample was 15%. The rate of inconclusive results in procedures performed with CNB was lower than FNA (2.7% vs. 50.7%, p<0.05). Among the 79 patients who underwent FNA as the initial procedure, the total cost was USD 16,901, whereas for the 221 women who initially underwent CNB, the cost was USD 9,930. Although the initial FNA procedure was less expensive (USD 1,755) than the initial CNB procedure (USD 8,185), FNA resulted in a 17-fold higher rate of inconclusive results, and the need for additional procedures made CNB more cost-effective overall. A scenario in which only FNA was available as the initial procedure resulted in 11% higher overall costs compared to the base-case scenario (USD 26,832). Conversely, if CNB were available to all patients as the initial procedure, overall costs would be USD 13,718, approximately 50% lower than the base-case scenario. Conclusion: core needle biopsy offers superior diagnostic performance and greater economic efficiency compared to fine-needle aspiration for lymph node evaluation in breast cancer. Prioritizing CNB as the first-line diagnostic approach could improve resource allocation in public healthcare systems, particularly in LMICs where cost-effectiveness is crucial to expanding access to high-quality cancer care.
Presentation numberPS1-06-04
Factors Associated with Receipt of Magnetic Resonance Imaging for Breast Cancer Screening Among Racially/Ethnically Diverse High-Risk Women
Alissa Michel, Columbia University Irving Medical Center, New York, NY
A. Michel1, J. Zahlan2, K. D. Crew1; 1Medical Oncology, Columbia University Irving Medical Center, New York, NY, 2Public Health, Columbia University Mailman School, New York, NY.
Introduction: Recent randomized controlled trials (RCTs) of breast MRI combined with mammography among high-risk women resulted in fewer interval breast cancer (BC) cases and earlier cancer detection compared to mammography alone. Despite this finding, supplemental breast MRI is underutilized. We aimed to identify factors associated with the receipt of supplemental breast MRI in a racially/ethnically diverse population of high-risk women.Methods: We conducted a retrospective cohort study among high-risk women undergoing screening mammography at Columbia University Irving Medical Center (CUIMC) in New York, NY, from 2007-2024. BC risk categories were classified based upon ICD-9/10 diagnostic codes or findings from breast radiology/pathology reports: 1) at high risk for BC, 2) family history of BC, 3) extremely dense breasts, 4) high-risk breast lesion (atypical hyperplasia [AH] or lobular carcinoma in situ [LCIS]), and 5) genetic predisposition for BC. Patients with a diagnosis of BC prior to 2007 were excluded. We collected information on demographic and clinical characteristics in the electronic health record (EHR), including age, race, ethnicity, primary health insurance, body mass index, mammographic density (BIRADS classification), benign breast biopsies, and family history of BC. Our primary outcome was receipt of supplemental breast MRI based upon radiology reports in the EHR. A multivariable logistic regression model was used to determine the association between breast MRI receipt and demographic and clinical characteristics.Results: Of the 54,122 women included in our analysis, the mean age was 58 years (SD, 10), including 29.0% non-Hispanic White women, 34.0% Hispanic, 12.0% non-Hispanic Black, 3.9% Asian, and 5.1% other or multiracial. In terms of BC risk factors, 4.5% of the women had extremely dense breasts, 16.0% had a family history of BC, 2.0% had AH or LCIS, and 1.6% had a genetic predisposition for BC. Overall, only 3.6% of women received a supplemental breast MRI. In multivariable analysis, younger age was significantly associated with breast MRI receipt, with women aged 25-34 demonstrating higher odds of undergoing supplemental breast MRI (odds ratio [OR]=2.59; 95% confidence interval [CI]=1.77-3.72) compared to those aged 45-54. Compared to non-Hispanic White women, Asian and Hispanic women had significantly lower odds of receiving a breast MRI (OR=0.72, 95% CI=0.63-0.82 and OR=0.66, 95% CI=0.51-0.84, respectively). Having extremely dense vs. less dense breasts on mammography (BIRADS D vs. B) was associated with higher breast MRI uptake (OR=1.99, 95% CI=1.35-2.92). Compared to women with a diagnostic code for at high risk for BC, those in other risk categories had significantly higher odds of receiving a screening breast MRI: family history of BC (OR=7.14, 95% CI=6.36-8.02), genetic predisposition to BC (OR=26.6, 95% CI=22.30-31.70), and high-risk breast lesions (OR=28.1, 95% CI=23.70-33.30). We did not observe differences in breast MRI use among Black vs. White women or based upon health insurance status (private vs. public).Conclusion: In a racially/ethnically diverse cohort of women at high risk for BC, only 3.6% received a supplemental breast MRI. Utilization varied significantly by age, race/ethnicity, and specific BC risk factors. These findings highlight persistent gaps in the uptake of guideline-concordant supplemental BC screening. We plan to study the impact of breast MRI use on early detection of local disease and screen-detected vs. interval BC. Future research should investigate patient-, provider- and system-level barriers to adoption of supplemental breast MRI among high-risk women to promote equitable access to advanced screening modalities.
Presentation numberPS1-06-05
The Early and Late Predictive Value of <sup>18</sup>F-FAPI-42 PET/MRI for Neoadjuvant Therapy Response in Breast Cancer: A Prospective Study
Sihua Liu, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
S. Liu1, Y. Zhang2, Y. Yang2, D. Zhai1, X. Kuang1, Y. Shi1, L. Yu1, Y. Zhang1, N. Shao1, X. Zhang2, Y. Lin1; 1Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, CHINA, 2Department of Nuclear Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, CHINA.
Objectives and rationale: Early response prediction during neoadjuvant therapy (NAT) for breast cancer (BC) enables response-guided precision treatment. Imaging-based post-NAT evaluation may reduce dependence on pathological complete response (pCR) confirmation and facilitate surgical de-escalation. Fibroblast activation protein inhibitor (FAPI) PET/MRI represents a promising molecular imaging modality targeting the tumor microenvironment. This study aimed to evaluate the efficacy of 18F-FAPI-42 PET/MRI for early and late prediction of pCR to NAT in BC. Methods: 45 consecutive patients with newly diagnosed BC who met the criteria for NAT were prospectively enrolled and underwent 18F-FAPI-42 PET/MRI before treatment initiation (PET1), after two cycles of NAT (PET2), and before post-NAT surgery (PET3). SUVmax was measured in the primary tumor and metastatic lymph nodes (MLNs). The tumor-to-background ratio (TBR) was calculated as the lesion SUVmax divided by the SUVmean of contralateral normal breast tissue. The percentage changes in SUVmax and TBR from baseline were calculated after 2 NAT cycles (ΔSUVmax1 and ΔTBR1) and before surgery (ΔSUVmax2 and ΔTBR2). Predictive performance of 18F-FAPI uptake was analyzed by receiver operating characteristic curve analysis and the area under the curve (AUC). The Youden index determined optimal cutoffs. Results: 18 patients (40%) achieved pCR, while 27 patients (60%) exhibited residual disease. Among 40 patients with MLNs, 16 (40%) attained axillary pCR. By molecular subtypes, pCR rates were 21.1% (4/19) in HR-positive/HER2-negative, 52.4% (11/21) in HER2-enriched, and 60.0% (3/5) in TNBC cases. The ΔSUVmax of primary tumors (r = 0.636; P < 0.001) and MLNs (r = 0.560; P < 0.001) were significantly correlated between PET2 and PET3. Patients achieving pCR demonstrated significantly lower absolute SUVmax and TBR values at both PET2 and PET3 compared to non-pCR cases (P < 0.05). Furthermore, greater reductions in both SUVmax and TBR at any time point were strongly associated with pCR (P < 0.05). In predicting pCR, the ΔSUVmax1 (AUC = 0.792; sensitivity = 55.6%; specificity = 92.6%) and ΔTBR1 (AUC = 0.774; sensitivity = 61.1%; specificity = 100%) of the primary tumor had the highest specificity; the TBR3 (AUC = 0.855; sensitivity = 94.1%; specificity = 76.0%) and ΔTBR2 (AUC = 0.815; sensitivity = 94.1%; specificity = 72.0%) of the primary tumor showed the highest sensitivity. In the HR-positive/HER2-negative subtype, parameters derived from PET2 (SUVmax2 and TBR2; AUC = 0.950; sensitivity = 100%; specificity = 86.7%) and all parameters derived from PET3 (SUVmax3, ΔSUVmax2, TBR3, and ΔTBR2; AUC = 1.000; sensitivity = 100%; specificity = 100%) of the primary tumor showed optimal AUC in predicting pCR; both the ΔSUVmax1 and ΔSUVmax2 of MLNs achieved 100% sensitivity for axillary pCR prediction. Conclusions: Early-phase 18F-FAPI-42 PET/MRI demonstrates high specificity in predicting pCR, which may facilitate the selection of alternative NAT regimens for non-pCR cases. Preoperative 18F-FAPI-42 PET/MRI shows high sensitivity for pCR prediction, which can identify potential candidates for surgery de-escalation. 18F-FAPI-42 PET/MRI exhibits excellent potential for both early and late-phase prediction of pathological response to NAT in HR-positive/HER2-negative BC.
Presentation numberPS1-06-06
Randomized phase III study comparing intensive follow-up with standard follow-up in post-operative high risk breast cancer <INSPIRE trial: JCOG1204>: the early analysis of Relapse-free survival and Onset of recurrence
Takashi Hojo, Saitama Medical Center, kawagoeshi, Japan
T. Hojo1, N. Masuda2, H. Iwata3, A. Yoshimura4, T. Ueno5, M. Tsuneizumi6, T. Onishi7, H. Matsumoto8, Y. Yanagita9, M. Sawaki10, H. Kawabata11, S. Takayama12, H. Shigematsu13, T. Fujisawa9, F. Hara4, R. Kajikawa14, K. Sasaki15, T. Shien16; 1Department of Breast Care, Saitama Medical Center, kawagoeshi, JAPAN, 2Department of Breast Surgery, Kyoto University,, Sakyo-ku, JAPAN, 3Department of Advanced Clinical Research and Development, Nagoya City University Graduate School of Medical Sciences, Nagoya, JAPAN, 4Department of Breast Oncology, Aichi Cancer Center, Nagoya, JAPAN, 5Department of Breast Center, Breast Oncology Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Koto, JAPAN, 6Department of Breast Surgery, Shizuoka General Hospital, Shizuoka City, JAPAN, 7Department of Breast Surgery, National Cancer Center Hospital East, Kashiwa, JAPAN, 8Department of Breast Surgery, Saitama Cancer Center, Kitaadachi-gun, JAPAN, 9Department of Breast Oncology, Gunma Prefecture Cancer Center, Ota, JAPAN, 10Department of Breast Surgery, NHO Nagoya Medical Center, Naka-ku, JAPAN, 11Department of Breast and Endocrine Surgery, Toranomon Hospital, Minato-ku, JAPAN, 12Department of Breast Surgery, National Cancer Center Hospital, Chuo-ku, JAPAN, 13Department of Breast Surgery, Hiroshima University Hospital, Minami-ku, JAPAN, 14Department of JCOG Operations Offic, Data Center, Chuo-ku, JAPAN, 15Department of JCOG Operations Office, Data Center, Chuo-ku, JAPAN, 16Department of Breast and Endocrine Surgery, Okayama University Hospital, Kitaku, JAPAN.
Background: For postoperative follow-up of breast cancer, only annual MMG is recommended, commonly in guidelines around the world. Early detection of metastasis and recurrence has been considered of little significance. However, there have been remarkable advances in the development of new drugs, and with the improved accuracy of imaging tests for early detection and recent innovations in MRD assay technology, the importance of early detection of metastasis and recurrence in other cancer tumors is being emphasized. Methods: The INSPIRE trial (Intensive follow (IF) vs. standard follow (SF) post-operative surveillance in high-risk breast cancer patients; JCOG1204) is a randomized phase III study due to confirm the superiority of IF in terms of overall survival (OS) in high-risk breast cancer patients after radical surgery. Eligible participants were from 20 to 70 years old and had undergone radical breast and axillary surgery. Patients were randomized 1:1 to the SF arm (annual MMG and tumor markers) or the IF arm (annual MMG, whole-body CT or PET/CT every 6 months until 3 years and every 12 months until 5 years after surgery, and tumor marker every 3 months until 3 years and 6 months until 5 years after surgery). Adjustment factors: ER and HER2 status, with/without preoperative chemotherapy, institutions. Primary endpoint: OS. Key secondary endpoints: disease free survival, relapse-free survival (RFS), distant metastasis-free survival (DMFS). Five-year follow-up was completed for all enrolled patients. Data cut-off is Jan.2025.Results: From Nov 2013 to Jan 2020, 1034 pts were enrolled. 516 and 518 were assigned to SF and IF, respectively. Median follow up time is 82.9 (range: 0.5-130.9) months. The patient’s characteristics were well balanced between two arms: median age 53 (range: 23-70) years, PS 0/1:1018/16, pre/post menopause: 486/548, median tumor size: 3.0 (range:0-23 cm), stage 1A/2A/2B/3A/3B/3C: 78/262/346/153/83/110, ER+/-: 617/417, HER2+/-: 218/816, preoperative chemo +/-: 596/438, 7Y RFS: 73.0% vs 73.6% in SF and IF arm, respectively (p=0.999, 95% confidence interval [CI]:0.79-1.26). 7Y DMFS: 74.1% vs 75.5% in SF and IF arm, respectively (p=0.974, 95% CI: 0.76-1.24). The duration to recurrence/distant recurrence is 29.3/31.7 and 24.0/24.1 months in SF and IF, respectively. Lead times (the difference between SF and IF) of all and each subtype are shown in Table.Conclusion: There was no significant difference in RFS/DMFS between IF and SF arm. Recurrence was detected earlier by IF compared to SF, and the median time to recurrence was longer in luminal type than in TN type. However, the lead times were similar. The results of continued follow-up are awaited to see if this will lead to an improvement in overall survival, which is the primary endpoint.
|
Standard follow (months) |
Intensive follow (months) |
Lead time (months) |
|
| Duration to recurrence | Median (range) | Median (range) | |
| all pts (n:138/136) | 29.3 (1.3-108.6) | 24.0 (0.7-114.1) | 5.3 |
| Luminal type (n:74/77) | 45.8 (2-108.6) | 39.5 (2.9-100.8) | 6.3 |
| HER2 type (n:27/22) | 16.1 (2.3-70.2) | 14.9 (0.7-96.4) | 1.2 |
| TN type (n:37/37) | 16.1 (1.3-89.2) | 10.8 (2.4-114.1) | 5.3 |
| Duration to distant recurrence | |||
| all pts (n:131/126) | 31.7 (2-108.6) | 24.1 (2.4-114.1) | 7.6 |
| Luminal type (n:72/74) | 45.8 (2-108.6) | 37.7 (2.9-100.8) | 8.1 |
| HER2 type (n:25/17) | 21.1 (2.3-70.2) | 28.1 (2.4-96.4) | -7.0 |
| TN type (n:34/35) | 16.6 (2.6-61.3) | 10.8 (2.4-114.1) | 6.4 |
Presentation numberPS1-06-07
Non-contrast detection of breast cancer using high-resolution restriction spectrum imaging
Stephane Loubrie, UC San Diego, La Jolla, CA
S. Loubrie1, J. Lim2, S. Batasin1, H. J. Yu1, J. Zou3, S. Ebrahimi1, C. C. Conlin1, A. Guidon4, T. M. Seibert5, A. M. Dale6, A. Wallace7, H. Ojeda-Fournier1, A. E. Rodriguez-Soto1, R. Rakow-Penner8; 1Radiology, UC San Diego, La Jolla, CA, 2Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong-si, KOREA, REPUBLIC OF, 3Herbert Werteim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA, 4Global MR and Workflow, GE Healthcare, Boston, MA, 5Radiology, Bioengineering & Radiation oncology, UC San Diego, La Jolla, CA, 6Radiology & Neurosciences, UC San Diego, La Jolla, CA, 7Surgery, UC San Diego, La Jolla, CA, 8Radiology & Bioengineering, UC San Diego, La Jolla, CA.
Problem: Breast MRI is recommended annually for high-risk individuals, typically using dynamic contrast-enhanced (DCE) MRI, which requires intravenous gadolinium. However, the added cost and invasiveness limit its use in average-risk populations. Developing a non-contrast MRI method would allow for expansion to broader screening. Diffusion-weighted imaging (DWI), a non-contrast technique, holds promise but suffers from low spatial resolution, limiting detection of small lesions. Restriction Spectrum Imaging (RSI), which uses multi-shell DWI, separates signal into restricted, hindered, free, and flow components without exogenous contrast, and has shown improved lesion characterization. This abstract presents a novel RSI model using multi-b-value input and high-resolution multi-band (MB) acquisition, achieving 2 mm isotropic resolution and improving small lesion detectability. Methods: Along with standard clinical DWI and DCE, high-resolution multi-shell (b-values (# of directions) 0 (1), 100 (6), 800 (6), 1500 (6), 3000 (12) s/mm2) DWI was collected using MB on 122 breast cancer and screening patients. RSI models follow a multi-exponential formula: S(b)= S0Σexp(-b*ADCi)=ΣCiexp(-b*ADCi). Tri- and tetra-exponential models (3C- and 4C-RSI) ADCi (fixed compartment apparent diffusion coefficients) were first calculated on large cancers (largest dimension > 2cm – 61 cancers). Ci signal contribution maps (C-maps) were estimated and provided spatial representation of each diffusion component. The products C1C2 and C2C3 were calculated as well. ADC maps from clinical DWI and DCE subtraction images (peak enhancement – pre-phase) were computed. To assess lesion conspicuity, contrast-to-noise ratios (CNR) were measured in C-maps on 28 lesions from a subset of 22 patients and compared to CNR from ADC and DCE. Finally, receiver operating characteristic (ROC) curves of each model and ADC were measured on the subset, separating malignant from benign lesions, by fitting a generalized linear model. Results: RSI models’ separation of diffusion contents are summarized on Table 1. Regarding CNR, 3C-RSI showed highest values in C1C2 (median: 3.42, IQR: 2.66) and C2C3 (2.72, 1.72). For 4C-RSI, C1C2 (3.15, 3.42) and C2C3 (3.66, 3.79) were also highest. DCE had the highest (3.86, 1.45) and ADC the lowest (1.29, 0.49). Of the 28 test lesions (median lesion size: 2 cm, min: 5 mm, max: 7 cm – largest dimension), 7 were benign. ADC had an AUC of 0.78 while 3C- and 4C-RSI had an AUC of 0.87 and 0.96 respectively. DCE is considered 100% sensitive as selected lesions are based on DCE findings and had a specificity of 75% (7 lesions are benign), while at 80% sensitivity ADC, 3C- and 4C-RSI had a specificity of 50, 83 and 100%, respectively. Conclusion: RSI models achieved comparable CNR to DCE, outperforming ADC. They also had the highest AUCs and specificities, highlighting RSI as a potential non-contrast method for early detection.
| model | ADCs (mm2/s) | Diffusion type | CNR (IQR) | CNR C1C2 (IQR) | CNR C2C3 (IQR) |
| 3C-RSI | ADC1,3=2.7×10-4 | Restricted | 1.86 (0.69) | ||
| 3C-RSI | ADC2,3=1.7×10-3 | Hindered/free | 2.11 (0.89) | 3.42 (2.66) | 2.72 (1.72) |
| 3C-RSI | ADC3,3=18×10-3 | Free/flow | 1.34 (0.83) | ||
| 4C-RSI | ADC1,4=0 | Highly restricted | 1.24 (0.52) | ||
| 4C-RSI | ADC2,4=5.8×10-4 | Restricted/hindered | 2.61 (1.32) | 3.15 (3.42) | 3.66 (3.79) |
| 4C-RSI | ADC3,4=1.9×10-3 | Hindered/free | 1.68 (1.02) | ||
| 4C-RSI | ADC4,4>>3×10-3 | Flow | 1.60 (0.78) | ||
| ADC | / | / | 1.29 (0.49) | / | / |
| DCE | / | / | 3.86 (1.45) | / | / |
Presentation numberPS1-06-09
Attenuated Total Reflectance-Fourier Transform Infrared spectroscopy for monitoring biochemical changes in breast cancer serum across tumor stages
Meagha T. K. Vithanage, Georgia state University, Atlanta, GA
M. T. K. Vithanage1, S. H. Ekanayake1, H. Ghimire1, M. K. Dayananda2, G. Qin3, R. Aneja4, E. A. Janssen5, A. G. Unil Perera1; 1Department of Physics and Astronomy, Georgia state University, Atlanta, GA, 2Department of Physics, Georgia state University Perimeter College, Dunwoody, GA, 3Department of Mathematics and Statistic, Georgia state University, Atlanta, GA, 4School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 5Department of Pathology, Stavanger University Hospital, Stavanger, NORWAY.
Attenuated Total Reflection Fourier-Transform Infrared (ATR-FTIR) spectroscopy offers a promising, minimally invasive, and label-free approach for serum analysis, capable of detecting molecular-level changes associated with breast cancer (BC). Few studies have examined the use of Infrared (IR) spectroscopy to monitor BC progression, and correlations between specific spectral changes and tumor development remain underexplored. This study provides results from a technique that uses ATR-FTIR spectroscopy of blood serum, combined with curve fitting analysis to identify molecular changes in five spectral biomarkers (SBMs) linked with BC progression across tumor stages. These include increases in β-sheet structures (1536 cm⁻¹) and disordered protein structures (1558 cm⁻¹), and a decrease in α-helix (1547 cm⁻¹), PO₂⁻ asymmetric stretching (1062 cm⁻¹), and C-O stretching of glycogen (1050 cm⁻¹). These SBMs showed statistically significant differences (p < 0.05 to p < 0.0001) between healthy controls (HC) and also between different BC Groups classified by tumor size. Among them, the β-sheet (1536 cm⁻¹), α-helix (1547 cm⁻¹), and PO₂⁻ asymmetric stretch (1062 cm⁻¹) SBMs demonstrated high diagnostic performance, each achieving 93% sensitivity and 100% specificity. These findings highlight utility of specific SBMs for monitoring BC and suggest that IR-spectroscopy in combination with curve fitting technique could serve as a robust approach for differentiating BC severity levels based on tumor size.
Presentation numberPS1-06-10
Screening Mammography and Breast MRI in High-risk Lactating Patients
Abigail Keller, University of Wisconsin School of Medicine and Public Health, Madison, WI
A. Keller, M. Elezaby, L. Salkowski, R. Strigel, A. Fowler; Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
Purpose: To evaluate abnormal interpretation/recall rates, imaging findings, and outcomes for asymptomatic lactating women at elevated lifetime risk of breast cancer screened with mammography (MG) and/or breast MRI. Methods: An IRB-approved, HIPAA-compliant retrospective chart review was conducted at a tertiary academic center (2013-2024). Patients were included if lactating and undergoing breast cancer screening due to a personal history of breast cancer, strong family history, or hereditary breast cancer predisposition. Screening was performed with MG, MRI, or both during lactation. Imaging findings were defined using American College of Radiology Breast Imaging Reporting and Data System (BI-RADS 5th ed.). Final outcomes were verified via histopathology or ≥2 years of clinical/imaging follow-up; patients without this were excluded. Results: Of 118 patients identified, 9 were excluded for insufficient follow-up. Among 109 included patients, 63 underwent MG (58 with tomosynthesis), and 45 had MRI. Mean age was 37 ± 4 years (range 27–47) for the MG group and 34 ± 3 years (range 26–43) for the MRI group. Overall, 84% had a first-degree family history of breast cancer, 13% were BRCA2+, 9% BRCA1+, and 8% had a prior breast cancer diagnosis. In the MG group, 30% were baseline screening exams and 70% were subsequent screening exams. Breast density was 54% extremely dense and 48% heterogeneously dense. BI-RADS assessments included 41 BI-RADS 1 (negative) and 16 BI-RADS 2 (benign). The recall rate was 9% (n=6), based on BI-RADS 0 assessments, for an asymmetry (n=1), mass (n=2), calcifications (n=2), and possible architectural distortion (n=1). Diagnostic imaging for the recalled cases yielded 4 benign/negative results, 1 BI-RADS 4A mass confirmed as fibroadenoma on US-guided biopsy, and 1 BI-RADS 3 finding with stable calcifications on follow-up imaging. In the MRI group, 40% were baseline screening exams and 60% were subsequent screening exams. Breast composition was 67% extremely dense and 29% heterogeneously dense. Background parenchymal enhancement (BPE) was marked in 62%, moderate in 16%, and mild/minimal in 22%. BI-RADS assessments were 41 BI-RADS 1 and 16 BI-RADS 2. The abnormal interpretation rate was 9% (n=4). Both BI-RADS 0 cases showed axillary lymphadenopathy: US was benign (BI-RADS 2) in one and suspicious (BI-RADS 4) in the other (biopsy-confirmed benign lymph node). One BI-RADS 4A case (focal non-mass enhancement) was benign breast tissue with dense stromal fibrosis on histopathology from MRI-guided biopsy. The other BI-RADS 4A case (mass) was benign lactational changes in accessory axillary tissue on histopathology from US-guided biopsy. No cancers were detected. Conclusion: Screening recall and abnormal interpretation rates were low in both the MG and MRI groups and fell within the acceptable range for screening MG performance (5-12%). Although no pregnancy-associated cancers were diagnosed, findings support continued adherence to high-risk screening during lactation. Screening high-risk lactating patients is potentially challenging due to physiologic changes in breast density and BPE which can cause false-positive and false-negative results. This study demonstrates that screening MG and breast MRI are practical tools in the high-risk lactating population.
Presentation numberPS1-06-11
Fibroblast activation protein (FAP)-targeted dedicated breast PET for primary invasive lobular carcinoma: A pictorial review
Natsuko Onishi, University of California, San Francisco, San Francisco, CA
N. Onishi1, R. Mukhtar2, T. Hope1, P. Metanat1, Y. Wang1, B. Joe1, K. Ray1, S. Choi2, L. Awni1, D. Heditsian3, D. Romer3, S. Brain3, I-SPY 2 Imaging Working Group, I-SPY 2 Investigator Network, J. Chien4, L. Esserman2, N. Hylton1; 1Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 2Department of Surgery, University of California, San Francisco, San Francisco, CA, 3I-SPY 2, Breast Cancer Advocacy Group, San Francisco, CA, 4Department of Medicine, University of California, San Francisco, San Francisco, CA.
Background Dedicated breast positron emission tomography (dbPET) is an innovative imaging technology specially designed to scan only the breasts, providing high spatial resolution and detailed characterization of tumor uptake levels, distribution, and textures. Fibroblast activation protein (FAP), predominantly expressed in cancer-associated fibroblasts within the tumor stroma, has recently gained significant attention as a promising target for cancer diagnostics and therapy. Recent whole-body PET/CT studies reported that novel FAP-targeted radiotracers showed higher sensitivity in the detection of primary and metastatic invasive breast cancer compared to the standard 18F-fluorodeoxyglucose (FDG) radiotracer. FAP-targeted radiotracers may offer unique advantages in depicting primary breast cancers, especially in invasive lobular carcinoma (ILC), which has unique imaging challenges due to its diffuse growth pattern and low cellularity. We present initial studies of FAP-targeted dbPET performed on the same day following whole-body PET/CT in patients with primary ILC lesions. Methods Patients with primary ILC were recruited from the participants of the Endocrine Optimization Program (EOP), a sub study of the I-SPY 2 TRIAL. After undergoing whole-body PET/CT with FAP-targeted radiotracer (6mCi +/-20% of 68Ga-FAP-2286) on a separate research protocol, they underwent dedicated breast imaging using the MAMMI dbPET scanner (OncoVision, Spain) without additional radiotracer injection. Due to the transportation of patients between two different PET scanner locations, the average interval between radiotracer injection and the start of dbPET scanning was approximately 2.5 hours. The dbPET scans were performed in the prone position, starting with the ipsilateral breast followed by the contralateral breast, with each scan taking approximately 15 minutes per breast. FAP-dbPET images were interpreted alongside DCE-MRI scans obtained closest to the FAP-dbPET scans. Results Four patients with primary ILC have underwent FAP-dbPET scanning to date: #1. 59 year old postmenopausal woman with Lt. ILC before treatment initiation #2. 78 year old postmenopausal woman with Rt. ILC before treatment initiation #3. 39 year old premenopausal woman with Lt. ILC after 4 weeks of neoadjuvant endocrine treatment #4. 51 year old premenopausal woman with bilateral ILC after 7 weeks of neoadjuvant endocrine treatment In all cases, known ILC lesions were clearly depicted on the dbPET as well as on the whole-bodyPET. Notably, for two patients, FAP-dbPET identified additional suspicious lesions with high uptake (pathology not confirmed) that were either uncertain or masked by background parenchymal enhancement in the DCE-MRI images. For individual patients, FAP-targeted dbPET will be presented along with whole-body PET/CT and DCE-MRI. Discussion Our preliminary study shows the promising capability of FAP-dbPET in visualizing tracer uptake of primary ILC with high resolution. These scans may provide better visibility of ILC lesions and provide a more accurate way to assess response to neoadjuvant treatment. In combination with novel tracers such as FAP that may provide unique insight into the biology of primary breast tumors and drug mechanism of action, dbPET provides a cost-effective and clinically-feasibly technology that will facilitate use of PET biomarkers in clinical trials testing novel drugs in the neoadjuvant setting. Based on these preliminary results, we plan to expand our FAP-dbPET study with advanced qualitative and quantitative analyses, over time, to better measure response to therapy and ultimately to help evaluate how best to target therapy in the setting of lobular cancer.
Presentation numberPS1-06-12
Early-stage breast cancer detection with a plasma cell-free RNA and AI-based liquid biopsy platform
Lee S. Schwartzberg, Reno Health, Reno, NV
L. Schwartzberg1, M. Karimzadeh2, A. Momen-Roknabadi2, T. B. Cavazos2, N. Chen3, J. Wang3, M. Multhaup2, J. Ku2, A. Krishnan4, M. Hernandez4, R. Hanna4, L. Fish5, M. Gebala4, H. Goodarzi2, H. Li3, F. Hormozdiari2, B. Behsaz3, A. Hartwig4, B. Alipanahi2; 1Pennington Cancer Institute, Reno Health, Reno, NV, 2Computational Biology and Algorithms, Exai Bio Inc., Palo Alto, CA, 3Engineering, Exai Bio Inc., Palo Alto, CA, 4Assay Development, Exai Bio Inc., Palo Alto, CA, 5Research and Early Development, Exai Bio Inc., Palo Alto, CA.
Background. Early detection of breast cancer saves lives, yet mammography—the current standard for screening—has well-documented limitations. Its sensitivity is notably reduced in women with dense breast tissue, where tumor tissue may be obscured by the naturally dense parenchyma, leading to false-negative findings. Emerging blood-based biomarkers for early cancer detection and monitoring have the potential to augment existing image-based screening technologies in high-risk populations including dense breasts. Tumor-derived RNA has two key advantages compared to tumor-derived DNA which favor its application in diagnosis of small tumors; i) each cell can produce multiple copies of RNA, while DNA content is limited to cellular ploidy, and ii) while cfDNA can originate from any genomic region, cfRNA can only originate from the subset of accessible chromatin undergoing active transcription. We have previously identified and developed a novel category of cancer-associated, small orphan non-coding RNAs (oncRNAs), and combined them with generative AI modeling to develop a liquid biopsy platform requiring a small volume of plasma. This platform has demonstrated high sensitivity and specificity for detection of early stage disease across several cancer types. Here we developed a cell-free RNA and AI-based assay to detect breast cancer across the range of tumor stages and sizes and analyzed it in a separate, independent test set. Methods. We utilized The Cancer Genome Atlas (TCGA) small RNA profiles to discover a library of pan-cancer oncRNAs that were significantly enriched among breast tumors compared to adjacent normal tissues. Our plasma-based training cohort comprised of 745 female patients with clinically diagnosed, pathologically confirmed untreated breast cancer and 258 age-matched women with no known history of cancer from five different sources (mean age 59.3 years ± 13.4 S.D.), including 593 stage I/II (79.6%), 147 stage III/IV (19.7%), and 5 unknown stage (0.7%) patients. The test set was obtained from a different sample source and consisted of blood samples from 92 female patients with breast cancer and 98 individuals with no known diagnosis of cancer (mean age 60 years ± 10.8 S.D.). The test-set included 60 stage I/II (65.22%) and 32 stage III/IV (34.78%) patients. We processed 1 mL of plasma for each sample through our universal, automated cell-free RNA workflow and sequenced at an average depth of 53 million ± 20 S.D. 100 bp single-end reads. We trained a generative AI model using 5-fold cross-validation (CV) to predict presence or absence of invasive cancer and then applied it on the independent test set. Results. Our model achieved an overall AUC of 0.88 (95% CI: 0.85-0.9) for predicting breast cancer versus controls in CV, and an overall AUC of 0.89 (0.83-0.93) in the independent test set. At 90% specificity, the model achieved overall cross-validated sensitivity of 71.8% (63.7%-77.6%) in the training data, and sensitivity of 71.7% (37.4%-79.5%) in the test set. For stage I breast cancer, the model had a CV sensitivity of 63% (57%-68%) in the training data, and 63% (44%-80%) in the test set, both at a 90% specificity. Conclusions. In an independent cohort, the oncRNA and AI-based plasma assay demonstrated high sensitivity for detecting early-stage invasive breast cancer. Given that 40-50% of women have dense breast tissue—where traditional imaging methods are less effective—and assay sensitivity of >70% in an independent cohort, our findings support the potential of oncRNA-based liquid biopsies for screening high-risk populations after negative mammography. A non-invasive blood test could complement current standard screening methodologies in women with dense breasts by stratifying women needing supplemental screening. A prospective study is planned to further validate these findings.
Presentation numberPS1-06-13
The Utility of DWIBS for Breast Cancer Screening: A Promising Alternative to Mammography
Nana Komatsu, Showa Medical University Northern Yokohama Hospital, Yokohama City, Japan
N. Komatsu1, T. Takahara2, H. Yano1, M. Matsuyanagi1, M. Oshi3, H. Ideguchi4, K. Taruno5, N. Hayashi5, T. Chishima1; 1Breast Surgery, Showa Medical University Northern Yokohama Hospital, Yokohama City, JAPAN, 2Radiology, Takahara Clinic – Innovative Scan, Tokyo, JAPAN, 3Breast Surgery, Yokohama City University Graduate School of Medicine, Yokohama City, JAPAN, 4Diagnostic Imaging, Kohoku NT Clinic, Yokohama City, JAPAN, 5Breast Surgical Oncology, Showa Medical University School of Medicine, Tokyo, JAPAN.
Background and Objectives: Diffusion-weighted whole-body imaging with background suppression (DWIBS) is a non-contrast MRI technique that visualizes the diffusion of water molecules while suppressing background signals, enabling the detection of malignant lesions. Due to its non-invasive nature and absence of radiation exposure, DWIBS has recently attracted increasing attention as a promising modality for breast cancer screening in Japan.Notably, no Japanese population-based data have demonstrated a mortality reduction from mammographic screening, and the utility of mammography remains a subject of debate, especially given that over 50% of Japanese women have heterogeneously or extremely dense breasts.This study aimed to assess the potential role of DWIBS as an alternative screening tool for breast cancer. Methods: We retrospectively analyzed 710 women who underwent DWIBS-based breast cancer screening at a dedicated MRI screening clinic in Yokohama, Japan, between April 2022 and January 2025. For those referred for further evaluation, clinical and mammographic data, including BI-RADS classification and breast density, were reviewed. DWIBS screening in this study was conducted as a self-paid service at a dedicated MRI screening clinic equipped with advanced diffusion-weighted imaging protocols. All scans were interpreted by radiologists with expertise in breast MRI. Results: Among the 710 women screened by DWIBS, 41 were referred for further evaluation (recall rate: 5.77%; national average for population-based mammographic screening in Japan: 8.4%). Of these, 30 underwent diagnostic work-up (follow-up rate: 73.2%; national average: 90.1%). Breast cancer was diagnosed in 7 individuals, yielding a cancer detection rate (CDR) of 0.99% (national average: 0.33%) and a positive predictive value (PPV) of 17.1% (national average: 5.4%).The median age of those requiring further evaluation was 46.5 years, with 18 of the 41 individuals (43.9%) under the age of 40, which is below the starting age for population-based breast cancer screening in Japan. Among the 7 diagnosed cases, 5 had a family history of breast or ovarian cancer in second-degree relatives, and 2 were under 40 years old with no prior history of mammography.Notably, 3 of the 7 confirmed cancer cases had BI-RADS 1 findings on mammography, indicating false-negative results. In addition, 5 individuals had extremely dense or heterogeneously dense breast tissue. Among the 11 individuals who did not undergo diagnostic follow-up despite referral, potential contributing factors included scheduling challenges, financial constraints, or limited awareness of the need for further evaluation. However, these factors were not formally investigated. Conclusion: DWIBS demonstrated a higher CDR and PPV than mammography and, with additional advantages such as being radiation-free, painless, and free from embarrassment, may serve as a promising tool for breast cancer detection, including among women at high risk or with dense breast tissue. To fully realize its potential as a screening modality, strategies to ensure diagnostic follow-up and further prospective validation are warranted.
Presentation numberPS1-06-15
Low‑Dose X‑Ray Diffraction Imaging with Real‑Time AI Classification Distinguishes DCIS from Invasive Breast Cancer: Hardware and Interim Clinical Performance from a First‑in‑Human Study
Aika Tanaka, EosDx, Inc., Van Nuys, CA
A. Tanaka1, A. Nguyen1, M. D. Pegram2, M. Khonkhodzhaev3, S. Shcherbakov4, D. Hoffman5, A. Lazarev6, B. Aram1, T. J. Lomis7, P. Chawla8, L. Mourokh3, P. Lazarev6; 1Clinical Operations and Development at EosDx, Inc., EosDx, Inc., Van Nuys, CA, 2Stanford Cancer Institute, Stanford University, Stanford, CA, 3Physics Department, Queens College of the City University of New York, New York, NY, 4Data Analytics Department, Matur UK Ltd., EosDx, Inc., Van Nuys, CA, 5Surgery, Daryl Hoffman Plastic Surgery, Campbell, CA, 6Software and Hardware Development, EosDx, Inc., Van Nuys, CA, 7Surgery, San Fernando Valley Cancer Foundation, Van Nuys, CA, 8Valley Breast Care Radiology, EosDx, Inc., Van Nuys, CA.
Low Dose X‑Ray Diffraction Imaging with Real Time AI Classification Distinguishes DCIS from Invasive Breast Cancer: Hardware and Interim Clinical Performance of the EosDx EoScan™ First‑in‑Human StudyAika Tanaka¹, Audrey Nguyen¹, Mark Pegram2, Masroor Khonkhodzhaev3,4, Slava Shcherbakov4, DarylHoffman5, Alexander Lazarev¹, Byron Aram¹, Thomas J. Lomis6, Prashant Chawla6, Lev Mourokh3,Pavel Lazarev¹¹ EosDx, Inc., Menlo Park, CA, USA ² Stanford Cancer Institute, Stanford University CA, USA ³ PhysicsDepartment, Queens College of the City University of New York, NY, USA, ⁴ Matur UK Ltd., London,UK, ⁵ Daryl Hoffman MD, Campbell, CA USA, 6 San Fernando Valley Cancer Foundation, Van Nuys, CA,USABackgroundXray diffraction detects nanoscale structural alterations in collagen, lipid, and water that heraldbreast neoplasia. EoScan™ is a diffraction breast scanner that captures Bragg signatures throughintact tissue and classifies malignancy using a convolutional neural network (CNN) trained on>12,700 spectra.First‑in‑Human HardwareThe IRB‑approved first‑in‑human unit (“Human‑1”, IRB #1381803, San Fernando Valley CancerCenter, Los Angeles, CA) comprises: (1) a micro‑focus Xray source with liquid cooling, (2) a80 µm slot collimator, (3) a 256×256‑pixel photon‑counting detector housed in a 2 mmPb‑equivalent shield. The system fits in <0.75 m², weighs 110 kg, runs standard 110V, withoutbreast compression.MethodsEx‑vivo cohort: Breast samples including malignant (n = 161) and benign (n = 131) obtained atValley Breast Care (Los Angeles, USA) and Keele University (Keele, UK). A total of 12,730diffraction patterns were collected at EosDx (Menlo Park, CA) and Queen Mary (London, UK)respectively. Malignant samples include infiltrating and in-situ types. Healthy control tissue wasdonated by patients undergoing reconstructive surgery performed by Dr. Daryl Hoffman(Campbell, CA); all samples were collected under the same IRB‑approved protocol governing thein‑vivo study. For all samples, pathology testing was conducted by the Valley Breast CarePathology Department to confirm the disease status. Standard immunohistochemistry testing isconducted for information on hormone receptor status. FISH results are also reported.In‑vivo cohort: ongoing NOVA first‑in‑human trial (n = 150) using the Human‑1 device atSan Fernando Valley Cancer Center; patients are guided into exam room after completing IRBapproved consent form and receive 3 scans on each breast. CNN outputs a malignancy index (0–100). Radiologic findings and pathology results are used to validate the malignancy index outputand calculate performance metrics, such as Sensitivity (Se), Specificity (Sp), PPV, and ROC-AUC. DCIS versus invasive carcinoma performance is evaluated separately.ResultsEx‑vivo: Se = 95.9 %, Sp = 93.5 %, PPV = 95.1 %, AUC = 0.96; DCIS vs invasiveaccuracy = 90 % In‑vivo interim: Se = 85 %, Sp = 70 % across 150 participants; collected results are analyzedthrough previously trained machine learning models and cloud‑based weekly model updatesimprove performance; no device‑related adverse events. Patients report high satisfaction and apositive user experience.ConclusionsEoScan delivers molecular level breast imaging at radiation doses ~25‑fold belowmammography. High ex‑vivo accuracy and encouraging interim in‑vivo performance, along withthe ability to distinguish DCIS from invasive cancer, support its potential as a front line screeningadjunct. Final NOVA results will be presented upon database lock.KeywordsX‑ray diffraction, breast cancer, DCIS, AI diagnostics, structural biomarkers, low‑dose imagingFundingSponsored by EosDx Inc.
Presentation numberPS1-06-16
FES-PET imaging in estrogen receptor-positive breast cancer: implications for clinical management
Nathaniel Wiest, Mayo Clinic, Jacksonville, FL
N. Wiest1, K. Bullock2, T. Pai3, M. Kornas4, B. McKinley5, E. Parent2, P. Advani1; 1Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, 2Department of Radiology, Mayo Clinic, Jacksonville, FL, 3Department of Hematology and Medical Oncology, Emory University, Atlanta, FL, 4Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, 5Division of Medical Oncology, Department of Internal Medicine, University of Utah, Salt Lake City, UT.
Background: 18F-Fluoroestradiol (FES) positron emission tomography (PET) is approved for non-invasive detection of estrogen receptor (ER)-positive breast cancer (BC) and recent updates to the National Comprehensive Cancer Center Network clinical practice guidelines include FES-PET for staging recurrent/metastatic ILC. Given the wide range of FES-PET indications, we retrospectively reviewed the management of ER+ BC patients after FES-PET at Mayo Clinic. Methods: We identified ER+ BC patients who underwent FES-PET from April 2021 to June 2025 across the Mayo Clinic. BC context at the time of FES-PET requisition was categorized as early (EBC), locally recurrent (LRBC), or metastatic (MBC). Management changes were classified as either affecting BC or non-BC care (not mutually exclusive). For pts who underwent 18F-flurodeoxyglucose (FDG) PET-CT and had a discrepant result with FES-PET, we assessed the clinical impact of these discrepancies. Finally, we examined the change in clinical management when FES-PET was ordered to investigate rising tumor markers CA 15-3 or CA 27.29. Results: 166 pts underwent ≥1 FES-PET scan. At diagnosis, 57% had invasive lobular carcinoma (ILC), 33% invasive ductal carcinoma (IDC), and 5.4% mixed histology. ER expression ≥ 50% was observed in 81.4% and 1.2% were HER2+. Overall, FES-PET led to a change in management in 54.5% (n=90) of pts with 43.0% (n=71) experiencing a change in BC care and 16.4% (n=27) in non-BC care. Changes in BC care occurred in 36% of EBC, 68% of LRBC, and 43.7% of MBC. The most common BC management changes after FES-PET were change in treatment strategy in 23.6% (n=39), additional BC-related imaging including further FES-PET scans in 23.0% (n=38), upstaging in 10.3% (n=17), change in surgical planning in 10.3% (n=17), local therapy in 4.2% (n=7), and biopsy avoidance in 3% (n=5). Surgical planning changes were mostly deciding to proceed with curative intent surgery (n=11), though some surgeries were aborted upon FES-PET upstaging (n=6). Non-BC management changes included additional imaging to evaluate incidental findings in 11.5% (n=19) and procedures in 6.7% (n=11), including 5 biopsies, 2 bronchoscopies, 2 endoscopies, 1 hysterectomy, and 1 cholecystectomy. FDG/FES discordancy was observed in 22.3% of the overall population (n=37). The most common pattern was FDG-/FES+ (n=19), leading to upstaging of BC in 8 and the sole use of FES-PET for future restaging in 13 BC patients. The second most common pattern was FDG+/FES- (n=18), attributed to non-BC related FDG uptake in 11 (false-positive FDG) or post-treatment ER loss in 7. Three FDG+/FES- patients were found to have additional cancers: two non-small cell lung cancers and one lymphoma. Non-malignant causes of false positive FDG were inflammation (n=3), silicone implants (n=1), thymic hyperplasia (n=1), and unknown (n=5). Six patients initially upstaged with FDG-PET imaging proceeded to curative-intent surgery after negative FES-PET confirmatory scans (FDG+/FES-). When FES-PET was ordered to investigate rising CA 15-3 or CA 27.29 tumor markers, this led to upstaging/progression in 3 of 21 cases (1 EBC, 2 MBC). The mean tumor marker increase was 57.0% in patients with upstaging/progression versus 19.0% in those without (p=0.02, two-tailed Student’s t-test). Conclusions: FES-PET altered clinical management in approximately half of BC cases in this study. Changes in BC-specific management were most frequent in LRBC, followed by MBC and EBC. One in six patients had a non-BC clinical management change after FES-PET. FDG/FES discordance provided value in upstaging FDG non-avid cancers and downstaging FDG-avid lesions unrelated to BC, influencing curative treatment decisions. FES-PET ordered for rising tumor markers rarely changed management, though larger tumor marker increases were associated with upstaging or progression.
Presentation numberPS1-06-17
A retrospective analysis of capecitabine-induced imaging hepatic steatosis in metastatic breast cancer: prevalence and resolution
Alessia Lucia Daverio, Gustave Roussy, Villejuif, France
A. Daverio1, T. Ben Ahmed2, J. M Ribeiro2, C. Bousrih2, E. Rassy2, M. Mokdad-Adi2, S. Morbelli3, B. Pistilli2, D. Deandreis1, A. Viansone2, T. Henry1; 1Department of Medical Imaging, Gustave Roussy, Villejuif, FRANCE, 2Department of Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 3Nuclear Medicine Division, AOU Città della Salute e della Scienza di Torino, University of Turin, Torino, ITALY.
Background: Capecitabine is a valid monotherapy option in multiple metastatic breast cancer (mBC) subtypes. While case reports of capecitabine-related hepatic steatosis have been published, its prevalence remains unknown. Traditional monitoring relies primarily on biochemical tests, which may not fully capture hepatic changes. Our aim was to evaluate the prevalence of capecitabine-related hepatic steatosis and determine whether routinely performed [¹⁸F]FDG PET/CT could quantify hepatic changes during capecitabine treatment, including in patients without visually evident steatosis. Methods: We conducted a retrospective analysis of patients with mBC treated with capecitabine between 2014 and 2021 at Gustave Roussy. Clinico-biological variables (age, BMI, metastatic sites, therapy line, dihydropyrimidine dehydrogenase (DPD) testing, biochemical liver tests) at baseline were collected. Baseline PET/CT, first evaluation (<180 days), and last PET/CT scans under capecitabine treatment were analyzed to assess hepatic steatosis prevalence. Hepatic steatosis was defined by visual assessment as a decreased liver attenuation relative to spleen and/or increased hepatic vessel visualization on unenhanced CT. Finally, hepatic changes were quantified using standardized uptake values (SUV) on PET images and Hounsfield units (HU) on CT images. Results: The study cohort included 183 patients with mBC. The median age at capecitabine initiation was 66.3 years (range 51-96), and median BMI was 24.2 (range 14.7-39.1). The most frequent metastatic sites were bone (67%), liver (40%) and lymph nodes (33%), with a median of 2 metastatic sites per patient. 86% of patients received capecitabine as monotherapy. Capecitabine was administered across multiple treatment lines: 1st line (14%), 2nd line (32%), 3rd line (31%) and >= 4th line (23%). When available, DPD testing revealed partial deficit in 16/138 patients (12%). Median treatment duration was 298 days (range 80-2162). Sequential imaging analysis was available for 122 patients with both baseline and first evaluation PET/CT scans, and 77 patients at capecitabine discontinuation. No patients showed hepatic steatosis at baseline. At first evaluation, hepatic steatosis was observed in 3/122 patients (2.5%). All 3 patients with steatosis underwent dose reduction due to other capecitabine toxicities (hand-foot syndrome in 2 patients, general condition deterioration requiring weight-based dose adjustment in 1 patient), resulting in resolution of CT steatosis in 2 patients. At final PET/CT, steatosis was present in 4/77 patients (5.2%) and resolved in 3 patients on follow-up imaging after changing treatment line. All patients who developed visible steatosis received capecitabine monotherapy.Quantitative analysis revealed small magnitude hepatic changes during capecitabine treatment: mean HU liver decreased by -2.3 units (95%CI -4.0 to -0.6, p=0.007) and SUVmean liver increased by +0.2 units (95% CI +0.1 to +0.2, p<0.001). These quantitative changes occurred independently of baseline liver metastases presence (p=0.470 for HU, p=0.881 for SUVmean). Among clinico-biological variables, only baseline bilirubin showed significant correlation with HU changes (p=0.044), while other liver function tests, BMI, and DPD status showed no significant association. Conclusions: This study provides a first estimate of capecitabine-induced hepatic steatosis prevalence, and initial evidence supporting the feasibility of using follow-up [¹⁸F]FDG PET/CT to assess its occurrence and reversibility after dose reduction or discontinuation. Further studies are needed to evaluate the potential impact of systematic steatosis evaluation on imaging follow-up, including shorter follow-up intervals and preemptive dose adjustments.
Presentation numberPS1-06-18
Does chemotherapy for early breast cancer affect cognition and brain structure? The Chemobrain Study
Laura Kenny, Imperial College London, London, United Kingdom
L. Kenny1, R. Vicidomini1, L. McLeavy1, P. Riddle2, S. Cleator3, E. Staples4, P. Gayani5, P. Edison6; 1Surgery and Cancer, Imperial College London, London, UNITED KINGDOM, 2Oncology, Chelsea and Westminster NHS Trust, London, UNITED KINGDOM, 3Oncology, Imperial College Healthcare NHS Trust, London, UNITED KINGDOM, 4Oncology, Barking Havering and Redbridge University hospital, London, UNITED KINGDOM, 5Oncology, Royal Wolverhampton NHS Trust, London, UNITED KINGDOM, 6Brain Sciences, Imperial College London, London, UNITED KINGDOM.
Background With improved survival in early breast cancer (EBC), attention is increasingly shifting toward long-term treatment sequelae. Chemotherapy-associated cognitive impairment (CICI or ‘chemobrain’) affects up to one-third of survivors yet remains under-recognized in routine care. Systemic treatments may induce structural and functional brain changes, particularly in regions linked to attention, memory, and executive function. Identifying vulnerable treatment regimens and patient profiles is crucial for tailoring survivorship care. This study aims to characterize the cognitive phenotype and neuroimaging correlates of CICI in EBC patients treated with anthracycline and/or taxane-based regimens, integrating cognitive assessments, longitudinal follow-ups, and multimodal brain MRI. Methods 328 patients who had completed anthracycline and/or taxane chemotherapy within the previous 12 months, along with 94 healthy volunteers, were enrolled from UK sites. Baseline and follow up evaluations assessing executive function, processing speed, attention, working and visuospatial memory were performed using an artificial intelligence-driven digital platform. Patients were followed up digitally every 3 months until 12 months post chemotherapy initiation. 18 patients with poorer performance and 19 age-matched controls underwent 3T brain MRI and in-person neuropsychological evaluation at Imperial College London. MRI scans were analysed using Region of Interest (ROI) and Voxel-Based Morphometry (VBM) to assess grey matter volumes and surface areas. Results At 12 months post-chemotherapy initiation, 27.3% breast cancer participants exhibited cognitive decline, 45.4% remained stable, and 27.3% showed fluctuations compared to baseline. Exploratory treatment-stratified observations revealed that cognitive decline at 12 months was more common with paclitaxel, especially without targeted therapies, while fluctuations in cognition were more frequent in patients receiving Epirubicin Cyclophosphamide + Docetaxel + Trastuzumab or Letrozole. The 18 patients selected for MRI were scanned on average 14.1 months post-chemotherapy; most patients had completed cytotoxic therapy and 89% were receiving endocrine therapy. MRI volumetric analysis revealed significant reductions in grey matter volume and surface area in left isthmus cingulate, orbitofrontal cortex, temporal pole, lingual gyrus, and precuneus – regions critically involved in working memory, semantic processing, attention, and visuospatial integration. Voxel-based analysis also showed reductions in cingulate, medial frontal, parietal, and periventricular areas. These were associated with lower Mini Mental State Examination scores and impairments in semantic and verbal fluency. Conclusions The Chemobrain preliminary data reveals objective cerebral morphological changes and cognitive impairment following anthracycline-taxane chemotherapy in a substantial number of patients at 12 months. However, long-term effects are yet to be determined. This underscores the urgent need to investigate chemotherapy-induced cognitive impairment in a larger cohort of patients to establish risk factors, preventive strategies and novel therapeutic approaches.
Presentation numberPS1-06-19
A COST-MINIMIZATION ANALYSIS OF VACUUM-ASSISTED BIOPSY (VAB) VERSUS SURGICAL BIOPSY (SB) IN WOMEN ELIGIBLE FOR BREAST CANCER SCREENING: BRAZILIAN PUBLIC HEALTH SYSTEM, HOSPITAL’S AND SOCIETAL PERSPECTIVES
Andressa Amorim, Hospital da Mulher SP, Sao Paulo, Brazil
A. Amorim1, M. Mattar2, M. Antonini3, M. Teixeira1, M. Ramos4, R. Lopes5, L. Damous5, L. Gebrim6; 1Mastology, Hospital da Mulher SP, Sao Paulo, BRAZIL, 2Mastology, Hospital da Mulher SP, São Paulo, BRAZIL, 3Mastololgy, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, BRAZIL, 4Mastology, Hospital Estadual Pérola Byington, Sao Paulo, BRAZIL, 5Mastology, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, Sao Paulo, BRAZIL, 6Mastology, Hospital Beneficiência Portuguesa de São Paulo, Sao Paulo, BRAZIL.
Introduction: Breast cancer is the leading cause of cancer-related morbidity among women globally and represents a significant burden on health systems, particularly in low- and middle-income countries (LMICs) such as Brazil. Surgical biopsy (SB) and vacuum-assisted biopsy (VAB) are both widely employed for the evaluation of suspicious breast lesions. While SB remains the more established and broadly implemented modality in public settings due to its lower acquisition cost and infrastructure requirements, VAB has gained attention for its enhanced diagnostic performance—enabling larger tissue sampling, improved lesion targeting, and reduced rates of diagnostic underestimation and repeat procedures. The economic implications of adopting VAB over SB remain insufficiently studied in LMICs, where healthcare budgets are constrained and decision-makers must balance cost containment with the pursuit of higher-value care.Objective: The objective of the study was to assess whether VAB or SB would provide better resource allocation in Brazil.Methods: Considering the Brazilian Public Health System (SUS), Hospital and Societal perspectives, a decision tree was modelled using retrospective data of women with breast cancer from 1,833 procedures performed at Pérola Byington Hospital (PBH), which included data on pathological outcomes and the proportion of patients requiring a subsequent lumpectomy. The costs were retrieved from SIGTAP platform, PBH and the societal perspective included costs from official sources of national average salary.Results: VAB kit costs was $382, while SB was considerably lower, ranging from $ 86 to $196 (HPB and Societal perspectives). Through modelling, the total average cost per patient was $268 for VAB and $492 for SB, considering the PBH perspective, resulting in average savings of $223 per patient. Considering the SUS perspective, the average cost was $208 for VAB and $62 for SB, representing a cost increase of $146. From the societal perspective, VAB led to savings of $353 per patient, largely due to fewer workdays lost compared to SB.Conclusion: VAB minimizes the costs against SB, considering the perspectives from PBH and society. While the SUS perspective shows a slight increase in cost, the overall findings support the economic benefit of using VAB, particularly in settings focused on efficiency and patient recovery.
Presentation numberPS1-06-20
Nipple Aspirate Cell-free DNA Methylation Signatures for Early Breast Cancer Detection
Minsun Jeon, Texas A&M Institute of Biosciences & Technology, Houston, TX
M. Jeon1, E. Sauter2, K. Zhang1; 1Center for Epigenetics & Disease Prevention, Texas A&M Institute of Biosciences & Technology, Houston, TX, 2Division of Cancer Prevention, National Cancer Institute, Rockville, MD.
Background: Cell-free DNA (cfDNA), shed by malignant tumor cells into extracellular fluid, provides valuable epigenetic information indicative of cancer status. Nipple aspirate fluid (NAF), a noninvasive liquid biopsy from at-risk women, contains nucleic acid and protein biomarkers from adjacent cancer cells, showing promise for breast cancer (BrC) detection. However, despite its potential, the application of cfDNA in NAF for BrC screening is still underexplored. This study aims to assess the utility and sensitivity of NAF cfDNA methylation signatures as biomarkers for early detection and risk stratification of BrC. Methods: A low-input cfDNA bisulfite sequencing (cfBS) protocol was developed and optimized for profiling genome-wide DNA methylation at single-base resolution using nanogram-scale cfDNA from NAF. A total of 76 NAF samples (17 BrC and 59 benign) were sequenced and analyzed. An epithelial-mesenchymal transition (EMT) methylation scoring metric was developed to quantify tumor phenotype. CpG methylation regions were identified using an in-house mean shift-based machine learning algorithm. Differentially methylated regions (DMRs) were identified using one-way ANOVA stratified by clinical stage. Predictive modeling was performed using random forest, LASSO regression, and support vector machine (SVM) classifiers. Results: When comparing cancer versus benign samples, differentially methylated regions were significantly enriched for EMT-related pathways. EMT scores were significantly elevated in BrC samples (p 90% accuracy in distinguishing BrC from benign cases, including early-stage tumors. These signatures consisted of 30 genes involved in the Gene Ontology biological process regulation of intracellular signal transduction. Conclusions: Our findings demonstrate that cfDNA methylation profiling from NAF is both technically feasible and biologically informative, offering a highly sensitive, noninvasive strategy for early BrC detection. By capturing EMT-associated epigenetic alterations, NAF cfBS has the potential to identify aggressive phenotypes at early stages. The high predictive accuracy achieved through machine learning models highlights its clinical utility as a complementary tool to existing screening modalities, especially for women with inconclusive mammograms, which is especially common in women with dense breast tissue. These results strongly support further validation in larger, prospective cohorts and suggest that NAF-based liquid biopsy could be integrated into personalized screening frameworks to improve early detection, reduce unnecessary biopsies, and guide preventative care. Financial Disclosures: The study is supported by the Texas A&M University Presidential Clinical Research Partnership program. Disclaimer: Opinions expressed by the authors are their own, and this material should not be interpreted as representing the official viewpoint of the US Department of Health and Human Services, the National Institutes of Health, the National Cancer Institute, or the Division of Cancer Prevention.
Presentation numberPS1-06-21
The impact of 16α-18F-fluoro-17β-estradiol (FES) positron emission tomography (PET) on the staging and management of invasive lobular carcinoma (ILC) of the breast: A Case Series
Alexander Crum, The Ohio State University, Columbus, OH
A. Crum, A. Oliver, M. Harrell, B. Facer, T. Andraos, J. Eckstein, R. Young, S. Jhawar, S. Beyer; Department of Radiation Oncology, The Ohio State University, Columbus, OH.
Background: Invasive lobular cancer (ILC) poses diagnostic challenges due to its diffuse growth pattern, often leading to upstaging at surgery and the potential for suboptimal initial management. 16α-18F-fluoro-17β-estradiol positron emission tomography (FES-PET) has shown utility in assessment of estrogen receptor (ER) positive disease. Given ILC’s diagnostic complexity, FES-PET may enhance the initial staging and management of newly diagnosed locally advanced ILC. This case series explores FES-PET’s potential role in optimizing the management of newly diagnosed locally advanced ILC. Methods: An institutional retrospective case series of 6 patients with newly diagnosed ILC between 4/2024-4/2025 was conducted. Three patients underwent FES-PET which altered or confirmed management, and 3 patients did not undergo FES-PET but were upstaged at the time of surgery. Data on patient demographics, standard of care (SOC) imaging (e.g. mammogram, MRI, CT C/A/P, bone scan and/or FDG-PET), FES-PET findings, pre-FES-PET management plans and post-FES-PET management plans were extracted from the medical records. Results: In Case 1 (ER: 100%), FES-PET detected occult supraclavicular (SCL) nodal involvement (SUV: 3.3) and confirmed residual chest wall (CW) disease (SUV: 5.4) post-neoadjuvant chemotherapy (NAC) and mastectomy, prompting extended radiation volumes and a 1000 cGy in 5 fraction boost to the SCL nodes and CW. In case 2 (ER: 95%), FES-PET detected osseous metastatic disease (SUV: 3.5) not detected on SOC imaging, shifting management to systemic therapy. In case 3 (ER: 100%), FES-PET confirmed no metastatic disease after completion of adjuvant chemotherapy for pT3N3a locally advanced disease; therefore, definitive local therapy with radiation was supported. Patients in cases 4-6 without FES-PET were significantly upstaged at the time of surgery despite SOC imaging; Case 4: (ER: 100%, cT2N1M0, pT3N3a), Case 5: (ER: 100%, cT1N0M0, pT2N2a), Case 6: (ER: 100%, cT1N1M0, pT2N3a), suggesting that FES-PET could have guided initial management. Discussion: FES-PET altered radiation dose/volumes (Case 1), shifted treatment to systemic therapy (Case 2), and confirmed initial treatment plan with local therapy (Case 3). In cases 4-6, FES-PET may have more accurately detected the extent of primary and nodal disease, thereby preventing upstaging at the time of surgery. Recent investigations have demonstrated the potential superiority of FES-PET in detecting ILC in the recurrent or metastatic setting. As a result, FES-PET is now recommended during the work-up for recurrent or metastatic ILC in the NCCN guidelines. However, there is limited data exploring the role of FES-PET in newly diagnosed locally advanced ILC. Unlike FDG-PET, FES-PET minimizes treatment-related inflammatory changes, potentially improving diagnostic accuracy. Future prospective clinical investigations are warranted to validate FES-PET’s role in in the management and outcomes for locally advanced invasive lobular cancer.
Presentation numberPS1-06-22
Impact of breast tumor size discrepancy between contrast-enhanced and conventional ultrasonography on axillary lymph node metastasis
Tomohiro Oshino, Hokkaido University Hospital, Sapporo, Japan
T. Oshino1, H. Shimizu2, M. Sato3, M. Nishida3, T. Horie3, S. Tsuneta4, F. Kato1, M. Hosoda1, I. Yokota5, K. Kudo4, M. Takahashi1; 1Department of Breast Surgery, Hokkaido University Hospital, Sapporo, JAPAN, 2Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, JAPAN, 3Diagnostic Center for Sonography, Hokkaido University Hospital, Sapporo, JAPAN, 4Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, JAPAN, 5Department of Biostatistics, Hokkaido University Graduate School of Medicine, Sapporo, JAPAN.
Normal 0 0 2 false false false EN-US JA X-NONE /* Style Definitions */ table.MsoNormalTable {mso-style-name:標準の表; mso-tstyle-rowband-size:0; mso-tstyle-colband-size:0; mso-style-noshow:yes; mso-style-priority:99; mso-style-parent:””; mso-padding-alt:0mm 5.4pt 0mm 5.4pt; mso-para-margin:0mm; mso-pagination:widow-orphan; font-size:12.0pt; font-family:”Calibri”,sans-serif; mso-ascii-font-family:Calibri; mso-ascii-theme-font:minor-latin; mso-hansi-font-family:Calibri; mso-hansi-theme-font:minor-latin; mso-bidi-font-family:”Times New Roman”; mso-bidi-theme-font:minor-bidi; mso-font-kerning:1.0pt; mso-ligatures:standardcontextual;} Background: Surgical procedures for breast cancer are determined by tumor size and axillary lymph node (ALN) metastasis. A size increase on contrast-enhanced ultrasonography (CEUS) compared with conventional ultrasonography (cUS) is often observed, for which the reasons are unclear. As vascular remodeling at the invasive front plays a critical role in metastasis, we hypothesized that this discrepancy reflects the metastatic potential. Purpose: This study investigated the association between size discrepancies on cUS and CEUS in relation to ALN metastasis in breast cancer. Methods: This retrospective study enrolled 259 patients who underwent surgery for breast cancer with sentinel lymph node biopsy or dissection of the ALN after preoperative cUS and CEUS examinations. The eligibility criteria included tumor size by cUS ranging from 20 to 50 mm and the absence of preoperative adjuvant therapy. Patients were grouped into a DISCR (i.e., tumor size discrepancy ≥ 4.0 mm between CEUS and cUS measurements) and other (non-DISCR) group. The primary outcome was ALN metastasis, defined by pathological evaluation, and mono-and multivariate analyses were performed. Secondary outcomes were the 5-year recurrence-free survival rates. Results: There were 94 patients in the DISCR and 165 in the non-DISCR groups. No size differences were observed between the DISCR and non-DISCR groups (p = 0.82), whereas the DISCR group had a significantly higher rate of ALN metastasis than the non-DISCR group (p < 0.01). Multivariate analyses showed a discrepancy of ≥ 4.0 mm was a-risk for ALN metastasis (relative risk: 5.836, 95% confidence interval [CI]: 2.52-13.5). The 5-year recurrence-free survival rate was lower in the DISCR (0.750, 95% CI: 0.632-0.868) than in the non-DISCR (0.924, 95% CI; 0.870 – 0.978) group. Conclusion: An increase in contrast-enhanced ultrasonography tumor size is helpful for assessing axillary lymph node metastasis and prognosis.
Presentation numberPS1-06-23
[¹¹C]-vorozole pet as a predictive imaging biomarker of aromatase inhibitor resistance in postmenopausal er-positive breast cancer
Lan Lei, Winship Cancer Institute of Emory University, Atlanta, GA
L. Lei1, G. Sharma2, J. Ying3, X. Zhang3, H. Wang3, J. Cohen4, A. Biegon5, S. Gandhi1, C. Huang3; 1Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 2Department of Medicine, Stony Brook University, Stony Brook, NY, 3Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 4Department of Hematology and Oncology, Stony Brook University, Stony Brook, NY, 5Department of Radiology, Stony Brook University, Stony Brook, NY.
Background:Aromatase inhibitors (AIs), such as letrozole and vorozole, block the final step of estrogen biosynthesis and have been associated with improved survival outcomes in postmenopausal women with hormone receptor – positive breast cancer (BC). Prior studies have validated the utility of [¹¹C]-Vorozole PET imaging in detecting aromatase expression in vivo in aromatase-expressing breast tumors. In this study, we aimed to further investigate: (1) differences in aromatase expression in affected and non-affected breasts between ER-positive (ER+) and ER-negative (ER-) patients at baseline and following letrozole administration, and (2) correlations between changes in [¹¹C]-Vorozole PET signal and clinical outcomes. Methods:Nineteen postmenopausal women with BCs underwent baseline [¹¹C]-Vorozole PET imaging, followed by a single 2.5 mg dose of letrozole. A repeat PET scan was obtained two hours post-ingestion. Mean standardized uptake values (SUVs) were measured at baseline and after letrozole administration in regions of interest including the bilateral breasts and thoracic spine. Clinicopathological characteristics and prior AI treatment history were extracted from the electronic medical record. Continuous variables were compared using the Wilcoxon rank-sum test, and correlations were assessed using Spearman’s rank correlation coefficient. A two-sided p-value < 0.05 was considered statistically significant. Results:The average patient age was 63.8 years. Of the 19 patients, 13 had invasive ductal carcinoma and 6 had invasive lobular carcinoma. Ten patients had early-stage disease, and nine had stage IV breast cancer. Fourteen tumors were ER+ and five were ER-. Most patients (n=16) had HER2-negative disease. SUV signals in diseased and healthy breasts were not significantly different in either ER+ or ER- patients at baseline or after letrozole. SUV suppression ranged from -25.8% to 49.6% in the diseased breast following letrozole intake. There was no difference in SUV signal in the whole thoracic spine between patients with and without metastases, but most biopsy-confirmed vertebral metastatic lesions presented high [¹¹C]-Vorozole signal. Among ER+ patients with concurrent FDG-PET, signal distribution was largely concordant between FDG and [¹¹C]-Vorozole. However, several FDG-negative lesions showed positive [¹¹C]-Vorozole uptake and were confirmed malignant by pathology. In early-stage ER+ disease, the average time to progression on AIs was 49.5 months versus 25.9 months in stage IV cases. Baseline SUV and post-letrozole suppression did not correlate with time to progression. Notably, patients with increased [¹¹C]-Vorozole uptake post-letrozole had a shorter average time to progression than those with decreased signal (20.9 vs. 30.4 months), though this difference did not reach statistical significance (P=0.63). Conclusions: [¹¹C]-Vorozole PET demonstrated higher sensitivity than FDG-PET in detecting certain metastatic lesions. An increase in [¹¹C]-Vorozole uptake following letrozole administration may signal resistance to AIs given unsuppressed aromatase expression, and correlate with shorter progression-free survival. These findings suggest that [¹¹C]-Vorozole PET has potential as a predictive imaging biomarker to identify patients who may benefit from early alternative treatments such as fulvestrant.
Presentation numberPS1-06-24
Single MIP Image Evaluation for Predicting Pathologic Response in Patients Receiving Combination Neoadjuvant Immunotherapy/Chemotherapy: A Reader Study
Robert Jared Weinfurtner, Moffitt Cancer Center, Tampa, FL
R. J. Weinfurtner, D. Ataya, S. Falcon, K. Funaro, B. Asha, B. Czerniecki, S. Hatem, H. Hyo; Radiology, Moffitt Cancer Center, Tampa, FL.
Purpose: The purpose of this study was to compare diagnostic accuracy of single-slice maximum intensity projection (MIP) image evaluation with standard MRI in determining pathologic response for patients undergoing combination neoadjuvant immunotherapy/chemotherapy (NAI/NAC). Materials and Methods: Four single-institution NAI/NAC trials were evaluated retrospectively for MRI and pathologic responses. Diagnostic reports were evaluated to determine MRI complete vs partial response (mCR vs mPR) in comparison to pathologic complete vs partial response (pCR vs pPR). Next, early vs late phase MRI analysis was performed with three breast-fellowship trained radiologists blinded to MRI and pathologic reports. The readers categorized cases as mCR vs mPR based on single, random-ordered, early or late-phase maximum intensity projection (MIP) subtracted T1 post-contrast images. Diagnostic accuracy was calculated, and inter-reader agreement was evaluated using Kappa analysis. Results: A total of 77 patients were included, with 154 cases (early and late) utilized for reader analysis. This included 20 patients with triple negative breast cancer and 57 patients with HER2+ breast cancer. Pathologic complete response was achieved in 44/77 (57%). MRI reports categorizing mCR as a true predictor of pCR demonstrated 75% accuracy (95% Confidence Interval, CI, 68-82%) with sensitivity 89% and specificity 58%. Averaged results from the MIP reader study demonstrated accuracy of 70% (CI 62-77%, reader accuracies 68-71%), not significantly different than the MRI report (kappa 0.620). Average reading time was 6.6 seconds per case. Comparison of diagnostic accuracy for early vs late MIP evaluations yielded similar results (accuracy 73% vs 68%) which were not significantly different (CI 61-82% vs 56-78%, respectively). Conclusion: Post-NAI/NAC pre-surgical MRIs demonstrated moderate accuracy in predicting pCR, despite the potential for confounding inflammatory changes with immunotherapy in the neoadjuvant regimen. In addition, accuracy was similar when evaluating a single early-phase MIP image, which could support investigation into abbreviated studies in the future. Clinical Relevance: Single-slice early-phase MIP image evaluation of post-NAI/NAC pre-surgical breast cancer patients demonstrates moderate accuracy in predicting pathologic response.
Presentation numberPS1-06-25
A new imaging approach for monitoring neoadjuvant chemotherapy
Elise Fear, University of Calgary, Calgary, AB, Canada
P. Mojabi1, R. Tsang2, J. Bourqui3, A. Garland1, Z. Lasemiimeni1, D. Deutscher1, B. Docktor4, E. Fear1; 1Electrical and Software Engineering, University of Calgary, Calgary, AB, CANADA, 2Medical Oncology, University of Calgary and Alberta Health Services, Calgary, AB, CANADA, 3Wave View Imaging, Wave View Imaging, Calgary, AB, CANADA, 4Radiology, University of Calgary, Calgary, AB, CANADA.
Background: Microwave imaging has been proposed as an alternative method of breast imaging that is low-cost and comfortable for women as it avoids compression. Microwave properties of tissues relate to water content and behaviour (Gabriel et al, 1996); specifically, low water content tissues (e.g. fat) have lower properties, while higher water content tissues (e.g. gland) have greater properties (Lazebnik et al, 2007). Diseased tissues with higher water content, such as cancer, exhibit increased microwave properties (e.g Sujitani et al, 2015). These differences in microwave signatures of healthy and diseased tissues provide the opportunity to detect tumors and monitor treatment progress. Previously, we have reported detection of tumors with a microwave imaging system in 15 patients (Mojabi et al, 2025a) and presented case-studies of 6 participants undergoing neoadjuvant chemotherapy (Mojabi et al, 2025b, conditionally accepted). Purpose: We explore the feasibility of tracking treatment-related changes in the breast with microwave imaging in a pilot study with women undergoing neoadjuvant chemotherapy for breast cancer. Methods: The prototype microwave imaging system consists of two plates which are placed in contact with the breast (Mojabi et al, 2024). Microwave transmitters and receivers are embedded in the plates; signals transmitted through the breast are used to estimate microwave frequency properties of tissues, and maps of these estimates form a 2D image. Images are collected in both craniocaudal (CC) and mediolateral oblique (MLO) views to facilitate comparison with mammography. 16 participants were recruited and provided written informed consent. Participants had pathologically confirmed diagnosis of early-stage or locally advanced breast cancer and received upfront neoadjuvant chemotherapy based on guidelines. Participants received standard of care treatment for 6-8 cycles and were scanned with the microwave imaging system prior to each treatment cycle. Participants also underwent mid-point imaging (primarily ultrasound), and relevant clinical data was obtained. Results: The images of the non-cancerous breast serve as an intra-patient reference; comparison of the bilateral images may be used to identify the presence of the tumor at baseline pre-treatment. In one group of patients (9/16), a single tumor was present in a location expected to be captured by the scanner. In these cases, increased average properties and/or localized responses suggest detection of the tumor in at least one view at baseline. Potential for tracking changes was suggested by trends in average properties for 7 patients; trends were less evident with changes in separation between the plates at serial scans and smaller breast sizes. The second group of patients (7/16) had multifocal tumors or other clinical factors that likely impacted the scans. One patient had breast implants, while separations between scanner plates for another were up to 4 cm greater for the breast with the tumor, resulting in poor images. Two patients had tumors in locations that were likely not positioned in the scanner. Of the 3 patients with multifocal tumours, detection was noted at baseline in two cases and potential to track changes in one case. Conclusions: Breast microwave imaging shows promise as an imaging biomarker of treatment response in patients receiving upfront neoadjuvant systemic therapy, when used serially. Scans of the cancerous and contralateral non-cancerous breast serve as an intra-patient control. Ongoing analysis is aimed at improving imaging algorithms to support automated detection of tumors. Further evaluation with a larger cohort of subjects is warranted.
Presentation numberPS1-06-26
Fewer cancers in post-treatment surveillance among patients initially staged using contrast-imaging
Gregory Bruce Mann, The Royal Melbourne and Royal Women’s Hospital, Parkville, Australia
T. W. Yang1, A. Rose2, J. Matheson1, K. Elder1, C. MacCallum1, C. Nickson3, G. B. Mann1; 1The Breast Service, The Royal Melbourne and Royal Women’s Hospital, Parkville, AUSTRALIA, 2Department of Radiology, The University of Melbourne, Parkville, AUSTRALIA, 3Melbourne School of Population and Global Health, The University of Melbourne, Parkville, AUSTRALIA.
Purpose: Women with a past history of breast cancer (PHBC) are at risk of local recurrence or new breast malignancy and guidelines recommend routine surveillance imaging. We previously reported our experience of contrast-enhanced mammography (CEM) for local staging of screen-detected cancer and for surveillance of women with a PHBC [1,2]. In the current study we evaluate whether including CBI at diagnosis is associated with different surveillance outcomes, including recall rates and cancer detection rates (CDR). Methods: A retrospective cohort study included women with early breast cancer or DCIS managed at a tertiary breast service in Australia who underwent surveillance involving contrast imaging. To maintain consistency in exposure to contrast-based surveillance, our analysis was restricted to patients who underwent their first surveillance CEM within 3 years of diagnosis. Outcomes were reported from the first round of CEM surveillance up to the first subsequent cancer diagnosis or 5 years from the index diagnosis, whichever occurred earlier. Outcomes were compared according to CBI use (either CEM or Magnetic Resonance Imaging (MRI)) at initial diagnosis. Outcome measures included overall CDR, surveillance tumour characteristics (stage, size, grade, laterality), recall rates, false positive rates, PPV1 (recalls leading to a cancer diagnosis) and PPV3 (biopsies leading to a cancer diagnosis). Statistical tests included two-sample tests of proportions and logistic regression. Results:The analysis included 1,107 women diagnosed between December 2014 and June 2022, comprising 594 who underwent CBI at diagnosis (355 CEM, 202 MRI, 37 both) and 513 without. The CBI group was slightly younger on average (mean 64.1 vs 66.6 years, p=0.0001), and more recently diagnosed (proportion diagnosed 2019-2022 75.6% vs 38.2%). Across the cohort, 3,879 surveillance episodes were evaluated, with patients undergoing an average of 3.6 rounds in the CBI group and 3.4 rounds in the non-CBI group (p=0.0497). During surveillance, 53 women were diagnosed with cancer, with a significantly lower proportion in the CBI group (3.5% vs 6.2%, p=0.0357) and this difference remained significant after adjusting for age and date at index diagnosis (OR 0.51 (95% CI 0.27-0.93, p=0.028)). Of these, 32 were invasive cancers and 21 were DCIS (one pleomorphic LCIS). Patients without CBI at diagnosis demonstrated a higher rate of surveillance-detected contralateral malignancy (2.5% vs 0.8%, p=0.0264). Surveillance-detected cancers did not differ by tumour behaviour (invasive vs DCIS, p=0.4), grade (p=0.2) or invasive size (p=0.8). Interval cancer rates were low (0.8/1000 episodes) and did not differ between groups. Recall rates after surveillance imaging were comparable between groups (5.3% in the CBI group vs 6.2% in the non-CBI group, p=0.234), as were false positive recall rates (4.2% vs 4.4%, p = 0.805), PPV1 (18.8% vs 29.4%, p=0.0648) and PPV3 (30.9% vs 40.0%, p=0.249). Conclusion: Women with early breast cancer who underwent CBI at diagnosis had significantly fewer cancers detected during early surveillance. This suggests that using more sensitive local staging at diagnosis may allow earlier identification and treatment of synchronous disease that would otherwise manifest as early ipsilateral or contralateral events. Accordingly, this will reduce the burden of breast cancer for patients, with expected improvements in longer-term outcomes. References1) MacCallum C, Elder K, Nickson C, et al. Contrast-Enhanced Mammography in Local Staging of Screen-Detected Breast Cancer. Ann Surg Oncol. 2024 Oct;31(10):6820-6830. 2) Matheson J, Elder K, Nickson C, et al. Contrast-enhanced mammography for surveillance in women with a personal history of breast cancer. Breast Cancer Res Treat. 2024 Nov;208(2):293-305.
Presentation numberPS1-06-27
The Use of Contrast Enhanced Mammography for Noncalcified Ductal Carcinoma Insitu for the Breast: Pre Surgical Staging and Planning
Julie L Shisler, Rose Imaging Specialists, Jupiter, FL
E. Winston1, J. L. Shisler2, R. L. Sexton3, S. L. Rose3; 1Radiology, Austin Breast Imaging, Austin, TX, 2Administration, Rose Imaging Specialists, Jupiter, FL, 3Radiology, Rose Imaging Specialists, Houston, TX.
Introduction: Ductal carcinoma in situ is a noninvasive breast cancer with a more favorable prognosis that is typically identified as calcifications on mammography. Less commonly diagnosed and more difficult to detect is non-calcified ductal carcinoma in situ (NCDCIS). NCDCIS represents 10% of all DCIS and may present as a clinical lump or present on imaging as a mass (67%) focal asymmetry (24%) or architectural distortion (15%). The ability to detect NCDCIS is important for cancer staging and surgical planning in the effort to obtain negative margins at lumpectomy. NCDCIS may not be detectable on diagnostic imaging as a structural abnormality and may only be discoverable with the use of contrast. Functional imaging with MRI or contrast enhanced mammography (CEM) typically shows NCDCIS as non-mass enhancement. CEM is a strong alternative to MRI for cancer staging. Advantages of CEM over MRI include lower false positive rates, lower cost and greater patient acceptance. A few small retrospective studies have shown that CEM detects pure DCIS and mixed DCIS and invasive disease, but there has not been a focus on NCDCIS. The purpose of our study is to evaluate the imaging and pathologic features of NCDCIS detected on CEM. Methods and Materials: This retrospective study included 3624 Hologic CEM exams, including 668 cancers and 2956 non-cancers in 3428 patients. The exams were interpreted by one of 15 MQSA certified Radiologists across two clinical sites from June 19, 2018 to December 31, 2024. The age of women ranged from 20 to 87 with median age of 51.5. Dense breast tissue identified in 67% (2445/3624) and 33% (1178/3624) were non dense, 1 patient was not evaluated for dense breast tissue due to post bilateral mastectomy. All cancers cases are biopsy proven and included in the study, the CEM exams were performed either pre or post biopsy. All exams included age, breast density, and histopathology results for biopsy cases from the medical record. Within the set of NCDCIS cases, image findings on the mammogram, patient reported symptoms and contrast enhancement type were obtained. The study assesses the overall prevalence of NCDCIS in the set of cancers and demonstrates the imaging and pathological findings with the addition of contrast to the standard mammogram. Results: In the set of 668 patients with a diagnosis of cancer, 408 were invasive ductal carcinoma, 64 invasive lobular carcinoma, 19 mixed ductal and in situ and 130 ductal carcinoma in situ. Of the 130 DCIS, 16 of the CEM exams identified NCDCIS. The overall prevalence of NCDCIS in the set of cancer cases is 3% (16/668) and 12% (16/130) of the DCIS cases. Age range was 37 to 75 with median of 49 and 69% (11/16) of women had dense breast tissue. Mammographic findings include asymmetry (7), mass (7), architectural distortion (2). Four of the 16 (25%) cases were non mass enhancement and mammographically occult and five of 15 (31%) were palpable. Conclusion: NCDCIS can be difficult to detect on pre-surgical imaging and may result in positive margins at lumpectomy. Contrast enhanced mammography has the ability to show NCDCIS as enhancement, even without an underlying structural abnormality such as a mass or architectural distortion. The use of CEM for cancer staging can assist breast surgeons by showing extent of disease and has the potential to perform at the level of MRI.
Presentation numberPS1-06-28
Non-invasive classification between benign and cancerous breast lesions via time-domain bioimpedance sensing in a first-in-human study
Marc Goldfinger, Zedsen Ltd, London, United Kingdom
M. Goldfinger1, J. Fernandez Vargas2, L. Honeyfield3, C. Colcol3, K. Wourms3, M. Taani2, S. Flais4, A. Antrobus2, A. Lim4; 1Clinical Sciences, Zedsen Ltd, London, UNITED KINGDOM, 2Algorithm Development, Zedsen Ltd, London, UNITED KINGDOM, 3Imaging Research, Imperial College NHS Healthcare Trust, London, UNITED KINGDOM, 4Radiology, Imperial College NHS Healthcare Trust, London, UNITED KINGDOM.
Introduction:Breast cancer is the most common cancer in women worldwide. While mammography remains the standard for early detection, radiation, discomfort and cost warrant development of novel screening tools and diagnostic approaches. One such approach is bioimpedance scanning, a non-invasive modality which measures electrical differences between healthy and cancerous tissue in the breast. In this study, we present a novel handheld device using time-domain bioimpedance sensing (TD-BIS) that infers dielectric properties of tissue from voltage time-series data. Analysing this data with machine learning methods, the TD-BIS technology was used to determine if lesions present in patients were either benign or cancerous in nature. Methods:68 participants undergoing routine mammography or breast ultrasound were enrolled post radiological review and diagnosed by biopsy. Prior to the biopsy, participants were scanned on each breast using the TD-BIS device at predefined locations on the breast. Scans from 7 standard positions were recorded from each breast, starting with one taken directly on the nipple (N) and the remaining scans taken at clock-face positions 2, 4, 6, 8, 10, and 12, with the nipple at the centre. For the right breast, these positions were labelled ‘inner above (IA), ‘inner below’ (IB), ‘below’ (B), ‘outer below’ (OB), ‘outer above’ (OA), and ‘above’ (A) respectively. OA/IA and OB/OB were swapped for the left breast. The range, mean, standard deviation, skewness and kurtosis for different groups of capacitances were used as input for a random forest model. Distinguishing between cancerous and benign lesion classification, the F1 score, balanced accuracy (BA), false positive rate (FPR), false negative rate (FNR), positive predictive value (PPV) and negative predictive value (NPV) were calculated. Models were trained using either one scan per patient from the above list (single scan), all scans combined (combined scans), or all scan data that was within 8 cm of the known lesion (close scans). Mann-Whitney tests (with a Bonferroni correction) were used to evaluate the significance between models. Independent t-tests were used to compare the performance of each scan position relative a random permutation of the labels. Results:62 breasts were categorized as cancerous (n=30) or benign (n=32) via radiological and biopsy evaluation. Results showed that the most significant and best (according to BA and F1) performing models in cancer/benign distinction were ‘Close’, A, IA, IB, N, and B models. However, using only one scan position resulted in high FNR. For close scan data, 12 of 30 benign lesions and 31 of 32 cancerous lesions were correctly identified, respectively. The performance for close scans was: BA:0.651, F1:0.623, FNR:0.039, FPR:0.660, PPV:0.609, NPV:0.895, all with p-value <0.05.when compared to Combined. When evaluating the combined data, 6/30 benign lesions and 31/32 cancerous lesions were correctly identified, with BA:0.567, F1:0.497, FNR:0.031, FPR:0.834, PPV:0.553, and NPV:0.828. Discussion: This initial evaluation of TD-BIS demonstrates its feasibility as a method for discriminating between benign and cancerous lesions. When evaluating spatially relevant data, the study shows that data closest to the lesion improves the cancer detection models when compared to non-spatial data. This implies that the TD-BIS device measures biologically relevant dielectric properties of the underlying tissue. The performance of the close model reveals an FNR less than 4%, suggesting this technology has the potential to be a non-invasive clinical tool to discriminate between benign and cancerous lesions. This conclusion is confirmed when the model input is expanded to include scans farther from the lesion site (e.g combined), which significantly reduces model performance.
Presentation numberPS1-07-01
Outcomes of 1-3 brain metastases treated with stereotaxy in metastatic breast cancer: a single institutional experience from eastern India
Anupam Datta, NCRI Hospital, Kolkata, India
A. Datta1, S. Majumdar1, P. Basu2, R. Jena2; 1Radiation Oncology, NCRI Hospital, Kolkata, INDIA, 2Medical Oncology, NCRI Hospital, Kolkata, INDIA.
Background Patients with metastatic breast cancer (MBC) behave differently with receptor status.Stereotaxy is recommended in treatment of 1-3 brain metastases usually in patients with disease specific graded prognostic assessment (DS-GPA) calculated median survival time (MST) of more than 6 months. Purpose To assess the outcomes of 1-3 brain metastases treated with stereotaxy in MBC in terms of Time to Progression (TTP) in brain and Overall Survival (OS) along with their relationship with receptor status and DS-GPA. Methods 22 MBC patients (from January 2020 – May 2025) with 1-3 brain metastases with DS-GPA (score 1.5 – 3.5) calculated MST more than 6 months of different receptor status (ER/PD and Her2) treated with stereotaxy 30 Gy/5 fractions were analyzed. Intracranial responses were recorded as per RANO-BM response assessment (Response Assessment in Neruro Oncology Brain Metastases) at 3 months post radiation with MRI of brain. Time to progression (TTP) in brain was calculated as time between completion of radiation to brain and radiologically detectable new brain lesion (in months). Overall survival was calculated as the time between completion of radiotherapy and death of the patient. Acute radiation toxicities were assessed as per RTOG (as per embedded in CTCAE v2.0) criteria. Results 2 patients were lost to follow up at 3 months of RT completion. Complete response (CR) was seen in 6 (30%) patients, partial response (PR) was seen in 12 (60%) patients, while stable disease (SD) and progressive disease (PD) was seen in 1 patient each at 3 months. Patients with ER/PR-positive tumors demonstrated non significant (p = 0.711) longer median time to brain progression (TTP: 14.0 months, 95% CI: 7.07–20.93) compared to ER/PR-negative patients (11.0 months, 95% CI: 2.51–19.49). Her2Neu-positive patients had longer median TTP (16.0 months, 95% CI: 8.42–23.58) compared to Her2Neu-negative (10.0 months, 95% CI: 4.16–15.84), with p = 0.067. Patients with fewer brain metastases (1 lesion) had longer median TTP (15.0 months) and OS (19.0 months) compared to those with 2 (TTP: 16.0; OS: not estimable) or ≥3 lesions (TTP: 6.0; OS: 12.0). Overall survival (OS) was higher in the ER/PR-positive group (median OS: 19.0 months, 95% CI: 7.12–30.88) vs ER/PR-negative (17.0 months, 95% CI: 10.36–23.64; p = 0.653). Median OS was notably higher in the Her2Neu-positive cohort 31.0 months versus 17.0 months for Her2Neu-negative patients (95% CI: 12.84–21.16), p = 0.304. Patients with DS-GPA 3.5 had the most favourable survival (median TTP: 20.0 months; OS: 31.0 months), whereas those with DS-GPA 1.5 had the poorest (TTP: 6.0 months; OS: 7.0 months). Toxicities were more frequent in patients ≥58 years (any grade: 4; grade ≥2: 2) compared to younger patients (3 and 1 respectively). Higher toxicity rates were also observed in ER/PR-positive and Her2Neu-positive groups (grade ≥2 toxicity: 2 in each) versus their negative counterparts (grade ≥2: 1 each). Conclusion The findings of the study suggest a favourable profile for the ER/PR positive subtype and Her2 targetted therapy settings. The findings reaffirm DS-GPA as a prognostic. While these findings are limited by sample size, they warrant attention in treatment planning for older or receptor-positive patients.
Presentation numberPS1-07-02
Precise study: postmastectomy radiotherapy evaluated in breast cancer patients achieving complete pathologic response following neoadjuvant chemotherapy and its impact on survival outcomes – a systematic review and meta-analysis
Marcelo Antonini, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, Brazil
M. Antonini1, F. P. Cavalcante2, A. Mattar3, F. P. Zerwes4, E. C. Millen5, F. P. Brenelli6, A. L. Frasson7, M. D. Teixeira3, M. Madeira8, H. L. Couto9, G. Facina10, R. Arakelian11, L. R. Soares12, A. D. Lima13, R. Freitas Júnior12, G. N. Garcia14, F. Bagnoli15, A. G. Amorim1, M. F. de Figueiredo3, G. T. Tosello16, L. H. Gebrim17, G. N. Marta18; 1Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, BRAZIL, 2Breast Surgery, Hospital Geral de Fortaleza, Fortaleza, BRAZIL, 3Breast Surgery, Hospital da Mulher SP, São Paulo, BRAZIL, 4Breast Surgery, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 5Breast Surgery, Americas Oncologia, Rio de Janeiro, BRAZIL, 6Breast Surgery, Universidade Estadual de Campinas, Campinas, BRAZIL, 7Breast Surgery, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 8Breast Surgery, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, BRAZIL, 9Breast Surgery, Redimama – Redimasto, Belo Horizonte, BRAZIL, 10Breast Surgery, Universidade Federal de São Paulo, São Paulo, BRAZIL, 11Oncology, Hospital da Mulher SP, São Paulo, BRAZIL, 12Breast Surgery, Universidade Federal de Goiás, Goiania, BRAZIL, 13Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 14Breast Surgery, Oncoclínicas, São Paulo, BRAZIL, 15Breast Surgery, Santa casa de misericórdia de São Paulo, São Paulo, BRAZIL, 16Breast Surgery, Unoeste, São Paulo, BRAZIL, 17Breast Surgery, Hospital Beneficiência Portuguesa de São Paulo, São Paulo, BRAZIL, 18Radiotherapy, Hospital Sírio Libanes, São Paulo, BRAZIL.
Background: Postmastectomy radiotherapy (PMRT) is well-established for high-risk breast cancer treatment. However, with the increasing adoption of neoadjuvant chemotherapy (NAC), the necessity of PMRT in patients achieving pathologic complete response (pCR) remains controversial. Recent studies, including the prospective NSABP B-51 trial, suggest minimal or no survival benefit in this population, questioning the routine use of PMRT and opening the door to potential treatment de-escalation. Objective: To evaluate whether PMRT provides an overall survival (OS) benefit in early-stage breast cancer patients who achieve nodal pathologic complete response (pCR) after NAC. Secondary objectives included assessing the impact of PMRT on locoregional control and disease-free survival (DFS). Methods: This systematic review and meta-analysis followed PRISMA guidelines and was registered in PROSPERO (CRD4202460366). A comprehensive search was conducted in PubMed, Embase, Cochrane Library, Scopus, and Web of Science for studies published between January 2005 and June 2024. Eligible studies included retrospective and prospective cohorts of women with breast cancer who received NAC, achieved nodal pCR, and underwent mastectomy with or without PMRT. The primary outcome was OS. Secondary outcomes included LRR and DFS. Pooled risk ratios and hazard ratios were calculated using random-effects models. Heterogeneity was assessed using the I² statistic and Cochran’s Q test. Publication bias was evaluated through funnel plots and Egger’s test. Methodological quality and risk of bias were assessed using the ROBINS-I tool for non-randomized studies. Results: Twenty studies published between 2007 and 2024 were included, comprising 36,380 patients across North America (45%), Asia (45%), and Europe (10%). Of these, 23,228 (63.8%) received PMRT, and 13,152 (36.2%) did not. Median follow-up ranged from 40 to 91 months. Study populations were heterogeneous, including patients with stages T1-T4, mean ages of 46-55 years, and multiple molecular subtypes. PMRT was associated with a statistically significant reduction in LRR (RR 1.02, 95% CI: 1.01-1.03; p = 0.005), indicating a 2% relative risk reduction. For DFS, HR was 1.05 (95% CI: 1.02-1.09; p = 0.004), favoring PMRT with a 5% improvement. No significant OS benefit was observed (RR 1.02, 95% CI: 0.99-1.05; p = 0.24). Heterogeneity was moderate for OS (I² = 50%) and DFS (I² = 54%), and low-to-moderate for LRR (I² = 32%). Egger’s test indicated possible publication bias for LRR (p = 0.0017) and DFS (p = 0.010), but not for OS (p = 0.188). Conclusion: This meta-analysis found no significant overall survival benefit from PMRT in patients with nodal pCR after NAC. Although PMRT was associated with statistically significant improvements in disease-free survival and locoregional control, the magnitude of these effects was small and likely clinically insignificant. These findings support the hypothesis that many patients may not require PMRT and could safely benefit from treatment de-escalation strategies. A cautious, individualized approach is recommended, and further prospective studies are needed to identify which subgroups may still derive meaningful benefit from PMRT in the era of effective systemic therapy.
Presentation numberPS1-07-04
Ct simulation as a missed opportunity? coronary artery calcifications predict risk of coronary events in older breast cancer patients
Inbal Golomb, Tel Aviv Medical Center, Tel Aviv, Israel
I. Golomb1, Y. Leshem1, E. Shachar2, V. Soyfer1; 1Institute of Oncology, Tel Aviv Medical Center, Tel Aviv, ISRAEL, 2Obstetrics and Gynecology, University of California, Los Angeles, Los Angeles, CA.
Introduction: Coronary artery calcifications (CAC) are frequently visible on CT simulation scans performed for radiotherapy planning in breast cancer patients. These women often have elevated cardiovascular risk, exacerbated by cancer therapies such as chemotherapy (including anthracyclines, and cyclophosphamide), aromatase inhibitors, trastuzumab, ovarian suppression and radiation. Despite this, routine cardiovascular evaluation is uncommon. As CT simulation may be the only imaging that reveals coronary anatomy, it offers a unique opportunity for radiation oncologists to identify high-risk patients, yet no guidelines exist on how to interpret or act upon these findings. Methods: We conducted a retrospective cohort study of breast cancer patients aged ≥65 treated with radiotherapy at a single tertial care cancer center 2017. Patients were excluded if they were <65 years of age, had metastatic disease, or<3 years of follow-up. CAC was graded semi-quantitatively on simulation CTs as 0 (none), 1 (mild), or 2 (moderate-to-severe), using a simplified visual scoring system. Clinical characteristics and cardiovascular outcomes were extracted from electronic medical records. Results: Among 112 eligible patients (age r 65-84; median 69), 58% (n=6558) had hypertension, 54% (n=60) had hypercholesterolemia, and 21% (n=24) had type II diabetes. CAC was absent in 45% (n=50), mild in 34% (n=38), and moderate-to-severe in 21% (n=24). Patients with moderate-to-severe CAC were significantly older than those without (mean age 72.1 vs. 68.3 years, P < 0.001). During a median follow-up of 7.5 years, 4.5% (n=5) experienced coronary events, at a median of 33 months (range 13-75) post-radiotherapy. All five patients had moderate-to-severe CAC (5/24, 21%), compared to none in the 88 patients with mild or no CAC (P= 0.0003). All had hypercholesterolemia, four treated with statins, three had hypertension, and three had type II diabetes. Notably, four of the five patients received right-sided radiotherapy, suggesting that breast cancer laterality was not a major contributor. Conclusion: Incidental CAC on CT simulation scans are strongly associated with subsequent risk of coronary events in older breast cancer patients. As CT simulation may be the first, and sometimes only, imaging that revealing coronary anatomy, this represents a missed but critical opportunity to identify and manage high-cardiovascular risk individuals. Systematic CAC reporting and communication with primary care providers are warranted to facilitate timely cardiovascular risk intervention.
Presentation numberPS1-07-05
2.5-year follow-up results of a five-fraction external beam radiotherapy accelerated partial breast irradiation for early-stage breast cancer after breast-conserving surgery
Nikolai Strusberg Fernandez, Miami Cancer Insittute, Miami, FL
R. A. Herrera1, N. Strusberg Fernandez1, Y. Weiss1, A. Botero1, C. Lopez-Penalver2, S. Mautner2, J. Mendez2, G. Giron2, J. Panoff1, M. M. Rodrigues1; 1Radiation Oncology, Miami Cancer Insittute, Miami, FL, 2Surgical Oncology, Miami Cancer Insittute, Miami, FL.
Background: External beam accelerated partial breast irradiation (APBI) has emerged as a convenient alternative to the conventional 4-6 weeks of whole breast irradiation following breast-conserving surgery. This study presents 2.5-year follow-up data assessing the feasibility, safety, and efficacy of the 5-fraction APBI Florence trial regimen in select patients with early-stage breast cancer. Methods and Materials: A retrospective analysis was conducted on patients who received an ultra-hypofractionated external beam APBI regimen of 30 Gy delivered in 5-fractions. Eligible patients were women aged ≥40 years with early-stage breast cancer (pTis or pT1-2), no axillary lymph node involvement, tumor size ≤30 mm, and no distant metastases. All had undergone breast-conserving surgery, had histologically confirmed invasive carcinoma or pure ductal carcinoma in situ (DCIS), and had tumor bed clips placed. Clinical data collected included patient and tumor characteristics, treatment parameters, oncologic outcomes, and acute and late toxicities assessed using CTCAE v5.0. Adverse events occurring within 90 days post-APBI were categorized as acute; those occurring thereafter were considered late toxicities. Results: Between April 2021 and December 2022, 54 female patients most commonly with pTis tumors (29.6%) underwent external beam APBI. The median age was 68.0 years (range, 50.0-85.0) and 98.1% had an ECOG performance status of 0-1. A body mass index ≥30.0 mg/kg2 was reported in 40.7% of patients. Tumor laterality was nearly evenly distributed, with 29 (53.7%) patients having left-sided and 25 (46.3%) right-sided breast cancer. The majority of tumors were localized in the upper outer quadrant (42.6%). Median pathological tumor size was 6.5 mm (range, 1.0-29.0). Histological subtypes included invasive ductal carcinoma (55.6%), DCIS (33.3%), and invasive lobular carcinoma (11.1%). Pathological staging was available for 52 of 54 patients: 16 (29.6%) were grade 1, 33 (61.1%) grade 2, and 3 (5.6%) grade 3. Estrogen receptor positivity was observed in 98.1%, progesterone receptor in 77.8%, and Her2Neu positivity in 1.9% of tumors. During the median follow-up of 37.7 months from diagnosis (range, 5.4-49.6) and 34.7 months from APBI (range, 0.6-46.8), majority of patients (87.0%) received adjuvant hormonal therapy post-APBI. Acute grade 1 toxicities were observed in 24.1% of patients, including breast pain (11.1%), dermatitis radiation (7.4%), and fatigue (7.4%). Late grade 1 toxicities were reported in 7.4% of patients, with breast pain (3.7%), and fatigue (3.7%) being the most common. No toxicities of grade 2 or higher have been reported. Breast tissue fibrosis occurred in 1 patient, while 3 patients developed fat necrosis at a median 18.9 months post-APBI (range, 4.3-21.2); all of which were asymptomatic and required no intervention. At the most recent follow-up, all patients remained alive, with no cases of locoregional recurrence or distant metastasis observed. Conclusion: External beam APBI after breast conservation surgery is a feasible treatment approach resulting in excellent tumor control and with minimal toxicities. The outcomes of this study are consistent with those of the external beam APBI Florence trial, further supporting its use as a standard treatment option for appropriately selected patients with early-stage breast cancer.
Presentation numberPS1-07-07
Assessment of Cutaneous Toxicity in Patients Treated with Ultra-Hypofractionated Radiotherapy in the Right Breast at the Pernambuco Cancer Hospital, Brazil
Carolina Souza Vasconcelos, Hospital Cancer of Pernambuco, Recife, Brazil
C. S. Vasconcelos, P. L. Quidute, M. Salgado, D. S. Viana, L. Torres, L. Mendonça; Breast Cancer, Hospital Cancer of Pernambuco, Recife, BRAZIL.
Abstract 1 Title: Assessment of Cutaneous Toxicity in Patients Treated with Ultra-Hypofractionated Radiotherapy in the Right Breast at the Pernambuco Cancer Hospital, BrazilBackground: Ultra-hypofractionated (UH) radiotherapy has emerged as a cost-effective and time-saving alternative for early breast cancer treatment. While randomized trials like FAST-Forward have demonstrated its safety and efficacy, real-world evidence in public healthcare settings lacking advanced radiotherapy technologies remains limited.Objective: To evaluate the degree of acute cutaneous toxicity in patients treated with ultra-hypofractionated radiotherapy (26 Gy in 5 fractions) for right-sided breast cancer in a Brazilian public hospital.Methods: A retrospective cross-sectional analytical study was conducted using medical records of 117 female patients treated between November 2021 and November 2023 at the Pernambuco Cancer Hospital. Eligible patients underwent adjuvant UH radiotherapy to the right breast. Exclusion criteria included incomplete data, treatment with conventional or moderately hypofractionated regimens, male sex, and left-sided tumors (due to cardiac exposure constraints without IMRT or DIBH). Skin toxicity was classified according to the Radiation Therapy Oncology Group (RTOG) scale. Statistical analysis included chi-square and Fisher’s exact tests with a 5% significance threshold.Results: The mean age of patients was 59.9 years, with 80.3% over 50 years old and 51.3% having systemic arterial hypertension. Most tumors were invasive ductal carcinoma (76.1%) with luminal B (44.4%) and luminal A (37.6%) subtypes. Quadrantectomy was performed in 95.7%, with 38.5% receiving chemotherapy. Acute radiodermatitis was observed in 66.7% of patients: Grade 1 in 54.7%, Grade 2 in 9.4%, Grade 3 in only 2.6%, and no Grade 4 cases. A statistically significant association was found between hypertension and the presence of radiodermatitis (p=0.050), suggesting a potential link with underlying vascular changes or obesity, warranting further investigation.Conclusion: This study reinforces that ultra-hypofractionated radiotherapy is a safe and feasible treatment for right-sided breast cancer, even in public hospitals lacking advanced radiotherapy technologies. The acute toxicity profile was consistent with international trials, notably FAST-Forward, with a predominance of mild radiodermatitis and minimal severe events. These findings support the broader implementation of UH regimens in resource-limited settings to optimize treatment access and reduce patient burden.Keywords: Breast Cancer, Radiotherapy, Ultra-Hypofractionation, Acute Skin Toxicity, Radiodermatitis, Public Health
Presentation numberPS1-07-08
Association of Prior Thoracic Radiation with Clinical Outcomes in Patients Hospitalized for Atrial Tachyarrhythmias
Aren S Saini, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
A. S. Saini1, S. Ghay2, R. M. Narasimhan1, K. Samimi1, R. P. Singh3, B. Kaur2, P. Ghay1, I. B. Dreyfuss1, B. Mahal1, C. Seldon Taswell1; 1Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, 2Department of Internal Medicine, Mount Sinai Medical Center, Miami Beach, FL, 3Department of Internal Medicine, Edward Via College of Osteopathic Medicine, Monroe, LA.
Introduction: Thoracic radiation therapy (TRT) is frequently utilized in the management of malignancies such as breast cancer, lymphoma, and lung cancer. While its cardiotoxic effects—particularly coronary artery disease—are well described, less is known about the long-term impact of TRT on arrhythmia-related hospitalizations. This is particularly relevant given the growing population of cancer survivors at risk for cardiovascular disease. Atrial fibrillation and flutter are among the most common sustained arrhythmias in this population, yet outcomes in patients with prior TRT remain poorly characterized. This study examines whether a history of thoracic radiation influences outcomes among patients hospitalized and admitted with atrial fibrillation or flutter. Methods: A retrospective cohort study was conducted using data from the National Inpatient Sample (2016-2022). Atrial tachyarrhythmia hospitalizations were identified using ICD-10 codes for atrial fibrillation and atrial flutter, which likely reflect cases of new-onset arrhythmia or rate-related decompensation. Patients with prior thoracic radiation were identified using corresponding diagnosis codes. Due to coding limitations, atrial tachycardias such as multifocal atrial tachycardia (MAT) and focal atrial tachycardia could not be reliably identified and were therefore excluded—an acknowledged limitation of this study. Multivariable logistic regression was used to calculate adjusted odds ratios (aORs) for binary outcomes, and linear regression was used for continuous outcomes. The primary endpoint was in-hospital mortality; secondary endpoints included total hospitalization charges and length of stay (LOS). Results: Among 3,198,304 patients hospitalized primarily for atrial tachyarrhythmias, 8,570 had a documented history of thoracic radiation. The mean age in the TRT group was 72.5 ± 6.21 years, with 52.9% identifying as male. Prior TRT was associated with a significantly elevated risk of in-hospital mortality (aOR 1.75; 95% CI: 1.27-2.40; p=0.001). Interestingly, these patients had significantly lower total hospital charges (-$9,356; 95% CI: -11,893 to -6,818; p<0.001), while LOS did not differ significantly (+0.06 days; p=0.479). Conclusion: A history of thoracic radiation is associated with increased in-hospital mortality among patients admitted for atrial fibrillation or flutter, highlighting the need for vigilant cardiovascular surveillance in this high-risk population. While hospitalization costs were paradoxically lower and LOS was similar, the elevated mortality risk suggests these patients may present with more advanced or treatment-refractory disease. Importantly, our analysis was limited to atrial fibrillation and flutter due to ICD-10 constraints and does not capture other forms of atrial tachycardia such as MAT and focal AT. Future studies with more granular clinical data are needed to fully characterize the spectrum of radiation-associated arrhythmias.
Presentation numberPS1-07-09
Germline microRNA-based variants predicting late radiation toxicity in breast cancer
Joanne Weidhaas, UCLA, Los Angeles, CA
J. Weidhaas1, K. McGreevy2, S. McCloskey3, J. Hegde1, M. Steinberg1, J. Baker4, M. Dinome5, A. Kusske6, J. Glaspy7, R. Callahan7, K. McCann7, M. Teshome7, N. Mcandrew7, A. Bardia7, D. Telesca8; 1Radiation Oncology, UCLA, Los Angeles, CA, 2Statistics, MiraDx, Los Angeles, CA, 3Radiation Oncology, UCLA, Santa Monica, CA, 4Surgical Oncology, UCLA, Los Angeles, CA, 5Surgery, Duke, Raleigh, NC, 6Breast Surgery, UCLA, Santa Monica, CA, 7Medical Oncology, UCLA, Los Angeles, CA, 8Department of Statistics, UCLA, Los Angeles, CA.
Background: Germline microRNA-disrupting variants (mirSNPs) predict late radiation toxicity, which generally presents as fibrosis, in several cancer types. mirSNP signatures of toxicity are radiation fractionation (size of delivered dose) dependent, meaning that identifying patients at risk of toxicity from one form of radiation treatment affords the opportunity to choose safer alternatives for their management. As radiation is an integral part of breast cancer management, with several fractionation approaches gaining acceptance, we tested the hypothesis that mirSNP signatures could predict late radiation toxicity in these patients. Methods: DNA was isolated from buccal swabs from 121 breast cancer patients treated with conventionally fractionated radiation (CFRT, 38%) or moderately hypofractionated radiation (16-20 fractions, 62%) and tested for over 120 mirSNPs. Acute, chronic and late radiation toxicity as well as clinical factors were collected, including post-mastectomy radiation or implant, chemotherapy treatment, alcohol and smoking history, autoimmunity, and DNA repair mutant status. Most of the cohort was White and non-Hispanic (81.8%), with 11.5% of women White and Hispanic, and 6.6% of women non-White and non-Hispanic. We applied a previously developed mirSNP signature of late grade > 2 CFRT radiation toxicity defined in prostate cancer patients [1]. We then explored the addition of clinical factors to this signature to see if any improved predictive accuracy. Finally, we incorporated germline mirSNPs marginally associated with late toxicity (Fisher or Jonckheere-Terpstra p < 0.1) in our breast cohort alongside the CFRT prediction in elastic net and boosted tree models.Results: The late CFRT radiation toxicity signature achieved robust predictive performance for late grade > 2 toxicity in our breast patient cohort, with a specificity of 0.957, a positive predictive value (PPV) of 0.79, and an overall AUC of 0.734. Notably, all patients with grade 0 late toxicity were accurately identified as low risk, and the model predicted grade 3 late toxicity more accurately than grade 2 (60% vs. 30%, p=0.317 for difference in proportions). As expected, the late CFRT radiation toxicity signature showed no predictive power for predicting acute or chronic toxicity in our cohort, adding to our recent research demonstrating different biological pathways involved in radiation induced acute, chronic, and late toxicity [2]. We found that none of the clinical factors improved the performance of the late CFRT radiation toxicity signature in our cohort, and in fact, the LOOCV AUC slightly decreased to 0.710. In contrast, adding just 6 additional mirSNPs, including MAT1A and KRAS, boosted the AUC to 0.810-0.837 and increased the sensitivity from 0.52 (CFRT alone) to 0.79 (CFRT + 6 SNPs). Interestingly, only 2 of these mirSNPs overlapped with the original CFRT model, and both MAT1A and KRAS variants appeared protective: the rate of late toxicity dropped from 38% to 4% (wild type to mutant MAT1A) and from 27% to 5% (wild type to heterozygous KRAS). Conclusions: These findings further highlight the power and generalizability of germline mirSNP signatures to meaningfully predict late radiation-induced toxicity, including in breast cancer patients. These are important steps towards meaningful biological insights into late radiation toxicity, as well as indicate the possibility of truly personalizing radiation recommendations in breast cancer.1.Kishan, A.U., et al., Germline variants disrupting microRNAs predict long-term genitourinary toxicity after prostate cancer radiation. Radiother Oncol, 2022. 167: p. 226-232.2.Kishan, A.U., et al., Validation and Derivation of miRNA-Based Germline Signatures Predicting Radiation Toxicity in Prostate Cancer. Clin Cancer Res, 2025. 31(12): p. 2530-2538.
Presentation numberPS1-07-10
Tumor response to ultra high dose rate radiation (FLASH) vs. conventional radiotherapy in normal and cGAS-knockout mice
Banita Verma, Stanford University, CALIFORNIA, CA
B. Verma1, A. Mutahar1, S. Melamenidis2, R. Verma3, K. Casey4, K. Horst2, E. Graves2, M. Clarke5, B. Loo6, F. Dirbas1; 1Department of General Surgery, Stanford University, CALIFORNIA, CA, 2Department of Radiation Oncology, Stanford University, CALIFORNIA, CA, 3Department of Neurosurgery, Stanford University, CALIFORNIA, CA, 4Department of comparative Medicine, Stanford University, CALIFORNIA, CA, 5Department of Institute for Stem Cell & Regenerative Medicine, Stanford University, CALIFORNIA, CA, 6Department of Radiaton Oncology, Stanford University, CALIFORNIA, CA.
Background: Radiotherapy (RT) plays a major role in eradicating residual disease in primary breast cancer (BC) and in palliating metastatic BC. While conventional RT (≤0.03 Gy/s) is effective, it often causes significant normal tissue toxicity—such as skin inflammation—limiting treatment intensity and patient quality of life. FLASH RT, a newer approach delivering ultra-high dose rates (>40 Gy/s), has shown similar tumor control with reduced normal tissue damage (the “FLASH effect”). However, the mechanism by which FLASH spares normal tissue and supresses tumor growth remains poorly understood. Ionizing radiation activates the cGAS-STING pathway through DNA damage, triggering proinflammatory cytokine production and tissue inflammation. Recent reports suggest that FLASH RT also activates the cGAS-STING pathway. In this study, we explored the role of the cGAS-STING pathway in mediating the effects of FLASH and CONV RT on tumor eradication and tissue toxicity using cGAS KO mice, to better understand the contribution of the host cGAS pathway to the FLASH effect and tumor suppression. Aim: This study aims to systematically compare the therapeutic efficacy and normal tissue toxicity of FLASH vs CONV RT in a syngeneic BC mouse model and to elucidate the role of the cGAS pathway in mediating radiation-induced inflammation and tumor response. Methods: PYMT117 BC cells were orthotopically injected into the 3rd mammary fat pad of 6-8-week-old female C57BL/6 and cGAS KO mice. Once tumors reached ~50 mm³, mice received a single 30 Gy dose of either FLASH or CONV RT. Tumor growth was monitored every other day with calipers, and mice were observed for skin toxicity (inflammation, cracking). Euthanasia was performed based on tumor burden or skin damage. Tumor volumes were analyzed using two-way ANOVA followed by Tukey’s post hoc test. Skin toxicity was graded (0-6). Each strain was analyzed separately, and p < 0.05 was considered significant. Results: Both FLASH and CONV RT significantly reduced tumor volume (p < 0.001, two-way ANOVA), with tumors becoming undetectable by day 14 post-irradiation compared to controls. Tumor recurrence began around day 25 in all treated groups. In C57BL6 mice, CONV RT showed a trend towards better tumor control with smaller recurrent tumors and some complete responses but caused severe skin toxicity, including inflammation and cracking, requiring euthanasia by day 50 (toxicity score: 6). FLASH RT, while slightly less effective in long-term tumor control, greatly reduced skin toxicity, with only mild lesions in 3 out of 10 mice (score: 4) in C57BL6 mice. Notably, cGAS KO mice showed no visible skin toxicity in either treatment group (score: 0). Conclusion: FLASH RT significantly reduces normal tissue toxicity compared to CONV RT, offering better tolerability with minimal skin damage. Although CONV RT showed stronger initial tumor control, its use was limited by severe skin toxicity. The absence of toxicity in cGAS KO mice suggests the cGAS pathway plays a key role in radiation-induced inflammation. These findings support FLASH RT as a promising, less toxic alternative and highlight the potential of targeting cGAS to improve radiotherapy outcomes in BC.
Presentation numberPS1-07-11
Vepdegestrant Overcomes Endocrine and CDK4/6i Resistance to Enhance Radiosensitivity in ESR1 mutant ER+ Breast Cancer
Priyanka S Rana, University of Alabama at Birmingham, Birmingham, AL
P. S. Rana1, E. Hochmuth2, R. Abou Zeidane3, B. Hauk4, A. Davis1, M. Endraws1, M. Tao2, V. Mercer1, C. Speers1; 1Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 2Radiation Oncology, Case Western Reserve University, Cleveland, OH, 3Radiation Oncology, University Hospitals, Cleveland, OH, 4Medical Scientist Training Program, Case Western Reserve University, Cleveland, OH.
Background:Resistance to endocrine therapy and CDK4/6 inhibitors (CDK4/6i) poses a major barrier to effective management of estrogen receptor-positive (ER⁺) breast cancer, especially tumors harboring activating ESR1 mutations such as Y537S. Previous work by our group has demonstrated that combining ER inhibition with CDK4/6i significantly enhances radiosensitivity in ER⁺ breast cancer models. Given this synergy, we hypothesized that targeted ER degradation using Vepdegestrant (ARV-471), a potent PROTAC-based ER degrader, would further augment radiosensitization in models resistant to CDK4/6 inhibition and endocrine therapy.Methods:We evaluated ARV-471 in ER⁺ breast cancer cell lines, including wild-type MCF-7, tamoxifen-resistant ESR1 Y537S-mutant MCF-7, and CDK4/6i-resistant variants, with and without radiation. Radiosensitization was quantified using clonogenic survival assays. Mechanistic effects on DNA double-strand break repair were assessed through non-homologous end joining (NHEJ) and homologous recombination (HR) reporter assays.Results:ARV-471 induced potent, dose-dependent (5 nM – 100 nM) ERα degradation across all cell lines, including tamoxifen-resistant and CDK4/6i-resistant variants. ARV-471 treatment (3 nM) significantly enhanced radiosensitivity (radiation enhancement ratio (rER) 1.23 – 1.35, p-value <0.05), even in the highly resistant ESR1-mutant and CDK4/6i-resistant models. Mechanistically, ARV-471 impaired DNA repair by reducing NHEJ efficiency by 30% and shifting DNA repair towards HR.Conclusion:ARV-471 effectively radiosensitizes ER⁺ breast cancer cells, including ERi and CD4/6i resistant models, through ER degradation and disruption of DNA repair mechanisms, overcoming critical resistance to endocrine therapy and CDK4/6 inhibitors. Building upon prior evidence of radiosensitization achieved by combined ER and CDK4/6i inhibition, these findings highlight ER degradation as a potentially effective therapeutic approach to enhance radiotherapy efficacy in resistant ER⁺ breast cancer, addressing an unmet clinical need, especially in women with metastatic, treatment-refractory breast cancer undergoing palliative RT.
Presentation numberPS1-07-12
Personalizing Radiotherapy for Left-Sided Breast Cancer: Identifying Candidates for Proton Beam Therapy Based on Cardiac Risk
Rasika Sangle, Tata Memorial Hospital, Mumbai, India
R. Sangle1, R. Krishnamurthy2, K. Bhandarwar1, P. Sharma1, N. Sharma1, S. Nair2, G. Yadav2, T. Wadasadawala2, R. Pathak1, S. Kale3, P. Sahoo3, S. Gadge3, R. Sarin1; 1Radiation Oncology, Tata Memorial Hospital, Mumbai, INDIA, 2Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, INDIA, 3Medical Physics, Tata Memorial Hospital, Mumbai, INDIA.
IntroductionYoung patients with left-sided breast cancer face a higher long-term risk of major cardiac events due to incidental cardiac radiation exposure and cardio-toxic systemic therapy agents. Photon-based radiotherapy delivering a mean heart dose (MHD) of ³3 Gy is associated with at least a 2% increase in the risk of major cardiac events (Kirby et al., 2023). This study aimed to stratify patients based on their risk and identify those most likely to benefit from proton beam therapy (PBT), a modality recently introduced at our tertiary care cancer center.ObjectiveTo identify patients with left-sided breast cancer who are most likely to benefit from proton beam therapy based on predicted cardiac exposure.MethodologyWe reviewed records of 737 patients with stage I to III or oligometastatic breast cancer treated with adjuvant radiotherapy at our center in 2023. A dosimetric audit was performed for all left-sided cases. Radiotherapy was delivered using IMRT or field-in-field 3DCRT, with or without DIBH. Fractionation regimens included 40 Gy in 15 fractions, 26 Gy in 5 fractions, and 28.5 Gy in 5 weekly fractions with boost when indicated. Patients were categorized according to the UK PARABLE trial criteria, which stratify cardiac dose thresholds based on age and the presence of risk factors. Statistical analysis was performed using IBM SPSS Version 28.0. Associations between groups were assessed using the Chi-square test, with a significance threshold of p < 0.05.Results The data for 444 patients was analyzed; 69.3% were under the age of 55. Cardiac comorbidities (22.3%) and diabetes (19.8%) were the most prevalent risk factors, while COPD (1.6%) and smoking (5.6%) were less common. Most patients (84.5%) had a BMI below 30.Fourteen patients (3.1%) did not achieve the age- and risk-adjusted desired MHD using photon techniques and could potentially benefit from proton therapy. Among those with risk factors, 6.5% did not meet their MHD thresholds. In contrast, only 1.1% of patients without risk factors failed to achieve the desired MHD. (Table 1) Younger patients (below 4 years) were disproportionately represented among those not achieving target MHD, particularly those with comorbidities. ConclusionProton therapy may offer significant benefit to a subset of left-sided breast cancer patients at high cardiac risk, particularly younger individuals with comorbidities or unfavorable anatomy who are unable to achieve guideline-recommended mean heart dose thresholds with photon techniques. Risk-adapted selection for PBT is a promising strategy to optimize cardiac sparing in this population.As PBT is a limited and high-cost resource, such data-driven selection is essential for equitable and cost-effective use, ensuring access for those most likely to benefit.
| Risk Factor | Age Group | Target MHD | Desired Dose Achieved | Not Achieved | Total | p | |||||||
| Yes | <44 | ≥2.5 Gy | 18 (66.7%) | 9 (33.3%) | 27 | <0.001 | |||||||
| 45-54 | ≥4.0 Gy | 60 (98.4%) | 1 (1.6%) | 61 | |||||||||
| 55-64 | ≥4.5 Gy | 52 (98.1%) | 1 (1.9%) | 53 | |||||||||
| 65+ | ≥5.5 Gy | 27 (100%) | 0 (0%) | 27 | |||||||||
| Subtotal | 157 (93.5%) | 11 (6.5%) | 168 | ||||||||||
| No | <44 | ≥4.0 Gy | 126 (97.7%) | 3 (2.3%) | 129 | 0.326 | |||||||
| 45-54 | ≥6.0 Gy | 91 (100%) | 0 (0%) | 91 | |||||||||
| 55-64 | ≥6.0 Gy | 38 (100%) | 0 (0%) | 38 | |||||||||
| 65+ | ≥6.0 Gy | 18 (100%) | 0 (0%) | 18 | |||||||||
| Subtotal | 273 (98.9%) | 3 (1.1%) | 276 | ||||||||||
| Total | 430 (96.8%) | 14 (3.2%) | 444(100%) |
Table 1: Mean Heart dose compliance by age and risk group per UK PARABLE criteria.
Presentation numberPS1-07-13
Single Fraction Boost Prior to Whole Breast Radiation Using Stereotactic Partial Breast Irradiation (S-PBI) with a Cobalt Stereotactic Unit for Early Stage Breast Cancer
Olivia Cong, University of Maryland School of Medicine, Baltimore, MD
O. Cong1, S. McAvoy1, A. Rahimi2, M. Guerrero1, S. Becker1, E. Nichols1; 1Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 2Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX.
Purpose/objective The GammaPod is a breast-specific stereotactic radiotherapy device capable of delivering ablative doses with sharp gradients through precise breast localization. We delivered a 8 Gy tumor bed (TB) boost using the GammaPod system followed by a hypofractionated course (40 Gy in 15 fractions or 42.56 Gy in 16 fractions) of whole breast radiation in patients with early stage breast cancers. We hypothesized that 8 Gy to the TB compared to 10 Gy with low energy photons or electrons prior to whole breast irradiation will achieve high patient cosmesis satisfaction, minimal toxicities, and comparable treatment outcomes. Materials/methods Patients diagnosed with an invasive or non-invasive breast cancer treated surgically by a partial mastectomy were eligible if they had AJCC clinical stage 0-II, tumor size < 4 cm, and negative margins. The clinical target volume (CTV), defined as the surgical cavity plus 5-mm, received 8 Gy and a 3-mm margin was added to create the planning target volume (PTV). Cosmesis was assessed at multiple follow-up time points using a physician-rated score (4 point scale excellent, good, fair, or poor). Treatment-related toxicity was graded with NCI CTCAE v5.0. Results From March 2019 to November 2023, 63 patients were treated. Histology included 35 DCIS and 28 invasive carcinomas. Of those with invasive carcinomas, 22/28 were ER+. 41/63 (65.1%) patients received endocrine therapy. Typically completed within a week of the single fraction tumor bed boost, 32 patients received 40 Gy in 15 fractions, 29 patients received 42.56 Gy in 16 fractions, and 2 patients received alternative fractionation regimens outside these two standard options.Median follow-up was 52.9 mo (1.9-93.5 mo). There were 27 acute Grade (gr) 1 toxicities and 5 gr 2 toxicities, with no gr 3 or higher acute toxicities (< 90 days) following the entire course of RT (boost + whole breast). Gr 2 toxicities included radiation dermatitis (2), breast pain (1), and fatigue (2). In the late period, there were 37 acute gr 1 toxicities and 6 gr 2 toxicities, with no gr 3 or higher late toxicities. Gr 2 toxicities included radiation dermatitis (2), breast pain (2), and fatigue (2).Physician scored cosmesis was excellent or good in 37/38 (97.4%), 40/42 (95.2%), 27/28 (96.4%), and 21/22 (95.5%) respectively at baseline, 12 mo, 24 mo, and 36 mo post S-PBI. There have been two local recurrences and no distant recurrences. Conclusion: Single fraction boost prior to whole breast radiation was safe and effective and reduced overall treatment time.
Presentation numberPS1-07-14
Five Fraction Stereotactic Partial Breast Irradiation (S-PBI) with a Cobalt Stereotactic Unit for Early Stage Breast Cancer
Olivia Cong, University of Maryland School of Medicine, Baltimore, MD
O. Cong1, S. McAvoy1, A. Rahimi2, M. Guerrero1, S. Becker1, E. Nichols1; 1Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, 2Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX.
Purpose/objective For early stage breast cancer, the GammaPod is a stereotactic radiotherapy device capable of delivering ablative doses with sharp gradients through precise breast localization. We hypothesize that 5-fraction stereotactic partial breast irradiation (S-PBI) will achieve high 3 year patient cosmesis satisfaction, minimal toxicities, and comparable treatment outcomes compared to external beam radiation therapy. We report combined results from patients treated with S-PBI at 30 Gy in 5 fractions or 40 Gy in 5 fractions as part of a multi-institutional phase II dose-escalation trial. Materials/methods Patient eligibility included DCIS or invasive epithelial histologies, AJCC clinical stage 0-II, tumor size < 3 cm, and negative margins. For those patients on the phase II trial protocol, 40 Gy dose was delivered in 5 fractions to the clinical target volume (CTV) with a minimum 30 Gy dose to the planning target volume (PTV). Cosmesis was assessed at multiple follow-up time points using a physician-rated score (4 point scale excellent, good, fair, or poor). Treatment-related toxicity was graded with NCI CTCAE v5.0. Results From November 2018 to June 2024, 100 patients were treated. Histology included 74 DCIS and 26 invasive carcinomas. Of those with invasive carcinomas, 23/26 were ER+. 83/100 (83.0%) patients received endocrine therapy. 16 patients received 40 Gy in 5 fractions, 71 patients received 30 Gy in 5 fractions, and 13 patients received alternative fractionation regimens outside these two standard options. Median elapsed time between the first and fifth treatment was 13 days (4-22 days). Median follow-up was 39.3 mo (1.6-72.8 mo). There were 30 acute Grade (gr) 1 toxicities and 4 gr 2 toxicities, with no gr 3 or higher acute toxicities (< 90 days). Gr 2 toxicities included radiation dermatitis (2), breast pain (1), and fibrosis (1). In the late period, there were 37 acute gr 1 toxicities with no gr 2 or higher late toxicities. Physicians scored cosmesis was excellent or good 79/80 (98.8%), 59/61 (96.7%), 33/34 (97.1%), and 29/30 (96.7%) respectively at baseline, 12 mo, 24 mo, and 36 mo post S-PBI. There have been two local recurrences and two distant recurrences. Conclusion: S-PBI was a safe and effective treatment and offers another treatment modality for PBI. Due to its sharp dose gradient and favorable side effect profile future studies will focus on further shortening treatment time for patients.
Presentation numberPS1-07-15
Ultra high dose rate radiation (FLASH) vs. conventional radiotherapy: normal tissue toxicity and long-term survival in cgas ko and wild-type mice
Adel Zaid Mutahar, Stanford University, California, CA
B. Verma1, A. Mutahar1, S. Melamenidis2, S. Dutt2, K. Casey3, K. Horst2, E. Graves1, M. Clarke1, B. Loo1, F. Dirbas1; 1Department of General Surgery, Stanford University, California, CA, 2Department of Radiation Oncology, Stanford University, California, CA, 3Department of Comparative Medicine, Stanford University, California, CA.
Background: Radiotherapy (RT) is central to cancer treatment, used either before or after surgery to reduce or eliminate remaining cancer cells. While effective, conventional radiotherapy (CONV. RT, ≤0.03 Gy/s) often causes significant side effects such as skin lesions, inflammation, and long-term tissue damage and this RT-induced toxicity further compromises quality of life. FLASH radiotherapy (FLASH RT, > 40 Gy/s) is an emerging approach, not yet used clinically, but gaining attention due to its “FLASH effect” the remarkable ability to spare normal tissues from radiation damage. RT-induced DNA damage activates the cGAS-STING pathway, triggering inflammation that can aid tumor clearance but also harm healthy tissue. Emerging studies suggest that FLASH may modulate this pathway differently than CONV RT. To better understand this, we studied the effects of FLASH and CONV RT on tissue toxicity and survival in mice with and without the cGAS gene, aiming to clarify how this pathway may influence the benefits of FLASH RT. Aim: To compare the normal tissue toxicity and healing response after FLASH and conv. RT in mice with and without the cGAS pathway, and to understand whether this pathway contributes to radiation-induced tissue damage. Methods: C57BL6 and cGAS KO mice (6-8 weeks) were irradiated unilaterally on the left chest with 30 Gy using CONV and FLASH RT. The mice were monitored regularly over several weeks for visible skin changes, including skin cracking, hair loss, and hair depigmentation development. Vetericyn was applied to help with any visible skin irritation. Observations focused on how quickly side effects appeared, how severe they were, and whether the skin started healing the mice. The skin toxicity was calculated by giving the score (1-6). Results: In the C57BL6 mice treated with CONV RT, skin cracking, hair loss, and hair depigmentation were observed within the first few weeks after radiation (Score 4). These effects were most noticeable early on but gradually improved (aside from hair depigmentation), with signs of healing by the end of the observation period. In the cGAS KO mice treated with CONV RT, side effects were milder mainly slight hair loss, and only a few mice showed very less skin changes (Score 2). In contrast, mice treated with FLASH RT showed no skin damage in C57BL6 as well as cGAS KO groups (Score 0). All mice stayed stable and healthy throughout the study, and none required euthanasia. These findings suggest that FLASH RT may mitigate normal tissue toxicity by avoiding full activation of the cGAS-STING pathway, similar to what is observed in cGAS-deficient mice, while still preserving tolerance and stability. Conclusion: CONV RT caused noticeable skin damage in C57BL6 mice, while cGAS KO mice showed milder side effects, suggesting that the cGAS pathway may contribute to early radiation-related inflammation and tissue damage. On the other hand, FLASH RT resulted in much lower or negligible skin toxicity in both mouse types and appears to avoid triggering the same damage. These results support the idea that FLASH RT is a promising, less toxic alternative to CONV RT for cancer treatment. Understanding how immune-related pathways like cGAS respond to radiation could help develop even better strategies for safer and more effective radiotherapy in the future.
Presentation numberPS1-07-16
Cosmesis Outcomes After Preoperative Radiation Boost for Early Breast Cancer
Mihir Patel, Rutgers Cancer Institute, New Brunswick, NJ
S. Mamidanna1, M. Patel1, K. Patel1, R. Noziere1, N. Ohri1, A. Grann2, D. Toppmeyer3, S. Kumar4, B. Haffty1; 1Radiation Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 2Radiation Oncology, RWJ Barnabas Health, Livingston, NJ, 3Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 4Surgical Oncology, Rutgers Cancer Institute, New Brunswick, NJ.
Purpose:Breast cosmesis is an important outcome for women after breast conservation surgery (BCS). Conventionally, radiation therapy (RT) is given after BCS, and often includes a boost to the lumpectomy cavity. However, in a recent phase 2 prospective trial, patients were treated with a preoperative RT boost of 1332 cGy in 4 fractions, followed by BCS and postoperative adjuvant whole breast RT of 3663 cGy in 11 fractions, resulting in a smaller boost volume than would be given with standard post-operative treatment. We hypothesize that there may be an improvement in patient- and physician-reported cosmesis when the boost is given preoperatively.Materials and Methods:Single institutional cross-sectional study evaluating patients enrolled in the preoperative boost trial who completed the assigned treatment and were available for post-treatment analysis. Standardized photographs of patients were taken after completion of adjuvant whole breast RT, and cosmesis was evaluated using standardized scoring systems. Physician-rated cosmesis assessment was performed by a radiation oncologist and a surgical oncologist specialized in breast cancer using the 4-point Harvard cosmesis scale. Patient-reported outcomes were captured by the Breast Cancer Treatment Outcome Scale (BCTOS) questionnaire at 6-month intervals.Results:Photographs of 27 patients (n=27) were taken and assessed. Median age was 68 years. 22% of the patients had ductal carcinoma in situ (DCIS) with median tumor size of 6.5 mm and 78% had invasive pathology and median tumor size of 9.5 mm. Median time from adjuvant RT completion to photographic assessment was 2.5 years. 14% of patients had bilateral oncoplastic reduction at the time of BCS. Perceived differences by patients between the affected breast and contralateral breast were minimal at 1 year (median [IQR] BCTOS score 22 [20-26], with lowest possible score of 17, highest score of 68, and higher scores indicating greater differences. Inter-rater agreement between radiation and surgical oncologists for cosmetic outcome was assessed using weighted Cohen’s kappa. The kappa value was 0.691 (95% CI: 0.42-0.96), indicating substantial agreement. Rating of cosmesis post-RT by radiation oncologist was: 59% excellent, 36% good, and 7% fair, and surgical oncologist assessment was: 63% excellent, 36% good, and 3% fair. None of the assessed patients had poor cosmetic outcomes.Conclusion:In this prospective phase 2 clinical trial, administering the radiation boost pre-operatively, analysis of physician and patient-reported outcomes demonstrated an overall positive experience and acceptable cosmesis for trial participants. This may facilitate in the decision-making process for patients who have early breast cancer suitable for breast-conserving surgery and are considering various treatment options with radiation therapy. Further randomized trials will be required to compare the long-term effectiveness of pre-operative compared to post-operative boost approach.
Presentation numberPS1-07-17
Real-world safety of concurrent pembrolizumab and radiotherapy in triple-negative breast cancer
Assile EL FAKIH, Institut Curie Paris, Paris, France
A. EL FAKIH1, P. LOAP1, L. CABEL2, S. ALLALI1, K. CAO1, M. MIRABEL3, J. PIERGA2, Y. KIROVA1; 1Radiation Oncology, Institut Curie Paris, Paris, FRANCE, 2Medical Oncology, Institut Curie Paris, Paris, FRANCE, 3Cardiology, Institut Mutualiste Montsouris, Paris, FRANCE.
Background: Triple-negative breast cancer (TNBC) accounts for approximately 15% of invasive breast cancers and is associated with a poor prognosis. The introduction of pembrolizumab in both neoadjuvant and adjuvant settings, as established by the KEYNOTE-522 trial, has improved event free survival and is now considered standard of care. Postoperative adjuvant radiotherapy remains essential in reducing recurrence and mortality. However, combining radiotherapy with pembrolizumab may increase the risk of toxicities, particularly cardiac, and its long-term safety profile remains poorly characterized. This study aims to assess the safety of this combination in TNBC patients.Methods: This monocentric retrospective study, conducted at Institut Curie in Paris, included patients with locally advanced TNBC treated according to the KEYNOTE-522 protocol—neoadjuvant chemotherapy and immunotherapy, followed by surgery and adjuvant therapy including radiotherapy with or without pembrolizumab. Patients were divided into two groups: those receiving concurrent radiotherapy and pembrolizumab (RT-P), and those receiving radiotherapy alone (RT). The primary endpoint was treatment tolerance. Secondary endpoints included overall survival and cancer-specific survival. A p-value < 0.05 was considered statistically significant.Results: A total of 89 patients were included, with a median follow-up of 16 months. Forty-one patients received radiotherapy alone, and 48 received concurrent radiotherapy and pembrolizumab. No significant differences were observed between groups in baseline characteristics or overall toxicity, except for grade 1 radiodermatitis, which was more frequent in the RT-P group (83.3% vs. 43.9%). No grade ≥3 toxicities were reported. Two cases of grade 1 pulmonary toxicity occurred in the RT-P group. The mean heart dose was 1.8 Gy [range: 0.01-7.9], with no cardiac toxicity attributable to radiotherapy.Conclusion: Adjuvant radiotherapy can be safely administered concurrently with pembrolizumab in TNBC patients without increasing radiation-related adverse events, supporting the continuation of systemic therapy in this high-risk population. Nevertheless, larger prospective studies are needed to assess long-term toxicity.
Presentation numberPS1-07-18
Assessment of the Risk of Humeral Head Fractures after Radiation Therapy
Maxmillian Tjauw, University of Oklahoma College of Medicine, Oklahoma City, OK
M. Tjauw, J. Oh, D. Han, P. Pitts, P. Pius, C. Henson, A. Johnston; Radiation Oncology, University of Oklahoma College of Medicine, Oklahoma City, OK.
Background: As 1 in 8 American women are diagnosed with invasive breast cancer in their lifetime and with the shift toward breast-conserving surgery, radiation therapy plays a critical role in breast cancer treatment. However, the humerus often receives off-target radiation, and post-radiation humeral head fractures are a rare but serious adverse effect of treatment, as surgical repair of hypovascularized bone is difficult. In addition, adjuvant aromatase inhibitors increase the risk of fracture. A previous study of 159 patients estimated that 6.06% of patients develop humeral head fractures ipsilateral to the treated breast. Therefore, we retrospectively analyzed 399 patients with breast cancer to identify risk factors for humeral head fractures. Methods: Our single-center study included patients with a diagnosis of in situ carcinoma or invasive breast cancer. The primary endpoint was development of humeral head fracture ipsilateral to the treated breast. We used logistic regression to elucidate associations between development of humeral head fractures and radiation exposure, usage of boost radiation, and hormonal therapy among other studied variables. In addition, we used Cox proportional hazards regression to temporally study the development of humeral head fractures. Results: Among the 399 patients, the mean age at biopsy was 61.04 (SEM: 0.61) with a mean follow-up time of 4.56 (SEM: 0.23). Of the 399 malignancies, invasive ductal carcinoma (IDC; 84%), invasive lobular carcinoma (ILC; 7%), and ductal carcinoma in situ (DCIS; 6%) were the most common subtypes. 2.5% of the 399 patients did not receive radiation. Of the patients receiving radiation, the mean dosage was 39.08 (SEM: 0.51). 42.6% received chemotherapy, and 79.2% received hormonal therapy. 41 patients (10.3%) developed humeral head fractures on the treated side, and 18 of the 41 had fractures were secondary to trauma.In multivariable analysis, women receiving radiation therapy were not significantly more likely to develop humeral head fractures (OR: 0.96; 95% CI: [0.92, 1.01], p=0.11). The presence of trauma was not associated with humeral head fractures. Patients with longer follow-up were more likely to develop humeral fractures (OR: 1.03, 95% CI: [1.003, 1.07], p=0.03). Likewise, on Cox proportional hazards test, radiation therapy was not associated with development of humeral head fractures (HR: 0.44, 95% CI: [0.18, 1.04], p=0.06). Usage of hormonal therapy (HR: 0.36, 95% CI: [0.52, 2.11], p=0.89) and chemotherapy (HR: 0.37, 95% CI: [0.29, 28.07], p=0.83) were also not associated with development of humeral head fractures. Likewise, lumpectomy (HR: 1.18, 95% CI: [0.54, 56.41], p=0.15) and mastectomy (HR: 1.17, 95% CI: [0.29, 28.07], p=0.37) were not associated with humeral head fractures. Conclusion: Our study suggests that radiation therapy, as well as surgical approaches such as lumpectomy and mastectomy, are not significantly associated with the development of humeral head fractures. Furthermore, the use of hormonal therapies, despite their known association with bone density loss, was not linked to increased fracture risk in our cohort. Overall, these findings support the safety of current standard therapies for breast cancer with humeral head fracture risk.
Presentation numberPS1-07-19
Comparing Utilization and Survival Outcomes of Post-mastectomy Radiation Therapy in Invasive Lobular versus Ductal Carcinoma: A National Cancer Database Analysis
Yanying Chen, University of California, San Francisco, San Francisco, CA
Y. Chen1, A. Shui2, C. Lee2, A. Vertido3, M. Kaur1, J. Moya3, E. Abeles4, A. Quirarte5, J. A. Mouabbi6, N. Prionas7, R. Mukhtar8; 1School of Medicine, University of California, San Francisco, San Francisco, CA, 2Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, 3Department of Surgery, University of California, San Francisco-East Bay, Oakland, CA, 4Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 5Carol Franc Buck Breast Care Center, University of California, San Francisco, San Francisco, CA, 6Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 7Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, 8Department of Surgery, University of California, San Francisco, San Francisco, CA.
Background: Post-mastectomy radiation therapy (PMRT) can improve locoregional control and overall survival in patients with invasive breast cancer, particularly those with high-risk features such as ≥4 positive lymph nodes or T3-T4 tumors. However, data on the utilization and effectiveness of PMRT across different histological subtypes remain limited. Invasive lobular carcinoma (ILC), the second most common histologic subtype of breast cancer, differs from invasive ductal carcinoma (IDC) in its features and clinical outcomes, with more hormone receptor positive status and lower grade, but typically higher stage at presentation, higher risk of positive surgical margins, and propensity for late recurrences. Given the unique features of ILC, including a characteristic diffuse growth pattern, we hypothesized that more patients with ILC would receive PMRT, and, that the use of PMRT might have relatively greater survival benefit in patients with ILC than those with IDC. Methods: We used the National Cancer Database (NCDB) to identify patients diagnosed with pathologic stage I-III breast cancer between 2010 and 2020 who underwent mastectomy, with or without adjuvant radiation therapy. Patients who received neoadjuvant radiotherapy or intraoperative radiotherapy were excluded. We considered the following criteria to be an indication for PMRT: T3 tumor with age <50 years, T3 tumor with lymphovascular invasion (LVI), N2 or greater nodal involvement, positive surgical margins, tumor grade 3 with LVI, T1-2N2 category with tumor grade 3 or LVI. PMRT utilization was compared between ILC and IDC using chi-square analyses, and stratified by age, tumor stage, and receptor subtype. Multivariable Cox proportional hazards models were used to evaluate the association between PMRT and overall survival (OS), adjusting for age, T category, N category, hormone receptor/HER2 subtype, and tumor grade in ILC and IDC cohorts; a test of interaction between histology and PMRT was included to assess differential benefit. Results: Of 1,774,249 patients with stage I-III breast cancer in the NCDB, 472,499 patients met our eligibility criteria, of whom 94,359 (20%) had ILC and 378,140 (80%) had IDC. In total, 32% patients received PMRT, with significantly higher utilization rates in patients with ILC compared to IDC (37% vs 30%, p<0.001). In stratified analysis, ILC patients had more PMRT across receptor subtypes and age groups, but only within the stage III cohort, suggesting that higher stage is driving increased PMRT in ILC patients. Among patients meeting the indication for PMRT, those who received radiation had improved overall 5-year survival than those who did not (log-rank p<0.001). In separate multivariable models within the subgroups of ILC and IDC, the adjusted hazard ratio for death within 5-years in ILC patients was 0.66 (95% CI: 0.62-0.70, p<0.001) in those receiving PMRT compared to those who did not, while in IDC patients it was 0.74 (95% CI: 0.72-0.76, p<0.001), with a significant test of interaction (p=0.01), suggesting differential survival benefit by histologic subtype. Conclusion: PMRT is more frequently utilized in patients with ILC than those with IDC. While PMRT is associated with improved overall survival in both subtypes, the relative benefit of PMRT appears greater in patients with ILC than IDC, perhaps reflecting the greater risk of occult residual disease in those with diffusely growing tumors like ILC. These findings highlight the importance of considering histologic subtype when evaluating the role of PMRT and the need to tailor radiation strategies in lobular breast cancer.
Presentation numberPS1-07-20
Outcomes of Once Daily and Twice Daily APBI Regimens in Hormone Receptor Positive Breast Cancer: A Single Institution Experience
Tanun Jitwatcharakomol, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
T. Jitwatcharakomol1, Y. Gokun2, S. Daniel2, M. Mestres-Villanueva2, R. Young2, J. Eckstein2, T. Andraos2, E. Healy3, J. White4, J. Bazan5, E. Cochran2, S. Beyer2, S. Jhawar2; 1Division of Radiation Oncology, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, THAILAND, 2Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, 3Department of Radiation Oncology, University of California, Irvine, Orange, CA, 4Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 5Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA.
Background: Accelerated partial breast irradiation (APBI) has demonstrated non-inferior in-breast recurrence rates compared to whole breast irradiation in low-risk hormone receptor positive breast cancer patients across multiple randomized clinical trials. Optimal external beam APBI fractionation, dosing, and technique, however, remain undefined. This study evaluated outcomes of two APBI regimens used at our institution: 28.5 Gy in 5 once-daily fractions on non-consecutive days and 38.5 Gy in 10 twice-daily (BID) fractions. Methods: This retrospective, single-institution study included post-menopausal women with ductal carcinoma in-situ (DCIS) and early-stage hormone receptor positive invasive breast cancer who underwent lumpectomy followed by APBI. Outcomes of interest include ipsilateral breast tumor recurrence (IBTR), disease-free survival (DFS), overall survival (OS), radiation toxicities (graded by CTCAE v5.0), and physician-reported (Harvard scale) cosmetic outcomes. Results: Patients treated with APBI between January 2017 and March 2025 were included. Total patients were 399: 257 in the 28.5 Gy group and 142 in the 38.5 Gy group. The median age was 66 years (IQR 59-72). Most patients were White (86.9%) and non-Hispanic (99.2%). The majority had T1 tumors (79.4%) or DCIS (18%), Grade 1-2 histology (93.5%), no lymphovascular invasion (96.5%), were ER-positive (98.5%), and PR-positive (93.5%). For invasive cases, HER-2 was negative in 98.8%. Baseline characteristics were similar between groups. The majority of patients received radiation using 3D-conformal technique (99.2%), in the prone position (96%), and started adjuvant endocrine therapy (86%). Based on 408 lesions, the median gross tumor volume (GTV), clinical target volume (CTV), and planning target volume (PTV) were 19.6 cc, 113.2 cc, and 194.3 cc, respectively. Larger CTV and PTV values were observed in the 38.5 Gy group compared to the 28.5 Gy group: CTV (118.2 cc vs. 108.3 cc, p = 0.02) and PTV (208.6 cc vs. 183.3 cc, p < 0.01).With a median follow-up of 3.4 years, the 3-year IBTR for 28.5Gy was 2% and 38.5Gy was 1%, p-value = 0.46. The 3-year DFS was 98% in the 28.5 Gy group and 99% in the 38.5 Gy group, p-value = 0.79 and the 3-year OS was 99% in both groups, p = 0.53.For acute toxicity, grade 1-2 dermatitis was significantly more common in the 38.5 Gy group (61.7% vs. 28.6%, p < 0.01). For late toxicity at 1-year post-treatment, grade 1-2 skin hyperpigmentation (79.5% vs. 32.4%, p < 0.01) and fibrosis (81.6% vs. 51%, p < 0.01) were significantly more common in the 38.5 Gy group than 28.5 Gy group, respectively. Radiation dose of 38.5 Gy in 10 BID fractions was associated with significantly higher odds of developing fibrosis (adjusted odds ratio 4.15, 95% CI: 1.7-10.1) compared to 28.5 Gy daily fractionation. No grade ≥3 acute or late toxicities were observed.Physician-reported (Harvard scale) cosmetic outcomes were similar between groups. At 1-year, cosmetic outcomes were available for 304 patients. Excellent or good cosmesis was observed in 98.1% of patients in the 28.5 Gy group and 99% in the 38.5 Gy group, p-value = 0.15. At 3-year, cosmetic outcomes were available for 90 patients. Excellent or good cosmesis was 94.1% (28.5 Gy group) and 97.2% (38.5 Gy group), p-value = 0.83. Conclusions: Both regimens—28.5 Gy in 5 once-daily fractions and 38.5 Gy in 10 twice-daily fractions—delivered using 3D-conformal technique in the prone position yielded excellent outcomes in patients with low-risk breast cancer. However, the 28.5 Gy regimen demonstrated a lower incidence of acute and late toxicity and may be preferable in this patient population given its lower skin toxicity and increased convenience. Longer follow-up is warranted to confirm these findings.
Presentation numberPS1-07-21
Germline Genetic Variants and Risk of Acute Radiation Dermatitis in Patients Receiving Curative-Intent Radiation Therapy for Breast Cancer
Nicholas Eustace, City of Hope National Medical Center, Duarte, CA
N. Eustace1, K. Ghaffarian2, T. Watkins1, K. Vo1, L. Reynaga3, J. Bonner3, S. Gruber3, T. Williams1, J. Bazan1; 1Radiation Oncology, City of Hope National Medical Center, Duarte, CA, 2Department of Medicine and Surgery, University College Cork, Cork, IRELAND, 3Precision Medicine, City of Hope National Medical Center, Duarte, CA.
Background: Multigene panel testing has increased the detection of pathogenic germline variants (PV) in patients with breast cancer. The impact of PVs and variants of unknown significance (VUS) on toxicity and clinical outcomes has largely been studied in patients with BRCA1 or BRCA2 mutations, with few data on patients with other genetic alterations. In addition, few studies have characterized the impact of VUS on toxicity and/or outcomes. At our institution, a prospective Precision Medicine study was started to characterize germline and somatic mutations in breast cancer patients. Here, we present a preliminary analysis of acute toxicity rates in breast cancer patients treated with curative intent radiation therapy based on germline mutation status. Materials/Methods: This retrospective, single-institution study utilized the Precision Medicine database at our institution to identify breast cancer patients with available genetic data. Among 4400 patients with stage I-IV breast cancer, 900 received curative-intent breast radiotherapy (RT). Clinicodemographic information, RT parameters, and germline mutation status – characterized as PV, VUS, or benign – were extracted from the database. RT toxicity was prospectively documented in the electronic medical record (EMR) during the study period. The primary endpoint was the incidence of acute grade 2 or higher radiation dermatitis (G2RD). Associations between germline mutational status and toxicity rates were evaluated using the chi-square test, with significance defined as p<0.05. Results: Of the 900 patients in the Precision Medicine database who received breast RT, 396 had complete clinical, RT, and genomic data available. The median age is 57 years (IQR 47-65), with 37% Hispanic, 40% non-Hispanic White, and 15% non-Hispanic Asian. Disease stage was 29% stage I, 50 % stage II, and 17% stage III. Breast cancer subtypes were 68% HR+/HER2-, 17% HER2+, and 15% triple negative. Breast-conserving therapy was performed in 65% of cases. Radiation was delivered to the whole breast in 46% (N=182), breast/chest wall plus regional nodes (PMRT/RNI) in 45% (N=177), accelerated partial breast irradiation (APBI) in 4%, and intraoperative radiation therapy (IORT) in 5% of patients. Hypofractionated RT (≥2.66 Gy/fraction) was delivered to 45% of patients. Germline testing identified 10% PV (N=39), 57% with VUS in at least one gene (N=224), and 33% Benign (N=133). The overall incidence of G2RD was 24% (N=95) and did not differ based on germline mutation status: 28.2% for PV, 22% for VUS, and 26% for benign status (p=0.64). Among patients with PV, BRCA1/2 mutations were the most common (N=11), with a G2RD rate of 18%, similar to the entire population. MUTHY mutations were the next most common (N=9), with 0% G2RD. The one patient with PALB2 mutation did not have G2RD and no CHK2 or ATM mutations were present in the analyzed cohort. Factors significantly associated with increased G2RD were the type of RT delivered (31% in the PMRT/RNI group, 21% for whole breast only, and 3% for APBI/IORT; p=0.0006) and fractionation schedule (31.9% for conventionally fractionated RT vs 14.5% in hypofractionated RT; p<0.0001). Conclusion: In this preliminary analysis, germline pathogenic mutations or variants of unknown significance were not associated with an increased risk of acute radiation dermatitis compared to those with benign mutation status. As expected, only factors related to RT delivery were associated with the development of grade 2 dermatitis. Ongoing analysis will assess these findings in the complete dataset and evaluate local-regional recurrence by germline and somatic mutation status.
Presentation numberPS1-07-23
Outcome After Locoregional Recurrences For Definitively Treated Non-Metastatic Inflammatory Breast Cancer
Ekene Onwubiko, MD Anderson Cancer Center, Houston, TX
E. Onwubiko1, M. Kai2, M. Stauder1, S. Sun3, C. Goodman1, A. Lucci3, H. Johnson3, R. Layman2, W. Woodward1, B. Lim2, A. Nasrazadani2, S. Saleem4, V. Valero2, G. Whitman5, M. Patel5, H. Le-Petross5, Y. Lu6, M. Alexander2, A. Alexander2, C. Yajima2, L. Villarreal2, H. Lopez2; 1Radiation Oncology – Breast, MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, 4General Oncology, MD Anderson Cancer Center, Houston, TX, 5Breast Imaging, MD Anderson Cancer Center, Houston, TX, 6Nuclear Medicine, MD Anderson Cancer Center, Houston, TX.
Purpose: This study examines the clinical outcomes for Inflammatory Breast Cancer (IBC) patients who develop locoregional recurrence (LRR) after tri-modality therapy (TMT), including systemic therapy, modified radical mastectomy and post-mastectomy radiation therapy (PMRT). While LRR is uncommon after standard trimodal therapy targeting all pre-systemic therapy disease, we sought to characterize patient demographics, tumor characteristics, recurrence patterns, and survival outcomes after LRR in IBC.Methods: This data set was derived from patients enrolled in a prospective IBC registry diagnosed from 2010 to 2017. Among 140 M0 patients who had TMT, 9 with LRR were identified. Clinical data were extracted for the 9 patients with LRR using the Electronic Health Record. Descriptive statistics were used to summarize patient characteristics. ER/PR < 10% was considered triple negative. Kaplan-Meier survival curves assessed time to LRR from the end of radiation and overall survival after LRR diagnosis.Results: The average age at diagnosis was 59 years (range 23-96 years). All patients were White. Six were not Hispanic or Latino, three were of unknown ethnicity. Four cancers were HR-HER2-, three were HR+HER2-, and two were HR-HER2+. All cancers were node-positive, four had N3 disease.The average time to LRR development after treatment was 17 months. Isolated LRR occurred in three cases. Two patients had LRR prior to and after radiation completion. One had palliative dose PMRT (30Gy/ 10 fractions) and progressed on the chest wall less than a month after PMRT. Another developed LRR approximately one month after radical mastectomy before radiation initiation, which was managed with surgical re-excision followed by completion of PMRT. A subsequent LRR occurred on the chest wall at 82 months. Overall survival after LRR was 14% at five years, with one patient alive without distant disease at 89 months. Chest wall recurrence was the most common LRR site, occurring in four of patients. Supraclavicular lymph node recurrence occurred in two patients, and axillary lymph node recurrence occurred in one patient.Conclusion: Low LRR among non-metastatic IBC patients after trimodality therapy has been previously reported ( < 10% at 5 years). LRR occurred across molecular subtypes and was most often concurrent with distant disease, although isolated LRR did occur. We plan to analyze the radiation treatment plans and parameters to correlate radiation doses delivered to areas of recurrence, which may identify technical factors contributing to LRR development and inform future treatment optimization strategies.
Presentation numberPS1-07-24
External beam accelerated partial breast irradiation versus whole breast irradiation for invasive lobular carcinoma: a retrospective, single-institution study
Tanun Jitwatcharakomol, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
T. Jitwatcharakomol1, R. Young2, D. L. Handley3, S. Beyer2, T. Andraos2, J. Eckstein2, D. Agnese4, S. Obeng-Gyasi4, R. Wesolowski5, K. Johnson5, S. Jhawar2; 1Division of Radiation Oncology, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, THAILAND, 2Radiation Oncology, The Ohio State University/James Comprehensive Cancer Center, Columbus, OH, 3Center for Biostatistics, The Ohio State University/James Comprehensive Cancer Center, Columbus, OH, 4Surgical Oncology, The Ohio State University/James Comprehensive Cancer Center, Columbus, OH, 5Medical Oncology, The Ohio State University/James Comprehensive Cancer Center, Columbus, OH.
Introduction Accelerated partial breast irradiation (APBI) for invasive lobular carcinoma (ILC) is conditionally not recommended in the 2024 ASTRO Clinical Practice Guidelines. This recommendation stems from the limited number of women with ILC, as opposed to ductal carcinoma, included in landmark APBI trials, and lack of subtype-specific data. This study evaluates APBI in patients with low-risk ILC. Methods and Materials This single-center retrospective study included early-stage breast cancer patients who underwent lumpectomy for ILC or carcinoma with a lobular component. Patients comprised two groups: those who received APBI and those who received whole breast irradiation (WBI). Log-Rank tests were used to compare WBI vs. ABPI for ipsilateral breast tumor recurrence (IBTR), disease-free survival (DFS) and overall survival (OS), while Fisher’s exact tests were used to compare WBI vs. ABPI for toxicities (CTCAE v5.0) and physician-reported cosmesis (Harvard Scale). Results Among patients treated from January 2015 to March 2025, 200 patients met inclusion criteria, 36 (APBI) and 164 (WBI). The median age was 66 years (IQR: 58-71). Pure ILC was present in 179 patients (89.5%), while 21 (10.5%) had mixed lobular and ductal components. Most patients had pT1 tumors (77.5%), grade 1-2 (96.5%), negative margins (98%), estrogen receptor-positive tumors (98%), received endocrine therapy (91.5%), and had no lymphovascular invasion (98.5%). All patients were clinically node-negative, and the median Oncotype score was 18. APBI patients were older (median 69 vs. 64 years; p < 0.01) and more likely to have T1 tumors (91.7% vs. 74.4%; p = 0.03). Baseline characteristics, such as tumor grade and receptor status, were similar between groups. 3D conformal RT was used exclusively in both groups. Most patients (92.5%) were treated in the prone position. APBI regimens included 28.5 Gy in 5 fractions on non-consecutive days (61.1%) and 38.5 Gy in 10 twice-daily (BID) fractionation (38.9%). WBI regimens included 40 Gy in 15 fractions (33.5%) and 42.56 Gy in 16 fractions (53.7%). 74.5% of WBI plans used a tumor bed boost. The median follow-up was 4.7 years. The 5-year IBTR was 2.8% in the APBI group and 0.6% in the WBI group (p = 0.26). The 5-year DFS was 97.2% (APBI) and 97.5% (WBI), p = 0.50, and the 5-year OS was 100.0% in APBI group and 97.6% in WBI group (p = 0.45). No Grade 3 or higher acute toxicities occurred. Common Grade 1-2 acute toxicities were fatigue (63.9%) and dermatitis (77.0%). Dermatitis was significantly more common in WBI group (86.6%) than in APBI group (34.3%; p < 0.01). Grade 2 dermatitis was observed in 15.4% with WBI vs. 0% with APBI. Among those patients with Grade 1-2 late toxicities at 1 year, common findings in both groups included skin induration (70.9%), hyperpigmentation (55.7%), and breast atrophy (53.5%). One WBI patient experienced Grade 3 breast atrophy. Skin hyperpigmentation was more common in WBI group than in the APBI group (61.8% vs 35.0%, p-value=0.04). One- and 3-year physician-reported cosmesis were similar. Data were available for 138 patients at 1 year. Excellent or good cosmetic outcomes were reported in 93.1% (APBI) and 93.6% (WBI), p = 0.67. At 3-years, cosmetic outcomes were available for 74 patients. Excellent or good cosmesis was 90% (APBI) and 89.1% (WBI), p-value = 0.20. Conclusions In our experience, APBI appears to be a safe and effective treatment option for select patients with low-risk ILC who undergo endocrine therapy. In addition, cosmesis appears equivalent at 1- and 3-years follow up. However, given the potential for late recurrences in hormone receptor-positive disease, longer follow-up is necessary. We plan to continue this investigation with additional patient data on IBTR and survival rates, as well as long-term toxicity and cosmesis.
Presentation numberPS1-07-25
The Impact of Post Mastectomy Radiotherapy(PMRT) on T1-2N0-1 Male Breast Cancer(MBC) and establishment of an artificial neural network(ANN) predicting model: Population-Based Study
Kaiyan Huang, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
K. Huang1, Y. Yu2, X. Li2, Y. Liu2, K. Huang1, X. Wang1, J. Zhang2; 1Department of Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, CHINA, 2Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA.
Objective: The objective of this study was to analyze the impact of post-mastectomy radiotherapy (PMRT) in male breast cancer (MBC) patients and develop an artificial neural network (ANN) model to identify a potential PMRT benefit population. Methods: Data from a total of 2,247 MBC patients with T1-2N0-1M0 who underwent total mastectomy between 1998 and 2016 were enrolled from the SEER database. Propensity score matching was used to reduce covariate imbalances. Cox regression analysis was conducted to compare overall survival (OS) between the PMRT and no-PMRT groups. The hypothesis was that patients who had undergone PMRT and lived longer than the median OS of the no-PMRT group could benefit from PMRT. An ANN model was then developed to predict PMRT benefit population. Results: Multivariate Cox regression analysis demonstrated better OS in the PMRT group compared to the no-PMRT group of matched patients. This survival benefit was particularly significant in patients with grade III or T2N0 and T2N1 disease, while no significant difference was observed in patients with grade I/II or T1N0 and T1N1 disease. An ANN model was established to predict PMRT benefit population based on patients with T2N0/T2N1. The optimal cut-off value for the model predicted probability was 0.51. Survival curves indicated that a score of 0.51 could accurately distinguish potential PMRT benefit population. Conclusions: For MBC patients with T2N0, T2N1, and grade III, PMRT would improve survival. The ANN model would be used to identify patients who are likely to benefit from PMRT and aid in clinical decision-making.
Presentation numberPS1-07-26
Partial Breast Adaptive Radiation Therapy Workflow Decreased Planning Time with Increased Institutional Experience
Narine Wandrey, UT Southwestern, Dallas, TX
N. Wandrey, D. Parsons, J. Wang, M. Arbab, P. Alluri, X. Li, A. Rahimi; Radiation Oncology, UT Southwestern, Dallas, TX.
Background: Adaptive radiation therapy (ART) is a personalized radiation treatment approach which adjusts the treatment plan during the radiation course to account for anatomic changes and tumor response. Real-time adjustment allows for smaller margins, particularly in stereotactic partial breast irradiation ([SPBI], 1). We have implemented a radiation therapy technologist (RTT) driven workflow at our institution which was previously shown reduces treatment demand on physicians without compromising plan quality (2). Here we report with more experience the adaptive stereotactic partial breast irradiation (A-SPBI) workflow is more efficient and total treatment time is quicker for patients. Objectives: Describe our institutional experience with A-SPBI and demonstrate that with increased ART experience, treatment time is quicker while maintaining plan quality and delivery compared to the inception of the ART program. Methods: We retrospectively reviewed patients with early stage breast cancer treated with A-SPBI between 01/2023-03/2025. Patients received SPBI on a cone beam computed tomography-based ART platform. Thirty Gy in 5 fractions was prescribed to the planning target volume (PTV) delivered twice weekly on non-consecutive days. A total of 867 fractions were evaluated. RTTs prospectively collected treatment planning time which was defined as time taken from the initial cone beam to time at which the plan was signed by the physician.Results: Eight hundred and sixty-seven A-SPBI fractions were evaluated from 01/2023-03/2025. From 01/2023-06/2023, ART was physician-driven where clinicians contoured in-person organs at risk (OARs) and target volumes for each fraction with an average planning time of 17.4 minutes. During this time, RTTs were present to review contours and concurrently trained for ART certification, the process for certification which has previously been described (2). From 07/2023-04/2024, RTTs contoured OARs and target volumes under direct supervision from a physicist or physician with an average planning time of 22.2 minutes. Importantly during this time, 95% of fractions had no major edits or only stylistic changes recommended by the reviewing physician. From 10/2024 – 03/2025, A-SPBI was primarily RTT-driven. RTTs generated OARs and target volumes with remote physician review with an average planning time of 19.8 minutes. From 10/2024-03/2025, this was reduced to 17.0 minutes. Comparing 03/2025 to 07/2023, RTTs have decreased treatment
delivery time by 3.3 minutes per fraction (p=0.0004 welch’s t-test) while allowing physicians to review contours and plans remotely, and also doubling total number of delivered fractions. Conclusions: With 27 months of follow up and more experience, A-SPBI treatment delivery time has decreased while still maintaining plan quality with an RTT-driven workflow platform. Sources: 1. Montalvo SK, Kim N, Nwachukwu C, Alluri P, Parsons D, Lin MH, Cai B, Zhuang T, Hrycushko B, Chen L, Timmerman R, Rahimi A. On the feasibility of improved target coverage without compromising organs at risk using online adaptive stereotactic partial breast irradiation (A-SPBI). J Appl Clin Med Phys. 2023 Feb;24(2):e13813. doi: 10.1002/acm2.13813.2. Parsons D, Domal S, Chambers E, Salazar D, Visak J, Arbab M, Holgado F, Li X, Okoro C, Wandrey N, Iqbal Z, Alluri P, Cai B, Keen H, Cleaton J, Godley A, Sher D, Badiyan S, Rahimi A, Lin MH. Feasibility and Impact of a Radiation Therapy Technologist-Driven Workflow for Cone Beam Computed Tomography Guided Partial Breast Adaptive Radiation Therapy. Int J Radiat Oncol Biol Phys. 2025 Jul 15;122(4):893-901. doi: 10.1016/j.ijrobp.
Presentation numberPS1-07-27
A Single-Institutional Analysis of Survival Outcomes and Predictors in pN1 BreastCancer: Is There a Role for Post-Mastectomy Radiation?
Raksha Madhu Narasimhan, University of Miami Miller School of Medicine, Miami, FL
R. M. Narasimhan1, X. Zhao2, S. Han2, A. S. Saini1, K. Samimi1, I. Ogobuiro1, C. S. Taswell1, C. Takita1; 1Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL, 2Radiation Oncology, Sylvester Comprehensive Cancer Center, Miami, FL.
Purpose/Objectives: Post-mastectomy radiation therapy (PMRT) is known to benefit patients with pathologic N2+ breast cancer, but its impact on pN1 disease remains uncertain. The primary objective of this study was to examine whether PMRT improves recurrence-free survival (RFS) and overall survival (OS) in cN1 breast cancer patients. Various clinical factors were also assessed for associations with survival outcomes. Materials/Methods: A retrospective review of pT1N1M0 and pT2N1M0 breast cancer patients treated at one institution from 2016-2022 was conducted. RFS was defined as the elapsed time from diagnosis to recurrence, death, or last follow-up. OS was defined as the elapsed time from diagnosis to death or last follow-up. The Kaplan-Meier method was used to estimate RFS and OS, and groups were compared using the log-rank test. Cox proportional hazards regression models assessed associations between clinical factors—including age, race, receptor status, tumor size, laterality, and adjuvant therapies—and RFS and OS. Results: Among 57 patients, 22 (38.6%) received PMRT and 35 (61.4%) did not. There were no significant differences in baseline demographic or clinical characteristics between the two groups (p > .05). Median RFS and OS were higher in the PMRT group (133 months and not estimable [NE]) than in the non-PMRT group (120 and 195 months); however, the differences were not statistically significant (p = 0.256 and p = 0.154, respectively).Univariable Cox analyses across the entire cohort found hormone therapy to be significantly associated with improved RFS (HR = 0.43, 95% CI: 0.21-0.90, p = 0.026) and OS (HR = 0.13, 95% CI: 0.04-0.50, p = 0.003). Receptor subtype (ER+/PR+/HER2− vs. triple negative) was also associated with longer OS (HR = 0.25, 95% CI: 0.08-0.80, p = 0.019), but not with RFS (p = 0.839). In subset analyses of 25 non-PMRT patients, tumors located in the left breast were significantly associated with improved RFS (HR = 0.25, 95% CI: 0.09-0.73, p = 0.011) and OS (HR = 0.22, 95% CI: 0.05-0.89, p = 0.033), compared to right-sided tumors. Additionally, having two or more positive lymph nodes (vs. one: HR = 2.89, 95% CI: 1.15-7.27, p = 0.024) and hormone therapy (HR = 0.15, 95% CI: 0.03-0.70, p = 0.016) were significant predictors of RFS and OS, respectively. Conclusions: PMRT did not significantly improve RFS or OS in patients with N1 breast cancer, though a trend toward longer survival was observed. Hormone therapy, receptor subtype, tumor laterality, and nodal burden emerged as predictors associated with survival outcomes. These findings highlight the importance of individualized treatment decisions based on tumor biology and the adjuvant therapy context. Further investigation with a larger sample size is required to clarify the role of PMRT in patients with pathologic N1 breast cancer.
Presentation numberPS1-07-29
Use of cream with 5% panthenol and Centella asiatica extract in patients undergoing breast radiotherapy: a randomized study in Uruguay.
Federico Lorenzo, Hospital de Clinicas, Montevideo, Uruguay
J. C. Ferrer, S. Guerreros, M. Torres, M. Vola, E. Rivero, L. Ricagni, A. Notejane, M. Giordano, P. Fernandez, S. Roldan, F. Lorenzo; Montevideo, Hospital de Clinicas, Montevideo, URUGUAY.
Background: Breast cancer is the most common malignancy in Uruguay, with approximately 75% of women treated for this disease receiving adjuvant radiotherapy. Radiation-induced skin toxicity (radiodermatitis) significantly impacts treatment tolerance, yet no robust evidence supports a single standard topical treatment to mitigate these adverse effects. This study aims to evaluate whether a topical cream containing 5% panthenol and Centella asiatica extract can effectively reduce radiodermatitis compared to standard care in breast cancer patients undergoing radiotherapy. Methodology: This prospective, randomized, open-label clinical trial will be conducted at Hospital de Clínicas Dr. Manuel Quintela in Uruguay. Eighty patients with breast cancer requiring adjuvant radiotherapy using hypofractionated schemes (240-340 cGy per fraction) will be randomized in a 1:1 ratio to receive either: daily water washing plus twice-daily application of a cream containing Centella asiatica extract and 5% panthenol (Group A), or daily water washing alone (Group B). Weekly clinical evaluations during treatment will be performed by radiation oncologists using the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. Consenting patients will also have photographic documentation evaluated by an independent, blinded dermatologist. The primary endpoint is radiodermatitis-free time measured in days. Secondary endpoints include the proportion of patients experiencing radiodermatitis of any grade and the proportion developing grade ≥2 radiodermatitis. Statistical analysis will use Kaplan-Meier survival curves with log-rank tests for univariate analysis and Cox regression for multivariate analysis, using SPSS version 29.0. Sample size was calculated to detect a 12-day difference in radiodermatitis onset with 80% power and significance level of 0.05. Expected outcomes: This study will determine whether a cream containing 5% panthenol and Centella asiatica extract can delay radiodermatitis onset and reduce its incidence during breast radiotherapy. The results will provide more the needed evidence to standardize skin care protocols in radiotherapy, potentially improving treatment tolerance and quality of life for breast cancer patients undergoing radiotherapy. Registration: This study has been registered with the Uruguayan Ministry of Public Health (Registration number: 9170979, registered April 21, 2025).
Presentation numberPS1-07-30
Cherenkov Imaging as a Real-Time Quality Improvement Tool in Breast Radiotherapy: A Case Series
Allison Leigh Matous, Dartmouth Cancer Center / Dartmouth Health, Lebanon, NH
A. L. Matous1, E. Chen2, L. Jarvis1; 1Department of Radiation Oncology and Applied Sciences, Dartmouth Cancer Center / Dartmouth Health, Lebanon, NH, 2Department of Radiation Oncology, Cheshire Medical Center, Keene, NH.
Title: Cherenkov Imaging as a Real-Time Quality Improvement Tool in Breast Radiotherapy: A Case Series Background: Radiotherapy is an essential component of definitive treatment for many patients with breast cancer.Cherenkov imaging is an emerging optical imaging technology that enables visualization of radiotherapy beams on the patient’s surface in real-time. The anatomy and treatment setup of breast radiotherapy make it particularly well suited for evaluation with this technique. While Cherenkov imaging typically confirms accurate treatment delivery, it has also revealed instances of irradiation to unintended tissues. We hypothesized that Cherenkov imaging could be used as a quality improvement tool to identify and minimize radiation dose to normal tissues among patients receiving radiotherapy for breast cancer. Methods: As part of a prospective clinical trial, Cherenkov images were reviewed daily for all patients receiving breast radiotherapy at 1 academic and 1 community medical center. When dose to anatomical regions outside the treated breast was identified on Cherenkov images, a root cause analysis was performed to determine contributing factors. Treatment plans were re-evaluated using the treatment planning system (TPS), and modifications were made when clinically appropriate. Results:•Case 1: Cherenkov imaging identified unexpected dose on the ipsilateral back of a patient treated in the prone position. TPS review revealed an anterior-posterior (AP) beam exiting through the heart. The plan was revised to eliminate the AP field, and institutional practice was updated to minimize heart and lung exposure in similar cases.•Case 2: Excess dose to the left arm was identified in a prone left breast treatment. Review suggested that adjusting the multi-leaf collimators (MLCs) could reduce exposure. A revised plan was implemented, resulting in reduced arm dose on subsequent fractions.•Case 3: Cherenkov imaging showed signal on the chin and neck of a young patient receiving supraclavicular irradiation. A more optimal setup was identified, the patient was re-simulated with modified positioning, and a new plan was created. Subsequent imaging confirmed reduced unintended exposure in the revised plan.•Case 4: A patient noted a sore throat 2 weeks into a course of breast and supraclavicular irradiation. Review of Cherenkov images and the TPS suggested contralateral extension of the right supraclavicular field due to a setup issue during treatment delivery, as breast anatomy and the supraclavicular field did not correlate well. MLCs were adjusted to minimize esophagus dose. •Case 5: Cherenkov imaging showed unexpected contralateral breast dose. Review of on-treatment imaging and the TPS suggested this was due to patient setup, and modifications were made in to setup in future treatments to minimize contralateral breast dose. Conclusions:Cherenkov imaging provides real-time visual feedback on beam delivery during breast radiotherapy and is a valuable tool for identifying and improving suboptimal deliveries. By increasing the salience of treatment plan features that may be overlooked with traditional plan evaluation strategies and enabling timely plan adjustments, this methodology has the potential to improve patient safety. Incorporating Cherenkov imaging into routine clinical workflows may enhance the quality of breast radiotherapy by minimizing the irradiation of off-target tissues.
Presentation numberPS1-08-01
Safety of Combining Radiotherapy With Sacituzumab Govitecan in Patients With Advanced Triple-Negative Breast Cancer: An International Multicenter Cohort Study
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
A. Polakiewicz-Gilowska1, M. Pieniążek2, M. Holánek3, Z. Bielčiková4, K. Winsko-Szczęsnowicz5, A. Konieczna6, I. Kolářová7, R. Soumarová8, T. Ciszewski9, J. Żubrowska10, H. Študentová11, M. Malejčíková12, A. Młodzińska6, M. Lisik-Habib13, A. Pękala13, D. Krejčí14, J. Šustr15, I. Danielewicz16, M. Szymanik-Resko16, L. Rusinova17, B. Czartoryska-Arłukowicz5, A. Łacko2, R. Pacholczak-Madej18, M. Jarzab1, M. Püsküllüoğlu19, M. Kubeczko1; 1Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 2Department of Oncology; Breast Unit Clinical Oncology Day Care Department, Wrocław Medical University; Lower Silesian Comprehensive Cancer Center, Wrocław, POLAND, 3Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, and Masaryk Memorial Cancer Institute, Brno, CZECH REPUBLIC, 4Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 5Department of Clinical Oncology, M. Skłodowska-Curie Bialystok Oncology Center, Białystok, POLAND, 6Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warszawa, POLAND, 7Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Králové, CZECH REPUBLIC, 8Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 9Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 10Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, POLAND, 11Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 12Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 13Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 14Department of Oncology, First Faculty of Medicine, Charles University in Prague; Bulovka University Hospital, Prague, CZECH REPUBLIC, 15Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University; University Hospital Pilsen, Plzen, CZECH REPUBLIC, 16Oddział Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 17Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 18Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND, 19Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND.
Background. Antibody-drug conjugates (ADC) such as sacituzumab govitecan (SG) have significantly improved outcomes in advanced triple-negative breast cancer (TNBC). However, many patients undergoing SG require radiotherapy (RT) for symptom control or oligoprogression. Clinical data on the safety of concurrent or sequential SG and RT are scarce. We aimed to evaluate the tolerability and survival outcomes associated with combining RT and SG in real-world clinical practice. Methods. We retrospectively analyzed 303 female patients treated for metastatic or unresectable locally advanced TNBC between August 2021 and May 2025 across multiple oncology centers in Poland, The Czech Republic, and Slovakia. All patients received SG; 66 (21.8%) received RT either prior to (n=33) or during (n=33) SG therapy. Progression-free survival (PFS) was defined as the time from SG initiation to disease progression or death. Overall survival (OS) was defined as the time from SG initiation to death from any cause or last follow-up. Survival was estimated using the Kaplan-Meier method; group comparisons were performed using Fisher’s exact test. Analyses were performed with Stata Statistical Software 19.0 (StataCorp LLC, College Station, TX, USA). Results. Among the 303 patients, the median PFS was 5.1 months in those who did not receive RT (237 patients, 78.2%) and 4.2 months in those who did (66 patients, 21.8%; p=0.42). The 6-month PFS rates were 42.0% (95% CI, 35.3-48.5%) and 36.9% (95% CI, 24.5-49.3%), respectively. Median OS was 11.9 months in the non-RT group versus 9.3 months in the RT group (p=0.14), with 12-month OS rates of 49.5% (95% CI, 42.1-56.4%) and 36.2% (95% CI, 23.7-48.8%), respectively. Patients in the RT group more frequently had central nervous system metastases (27.3% vs 4.6%, p<0.001), indicating higher baseline disease burden. SG dose reductions occurred in 34.9% of patients receiving RT and 38.4% of those not receiving RT (p=0.35). The most common reason for SG discontinuation in both groups was disease progression. Discontinuation due to adverse events or declining performance status was rare: 1.5% in the RT group and 2.1% in the non-RT group (p=1.00). There were no statistically significant differences in the rates of all grade treatment-related toxicities between groups: neutropenia (74.2% with RT vs 67.5% without RT, p=0.37), diarrhea (15.2% vs 20.7%, p=0.38), anemia (47.0% vs 37.6%, p=0.20), thrombocytopenia (16.9% vs 8.0%, p=0.06), or fatigue (43.3% vs 39.1%, p=0.56). The safety profile of SG was not adversely affected by the addition of RT. Conclusions. In this large real-world multicenter cohort, the addition of radiotherapy to sacituzumab govitecan appeared safe and was not associated with increased toxicity, dose reductions, or treatment discontinuation. The modest differences in survival outcomes likely reflect greater disease burden, particularly CNS involvement, in patients receiving RT. Prospective studies are needed to optimize the integration of RT with ADC-based therapies in advanced TNBC.
Presentation numberPS1-08-02
Feasibility of accelerated partial breast irradiation (APBI) following neoadjuvant systemic therapy and lumpectomy
Ari Azad Kassardjian, City of Hope, Duarte, CA
A. A. Kassardjian, J. Nall, T. Reilly, C. Wong, S. Yoon, T. Watkins, J. Bazan; Radiation Oncology, City of Hope, Duarte, CA.
Purpose/Objectives: Response to neoadjuvant systemic therapy (NST) is often associated with favorable outcomes in breast cancer patients. We have demonstrated that patients with pathologic complete response (pCR) or minimal residual disease following breast conserving surgery (BCS) and treated with whole breast irradiation (WBI) have in-breast recurrence rates low enough that de-escalation of therapy with accelerated partial breast irradiation (APBI) may be safe. However, the role of APBI in this population remains poorly described. Herein, we retrospectively evaluated the technical feasibility of APBI in women who received NST and BCS. Materials/Methods: We have an established database of 321 breast cancer patients at a single comprehensive cancer center treated with NST, BCS, and WBI from 2017-2021. From this group, we identified a subset of patients that had pre-chemotherapy magnetic resonance imaging (MRI) of the breasts and had not undergone oncoplastic reduction mammoplasty at the time of BCS. On the radiation treatment planning CT scans, we contoured the surgical bed gross tumor volume (GTVsb). A 1.0-1.5 cm margin was added to create the surgical bed clinical target volume (CTVsb), and a final 0.5 cm margin was added to create the surgical bed planning target volume (PTVsb_Eval), which was cropped 0.4 cm from the skin and did not extend outside of breast tissue. We also contoured the ipsilateral breast (IB) to calculate the ratio of the PTVsb_Eval volume to the IB volume (PTV/IB-R). Technical feasibility of APBI was defined as GTVsb location within the same quadrant as the primary tumor on MRI and PTV/IB-R≤30%. APBI treatment plans using volumetric modulated arc radiotherapy (VMAT) were retrospectively created for patients with PTV/IB-R in the range of 25-30% using the dose/fractionation schedule of 3000 cGy in 5 fractions and the same planning constraints as the University of Florence APBI trial. Our primary endpoint was to establish the technical feasibility rate of APBI in this patient population with a rate of ≥70% defined as worthy of moving forward with development of a prospective trial. Results: From our cohort, we identified 40 women who received NST followed by BCS and WBI. Median age was 53 years (IQR 47-61 years), with most patients being Hispanic White (50%), non-Hispanic White (22.5%), Black or African American (10%), or Asian (7.5%). Median body mass index (BMI) was 30 kg/m2 (IQR 26.9-35.2 kg/m2), and 35% were pre-menopausal. When delineating the GTVsb, 92.5% had visible metallic clips, 52.5% had no associated seroma, and 10% underwent local tissue rearrangement during BCS. All GTVsb volumes were concordant with the same quadrant as the primary tumor visualized on pre-treatment MRI. The median margin from GTVsb to CTVsb was 1.0 cm (range, 1.0-1.5 cm). Median PTV_eval volume was 138.2 cc (IQR 98.3-232.5 cc), and median IB volume was 993.6 cc (IQR 708.3-1587.7 cc). The median PTV-IB/R was 15.3% (IQR 11.6-19.5%). Three patients had PTV/IB-R values above 25% (29.2%, 25.5%, and 25.5%). All 3 corresponding VMAT plans met all dosimetric planning objectives and normal tissue constraints for acceptable APBI plans. Therefore, the technical feasibility rate of APBI in this patient cohort was 100%. Conclusion: Our current analysis indicates that APBI is technically feasible in patients that receive NST followed by BCS. Pre-chemotherapy MRI is an important prerequisite to ensure that the GTVsb is accurately defined. Coupled with recent evidence from our institution demonstrating that patients with early-stage (cT1-3 cN0-1) breast cancer achieving pCR or lymph node-negative with minimal residual disease in the breast (cN0/ypN0 and ≤ypT1c) have low rates of in-breast tumor recurrence after WBI, the current study supports prospective evaluation of APBI in this favorable patient population.
Presentation numberPS1-08-03
Three-year outcomes following five-fraction sbrt for apbi in early-stage breast cancer: cosmesis, functional status, and local control
Melinda Gevorgian, Alabama College of Osteopathic Medicine, Dothan, AL
M. Gevorgian1, V. Reddy2, T. Mathew3, Y. Liu4, C. Veale5, D. Hablitz4, H. Krontiras6, A. Dalton4, K. Meyers4, M. Dobelbower4, R. Lancaster6, M. Bredel4, C. Parker6, K. Keene4, E. Thomas7, D. H. Boggs4; 1Student, Alabama College of Osteopathic Medicine, Dothan, AL, 2Student, University of Alabama at Birmingham, Birmingham, AL, 3Internal Medicine, Southeast Health, Dothan, AL, 4Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 5Radiation Oncology, Columbus Community Hospital, Columbus, NE, 6Surgery, University of Alabama at Birmingham, Birmingham, AL, 7Radiation Oncology, Renaissance Institute of Precision Oncology & Radiosurgery, Winter Park, FL.
Background:For many women with early-stage breast cancer, accelerated partial breast irradiation (APBI) offers comparable tumor control to whole-breast radiation with a shorter treatment course. However, cosmetic outcomes vary by technique. Stereotactic body radiotherapy (SBRT) is a promising external beam method allowing highly conformal dose delivery with tight margins, potentially improving cosmesis by sparing normal tissue. We present final three-year results from a prospective trial evaluating a five-fraction SBRT regimen for APBI, focusing on long-term safety, cosmesis, functional status, and local-regional control.
Methods:Twenty-three patients with early-stage breast cancer (24 treatment plans) received 30 Gy in five daily fractions. A bioabsorbable 3D tissue marker (BioZorb™) was placed at lumpectomy for cavity delineation. Clinical target volume (CTV) included the cavity plus 1-cm margin; planning target volume (PTV) added 3 mm and was limited to ≤124 cc. Median PTV volume was 76.0 cm³ (range, 50.3–211.7). Treatment was delivered using 10-MV flattening filter-free beams on a Varian Edge system with VMAT planning and daily cone-beam CT. Positioning was prone or supine per anatomy; two patients had 9- and 11-day courses due to non-clinical delays. Zubrod status, patient- and nurse-reported cosmesis (4-point scale), adverse events, and imaging (mammography, ultrasound, MRI) were evaluated through 36 months.
Results:At 36 months, 20 of 23 patients remained on protocol. One experienced biopsy-confirmed nodal recurrence at 6 months; two were lost to follow-up with benign or stable imaging. No ipsilateral breast tumor recurrences were observed. Zubrod status remained 0 in 90% at 36 months, with no grade ≥2 functional decline. Patient-reported cosmesis was excellent or good in 75% at 36 months (down from 85% baseline); nurse-reported was 80%, with stability across intervals. Poor cosmesis was reported by ≤2 patients at any timepoint. Fat necrosis affecting cosmesis occurred at 18 months but resolved. Erythema, pigment change, retraction, and volume loss were noted at 24 months but were typically non-cosmetic. Excellent/good cosmesis was maintained in ≥75% of patients across all intervals.
Conclusion:This five-fraction SBRT approach to APBI was safe, well-tolerated, and preserved long-term cosmesis and function. Among patients completing 36-month follow-up, no ipsilateral breast tumor recurrences occurred. SBRT is a promising, efficient alternative to whole-breast radiation in carefully selected early-stage patients.
Figure 1. Patient-scored cosmesis transitions from baseline through 36 months.Cosmesis scores (Excellent, Good, Fair, Poor) were reported at standard follow-up intervals. The majority of patients maintained or improved their cosmesis rating over time, with minimal deterioration. Most transitions remained within the “Excellent” or “Good” categories.
Figure 2. Nurse-Reported Cosmesis Over TimeFigure 2. Nurse-scored cosmesis transitions from baseline through 36 months.Nurse assessments followed a similar trend, though with slightly lower ratings compared to patient self-reports. A stable trend was observed across the 3-year span, with the majority of patients rated “Excellent” or “Good” throughout follow-up.
Presentation numberPS1-08-04
Acute skin toxicity with ultra-hypofractionated whole breast radiation therapy after lumpectomy for breast cancer: a single institutional experience
Samantha Juang, NYITCOM, Old Westbury, NY
S. Juang1, J. Neiswander2, A. Ragupathi1, D. Zhang1, M. M. Plummer1, R. Rajasree3, E. Obedian3; 1Department of Biomedical Sciences, NYITCOM, Old Westbury, NY, 2Department of Biomedical Sciences, NYITCOM, Jonesboro, AR, 3The Cancer Institute, Saint Francis Hospital, East Hills, NY.
Intro: Randomized control trials have shown that adjuvant breast radiation therapy (RT) reduces local recurrence after lumpectomy for breast cancer. The most common adverse effect of RT is radiation dermatitis and is seen in 90% of patients. Hypofractionated (HF) dosing leads to a lower degree of acute radiodermatitis and better patient outcomes compared to conventional radiation, and we aimed to see if ultra-hypofractioned radiation therapy (UHF-RT) continues this trend.1 The current standard of care has changed from treatment over 5 – 6 weeks to a more accelerated HF approach typically over 3 – 4 weeks. Based on the FAST-FORWARD trial, a 5-fraction course of UHF-RT is evolving into a new standard of care to significantly reduce costs and increase access, compliance, and adoption of adjuvant radiotherapy.2 The purpose of this study is to review the acute skin toxicity in a cohort of patients who have undergone UHF-RT. Methods: We conducted a retrospective chart review of 20 patients who have undergone post-lumpectomy UHF-RT. All 20 patients received 2600 cGy to the whole breast in 5 fractions, and 8 of those patients received a single fraction boost dose of 520 cGy. Radiation plans were delivered as field in field tangential plans using 6 MV photons with a dose homogeneity within 105%. We graded skin toxicities G0 to G5, according to NCI-CTCAE criteria, at 1-month and 6-months post-treatment based on follow-up examinations. Results: All patients were females between the ages of 50 and 95 years old. 85% were ER+, 85% were PR+, and 5% were HER2+. The Ki-67 percentages ranged from 10% to 70%. There were no known ATM mutations in our cohort. T stages were as follows: Tis (20%), T1 (65%), and T2 (15%). Lymph node status showed: N0 (85%), N1 (10%), and NX (5%). The radiation fields treated breast only in 95% (n=19) and breast and axillary nodes in 5% (n=1). In addition to UHF-RT, 10% (n=2) of patients received chemotherapy and 80% (n=16) received anti-estrogen therapy. Skin toxicity at 1 month was G0 for 4/20 and G1 for 16/20 patients. For patients with a minimum 6-month follow-up (n=16), skin toxicity observed was G0 for 11/16 and G1 for 5/16 patients. Of the 20 patients who received UHF-RT, 40% (n=8) received a boost dose. At 1-month, 8/8 of boost dose patients were G1. Boost dose patients with a 6-month follow-up (n=6) showed G1 for 4/6 and G0 for 2/6. None of the 20 patients developed worse than a G2 skin toxicity, and all patients completed their treatment as planned, with no breaks in treatment. Conclusion: UHF-RT significantly reduces the costs of treatment and therefore can improve patient compliance and access. This study shows an additional benefit of decreased skin toxicity severity after UHF-RT, even with a boost dose administered, compared to HF radiation protocol. Though these results are promising, further follow-up is necessary to confirm these findings and to assess potential long-term effects (> 6 months) of UHF-RT after lumpectomy for breast cancer. References: 1. Borm, K. J., et. al. (2021). A Comprehensive Prospective Comparison of Acute Skin Toxicity after Hypofractionated and Normofractionated Radiation Therapy in Breast Cancer. Cancers, 13(22), 5826. https://doi.org/10.3390/cancers13225826 2. Brunt, A. M., et al. (2020). Hypofractionated breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-year efficacy and late normal tissue effects results from a multicentre, non-inferiority, randomised, phase 3 trial. The Lancet, 395(10237), 1613-1626. https://doi.org/10.1016/s0140-6736(20)30932-6
Presentation numberPS1-10-01
Updated analyses from the retrospective rAMBER study exploring abemaciclib monotherapy after prior CDK4/6 inhibitor progression in metastatic hormone-receptor positive breast cancer
Sahar Shahamatdar, Massachusetts General Hospital, Boston, MA
S. Shahamatdar1, K. Clifton2, H. Maynard3, U. Joshi4, J. Wu1, A. Dedeoglu1, A. Putur1, I. Kuter1, A. Bardia5, D. Juric1, L. Spring1, K. Harris1, B. Moy1, J. Shin1, N. Vidula1, A. Medford1, T. Mulvey1, A. Brufsky3, H. Han4, C. Ma2, S. Wander1; 1Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, 2Medical Oncology, Washington University in St. Louis, St. Louis, MO, 3Division of Malignant Hematology and Medical Oncology, University of Pittsburgh, Pittsburgh, PA, 4Hematology/Oncology, Moffitt Cancer Center, Tampa, FL, 5Department of Medicine, Division of Hematology/Oncology, UCLA, Los Angeles, CA.
Introduction: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) have become the standard of care for patients with advanced hormone-receptor positive (HR+)/HER2- breast cancer. The phase III postMONARCH trial demonstrated a modest improvement in progression free survival with fulvestrant and abemaciclib in the second-line advanced setting after prior CDK4/6i progression. However there remains uncertainty regarding the efficacy of CDK4/6i re-introduction. Here, we provide updated data from the rAMBER (retrospective multi-center, biomarker study to evaluate efficacy of Abemaciclib Monotherapy Beyond first line CDK4/6 inhibition in ER+ breast cancer) study exploring clinical outcomes with abemaciclib monotherapy after prior CDK4/6i progression. Methods: We collected retrospective data at 4 academic cancer centers (Massachusetts General Hospital, Washington University in St. Louis, University of Pittsburgh Medical Center, Moffitt Cancer Center) in accordance with site-specific IRB protocols. We identified patients with metastatic HR+/HER2- breast cancer with disease progression on CDK4/6i-based therapy who later received abemaciclib monotherapy. We abstracted clinical outcomes, analyzed radiographic data using Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and explored genetic correlates of response and resistance from cell free DNA retrospective data. Results: A total of 30 patients were treated with abemaciclib monotherapy following progression on CDK4/6i-based regimens (all included palbociclib). The median duration of palbociclib treatment was 7.9 months (95% CI: 4.6-12.7 months), with a median number of 2 intervening lines of therapy prior to abemaciclib initiation. In this cohort, 23 patients remained on abemaciclib until disease progression or death, while 7 discontinued treatment due to adverse effects, including gastrointestinal issues and bone marrow suppression. The median duration of abemaciclib treatment was 4.0 months (95% CI: 2.8-13.9 months). Notably, 11 patients continued abemaciclib for more than 180 days before experiencing progression or death, whereas 6 patients had disease progression within 90 days of initiation. The majority (22) of patients had visceral and bony disease, while 6 patients had bone-only disease and 2 only had visceral disease. Preliminary review of radiographic images with RECIST criteria from 16 patients revealed 1 patient with partial response, 8 patients with stable disease, and 7 patients with progressive disease. Preliminary analysis of cell free DNA sequencing from 10 patients revealed RB1 alterations in 4 patients; 3 patients had rapidly progressive disease while 1 patient who initially responded to abemacliclib had an the RB1 alteration discovered at time of progression. Ongoing analyses will be presented at the meeting, including characterization of radiographic response across the entire cohort and exploration of genomic sequencing. Conclusions: Approximately one third of patients derived benefit from abemaciclib monotherapy, tolerating the regimen for > 180 days, despite progression on palbociclib. In addition, preliminary genetic analyses revealed enrichment in RB1 alterations in patients with rapid disease progression. The rAMBER study is the first effort to evaluate the effectiveness of abemaciclib monotherapy following resistance to prior CDK4/6i, offering new perspectives on the role of sequential CDK4/6-targeted treatments. These results highlight potential benefit for continued CDK4/6 blockade and ongoing efforts may elaborate new insights related to genetic mechanisms of response and resistance and help identify patients who are likely to benefit from similar clinical strategies.
Presentation numberPS1-10-03
The Aurora Kinase A Inhibitor Alisertib Alone or in Combination with Endocrine Therapy for Patients with Estrogen Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative (ER+/HER2-) Metastatic Breast Cancer (MBC)
Tufia C Haddad, Mayo Clinic Rochester, Rochester, MN
T. C. Haddad1, K. McCann2, P. Advani3; 1Medical Oncology, Mayo Clinic Rochester, Rochester, MN, 2Medical Oncology, University of California Los Angeles, Los Angeles, CA, 3Medical Oncology, Mayo Clinic Jacksonville, Jacksonville, FL.
Background: Aurora kinase A (AURKA) plays an integral role in the mitotic cell cycle, regulating spindle assembly, centrosome function, chromosome alignment, and mitotic entry. In addition to its mitotic role, activated AURKA has been shown to induce epithelial-mesenchymal transition reprogramming and contribute to endocrine resistance in ER+ breast cancer (BC) models. Amplification/overexpression of AURKA is linked to poor prognosis across multiple tumor types, including ER+/HER2- BC. Alisertib is a selective, reversible, ATP-competitive AURKA inhibitor that has been shown to disrupt spindle formation, leading to mitotic defects and cell death. In xenografts, the potent inhibition of AURKA by alisertib led to antitumor activity across a variety of tumor types. Preliminary clinical data with alisertib have shown significant antitumor activity and a manageable safety profile in patients with various cancers, including ER+/HER2- BC. This study aims to better characterize the role of alisertib in patients with BC to provide a rationale for future clinical studies. Methods: Three studies evaluating alisertib in patients with ER+/HER2- MBC have been completed, including a phase I study of alisertib + fulvestrant (Haddad 2018), a phase II alisertib monotherapy study (Melichar 2015), and a phase II study of alisertib ± fulvestrant (TBCRC 041; Haddad 2023). Descriptive summary statistics were utilized for patient characteristics, as well as clinical safety and efficacy data. Results: A total of 126 patients were enrolled and evaluable in the 3 clinical studies, as per Table 1. All patients had MBC and received prior endocrine therapy and chemotherapy. All patients in TBCRC 041 also received a prior CDK4/6 inhibitor (CDK 4/6i). In these heavily pretreated patients, tumor objective response rates (ORR) were ~20%, and 6-month clinical benefit rates (CBRs) were 29-78%. The most common grade 3/4 adverse events (AEs) across studies were hematologic AEs (neutropenia, leukopenia, and anemia), and fatigue. In TBCRC 041, ctDNA- and tumor-derived RNA-Seq-based biomarker assessments are ongoing to investigate associations with clinical benefit from alisertib alone or combined with fulvestrant. Conclusions: Alisertib has shown efficacy and safety across multiple clinical studies in patients with ER+/HER2- MBC and could provide a novel option for treating those who have progressed on endocrine therapy and a CDK 4/6i. These findings provide a strong rationale for further development of alisertib in this patient population. The ongoing phase II ALISCA-Breast1 study (NCT06369285) is investigating the optimal dose of alisertib in combination with standard endocrine therapies in this setting.
| Table 1. Summary of data from 3 studies of alisertib ± fulvestrant in patients with ER+/HER2- MBC | |||||||
| Phase II Alisertib monotherapy Melichar et al, 2015 | Phase I Alisertib + fulvestrant Haddad et al, 2018 | Phase II, TBCRC041 Alisertib ± fulvestrant Haddad et al, 2023 | |||||
| No. of patients | 26 | 9 | 91 | ||||
| Regimen | Alisertib (50 mg twice daily on days 1-7 of a 21-day cycle) | Alisertib (40 or 50 mg twice daily on days 1-3, 8-10, and 15-17 of a 28-day cycle) + fulvestrant | Arm1: Alisertib 50 mg* Arm 2: Alisertib 50 mg* + fulvestrant *Dose/schedule per prior phase I study | ||||
| Allowable prior lines of therapy for MBC | Unlimited endocrine and ≤4 chemo regimens | ≥1 prior endocrine and ≤2 chemo regimens | Unlimited endocrine (fulvestrant required) and ≤2 chemo regimens | ||||
| Prior systemic therapy for MBC | 100% had ≥3 systemic therapies | 100% prior endocrine and 67% prior chemo | 100% prior endocrine; 48% and 69% prior chemo in Arms 1 and 2, respectively | ||||
| Prior fulvestrant | 62% | 67% | Arm1: 98% Arm 2: 100% | ||||
| Prior CDK4/6i | 0% | 0% | 100% | ||||
| Clinical efficacy | ORR: 23% DOR: 4.2 months CBR: 54% mPFS: 7.9 months | 2 of 3 patients with measurable disease had PR lasting 11.1 and 25.9 months. 6-month CBR: 78% mPFS: 12.4 months | Arm 1 ORR: 20% DOR: 15.1 months 24-week CBR: 41% mPFS: 5.6 months Arm 2 ORR: 20% DOR: 8.5 months 24-week CBR: 29% mPFS: 5.4 months | ||||
| Grade 3 or 4 AEs in ≥15% of patients (treatment-related AEs in Haddad et al 2023) | Overall MBC cohort: Neutropenia, 57%; Leukopenia, 36%; Stomatitis, 15%; Fatigue, 11% | Neutropenia, 22%; Leukopenia, 22%; Lymphopenia, 11% Fatigue, 11% | Arm 1 Neutropenia, 43%; Leukopenia, 17%; Anemia, 20%. Arm 2 Neutropenia, 42%; Leukopenia, 31%; Lymphopenia, 16%; Fatigue, 11%. | ||||
| Discontinuation due to AEs | n=2 (8%) | n=1 (11%) | Arm 1: n=2 (4%) Arm 2: n=6 (13%) | ||||
| DOR, duration of response; mPFS, median progression-free survival; PR, partial response. | |||||||
Presentation numberPS1-10-04
Metronomic Chemo-Endocrine Therapy with Fulvestrant and Polychemotherapy in Heavily Pretreated ER+ Breast Cancer Patients: A Single-Center Cohort Study”
Piotr Wysocki, Jagiellonian University-Medical College Hospital, Krakow, Poland
P. Wysocki1, A. Buda-Nowak1, &. Kwinta1, A. Michalowska-Kaczmarczyk1, K. Konopka1, M. Lubas1, M. Lobacz1, M. Jurczyk1, P. Potocki1, M. Koniewski2; 1Dept. of Oncology, Jagiellonian University-Medical College Hospital, Krakow, POLAND, 2Institute of Sociology, Jagiellonian University, Krakow, POLAND.
Introduction: There is no doubt that the combination of endocrine drugs with CDK4/6 inhibitors, which act as phase-specific antiproliferative agents, demonstrated a striking clinical activity and became a treatment of choice in advanced ER+ breast cancer. Therefore, we have hypothesized that combining ER degrader with metronomic polychemotherapy can improve clinical outcomes in advanced, endocrine-resistant ER+ breast cancer patients. Material and methods: The treatment (FulVEC) consisted of a combination of fulvestrant (500 mg i.m. on days 1, 14, and 28, and every 4 weeks thereafter) with a VEC regimen (vinorelbine 40 mg p.o. three times a week, capecitabine 500 mg p.o. three times a day, cyclophosphamide 50 mg p.o. once a day). After local ethical committee approval, this treatment was offered initially as a salvage therapy for advanced ER+ BC patients who had exhausted all available treatment options. Further, after promising results, also for patients who refused or were ineligible for standard intravenous chemotherapy. All patients had previously received at least one line of palliative systemic therapy (40% received > 3 lines and 22% received> 5 lines). Most (53%) have previously failed fulvestrant, and 34%, 32%, and 29% have previously failed palliative treatment with capecitabine, vinorelbine and cyclophosphamide, respectively. CDK4/6i have been previously administered to 47% of patients. Most patients presented with bone (82%) and liver (66%) metastases. Three patients (8%) had OUN metastases. The median duration of previous palliative systemic treatment was 23 months. All patient’s data were collected prospectively. Results: Between 2017-2025 – 68 patients (median age 49.5 years) were treated with the FulVEC regimen in a single-center cohort study. In the general population, administration of FulVEC led to at least disease stabilization in 87% of patients and was associated with median PFS and OS of 8.5 months and 21.5 months, respectively. The 12- and 24-month PFS and OS rates were 24%, 6% and 65%, 28%, respectively. Previous treatment with CDK4/6i, fulvestrant, vinorelbine, or cyclophosphamide had no impact on patient outcomes; however, previous use of capecitabine was associated with decreased median PFS (6.5 months, HR=1.58, p=0.12). Liver metastases were associated with significantly increased risk of death (HR=2.11, p=0.02). Median OS and PFS for patients who received 3-4 and ≥5 lines of previous treatment was 7.5 and 15.6 months, and 6.5 and 16.5 months, respectively. Three patients with symptomatic OUN metastases (2 with leptomeningeal dissemination) treated with FulVEC responded to the treatment with median PFS and OS of 11.6 and 25.8 months, respectively. The FulVEC regimen was generally well tolerated, with no treatment-related toxicity necessitating cessation. Temporary treatment interruption was required in 18% of patients due to G3-4 myelotoxicity, primarily neutropenia. Dose reduction, needed in 47% of patients, was due to myelotoxicity, capecitabine-induced hand-foot-syndrome, and cyclophosphamide-induced cystitis. Conclusion: Chemo-endocrine therapy (FulVEC) comprising fulvestrant and metronomic polychemotherapy demonstrated high activity in pretreated, endocrine-refractory breast cancer patients. The activity of the FulVEC regimen compares favorably to available novel anti-cancer strategies used after failure of endocrine therapies. One of the most critical aspects of this therapy, besides its antitumor activity and safety, is its cost, which is minimal compared to novel therapeutic options considered for this patient population. A phase III trial comparing FulVEC with physician treatment of choice in patients who failed first line HTH+CDK4/6i is warranted.
Presentation numberPS1-10-05
Efficacy and Safety of PI3K Inhibitors in Hormone Receptor Positive, HER 2 Negative Advanced Breast Cancer: A Meta-Analysis of Phase III Randomized Controlled Trials
Sara Albagoush, Creighton University, Omaha, NE
S. Albagoush1, T. Agarwal2, Z. Coy3, K. Mohanraj4, Y. Elmasry5, D. Masih6, P. Silberstein7; 1Internal Medicine, Creighton University, Omaha, NE, 2Internal Medicine, Kastruba Medical College, Manipal, INDIA, 3Medical school, Universidad CES, Medellin, COLOMBIA, 4medical school, Indira Gandhi Medical College and Research Institute, Puducherry, INDIA, 5Hematology oncology, SSM Health St. Louis University, Saint Louis, MO, 6Hematology oncology, University of Nebraska Medical center, Omaha, NE, 7Hematology oncology, Creighton University, Omaha, NE.
Background: Phosphatidylinositol 3-kinase (PI3K) inhibitors target the phosphoinositide 3-kinase pathway, which is crucial for cell growth, survival, and proliferation. The efficacy of PI3K inhibitors in Hormone receptor -positive, HER 2 – negative advanced breast cancer is an area of active investigation. While promising, concerns about safety, and side effects remain. This study aims to analyze the current data on the safety and efficacy of PI3K inhibitors.Methods: PubMed, Embase and Cochrane database were systematically searched for randomized controlled trials (RCTs) phase III comparing PI3K inhibitors plus standard of care to placebo plus standard of care along in patients with hormone receptor-positive, HER2-negative advanced breast cancer. Studies reporting outcomes such as progression-free survival (PFS), overall survival (OS), response rates (RR), clinical benefit rate (CBR), and adverse events (AEs) were included. A random-effects model was used to synthesize pooled hazard ratio (HR)/risk ratio (RR) with 95% confidence intervals (CIs).Results: Five phase III RCTs with 2992 patients were identified, with 1650 (55.1%) receiving PI3K inhibitors. The pooled Hazard ratio (HR) for PFS was statistically significant in PI3K inhibitors group (HR 0.74; 95% CI 0.68-0.81; p<0.00001, I2 0%), with similar results were seen in PI3K-pathway-activated population (HR 0.57;95%CI 0.47-0.68; p<0.00001, I2 52%). Overall survival (OS) analysis was not statistically significant between the groups (HR 0.89;95% CI 0.79-1.00; p 0.06; I2 0%). Overall response rate (ORR) analysis and Clinical benefit rate (CBR) were higher in PI3K inhibitors group (RR 2.57; 95% CI 1.84-3.58; p< 0.00001; I2 36%), (RR 1.62; 95% CI 1.38-1.91; p<0.00001, I2 63%) respectively. The most frequently observed all‐grade adverse events (AEs) in patients treated with PI3K inhibitors were hyperglycemia (50.7 %), diarrhea (44.6%), nausea (34.8%), rash (32.4%), and stomatitis (28.9%). The most common grade 3 or more toxicities were hyperglycemia (16.5%), diarrhea (5.8%), and rash (4.1%).Conclusion: PI3K inhibitors demonstrate a favorable efficacy profile but associated with notable toxicity. Future research is needed to refine their clinical application, maximize therapeutic benefits and optimize their safety profile.
Presentation numberPS1-10-07
Progression free survival and exploratory endpoints from PALVEN: a phase 1b study of palbociclib, letrozole and venetoclax in ER and BCL2-positive metastatic breast cancer
Geoffrey J Lindeman, Peter MacCallum Cancer Centre, Melbourne, Australia
F. C. Martin1, C. Muttiah2, L. Lal2, M. Christie3, S. Dawson1, J. Desai1, A. Grobler4, K. Moodie5, A. Murugasu3, P. Phan1, M. R. Rosenthal1, A. Travers1, J. E. Visvader2, J. Whittle1, B. Yeo6, G. J. Lindeman1; 1Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 2Cancer Biology & Stem Cells, The Walter & Eliza Hall Institute of Medical Research, Parkville, AUSTRALIA, 3Department of Pathology, The Royal Melbourne Hospital, Parkville, AUSTRALIA, 4Royal Children’s Hospital, Murdoch Children’s Research Institute, Parkville, AUSTRALIA, 5Department of Radiology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 6Department of Medical Oncology, Olivia Newton John Cancer Research Institute, Heidelberg, AUSTRALIA.
Background: CDK4/6 inhibitors induce growth arrest/senescence in breast cancer cells, which reduces their susceptibility to apoptotic cell death. In preclinical models, the BCL2 inhibitor venetoclax augmented tumor response to endocrine and CDK4/6 inhibitor (CDK4/6i) therapy by triggering apoptosis. PALVEN is a phase 1b dose escalation study in patients (pts) with ER+ and BCL2+ metastatic breast cancer (MBC). The maximum tolerated and recommended phase 2 dose was defined as letrozole 2.5 mg (d1-28), palbociclib 75 mg (d1-21) and venetoclax 400 mg (d1-21). Here, we report the progression free survival (PFS), overall survival (OS), updated treatment-related toxicity and exploratory endpoints at 48 wks. (NCT03900884) Methods: Postmenopausal women with ER+/HER2- (ASCO/CAP) and BCL2+ MBC who had received ≤2 prior lines of systemic therapy for MBC were included. Tumor assessment occurred every 8 wks (every 12 wks after 24 wks). Secondary endpoints were analyzed once the final pt had been on the study for 48 wks and included PFS and OS. Pre-defined exploratory endpoints included Clinical Benefit Rate (CBR) for: 1st vs 2nd/3rd line metastatic therapy; visceral vs non-visceral metastases; disease-free interval (DFI) following adjuvant endocrine therapy (ET) (relapse on or within 12 mo vs ≥12 mo following completion of ET). Adverse events (AEs) were reported using CTCAE v5.0. Tumor biopsies were collected at baseline and on day 15 of triplet therapy. Results: As of 31 January 2025, 16 pts had received venetoclax, palbociclib and letrozole treatment and 15 were DLT-evaluable (median age 50 yrs [range 36-67]), with a median follow-up of 37.6 months. Eight pts had de novo MBC and 8 pts had relapsed MBC. Seven pts had previously received adjuvant ET with 3 pts having primary endocrine resistance. Four pts had received prior therapy in the metastatic setting; 1st line aromatase inhibitor (3 pts) and chemotherapy and tamoxifen (1 pt). Five pts had visceral metastases and 11 pts had non-visceral or bone-only metastases. The CBR (rate of confirmed complete or partial response or stable disease at 24 wks) was 93% (14/15 pts). CBR for pts treated in the 1st line setting was 12/12 vs 2/3 for pts treated in the ≥2nd line; CBR for pts with visceral disease was 5/5 vs 9/10 for non-visceral disease; 3/3 for pts with primary endocrine resistance versus 3/3 for pts who relapsed >12 mo post-adjuvant ET. Median PFS was 29.2 mo (95% CI: 18.4-not estimable). OS was not mature. Seven pts remained on study therapy including 4 pts who had completed ≥32 months of triplet therapy. Paired tumor samples were available for 6 pts. Following 14 days of triplet therapy all tumors retained ER and BCL2 positivity however a reduction or loss in PR staining was observed in all samples. Ki67 was reduced by at least 50% for all samples with a baseline Ki67 of ≥10%. No additional TRAEs were observed from those previously reported at 24 wks. Grade ≥3 hematological TRAEs were decreased neutrophil count (63%), reduced white cell count (44%) and decreased lymphocyte count (19%). G3 neutropenia (without associated infections) was the main dose-limiting feature of combination therapy. Conclusion: Promising clinical activity has been observed using triplet therapy with palbociclib, letrozole and venetoclax in pts with ER+ MBC selected for BCL2-positive tumor expression. Studies combining ET with potent inhibitors of BCL2 and CDK4/6 (or a CDK4 inhibitor to mitigate CDK6i-associated neutropenia) merit further investigation.
Presentation numberPS1-10-09
Dose optimization of PF-07248144, a first-in-class KAT6 inhibitor, in patients with ER+/HER2− metastatic breast cancer: results from phase 1 study to support the recommended phase 3 dose
Yeon Hee Park, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea, Republic of
P. M. LoRusso1, T. Mukohara2, D. Sommerhalder3, K. Yonemori4, E. Hamilton5, R. M. Layman6, S. Kim7, S. Im8, H. S. Rugo9, T. Yamashita10, F. Yan11, F. Hara12, G. Kim13, S. Wang14, S. Kent15, L. Liu16, A. Skoura17, K. Kowalski16, M. Li18, Y. Park19; 1Medical Oncology, Yale School of Medicine, New Haven, CT, 2Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, JAPAN, 3Oncology, NEXT Oncology, San Antonio, TX, 4Department of Medical Oncology, National Cancer Center Hospital, Chuo City, JAPAN, 5Medical Oncology, Sarah Cannon Research Institute, Nashville, TN, 6Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 7Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, KOREA, REPUBLIC OF, 8Department of Internal Medicine, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 9Department of Medical Oncology & Therapeutics Research, City of Hope (Previously at University of California at San Francisco), Duarte, CA, 10Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Yokohama, JAPAN, 11Breast Medical Oncology, Swedish Cancer Institute, First Hill-True Family Women’s Cancer Center, Seattle, WA, 12., The Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, JAPAN, 13Department of Internal Medicine, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 14Department of Medical Oncology, Sun Yat-sen University, Guangzhou, CHINA, 15Oncology, Pfizer Inc., Cambridge, MA, 16Oncology, Pfizer Inc., San Diego, CA, 17Oncology, Pfizer Inc., Collegeville, PA, 18Oncology, Pfizer Inc., San Francisco, CA, 19Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF.
Methods: Patients with ER+/HER2− mBC after prior CDK4/6i and endocrine therapy (ET) received PF-07248144 at recommended doses for expansion (RDEs) of 5 mg QD alone, 5 mg QD plus FUL, or 1 mg QD plus FUL (N = 107) and were followed up (at least 6 months across all cohorts) to assess for safety and efficacy. Primary objective was safety/tolerability per CTCAE 5.0 and RDE selection. Other objectives included antitumor activity per RECIST 1.1, pharmacokinetics (PK), pharmacodynamics (PD), and predictive biomarkers. Results: 5 mg QD was identified as the RDE for both PF-07248144 monotherapy (35 patients treated) and FUL combination (43 patients treated) based on safety, PK, PD, and antitumor activity. 1 mg PF-07248144 plus FUL (29 patients treated) was selected as the lower RDE based on a distinguishable PK and safety profile while achieving maximal blood and tumor PD marker reduction and efficacious concentrations supported by preclinical models. As of Oct 11, 2024, a total of 107 patients were treated at RDEs. Baseline patient characteristics from the two RDEs plus FUL were comparable. All patients received prior CDK4/6i and ET in the metastatic setting. Positive dose-response relationships were identified for both safety (neutropenia) and efficacy (objective response rate [ORR]) endpoints. At 5 mg and 1 mg doses plus FUL, the most common treatment-related adverse event (TRAE) was dysgeusia (Grade [G]1+G2: 83.7% vs 89.7%). The most common G≥3 TRAE was neutropenia (G3: 39.5% vs 20.7%; G4: 7.0% vs 0.0%). The neutropenia was reversible and manageable with dose modifications. No febrile neutropenia was observed. The safety profile of 5 mg PF-07248144 monotherapy was consistent with 5 mg RDE plus FUL. No events of pneumonitis were reported in the 107 patients treated. For FUL plus 5 mg and 1 mg PF-07248144, ORR was 37.2% (95% CI: 23.0-53.3) vs 24.1% (10.3-43.5); median duration of response was 15.8 months (9.2-not estimable [NE]) vs 4.6 months (3.4-NE); clinical benefit rate was 55.8% (39.9−70.9) vs 37.9% (20.7-57.7). With median duration of follow-up 21.9 months and 11.0 months for patients receiving FUL plus 5 mg and 1 mg PF-07248144, the median progression-free survival was 10.7 months (95% CI: 5.3−13.8) vs 3.6 months (1.8-5.6), respectively. Conclusions: Based on a thorough benefit-risk assessment of two pharmacokinetically distinguishable doses with sufficient number of patients and follow up, 5 mg QD PF-07248144 was identified as the optimal dose in combination with FUL with acceptable safety and encouraging activity. A pivotal phase 3 trial is ongoing to address the high unmet medical need in ER+/HER2− mBC after progression on CDK4/6i plus ET.
Presentation numberPS1-10-10
Chidamide combined with fulvestrant in the treatment of HR-positive,HER2-negative advanced breast cancer after failure of previous endocrine therapy: A single-arm, single-center, phase 2 study
La Zou, Chongqing University Cancer Hospital, ChongQing, China
L. Zou, T. Xianjun, W. Li, X. Zeng, P. Deng, Y. Liu, T. Yang, X. Tan; breast cancer center, Chongqing University Cancer Hospital, ChongQing, CHINA.
Background: Chidamide is an oral subtype-selective histone deacetylase inhibitor previously utilized as an anti-cancer agent. The ACE study demonstrated that chidamide combined with exemestane significantly improved progression-free survival compared to placebo plus exemestane in patients with advanced, hormone receptor-positive(HR-positive), HER2-negative (HER2-) breast cancer who had experienced disease progression following prior endocrine therapy. This phase 2 study aimed to assess the efficacy and safety of chidamide in combination with fulvestrant for the treatment of HR-positive , HER2-negative advanced breast cancer that had progressed after previous endocrine therapy.Methods: Eligible patients were women aged 18 to 75 years with histologically confirmed HR+/HER2- advanced invasive breast cancer, whose disease had relapsed or progressed following at least one prior endocrine therapy, with or without a CDK4/6 inhibitor (CDK4/6i), administered in the advanced, metastatic, or adjuvant setting. Patients received oral chidamide (30 mg twice weekly for four consecutive weeks in a 4-week cycle) combined with intramuscular fulvestrant (500 mg on days 1 and 15 of cycle one, followed by day 1 of each subsequent 28-day cycle) until disease progression or intolerance. Premenopausal women were additionally treated with a concomitant GnRH analogue. The primary endpoint was progression-free survival (PFS) ,while secondary endpoints included overall response rate (ORR), duration of response (DoR), disease control rate (DCR), overall survival (OS), and safety.Results: Between March 8, 2021, and March 20, 2024, a total of 30 patients were enrolled. The median age was 54.5 years (range:31-70), with all 30 (100%) patients having an Eastern Cooperative Oncology Group Performance Status (ECOG PS) of 1. Additionally, visceral metastases were present in 80% (n = 24) of cases, and metastases involving more than three anatomical sites were observed in 50% (n = 15). The median number of prior chidamide treatments was two (range one to four). Overall, chemotherapy had been administered to 90% (n = 27) of the patients; primary endocrine resistance was noted in 10% (n = 3), and prior CDK4/6 inhibitor therapy had been utilized in 40% (n = 12) .At the data cutoff date on September 15, 2024, five patients (17%) remained on the treatment regimen. Among all efficacy-evaluablepatients n= 30 the objective response rate (ORR) was reported as being at a level equivalent to ten percent while disease control rates stood at eighty-three point three. Conclusions: Chidamide combined with fulvestrant demonstrated promising antitumor efficacy and manageable toxicity in patients with HR+/HER2- advanced breast cancer who experienced disease progression following prior endocrine therapy.
Presentation numberPS1-10-11
Safety of CDK 4/6 inhibitors Combined with Radiotherapy in HR+/HER2- Metastatic Breast Cancer: A Meta-Analysis
Khalid Ahmad Qidwai, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
K. A. Qidwai, V. Andion Camargo, F. Akkoc Mustafayev, M. Jaramillo, Z. Sarfraz, L. S. Spiegelman, M. A. Ganiyani, M. S. Ahluwalia, R. L. Mahtani; Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) are crucial in the treatment of hormone-positive, HER2-negative (HR+/HER2-) metastatic breast cancer (MBC), with radiotherapy (RT) remaining an important modality for symptom control. With increasing clinical use of CDK4/6i in combination with RT, questions have emerged regarding its safety. Although formal prospective guidelines are lacking, increasing retrospective data and expert consensus has begun to inform practice, particularly around timing and toxicity management. This meta-analysis evaluates its safety and feasibility. Methods: We systematically searched PubMed, Embase, Scopus and Cochrane CENTRAL (as of June 2025) to identify studies evaluating the safety of CDK4/6i + RT in HR+/HER2- MBC patients (pts). Eligible studies included those reporting safety outcomes such as grade ≥3 adverse events (AEs), dose reduction, or treatment discontinuations. Data were pooled using a random-effects model. We calculated pooled proportions (PP) and odds ratios (ORs) with 95% CIs. Statistical significance was set at p 50%). The protocol was registered with PROSPERO (CRD420251080439). Results: Nineteen studies comprising 843 HR+/HER2- MBC pts treated with CDK4/6i and RT were included. The median follow-up was 18 months (IQR: 14.7-19.0), and median age was 58.8 years (IQR: 57.15-60.13). The PP of all hematological toxicity was 27.8% (95% CI: 15.6-44.5, I²=85.7%, p< 0.0001), with neutropenia in 24.3% of pts (95% CI: 14.9-37.0, I²=79.8%, p< 0.0001). Non-hematological toxicities were infrequent, with a PP of 5.4% (95% CI: 3.8-7.7, I²=0.0%, p=0.986). Severe AEs (grade ≥3) occurred in 29.9% (95% CI: 19.8-42.5; I²=79.3%; p<0.0001). Dose reduction due to toxicity occurred in 26.7% of pts (95% CI: 17.8-38.1, I²=77.7%, p< 0.0001), while treatment discontinuation occurred in 6.3% (95% CI: 4.4-8.9, I²=0.0%, p=0.69). When compared to CDK4/6i therapy alone, the addition of RT was not associated with significantly increased odds of neutropenia (1.13; 95% CI: 0.35-3.68; I²=64.8%; p=0.85), dose reduction (1.06; 95% CI: 0.81-1.40; I²=0%; p=0.58), or treatment discontinuation (0.75; 95% CI: 0.07-8.26; I²=19.5%; p=0.66). Conclusion: CDK4/6i + RT appear to be safe and generally well tolerated in HR+/HER2- MBC, with no significant increase in severe toxicities, dose reductions, or treatment discontinuation vs CDK4/6i alone, supporting feasibility in clinical practice. However, the evidence is limited to retrospective studies with heterogeneous designs, small cohorts, and short follow up, which may underestimate late or site-specific toxicities. Prospective studies with standardized toxicity reporting and stratification by radiation site, dose, and sequencing are needed to better assess long-term safety.
| Analysis | Effect Size (95% CI) | P-Value | I² (%) | Heterogeneity |
| Proportion | ||||
| All hematological AE | 27.80% (15.6%–44.5%) | < 0.0001 | 85.70 | High |
| Neutropenia | 24.30% (14.9%–37.0%) | < 0.0001 | 79.80 | High |
| Non-hematological AE | 5.40% (3.8%–7.7%) | 0.9861 | 0.00 | Low |
| All severe AE | 29.90% (19.8%–42.5%) | < 0.0001 | 79.30 | High |
| Dose reduction | 26.70% (17.8%–38.1%) | < 0.0001 | 77.70 | High |
| Dose discontinuation | 6.30% (4.4%–8.9%) | 0.6927 | 0.00 | Low |
| Odds Ratio | ||||
| Neutropenia | 1.131 (0.3479–3.6775) | 0.6972 | 64.80 | High |
| Dose reduction | 1.0628 (0.8057–1.4019) | 0.5746 | 0.00 | Low |
| Dose discontinuation | 0.7481 (0.0677–8.2623) | 0.6549 | 19.50 | Low |
Presentation numberPS1-10-12
Impact of Glucagon-like peptide-1 receptor agonists Use on Survival and Metabolic Outcomes in Metastatic Estrogen Receptor Positive Breast Cancer: A Real-World Cohort Study
Maha Zafar, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY
F. Khan1, M. Zafar1, B. Singeltary2; 1Hospice and Palliative Medicine, University at Buffalo Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Hematology and Oncology, TriHealth Good Samaritan Hospital, Cincinnati, OH.
Background: Glucagon-like peptide-1 receptor agonists (GLP-1a), commonly used for type 2 diabetes and obesity, have shown potential anti-cancer effects in preclinical models. However, their impact on outcomes in patients with estrogen receptor-positive (ER+) metastatic breast cancer (MBC) remains unclear. This study evaluates the association between GLP-1a use and survival & metabolic outcomes in a real-world cohort of patients with ER+ MBC. Methods: We conducted a retrospective cohort study using the TriNetX US Collaborative Network. Two cohorts were defined: (1) ER+ MBC patients treated with GLP-1a (tirzepatide, dulaglutide, semaglutide, or liraglutide) and aromatase inhibitors (letrozole, anastrozole, or exemestane), excluding metformin users; and (2) ER+ MBC patients treated with aromatase inhibitors without GLP-1a or metformin. Cohorts were matched 1:1 using propensity scores based on demographics, comorbidities including type 2 diabetes mellitus and Obesity; and concurrent medications, including endocrine therapies (letrozole, anastrozole, exemestane) and other systemic treatments such as CDK4/6 inhibitors, mTOR inhibitors, and HER2-targeted agents. Kaplan-Meier survival analyses were performed for overall survival and adverse outcomes over a 2-year follow-up. Results: After propensity score matching, each cohort included 1,793 patients. GLP-1a use was associated with significantly improved 2-year overall survival (HR 0.612, 95% CI: 0.484-0.773, p<0.001). Risk of hospitalization was also lower in the GLP-1a group (HR 0.559, 95% CI: 0.412-0.759, p<0.001). GLP-1a use was associated with reduced risk of Anemia (HR 0.769, 95% CI: 0.601-0.983, p=0.035), Thrombocytopenia (HR 0.682, 95% CI: 0.487-0.955, p=0.025) and Severe sepsis (HR 0.633, 95% CI: 0.411-0.975, p=0.037). No significant differences were observed for Neutropenia (HR 0.712, 95% CI: 0.501-1.012, p=0.057), Nausea/vomiting (HR 1.204, 95% CI: 0.958-1.512, p=0.111), Outpatient visits (HR 0.795, 95% CI: 0.159-3.970, p=0.779), Heart failure (HR 1.150, 95% CI: 0.818-1.616, p=0.421), Cardiomyopathy (HR 0.777, 95% CI: 0.460-1.312, p=0.344), Diarrhea (HR 0.998, 95% CI: 0.773-1.288, p=0.987), HFrEF (HR 0.881, 95% CI: 0.505-1.537, p=0.656), Pneumonia (HR 0.925, 95% CI: 0.680-1.258, p=0.620), Peripheral neuropathy (HR 0.893, 95% CI: 0.677-1.178, p=0.423). Conclusions: This large retrospective cohort analysis provides evidence suggesting that GLP-1a use in patients with ER+ metastatic breast cancer may offer survival benefits beyond their established metabolic indications. The association between GLP-1a use and improved two-year overall survival, as well as reduced hospitalizations and lower incidence of several treatment-related adverse events, is clinically significant and suggests a possible dual role for GLP-1a in addressing both metabolic and oncologic challenges in this population. Importantly, GLP-1a therapy was not associated with increased gastrointestinal or cardiac toxicity, reinforcing its safety profile when used alongside standard endocrine and targeted cancer therapies. These findings align with emerging data that metabolic interventions can impact tumor biology and patient outcomes, and they highlight the value of a multidisciplinary approach that addresses both oncologic and comorbid metabolic conditions. Future prospective studies and randomized clinical trials are needed to validate these results and further elucidate the underlying biological mechanisms by which GLP-1a may exert anti-tumor effects in ER+ MBC. This study signifies the evolving landscape of cancer care, where holistic, patient-centered strategies are increasingly recognized as essential for optimal outcomes.
Presentation numberPS1-10-13
Fulvestrant Post-CDK Inhibitor in Hormone Receptor-Positive Breast Cancer: A Pooled FDA Analysis
James Buturla, U.S. Food and Drug Administration, Silver Spring, MD
J. Buturla1, Y. Zhang1, E. Chang1, P. Narayan1, M. Shah1, C. Osgood1, M. Fiero1, A. De Claro2, R. Pazdur2, L. Amiri-Kordestani1; 1Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 2Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD.
Background: Fulvestrant monotherapy has served as a control arm option in many later-line clinical trials enrolling patients with metastatic hormone receptor-positive (HR+) breast cancer. Since CDK4/6 inhibitors (CDKi) have become a standard initial therapy in the United States, such trials now enroll patients with disease progression on or after CDKi-based therapy. Moreover, recent trials have identified biomarkers that may affect the choice of therapy and disease prognosis in this treatment space. These developments call for a close reevaluation of the role of fulvestrant monotherapy in the treatment of patients with HR+ breast cancer, after progression on initial therapy. Methods: We pooled patient-level data from four randomized phase 3 trials: SOLAR-1, EMERALD, CAPItello-291, and EMBER-3. For patients in the control arm who received single-agent fulvestrant, we descriptively compared objective response rate (ORR) in patients who had received prior CDKi versus patients with no prior CDKi, and evaluated progression-free survival (PFS) and overall survival (OS) in the patients with prior CDKi. In the post-CDKi population with ESR1/PI3K-pathway alterations, we assessed OS with fulvestrant against biomarker-informed therapy on the trial comparator arm. Last, we developed multivariate Cox regression models adjusting for baseline characteristics to explore the impact of ESR1 and PI3K pathway alterations on the prognosis of patients treated with fulvestrant post-CDKi. Results: Among 1099 patients receiving fulvestrant, the ORR in 603 patients with prior CDKi was 5.1% (95% CI: 3.5, 7.2) compared to 12.7% (95% CI: 9.9, 16.0) in the 496 patients without prior CDKi. In patients with prior CDKi, median PFS was 2.8 months (95% CI: 2.0, 3.6), with 11.2% (95% CI: 8.5, 14.2) progression-free at 1 year. Median OS in these patients was 26.8 months (95% CI: 20.8, NE). Among 572 patients with a treatment-eligible biomarker and prior CDKi, randomization to biomarker-informed therapy (N=303) was associated with favorable OS, compared to fulvestrant (N=269), with hazard ratio (HR) 0.61 (95% CI: 0.46, 0.82). Among the 603 patients treated with fulvestrant post-CDKi, we observed inferior outcomes in patients with an ESR1 mutation or a PI3K-pathway alteration. In multivariate Cox regression analyses, PI3K pathway alterations were associated with shortened OS (HR = 1.66; 95% CI: 1.18-2.33), while ESR1 mutations were associated with shortened PFS (HR = 1.31; 95% CI: 1.01-1.70). Discussion: In this exploratory pooled analysis of patient-level data, the activity of fulvestrant as reflected by ORR was reduced in patients with prior CDKi treatment. Patients with prior CDKi also experienced a generally short progression-free interval on fulvestrant, suggesting limited treatment benefit. Tumor-based metrics such as ORR and PFS can underestimate the value of a less toxic therapy, so we evaluated the pooled OS comparison against biomarker-directed management among eligible patients, showing favorable OS for biomarker-directed management over fulvestrant. To evaluate the importance of ESR1 and PI3K-pathway alterations in defining the post-CDKi patient population, we assessed whether they had prognostic impact in this setting, in patients receiving fulvestrant. The presence of either biomarker was independently associated with worse outcomes. Given its diminished activity, short intervals to progression, and unfavorable survival comparison in recent trials, single agent fulvestrant is not a reasonable option for biomarker-positive or biomarker-unselected populations post-CDK4/6 inhibitor-based therapy. The current data are not well suited to evaluate its role in selected biomarker-negative populations, which appear distinct prognostically.
Presentation numberPS1-10-14
Defining the prognosis of estrogen receptor low-positive de novo metastatic breast cancer
Madelyn Klugman, Johns Hopkins School of Medicine, Baltimore, MD
M. Klugman1, M. Aboumrad2, R. Chen1, C. H. Marshall1, J. V. Canzoniero1, A. C. Wolff1, K. Visvanathan3; 1Medical Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 2Epidemiology, Bloomberg School of Public Health at Johns Hopkins University, Baltimore, MD, 3Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD.
Background: Recent studies have reported the prognosis and heterogeneity of estrogen receptor (ER) low-positive early-stage breast cancer. Survival and treatment outcomes have not been reported for patients with ER low-positive, metastatic breast cancer. The primary objective of this study was to compare overall survival (OS) among patients with ER low-positive de novo metastatic breast cancer (dnMBC), ER-negative dnMBC, and ER-positive dnMBC. The secondary objective was to determine if progesterone receptor (PR) status can define two prognostic groups among ER low-positive dnMBC patients. Methods: A retrospective cohort of patients with dnMBC was created using data from the National Cancer Database (NCDB). Adults diagnosed with human epidermal receptor 2 (HER2) negative dnMBC between 2018-2021 were included. The primary exposures were ER status, based on immunohistochemistry and defined as negative, <1%, low-positive, 1-10%, or positive, 11-100%, and PR status (negative, <1% and positive, 1-100%). Cox regression models were used to compare OS by ER-PR status in age-adjusted and multivariable models adjusting for metastatic disease sites, race, ethnicity, comorbidities, insurance, and treatment receipt (see Table for ER-PR status classification). An exploratory survival analysis by first-course treatment (chemotherapy and/or endocrine therapy) was conducted among ER low-positive patients. We distinguished cytotoxic chemotherapy from cyclin-dependent kinases 4 and 6 inhibitors (CDK4/6i) receipt based on timing of endocrine and chemotherapy treatment. Results: Among 27,672 HER2-negative dnMBC patients, 2.5% were ER low-positive, 20.8% ER negative, and 76.7% ER positive. Patients with ER low-positive dnMBC had a similar OS to those with ER negative dnMBC in multivariable analysis [hazard ratio, 95% confidence interval [HR(95%CI)]: 1.04 (0.94-1.14)], while patients with ER positive dnMBC had improved OS [HR(95%CI)]: 0.50 (0.48-0.52)] (see Table below). Patients with ER low-positive PR positive dnMBC had superior OS to patients with ER negative dnMBC in multivariable analysis [HR(95%CI): 0.84 (0.71-1.00)]. In contrast, patients with ER low-positive, PR negative dnMBC did not have improved OS compared to ER negative patients. ER low-positive patients, irrespective of PR status, who received chemotherapy and endocrine therapy (with or without CDK4/6i) or endocrine therapy with CDK4/6i alone had improved or similar OS compared to ER low-positive patients who received chemotherapy alone. Conclusions and Relevance: Our findings suggest PR positivity identifies a subgroup among ER low-positive dnMBC patients with superior OS than ER negative patients. Further, first-line treatments incorporating endocrine therapy may be an option for patients with ER low-positive dnMBC. This promising finding warrants further investigation.
| Age-adjusted analysis | Multivariable analysis | |
| HR (95%CI) | HR (95%CI) | |
| Survival by ER status | ||
| ER negative, PR positive or PR negative | 1 (ref) | 1 (ref) |
| ER low-positive, PR positive or PR negative | 0.91 (0.83-1.00) | 1.04 (0.94-1.14) |
| ER positive, PR positive or PR negative | 0.37 (0.36-0.38) | 0.50 (0.48-0.52) |
| Survival by ER-PR status | ||
| ER negative, PR positive or PR negative | 1 (ref) | 1 (ref) |
| ER low-positive, PR negative | 1.04 (0.93-1.16) | 1.14 (1.02-1.27) |
| ER low-positive, PR positive | 0.70 (0.59-0.83) | 0.84 (0.71-1.00) |
| ER positive, PR positive or PR negative | 0.37 (0.36-0.38) | 0.50 (0.48-0.52) |
Presentation numberPS1-10-15
Impact of modern imaging on survival in a retrospective and consecutive cohort of 145 HR+/HER2 negative oligometastatic breast cancer
Jean Louis Lacaze, Oncopole Claudius Regaud, Toulouse, France
J. Lacaze1, F. Dalenc1, E. De Maio1, B. Cabarrou2, T. Cassou Mounat3, C. Massabeau4, V. Nicolai1, G. Selmes1, E. Jouve5, M. Ung1, C. Korenbaum1; 1Medical Oncology, Oncopole Claudius Regaud, Toulouse, FRANCE, 2Biostatistics & Health Data Science Unit, Oncopole Claudius Regaud, Toulouse, FRANCE, 3Imaging, Oncopole Claudius Regaud, Toulouse, FRANCE, 4Radiotherapy, Oncopole Claudius Regaud, Toulouse, FRANCE, 5Surgery, Oncopole Claudius Regaud, Toulouse, FRANCE.
Background To date, no prospective randomized trials have been published to validate a curative-intent strategy combining standard-of-care systemic therapy (SOC ST) with local ablative treatment (LAT) in patients with oligometastatic breast cancer (OMBC). Available evidence derives solely from retrospective series. These series can sometimes be criticized for their short follow-up periods, the biological and anatomical heterogeneity of the patients’ tumors, the use of suboptimal or insufficiently detailed imaging modalities, and the non-consecutive nature of their recruitment. Our objective was to identify prognostic factors for progression-free survival (PFS) and overall survival (OS) in patients with HR+/HER2- OMBC. We specifically evaluated the impact of modern versus standard imaging techniques and the contribution of LAT. To minimize selection bias, we enrolled all eligible patients consecutively, and to ensure biological homogeneity we therefore restricted inclusion to the HR+/HER2- phenotype. Methods We retrospectively reviewed all patients consecutively diagnosed and treated from 2012 to 2020 at our institution for HR+/HER2-negative OMBC. OMBC was defined as breast cancer with 1 to 5 distant metastases. Cases were categorized as de novo or metachronous. Surrogate intrinsic subtype – Luminal A-like (Lum-A) and Luminal B-like (Lum-B) were defined according to St Gallen consensus criteria. Imaging were classified as modern imaging methods (MIMs: PET-CT or whole-body MRI) versus standard imaging methods (SIMs: bone scintigraphy or thoraco-abdomino-pelvic (TAP) CT). Number and location of metastases, treatments – SOC ST vs SOC ST plus LAT (surgery or SBRT) were collected. Survival analyses were performed using the Kaplan-Meier method, the Logrank test, and the Cox model including time-dependent variable. The use of time-dependent models allowed adjustment for the timing of LAT and helped mitigate immortal time bias. Results Median age was 57.0 years [29.0; 90.0]; 23% patients had Lum-A and 77% had Lum-B tumors. OMBC was de novo in 34%, and metachronous in 65%. One organ was involved in 94% and 1 to 3 metastases in 88% cases. Metastatic sites included bone in 98 (68%), lymph node in 20 (14%), liver in 18 (12%), lung in 10 (7%), and brain in 7(5%) cases. MIMs were applied for staging in 54% of patients. No patients with four or five metastases received LAT (surgery or SBRT). Unfortunately, by lake of data, systemic treatment could not be analyzed. The median follow-up was 82.8 months. 73% relapsed and 51% died. Median OS was 81.1 months (95%CI [60.0-94.1]). Median PFS was 26.5 months (95% CI [21.0-33.8]). In univariable analysis, worse OS was associated with 4-5 metastases, liver metastases, Lum-B and SIMs (HR=2.56, 95% CI [1.58-4.13], p<0.0001). LAT was associated with improved PFS (HR=0.34, 95% CI [0.20-0.58], p<0.0001) and OS (HR=0.32, 95% CI [0.16-0.63], p=0.0009). In multivariable analysis: worse OS was associated with Lum-B, liver metastases and SIMs (HR=1.64, 95% CI [1.12-3.16], p=0.017). LAT improved PFS (HR=0.52, 95% CI [0.29-0.91], p=0.0233), showed a non-significant trend toward better OS (HR=0.52, 95% CI [0.25-1.08], p=0.078). In the subgroup staged with MIMs, in univariable analysis, LAT was significantly associated with improved PFS (HR=0.40, 95% CI [0.21-0.77], p=0.0064) and OS (HR = 0.29, 95% CI [0.12-0.73], p= 0.008). Conclusion This retrospective study reports prognostic data from a biologically homogeneous and unselected cohort of HR+/HER2- OMBC patients, with extended follow-up. In this study, LAT significantly improved PFS and showed a strong trend toward improved OS. In patients staged with MIMs, LAT was significantly associated with both PFS and OS, underscoring the importance of advanced imaging in selecting candidates for curative-intent strategies.
Presentation numberPS1-10-16
Longitudinal genomic profiling of circulating tumor DNA in metastatic invasive lobular (ILC) and no special type (NST) breast cancer
Daisong Liu, University of Pittsburgh, Pittsburgh, PA
D. Liu1, R. Kumar1, J. Hooda1, A. C. Chang1, L. Miller1, M. Rosenzweig2, J. M. Atkinson1, A. Stalnaker1, V. Gao3, S. Oesterreich1, A. V. Lee1, M. Balic1, J. Foldi1; 1Women’s Cancer Research Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 2School of Nursing, University of Pittsburgh, Pittsburgh, PA, 3UPMC Magee Woman’s hospital, UPMC, Pittsburgh, PA.
Longitudinal genomic profiling of circulating tumor DNA in metastatic invasive lobular (ILC) and no special type (NST) breast cancer Background Liquid biopsy enables minimally invasive cancer monitoring through blood circulating tumor DNA (ctDNA) sequencing. We analyzed ctDNA sequencing data of 127 blood samples from 27 patients with hormone receptor positive (HR+) metastatic invasive lobular breast carcinoma (ILC) and 10 patients with HR+ no special type/invasive ductal carcinoma (NST/IDC) from the UPMC Hillman Cancer Center APOLLO biobank collection. In APOLLO collection, patients’ blood is collected at each disease progression, enabling longitudinal analysis of ctDNA dynamics. In this study, we focused on longitudinal analysis of ctDNA dynamics in patients with ILC. Methods We analyzed 31 longitudinal NST samples (2-6 per patient, median 3) and 96 ILC samples (2-7 per patient, median 3). DNA was extracted from plasma and sequenced using shallow whole genome sequencing, with a mean coverage of 0.21x. We used ichorCNA to determine the tumor fraction (TFx) of samples, and used CNVkit to annotate gene level copy number variation (CNV) and filtered with a list of genes that have most frequent copy number aberration in breast cancer. GISTIC2.0 was applied to compare chromosomal segment aberration of ILC and NST cases. Gene level CNVs were collapsed to each patient, and continuity index (1 – [number of transition/maximum possible transitions]) was applied to measure the longitudinal consistency of gene CNVs. Results Patients with ILC had a median overall survival of 43 months (range 6-124 months) with a median of 5 treatment lines (range 2-11). IchorCNA analysis showed 85 samples (91.4%) had TFx >3% (mean TFx 12.5%). Tumor fraction fluctuates across time, and shows correlation with the patient treatment scheme. Patient-level analysis demonstrated predominant copy number gains (copy number ≥3) versus losses (copy number <2). MDM4, ELF3, IKBKE and IL10 were the four most gained genes across 88% of patients for each gene. ESR1 was amplified in 40% and ERBB2 in 32% of patients. The top copy number loss gene was ZFHX3 (68% of patients), followed by TP53, MAP2K4 and NCOR1 (totaling 64% of patients). We compared ILC cases to NST/IDC cases in genomic CNV. We discovered a chromosome 11q13.3 amplicon enriched in ILC cases. This region showed preferential gain in genes CCND1, FGF3, FGF4, and FGF19, suggesting a potential ILC oncogenesis driver locus. Additional ILC and NST/IDC samples are being prepared for sequencing to validate these findings. Conclusions This study demonstrates the dynamics of ctDNA in metastatic ILC, which can be used to infer tumor evolution in concordance with treatment scheme. ILC-enriched chromosomal signatures may suggest potential unique oncogenic drivers. We are also developing a transformer-based model to predict gene expression pattern via genomic copy number changes using public databases, and fine-tune it with in-house metastatic breast cancer sequencing dataset. We will validate the model with liquid biopsy sequencing data and paired tumor sequencing data to identify robust predictors that can stratify patients’ prognosis. Our study highlights the potential clinical utility for ctDNA-based cancer monitoring to advance precision oncology in metastatic breast cancer.
Presentation numberPS1-10-17
Progression-free Survival by Local Investigators vs. Blinded Independent Central Review in Hormone-Positive, HER2-Negative Metastatic Breast Cancer: A Systematic Review and Meta-analysis
Lis Victoria Ravani, University of Sao Paulo School of Medicine, São Paulo, Brazil
L. Ravani1, Z. Bagheri2, K. Kalinsky3, H. Burstein4, A. Bardia5, J. A. Mouabbi6, R. O’Regan7, S. A. Wander8; 1Medicine, University of Sao Paulo School of Medicine, São Paulo, BRAZIL, 2Biostatistics, Harvard Medical School, Boston, MA, 3Medical Oncology, Emory University, Atlanta, GA, 4Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, São Paulo, MA, 5Medicine, University of Sao Paulo School of Medicine, Los Angeles, CA, 6Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 7Medicine, University of Sao Paulo School of Medicine, Rochester, NY, 8Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, MA.
INTRODUCTION: Progression-free survival (PFS) is a widely accepted primary endpoint in breast cancer trials; however, it remains susceptible to assessment bias. To reduce this risk, blinded independent central review (BICR) is often incorporated into trial designs. Emerging evidence has suggested discordance between investigator- and BICR-assessed PFS, with BICR often producing more optimistic estimates, raising concerns about BICR-PFS as a definitive primary endpoint. We conducted a meta-analysis comparing BICR- and investigator-PFS in RCTs of patients with hormone receptor-positive, HER2-negative (HR+/HER2-) metastatic breast cancer (mBC) following progression on CDK4/6 inhibitor (CDK4/6i) therapy. METHODS: We conducted a systematic review and meta-analysis searching PubMed, Embase, Cochrane, and major conference proceedings for phase II-III RCTs including patients HR+/HER2- mBC with progression following CDK4/6i. Eligible studies required or permitted prior CDK4/6i use and reported PFS assessed by both local investigators and BICR. For each trial, we calculated a discrepancy index (DI)—the ratio of hazard ratios (HR) for BICR- vs investigator-PFS—and the absolute difference in median PFS (mPFS) between the two assessments. A pooled DI was estimated using a mixed-effects model. Subgroup analysis of trials requiring prior CDK4/6i and meta-regression by bone-only disease, visceral metastases, control arm type (endocrine/targeted therapy vs chemotherapy), study design (open-label vs double-blinded), and primary endpoint (investigator- vs BICR-PFS) were performed. RESULTS: Of 2,807 identified records, 19 RCTs with 7,599 patients met inclusion criteria. No statistically significant difference was observed between investigator- and BICR-PFS overall, with a pooled DI of 1.01 (95%CI, 0.93-1.01; p=0.66). In studies requiring prior CDK4/6i treatment, the pooled DI was 0.94 (95% CI, 0.80-1.11; p=0.81). Findings were consistent across meta-regression analyses (all p>0.05). The pooled PFS difference was 0.07 months (95%CI 0.38-0.52; p=0.76) in interventional arms and 0.33 months (95%CI 0.02-0.68; p=0.06) in control arms. Although major outliers, postMONARCH, SERENA-2, and SOLAR-1 did not exhibit statistically significant discrepancy. CONCLUSION: Although several recent RCTs in HR+/HER2- mBC have shown a trend toward longer mPFS with BICR—likely due to investigators incorporating additional clinical data, leading to censoring and exclusion from BICR analysis—our meta-analysis found no statistically significant differences between the two methods in this patient population. This consistency, demonstrated across meta-regression analyses adjusting for potential effect modifiers and in outlier trials, suggests that both approaches yield comparable PFS estimates.
Presentation numberPS1-10-18
Genomic analysis of ER+/HER2- metastatic invasive lobular carcinoma (ILC) and clinical outcomes comparison with metastatic invasive carcinoma of no special type (NST)
Kristina Fanucci, Dana-Farber Cancer Institute, Boston, MA
K. Fanucci, S. Challa, Y. Li, G. Nader-Marta, A. Martin, M. Hughes, B. E. Johnson, L. Sholl, D. Dillon, B. Carroll, S. M. Tolaney, A. Cherniack, N. U. Lin, R. Jeselsohn; Breast Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: ILC is the second most common histologic subtype of breast cancer (BC) comprising about 15% of invasive BCs. Despite differences in clinical behavior compared to NST, patients (pts) with these histologies are treated the same. Genomic analyses may provide insights into treatment strategies for this subset of BCs. The genomic landscape of early-stage ILC has been previously described. In this analysis we describe the genomic alterations of pts with metastatic ILC and clinical outcomes in the metastatic setting compared to pts with NST histology. Methods: Pts with metastatic, ER+/HER2- BC seen at least once at Dana-Farber Cancer Institute between 7/1/2001 and 8/2/2022 who had targeted tumor only DNA sequencing (OncoPanel) of ≥1 archival tissue sample collected within 3 months (mo) of diagnosis of distant recurrence/de novo metastatic disease were included. Clinical annotation for these pts was available and median time to next treatment (mTTNT) for pts receiving endocrine therapy (ET) + CDK4/6 inhibitor as first-line treatment and median overall survival (mOS) were compared between ILC and NST cohorts. Genes with oncogenic single nucleotide variants (SNV) and copy number variants (CNV) occurring at a frequency above 0.03 were tested via Fisher enrichment, with a Benjamini-Hochberg correction, for differences across clinicopathological characteristics. Kaplan-Meier analysis and Cox proportional hazards models were used to assess significant predictors of survival across and within ILC and NST cohorts. Results: 533 pts met criteria for inclusion in the final analytic cohort (ILC n=125; NST n=408, mixed histology excluded). In the ILC cohort, at the time of initial diagnosis 8.0% of pts had stage I disease, 22.4% stage II, 27.2% stage III, and 40.8% de novo metastatic disease. The median disease-free interval was 5.4 years (yr) (range 1.0-30.9). Genomic analyses of ILC samples revealed 16 genes mutated and 2 genes with high amplifications (amp) or deep deletions in ≥3% of ILC tumors. The most commonly altered genes with SNVs or INDELS were CDH1 (76%), PIK3CA (58%), TP53 (14%), ESR1 (10%), PTEN (6%), NF1 (6%), MAP3K1 (6%), ERBB2 (6%), and AKT1 (5%). The genes with most frequent CNVs were CCND1 amp (16%) and FGFR1 amp (7%). Comparing pts with early (0-5 yr, n=36) vs. late (5+ yr, n=38) recurrence showed CDH1 and ERBB2 SNVs were associated with early recurrence (odds ratio (OR)=0.18, p=0.0037, q=0.0394 and OR=0, p=0.0046, q=0.0394 respectively). Pts with liver metastases (mets) at the time of metastatic diagnosis (n=26) were more likely to have ERBB2 alteration (OR=14.1, p=0.001, q=0.0171). Comparing prevalence of gene alterations between ILC and NST, alteration in CDH1, PIK3CA, ERBB2, and MEN1 was more common in ILC while GATA3, TP53, FGFR1, ZNF217, CDKN2A, and IGF1R was more common in NST. mTTNT was not significantly different in pts who received ET + CDK4/6 inhibitor in the first line setting comparing ILC (9.5 mo [95% confidence interval (CI): 6.8-21.0]) to NST (10.2 mo [95% CI: 9.0-14.1]) (p=0.78) and mOS did not significantly differ between ILC (43.4 mo [95% CI: 38.3-not reached (NR)]) and NST (57.2 mo [95% CI: 41.8-78.8]) in this subset (p=0.74). There was no significant difference in mOS comparing all pts with ILC [4.2 yr (95% CI: 3.5-5.3)] to NST [4.3 yr (95% CI: 3.9-5.3)] (p=0.54). The presence of liver mets at diagnosis was associated with significantly shorter mOS in both ILC and NST (p<0.001) but there was no difference in mOS comparing pts with liver mets with ILC vs. NST (31.3 mo (95% CI 14.36-NR) vs 35.0 mo (95% CI: 28.5-41.4), [p=0.22]). Conclusion: Metastatic ILC has a unique genomic landscape which provides avenues for development of treatments designed specifically for this population. Pts with metastatic ILC had similar clinical outcomes to those with NST.
Presentation numberPS1-10-19
Palbociclib, pembrolizumab, and endocrine therapy in HR+/HER2- metastatic breast cancer: Updated results from a phase I/II trial
Alexis LeVee, City of Hope Comprehensive Cancer Center, Duarte, CA
A. LeVee1, C. Egelston2, S. E. Yost1, P. Frankel3, N. Ruel3, C. Ruel3, D. Schmolze4, P. Lee2, C. Yeon1, Y. Yuan5, J. Waisman1, J. Mortimer1; 1Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, 2Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, 3Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 4Department of Pathology, City of Hope Comprehensive Cancer Center, Duarte, CA, 5Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA.
Background: The role of immune checkpoint inhibitors (ICIs) in HR+/HER2- metastatic breast cancer (MBC) has yet to be defined. We enrolled patients (pts) in a clinical trial testing the addition of pembrolizumab to palbociclib and endocrine therapy (ET) with the ICI administered concurrently or sequentially. Here, we present updated trial results for the addition of pembrolizumab to palbociclib and ET. Methods: In this single-center, open-label, phase I/II study, pts with HR+/HER2- MBC were treated with palbociclib, pembrolizumab, and ET. Cohort 1 enrolled pts with stable disease on palbociclib + ET for at least 6 months (Cohort 1 closed early due to limited accrual, data previously reported). Cohort 2 and 3 enrolled pts in the first-line (1L) setting to ET + palbociclib with concurrent ICI (Cohort 2, data previously reported) or sequential ICI following a 28 day lead-in. The primary endpoint was overall response rate (ORR). Secondary endpoints included progression free survival (PFS), overall survival (OS), and treatment-related adverse events (TRAEs). Results: Between March 2017 and November 2024, 47 pts were accrued, with 4, 19, and 24 pts in cohorts 1, 2 and 3, respectively. All but 1 were female, with a median age of 55 years (range 33-75). With a median follow-up of 42.6 months (95% CI, 30.8-53.4) for patients treated in the 1L setting, the ORR was 62.8% (95% CI, 47.7-77.8). ORR of cohorts 2 and 3 was 57.9% and 66.7%, respectively. The median PFS of patients treated in the 1L setting was 27.1 months (95% CI, 13.9-42.5). PFS of cohorts 2 and 3 was 25.2 months and 29.1 months, respectively. The median OS in the 1L setting was 56.8 months (95% CI, 37.6-80.6). In the 1L setting, grade 2 or higher TRAEs occurred in 97.7% of pts, with the most common being leukopenia (95.3%), neutropenia (93.0%), lymphopenia (62.8%), fatigue (27.9%), upper respiratory infection (27.9%), anemia (25.6%), elevated alanine aminotransferase (ALT, 25.6%), and elevated aspartate aminotransferase (AST, 23.3%). In the 1L setting, grade 3 hematologic and non-hematologic TRAEs occurred in 90.7% and 30.2% of pts, with the most common non-hematologic TRAEs being pneumonitis (14.3%), nausea (9.5%), and diarrhea (9.5%). Grade 4 hematologic TRAEs occurred in 16.3% of pts in the 1L setting. There were no non-hematologic grade 4 TRAEs or treatment-related deaths. Conclusions: The combination of palbociclib, pembrolizumab, and ET in the 1L setting was tolerable in pts with HR+/HER2- MBC with no unexpected TRAEs. We report an improved ORR compared to PALOMA-2 of 62.8% vs. 55.3%, respectively; with a comparable PFS of 27.1 months vs. 24.8 months, respectively. Correlative studies are currently underway to explore immune biomarkers including tumor-infiltrating lymphocytes scoring and tumor immune microenvironment composition to identify pts who may have an improved response to this combination treatment leading to improved PFS and OS rates in some pts.
Presentation numberPS1-10-20
Free-her: discontinuation of maintenance her-2 directed therapy in long-term survivors of metastatic her-2 positive breast cancer: a trial update
Elisa Krill Jackson, U of Miami, Miami, FL
E. Krill Jackson, F. valdes, C. Calfa, T. King, A. Perez; Sylvester Cancer Center, U of Miami, Miami, FL.
Introduction:While metastatic breast cancer (BC) is considered an incurable disease, with the advent of HER-2directed therapy, a subset of patients with metastatic HER-2 positive (HER2+) BC maintain a completeremission for years. The current standard of care in the US for these patients is to continue anti-HER-2therapy indefinitely, with its associated toxicity, cost, and inconvenience. This study seeks to determinethe feasibility of discontinuing Her-2 directed therapy while maintaining remission and the possibility ofcure in patients who have remained in complete radiographic remission (CRR) of their metastatic HER2+BC for at least 36 months. Here, we provide a trial update.Methods:This is an IRB-approved, single-site pilot study that seeks to enroll 20 subjects with stage IV HER2+ BCin CRR for at least 36 months who have negative peripheral blood ctDNA at the time of enrollment.Financial toxicity and QOL assessments are also being performed. Study patients are monitored withctDNA testing at 6 and 12 weeks after treatment discontinuation, and once every 12 weeks thereafter.Standard of care radiologic imaging also occurs at 3 months after treatment discontinuation and thenevery 6 months thereafter. Patients are continued on study for a total of three years or until evidence ofdisease recurrence, defined as ctDNA positivity or radiographic progression of disease. If patients hadhormone receptor positive (HR+) disease, they are recommended to continue hormonal treatment duringthe trial.Results:At the time of this report, 14 patients have been enrolled. The median subject age at enrollment is 64.5(range 41-86), and the median duration of CRR prior to enrollment was 6 years (range 3-17). Eightpatients had de novo metastatic disease, and 5 had relapsed metastatic disease, with a median time of 6years between diagnosis of localized disease and the development of metastatic disease. Eleven of the14 patients were HER2 3+ on IHC, and 3 were 2+ on IHC and FISH positive. Ten of the 14 were alsoHR+. Only 6 patients had adequate tissue for the tumor-informed Signatera assay. The other 8 patientswere evaluated using Guardant Reveal assay.At baseline, 10 of the 14 had oligometastatic disease, defined as fewer than 5 metastatic sites. Sites ofmetastases included bone (8), distant lymph nodes (3), lung (2), liver (2), and brain (2). The majority ofpatients (11) had induction therapy with docetaxel, trastuzumab and pertuzumab (HP) and maintenanceHP, but 3 patients had no cytotoxic therapy and only targeted therapy for their metastatic disease.Of the 9 patients with de novo metastatic disease, 7 had local therapy to the primary breast lesion. Twopatients had local therapy (surgery and/or radiation) to all sites of disease; three had no local therapy atall.At the time of this report, only 2 patients have developed a positive ctDNA with one detected at 12 weeksoff therapy and one at 60 weeks. Both patients had oligometastatic disease. The patient relapsing at 12weeks had been in CRR for 4.5 years and had ER negative disease. After re-initiating anti-HER-2therapy (HP), she has remained ctDNA intermittently positive but in CRR for 18 months. The patient with ctDNA positivity at 60 weeks reverted to ctDNA negative without re-initiating therapy and has remainedctDNA negative with no clinical evidence of disease for 9 months.The maximum length of time on trial for any patient in the study is 24 months. No deaths have occurredon trial.Conclusions:Our current data suggest that HER2-directed maintenance therapy can safely be discontinued in patientswith metastatic HER2+ BC who have remained in CRR for at least 36 months. Baseline ctDNA testingmay be useful to select patients who may safely discontinue therapy and to closely monitor those patientsfor continued remission.
Presentation numberPS1-10-21
Switching CDK4/6 inhibitors due to Toxicity in first-line treatment of HR+/HER2- metastatic breast cancer: Incidence and Outcomes from the ESME Cohort
Thomas Papazyan, ICO Nantes, Saint Herblain, France
T. Papazyan1, A. Lusque2, W. Jacot3, T. Grinda4, A. Mailliez5, E. Brain6, T. Bachelot7, C. Levy8, M. Arnedos9, A. Goncalves10, V. Massard11, M. Mouret-reynier12, T. De la Motte Rouge13, T. Petit14, A. Savoye15, I. Desmoulins16, M. Leheurteur17, L. Bosquet18, M. Campone1, J. Frenel1; 1Medical oncology, ICO Nantes, Saint Herblain, FRANCE, 2Medical oncology, Institut Claudius Regaud, Toulouse, FRANCE, 3Medical oncology, ICM, Montpellier, FRANCE, 4Medical oncology, Institut Gustave Roussy, Villejuif, FRANCE, 5Medical oncology, Centre Oscar Lambret, Lille, FRANCE, 6Medical oncology, Institut Curie, Paris, FRANCE, 7Medical oncology, Centre Leon Berard, Lyon, FRANCE, 8Medical oncology, Centre Francois Baclesse, Caen, FRANCE, 9Medical oncology, Institut Bergonié, Bordeaux, FRANCE, 10Medical oncology, IPC, Marseille, FRANCE, 11Medical oncology, Institut de Cancérologie de Lorraine (ICL) – Alexis Vautrin, Vandœuvre-lès-Nancy, FRANCE, 12Medical oncology, Centre Jean Perrin, Clermont-Ferrand, FRANCE, 13Medical oncology, Centre Eugene Marquis, Rennes, FRANCE, 14Medical oncology, ICANS, Strasbourg, FRANCE, 15Medical oncology, Institut Gaudinot, Reims, FRANCE, 16Medical oncology, CGFL Dijon, Dijon, FRANCE, 17Medical oncology, CHB, Rouen, FRANCE, 18Department of Health Data and Partnerships, UNICANCER, Paris, FRANCE.
Background: CDK4/6 inhibitors (CDK4/6i) have markedly changed the treatment landscape for hormone receptors-positive (HR+)/HER2-negative (HER2-) metastatic breast cancer (MBC). According to current European guidelines, switching between CDK4/6i is advised when patients experience unacceptable toxicity during therapy. This study aimed to examine patterns of CDK4/6i use and switching strategies due to toxicity in the first line (L1) setting, using data from the ESME cohort, a large real-world database. Methods: The ESME MBC cohort is a national cohort collecting individual-level patient data from 18 French Comprehensive Cancer Centers (NCT03275311). For this study, we included all patients aged 18 years or older with newly diagnosed HR+/HER2- MBC who began L1 endocrine treatment (ET) combined with a CDK4/6i. Among these, patients who discontinued CDK4/6i due to toxicity and subsequently reinitiated a different CDK4/6i were selected to evaluate progression-free survival (PFS) following the second CDK4/6i exposure. Results: Between January 1, 2013, and December 31, 2023, 22,965 women treated for HR+/HER2- MBC were enrolled in the ESME database. Among them, 5,553 patients (24%) received L1 ET combined with a CDK4/6i. At the time of analysis, 4,058 patients had discontinued their initial CDK4/6i therapy, 67%, 16% and 17% due to disease progression, toxicity, or other reasons, respectively. Of these, 2,744 (68%), 787 (19%) and 527 (13%) were treated with palbociclib, ribociclib, and abemaciclib, respectively. Aromatase inhibitors were the most commonly used ET partner (78%), followed by fulvestrant (20%). During L1, 231 patients (5.7%) switched CDK4/6i due to toxicity, after a median treatment duration of 2.9 months (range: 0.1-57.6), occurring under palbociclib, ribociclib and abemaciclib in 50 (1.8%), 108 (13.7%), and 73 (13.8%) patients, respectively. The leading causes of discontinuation were gastrointestinal toxicity (28%), hematologic toxicity (26%), and liver toxicity (21%). The median age at the time of CDK4/6i switch was 66 years (range: 32-90), 37% having de novo MBC and 34% presenting with visceral metastases. At the time of analysis, 95 patients were still on treatment while 136 (59%) had discontinued the second CDK4/6i, 40 (29%) due to toxicity after a median rechallenge duration of 3.4 months (range: 0.1-40.0). The toxicity profile was consistent with that observed during L1 therapy, primarily hematologic toxicity (30%), gastrointestinal toxicity (22%), and liver toxicity (10%).With a median follow-up of 28.3 months [95% CI: 23.3-32.9], the median PFS on the second CDK4/6i was 17.9 months [95% CI: 14.7-21.9], and the 3-year overall survival rate was 71.7% [95% CI: 63.1-78.6]. Conclusion: This extensive multicenter retrospective study offers real-world evidence that switching to a second CDK4/6i after toxicity in L1 treatment is feasible. However, one third of this patients’ population discontinued the second CDK4/6i due to toxicity.
Presentation numberPS1-10-22
Tumor-infiltrating lymphocytes in advanced ER-positive, HER2-negative breast cancer prior to treatment with palbociclib and endocrine therapy: a retrospective analysis of the PROMISE study
Ciara C O’Sullivan, Mayo Clinic, Rochester, MN
C. C. O’Sullivan1, A. M. Moyer2, A. Nassar3, D. M. Zahrieh4, V. Suman5, X. Tang4, K. J. Thompson6, R. C. Hentz4, J. J. Harrington7, A. Moreno-Aspitia8, D. W. Northfelt9, T. C. Haddad1, S. Chumsri8, P. P. Peethambaram1, K. J. Ruddy1, K. V. Giridhar1, R. A. Leon-Ferre1, R. Weinshilboum10, L. Wang11, K. R. Kalari12, M. P. Goetz1; 1Medical Oncology, Mayo Clinic, Rochester, MN, 2Laboratory Genetics and Genomics, Mayo Clinic, Rochester, MN, 3Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 4Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 5Quantitative Health Science, Mayo Clinic, Rochester, MN, 6Computational Biology, Mayo Clinic, Rochester, MN, 7Center for Individualized Medicine, Mayo Clinic, Rochester, MN, 8Hematology/Oncology, Mayo Clinic, Jacksonville, FL, 9Hematology/Oncology, Mayo Clinic, Phoenix, AZ, 10Pharmacology, Mayo Clinic, Rochester, MN, 11Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 12Quantitative Health SciencesQHS, Mayo Clinic, Rochester, MN.
Background: Tumor-infiltrating lymphocytes (TILs) in HER2+ and triple-negative breast cancer (BC) are prognostic and associated with therapy response; however, there are few data regarding distribution and significance of TIL values in ER+ HER2-negative (HER2-) MBC patients (pts) treated with CDK4/6 inhibitors (CDK4/6i). CDK4/6 inhibition may promote antitumor immunity by production of type III interferons and tumor antigen presentation, and by inhibition of regulatory T cells (Goel, Nature 2017). Further, the addition of immune checkpoint therapy to palbociclib numerically prolonged PFS in the 2nd line (2L) setting (Mayer, JCO 2024). We aimed to investigate the prognostic role of TILs derived from fresh metastatic tumor biopsies in pts with ER+HER2- MBC prior to starting CDK4/6i and endocrine therapy (ET) in the first line (1L) and 2L setting. Methods: PROMISE (NCT0281902) is a multicenter, prospective study that enrolled women commencing the CDK4/6i palbociclib (Pb) with letrozole (1L) or fulvestrant (2L) for HR+HER2- MBC. The primary goal was to identify novel genomic variants and pathways associated with PFS. Here, we performed a secondary analysis to quantitate stromal TILs using protocol mandated pretreatment tumor biopsies and determine their association with PFS. Using the Kaplan-Meier method, we reported median PFS (mPFS) and 3-year PFS percentage (%) (95% confidence intervals [CIs]) by TILs [≥20% vs <20%] separately within 1L and 2L. As sample TIL values were generally concentrated at zero (27/58 = 46.6%), coupled with wide spread of sample TIL values ≥20% [range: 20%- 80%], we applied a ≥20% threshold for PFS analysis (high vs low TIL group) as described by Luen, Salgado et al. Lancet Oncol 2017. Statistical significance was based on the Wilcoxon test of the null hypothesis that there was no difference in the PFS curves by TILs. PFS is reported at median follow-up of 21 months. Results: Pretreatment tumor biopsies from 58/68 (85.3 %) pts were evaluable (42 in 1L and 16 in 2L). Median [Q1, Q2] TIL values were higher in 1L (5% [0, 10]) vs 2L (0% [0, 15]). The most common biopsy sites were bone (44.8%), lymph node (17.2%), liver (17.2%) and breast (12%). Overall, TILs frequency (>20%) by the 4 most common biopsy sites were bone (2/20, 10%), lymph node (4/7, 57%), liver (2/6, 33%), and breast (1/7, 14%). In 1L, although PFS was not significantly different (P = 0.237), mPFS (44 vs 34 months) and 3-year PFS 75.0%, [95% CI: 50.3%, 100%] vs 46.4% [95% CI: 31.0%, 69.5%]) were longer in pts with high versus low TILS, respectively. In 2L, the 3-year PFS was numerically similar comparing high and low TILs groups, (25.0%, [95% CI: 4.6%, 100%] vs 22.5% [95% CI: 6.7%, 76.1%]) with no significant difference in PFS (P = 0.761). Conclusions: In pts with ER+HER2- MBC treated with Pb and ET, PFS was numerically longer comparing high vs low TIL values in 1L but not 2L settings. Future studies evaluating TILs in larger datasets are needed to validate these findings. Genomic analysis (DNA/RNA) and its association with TIL levels is underway and will be presented at the meeting. Funding: The Center for Individualized Medicine, Mayo Clinic; Mayo Clinic Breast Specialized Program of Research Excellence (SPORE P50CA116201); Mayo Clinic Cancer Center (CA15083-40A2); TEMPUS (https://www.tempus.com/) and Conquer Cancer Foundation of ASCO (Career Development Award 2018, Dr. O’Sullivan)
Presentation numberPS1-10-23
Real-world analysis of unmet need among patients in the US with HR+/HER2- mBC following endocrine therapy
Joyce O’Shaughnessy, Baylor University Medical Center, Texas Oncology, Sarah Cannon Research Institute, Dallas, TX
J. O’Shaughnessy1, M. Rehnquist2, N. Sadetsky3, W. Verret4, P. Cinar4, N. Sjekloca5, L. Nguyen6, R. Nanda7, P. Sharma8; 1Oncology, Baylor University Medical Center, Texas Oncology, Sarah Cannon Research Institute, Dallas, TX, 2Real World Evidence, Gilead Sciences, Inc., Foster City, CA, 3Real World Evidence, Oncology, Gilead Sciences, Inc., Foster City, CA, 4Clinical Development, Gilead Sciences, Inc., Foster City, CA, 5Global Medical Affairs, Gilead Sciences Europe Ltd, Uxbridge, Middlesex, UNITED KINGDOM, 6US Medical Affairs, Gilead Sciences, Inc., Foster City, CA, 7Breast Oncology, The University of Chicago Medicine, Chicago, IL, 8Medical Oncology, University of Kansas Medical Center Westwood, Westwood, KS.
Background: Patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative (immunohistochemistry 0, 1+, or 2+/in situ hybridization-negative) metastatic breast cancer (HR+/HER2- mBC) are treated with endocrine therapy (ET) ± CDK4/6 inhibitor as the preferred option in the first-line setting followed by ET ± targeted therapy. Most HR+ disease will eventually become ET resistant, at which point treatment is primarily limited to cytotoxic therapy. This study describes characteristics, treatment patterns, and real-world outcomes among patients with HR+/HER2- mBC in the post ET-based treatment setting. Methods: This retrospective, observational cohort study utilized electronic health record-derived de-identified data from the US nationwide Flatiron Health Research Database. Women aged ≥ 18 years with HR+/HER2– mBC who received post ET-based treatment from January 1, 2015, to December 31, 2024 were included. Patient characteristics, treatment patterns, time to next treatment or death (TTNTD), and real-world overall survival (rwOS) were described; rwOS was assessed by Kaplan-Meier methods. Results: 5012 patients were included, with a median age of 63 years at mBC diagnosis. Most were Caucasian (62%), treated in the community setting (82%), had ECOG PS 0-1 (78%) at start of first post ET-based treatment, were diagnosed with recurrent mBC (68%), and had ≥ 2 prior lines of ET-based treatment in the mBC setting (65%) The median time from mBC diagnosis to first post ET-based treatment was 19 months. The median follow-up after the first post ET-based treatment in the mBC setting was 12 months. Ninety-six percent of patients received chemotherapy-based regimens as their first post ET-based treatment; capecitabine monotherapy was the most prescribed chemotherapy (34%) followed by taxane monotherapy (18%). Thirty percent of patients who initiated a post ET-based treatment did not survive to the next line of treatment. The median (95% CI) rwOS from start of first post ET-based treatment was 13.4 (12.9-13.9) months. There was a decrease in median (95% CI) rwOS with each subsequent line of treatment, ranging from 10.7 (10.2-11.1) months (second post ET-based treatment) to 7.4 (6.4-8.6) months (fifth post ET-based treatment) (Table). Similar trends were observed for TTNTD. Conclusions: These findings establish the high unmet need in patients with HR+/HER2– mBC treated with the standard of care regimens following ET-based treatment. For these patients, options are limited primarily to chemotherapy, with almost one-third not surviving to receive the second line of post ET-based treatment. This highlights the importance of making the most robust agents available for this patient population and demonstrates a clear and urgent need for new effective treatments in this setting.
| Index line | Total, N | Death prior to next line, n (%) | Median (95% CI) rwOS, months |
| First post ET-based treatment | 5012 | 1492 (30) | 13.4 (12.9-13.9) |
| Second post ET-based treatment | 3200 | 1023 (32) | 10.7 (10.2-11.1) |
| Third post ET-based treatment | 1878 | 624 (33) | 8.7 (8.2-9.3) |
| Fourth post ET-based treatment | 1070 | 387 (36) | 7.5 (6.7-8.6) |
| Fifth post ET-based treatment | 578 | 221 (38) | 7.4 (6.4-8.6) |
Presentation numberPS1-10-24
Compare the Data Differences between Dosing Groups in Patients with Advanced or Recurrent HR+/HER2- Breast Cancer Treated with Ecirciclib (BPI-1178) Combined with Endocrine Therapy
Feng Gao, Beta Pharma Inc., Princeton, NJ
Y. Du1, J. Zhang1, J. Wu2, C. Hu3, W. Chen4, Y. Li5, M. Yan6, H. Li7, W. Li8, Y. Teng9, T. Huang10, Y. Lu11, Y. Liu7, D. Zhang12, J. Peng12, F. Gao13, T. Wang14, W. Zhang14; 1Phase I Cinical, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 2Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 3Department of Medical Oncology, The First Affiliated Hospital Of Ustc West District/Anhui Provincial Cancer Hospital/Anhui Provincial Hospital West District, Hefei, CHINA, 4Breast Oncology Department, Nanchang Third Hospital, Nanchang, CHINA, 5Phase I ward (clinical research ward), Chongqing University Cancer Hospital, Chongqing, CHINA, 6Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, CHINA, 7Key Laboratory of Carcinogenesis and Translational Research, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, CHINA, 8Department of Oncology, The First Hospital of Jilin University, Changchun, CHINA, 9Department of Medical Oncology, The First Hospital Of China Medical University, Shenyang, CHINA, 10Department of thyroid and breast Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHINA, 11Department of Breast and Bone Soft Tissue Tumors, Guangxi Medical University Cancer Hospital & Guangxi Cancer Institute, Nanning, CHINA, 12Department of Drug Discovery, Beta Pharma Inc., Princeton, NJ, 13Department of Clinical Development, Beta Pharma Inc., Princeton, NJ, 14Department of Clinical Development, Beta Pharma (shanghai) co., Ltd., Shanghai, CHINA.
Background: Eciruciclib (BPI-1178) is a novel cyclin-dependent kinase (CDK) 2/4/6 inhibitor that has demonstrated potent inhibition of CDK2/4/6 expression in preclinical studies.Methods: Phase IIa had two cohorts, A and B. Cohort A included patients with HR+/HER2- advanced breast cancer (ABC) who progressed after endocrine therapy (ET) and were treated with eciruciclib and fulvestrant. In contrast, treatment-naive patients with HR+/HER2- ABC in cohort B received eciruciclib and letrozole. All patients got eciruciclib either on an intermittent (21 days on, 7 days off) or continuous (28 days on) schedule in a 28-day cycle until disease progression or unacceptable toxicity. Safety was evaluated by CTCAE 5.0. Efficacy endpoints like confirmed objective response rate (ORR), disease control rate (DCR), and progression-free survival (PFS) were assessed by investigators using RECIST 1.1. Results: The data released this time differs from that at ASCO 2025. As of Feb 25, 2025, cohort A enrolled 75 patients (73 evaluable for efficacy), with an ORR of 46.6% (95% CI, 34.80, 58.63), a DCR of 90.4% (95% CI, 81.24, 96.06), and a median PFS of 20.57 months. Cohort B enrolled 26 patients (25 evaluable for efficacy), with an ORR of 75.9% (95% CI, 56.46, 89.7), a DCR of 96.6% (95% CI, 82.24, 99.91), and the PFS not reached. In cohort A, the top three grade ≥ 3 TRAEs were neutrophil count decreased (38.7%), white blood cell count decreased (17.3%), and hypertriglyceridemia (14.7). In cohort B, they were neutrophil count decreased (23.3%), hypertriglyceridemia (20.0%), alanine aminotransferase increased (10.0%), and white blood cell count decreased (10.0%). Discussion: It is noteworthy that despite using the same combination drugs, there were differences in data between different dosing groups. The theoretical medication dosage per cycle was identical in both the intermittent dosing 400mg group and continuous dosing 300mg group of cohort A, both totaling 8400mg. The PFS of the continuous dosing 300mg group has not yet been reached, making comparisons currently unavailable, but its ORR is superior to the intermittent dosing 400mg group. Both groups exhibited comparable safety profiles, with no significant increase in the incidence of adverse reactions under the continuous dosing regimen.Conclusions: Under equivalent theoretical dosage, different administration regimens vary in efficacy but have comparable safety, offering valuable references for selecting the optimal dosing strategy in subsequent phases.
| Cohort A (n=73) | Cohort B (n=25) | |||||
|
400mga (n=20) |
300mga (n=16) |
300mgb (n=20) |
200mgb (n=17) |
400mga (n=19) |
300mga (n=6) |
|
| ORR, n (%) | 10 (50.0) | 7 (43.8) | 12 (60.0) | 5 (29.4) | 15 (78.9) | 6 (100.0) |
| DCR, n (%) | 17 (85.0) | 13 (81.3) | 20 (100.0) | 16 (94.1) | 18 (94.7) | 6 (100.0) |
| Median PFS, months (95% CI) | 18.27 (7.2, 22.08) | 7.33 (2.37, 23.89) | NR (12.81, NR) | NR | NR (20.7, NR) | NR |
| TRAEs with severity ≥ Grade 3 (%) | 13 (65.0) | 12 (70.6) | 14 (70.0) | 8 (44.4) | 11 (55.0) | 2 (33.3) |
| TRAE leading to permanent discontinuation, n (%) | 0 | 0 | 1 (5.0) | 0 | 0 | 0 |
| TRAE leading to death, n (%) | 0 | 0 | 0 | 0 | 0 | 0 |
| Median duration of exposure, weeks | 42.5 | 32.1 | 54.9 | 30.1 | 93.5 | 126.6 |
|
a intermittent dosing schedule; b continuous dosing schedule; NR, not reached. |
Presentation numberPS1-10-25
Pharmacodynamic effects of the first-in-class CDK4-selective inhibitor atirmociclib (PF-07220060) in combination with endocrine therapy in tumors of patients with HR+/HER2− metastatic breast cancer
Antonio Giordano, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
A. Giordano1, F. Liu2, W. Roh3, H. Zhang4, D. Fabiola Flores Diaz5, C. A. Hernandez6, X. Wu7, M. C. Sanchez2, J. Park8, M. Delioukina9, R. A. Moss3, T. A. Yap10; 1Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 2Clinical Pharmacology and Translational Sciences, Oncology, Pfizer Inc., La Jolla, CA, 3Oncology, Pfizer Inc., La Jolla, CA, 4Translational Biomarker Statistics, Pfizer Inc., New York, NY, 5., Instituto Nacional de Cancerologia, Mexico City, MEXICO, 6Hospital Regional Presidente Juárez, Institute for Social Security and Services for State Workers, Oaxaca, MEXICO, 7Department of Breast Surgery, Hubei Cancer Hospital, Wuhan, CHINA, 8Early Stage Clinical Development, Oncology, Pfizer Inc., Bellevue, WA, 9Early Stage Clinical Development, Oncology, Pfizer Inc., La Jolla, CA, 10Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: The cyclin-dependent kinase 4 (CDK4)-selective inhibitor atirmociclib (PF-07220060) in combination with endocrine therapy (ET) has demonstrated favorable tolerability and promising clinical activity, as well as strong pharmacodynamic effects in inhibiting serum thymidine kinase activity (TKa) and early ctDNA decrease in an ongoing phase 1 trial (NCT04557449). We performed further exploratory analysis of pharmacodynamic biomarker modulation in tumor biopsies collected from patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2−) metastatic breast cancer (mBC) treated with atirmociclib plus fulvestrant.Methods: Tumor biopsy samples were collected before the first dose (baseline) and at Cycle 2 Day 1 (C2D1) from patients with HR+/HER2− mBC treated with 100, 300, or 400 mg BID atirmociclib in combination with fulvestrant. Formalin-fixed paraffin-embedded tumor samples were analyzed for phospho-Rb (Ser807/811, pRb), Ki67, and FOXM1 expression by immunochemistry (IHC), and RNA expression by whole transcriptome RNA-seq. Gene set variation analysis (GSVA), PAM50 molecular subtype, and BayesPrism bulk RNA-seq deconvolution were performed with the RNA-seq data.Results: At data cutoff (Feb 21, 2025), results from baseline and C2D1 were available for IHC (pRb, Ki67, or FOXM1) in 14 patients and RNA-seq in 14 patients. Median changes of pRb, Ki67, and FOXM1 expression from baseline to C2D1 were −86%, −88%, and −100%, respectively. GSVA showed strong inhibition of cell cycle-related pathways (e.g., E2F, G2M, DNA repair, and Myc), largely in patients who achieved stable disease or better as best overall response. Atirmociclib plus fulvestrant treatment also resulted in upregulation of immune and inflammation response gene sets. Cell type fraction estimation based on BayesPrism bulk RNA-seq deconvolution showed a decrease in tumor cell fraction and an increase in cancer-associated fibroblasts (CAFs) fraction. An increase in macrophage fraction and expression of genes associated with antigen presentation was also observed. No significant change in CD4+ or CD8+ T-cell fraction was detected. Among 13 tumors with Luminal B or HER2-enriched PAM50 intrinsic molecular subtype at baseline, 10 (77%) switched to the less proliferative Luminal A subtype at C2D1.Conclusions: Atirmociclib, in combination with fulvestrant, showed strong effects in inhibiting cell cycle-related pharmacodynamic protein biomarkers and gene sets in HR+/HER2− mBC tumors, consistent with its mechanism of CDK4 inhibition. The analyses also revealed potential immune-modulatory effects of atirmociclib, including increased macrophage infiltration and enhanced antigen presentation pathway gene expression.
Presentation numberPS1-10-26
Comparative analysis of blood- and tissue-based <i>PIK3CA</i> mutation detection methods in the pivotal Phase 3 INAVO120 trial of palbociclib + fulvestrant ± inavolisib in hormone receptor-positive, HER2-negative advanced breast cancer
Kevin Kalinsky, Emory University, Atlanta, GA
S. Hilz1, K. Kalinsky2, D. Juric3, N. Turner4, K. Jhaveri5, S. Im6, S. Loibl7, S. Loi8, C. Saura9, P. Schmid10, E. Shaddox11, N. Pantoja Galicia11, C. Guo11, Y. Li11, W. Darbonne1, R. Desai1, M. Pagan12, P. Karavagul13, J. Aimi12, S. Royer-Joo14, J. Schutzmann14, T. Stout15, C. Song16, P. Bedard17, K. Hutchinson1; 1Translational Medicine Oncology, Genentech, Inc., South San Francisco, CA, 2Winship Cancer Institute, Emory University, Atlanta, GA, 3Mass General Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA, 4Royal Marsden Hospital, Institute of Cancer Research, London, UNITED KINGDOM, 5Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 6Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, KOREA, REPUBLIC OF, 7Center for Hematology and Oncology Bethanien, Goethe University and The German Breast Group, Frankfurt, GERMANY, 8Division of Cancer Research, Peter MacCallum Cancer Centre and The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, AUSTRALIA, 9Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology, Barcelona, SPAIN, 10Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 11Biometrics, Foundation Medicine, Inc., Boston, MA, 12Translational Medicine, Genentech, Inc., South San Francisco, CA, 13Product Development Global (PDG) Oncology, F. Hoffmann-La Roche AG, Basel, SWITZERLAND, 14gRED: Genentech Research and Early Development, Genentech, Inc., South San Francisco, CA, 15PDO Clinical Science, Genentech, Inc., South San Francisco, CA, 16Product Development Oncology, Genentech, Inc., South San Francisco, CA, 17Princess Margaret Cancer Centre – University Health Network, University of Toronto, Toronto, ON, CANADA.
BACKGROUND Detection of a PIK3CA mutation (PIK3CAmut) is a prerequisite for treatment with currently approved PI3K inhibitors in breast cancer (BC). However, this was only recently introduced in testing guidelines, and cross-assay concordance studies are limited. Using available blood and tissue specimens from participants (pts) screened for the Phase 3 INAVO120 trial (NCT04191499) of the recently approved PI3Kɑ inhibitor inavolisib, we compared the ability of various molecular tests to identify PIK3CAmut. METHODS Data from FoundationOne®Liquid CDx (F1LCDx®; Foundation Medicine, Inc.) profiling on circulating tumor (ct)DNA was available for 1422 pts, either from central screening for study eligibility (n = 1383) or post-enrollment testing of ctDNA from local blood / tumor tissue test eligible pts (n = 39). Due to data / sample sharing restrictions, 43 pts from China were excluded from an otherwise global population. Retrospective to enrollment, DNA extracted from available archival / fresh tumor tissue was assessed by FoundationOne®CDx (F1CDx®; n = 323) sequencing and / or the Cobas® PIK3CA Mutation Test (a polymerase chain reaction [PCR] test; n = 84). Depending on the sample data available, assay concordance was evaluated for samples from both enrolled and unenrolled individuals. For certain assay comparisons, the fraction of samples in which a PIK3CAmut was detected by both assays is reported; where otherwise appropriate, traditional positive (+), negative (-), and overall percent agreement (PPA, NPA, OPA) between assays are reported. RESULTS Of the 1383 individuals screened centrally with F1LCDx, 500 (36.2%) had an enrollment-eligible PIK3CAmut; 242 of these plus the 39 local test-identified pts (n = 281) were enrolled in the study and comprised the efficacy-evaluable population for this analysis. F1LCDx was performed on available ctDNA from 37 / 39 local test-identified pts; 27 / 37 samples (73.0%) had a PIK3CAmut identified by both assays. Of the ten discordances, most could be explained by low ctDNA tumor fraction (median 0.0% in discordant, 6.0% in remainder). F1CDx was performed on available tumor tissue from 29 local test-identified pts; a PIK3CAmut was identified in all 29 (100%). F1LCDx and F1CDx data were available for 315 pts (214 enrolled and treated, 101 screened but not enrolled / treated). For this F1LCDx vs F1CDx analysis, PPA ([F1LCDx+ and F1CDx+] / F1CDx+), NPA ([F1LCDx- and F1CDx-] / F1CDx-), and OPA ([F1LCDx+, F1CDx+, F1LCDx-, F1CDx-] / all data) were 93.0% (226 / 243), 73.6% (53 / 72), and 88.6% (279 / 315), respectively. Cobas PCR and F1CDx data were available for 84 pts (42 enrolled and treated, 42 screened but not enrolled / treated). PPA ([Cobas+ and F1CDx+] / F1CDx+), NPA ([Cobas- and F1CDx-] / F1CDx-), and OPA were 90.2% (46 / 51), 100% (33 / 33), and 94.0% (79 / 84), respectively. With regard to efficacy, pts enrolled by a local test (hazard ratio for progression-free survival [PFS] 0.7; 95% CI 0.3-1.5; n = 39) or the F1LCDx central test (hazard ratio for PFS 0.5; 95% CI 0.3-0.7; n = 242) showed a similar response to inavolisib plus palbociclib and fulvestrant over placebo plus palbociclib and fulvestrant. CONCLUSIONS This comprehensive concordance analysis utilizing INAVO120 specimens demonstrated the ability of both tissue- and blood-based PCR and next-generation sequencing assays to robustly identify a pt population with PIK3CA-mutated, hormone receptor-positive, HER2-negative advanced BC who may consistently benefit from this INAVO-based regimen.
Presentation numberPS1-10-27
Progression-free survival (PFS) and overall survival (OS) results from the phase 3 MONALEESA-3 trial of postmenopausal patients with hormone receptor-positive (HR+)/HER2-negative (HER2−) advanced breast cancer (ABC) treated with ribociclib (RIB) + fulvestrant (FUL): A subgroup analysis of patients with invasive lobular carcinoma (ILC)
Michelino De Laurentiis, National Cancer Institute “Fondazione Pascale”, Napoli, Italy
M. De Laurentiis1, J. Mouabbi2, S. Im3, M. Kruse4, S. Chia5, S. Brucker6, C. Villanueva7, G. Jerusalem8, M. Gao9, G. Sopher10, J. Wu11, J. Zarate12, J. Beck13; 1Breast and Thoracic Oncology, National Cancer Institute “Fondazione Pascale”, Napoli, ITALY, 2Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 4Hematology and Medical Oncology, Division of Breast Medical Oncology, Taussig Cancer Institute, Cleveland, OH, 5Medical Oncology, BC Cancer, Vancouver, BC, CANADA, 6Gynecology and Obstetrics, University Hospital Tuebingen, Tuebingen, GERMANY, 7Clinical Research, Clinique Clementville, Montpellier, FRANCE, 8Medical Oncology, CHU Liege and Liège University, Liège, BELGIUM, 9Global Medical Affairs, Novartis Pharma AG, Basel, SWITZERLAND, 10Medical Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, 11Biostatistician, Novartis Pharmaceuticals Corporation, East Hanover, NJ, 12Medical Affairs & Clinical Development, Novartis Pharmaceuticals Corporation, East Hanover, NJ, 13Medical Director of Research, Highlands Oncology, Springdale, AR.
Background: The phase 3 MONALEESA-3 trial reported a statistically significant improvement in PFS with RIB + FUL over placebo (PBO) + FUL as first-line (1L) or second-line (2L) treatment (tx) in postmenopausal patients with HR+/HER2- ABC at the primary analysis (median: 20.5 months vs 12.8 months; HR: 0.59; 95% CI: 0.48-0.73; P < 0.001). Additionally, a statistically significant OS benefit with RIB + FUL was seen at the final protocol-specified analysis (HR: 0.72; 95% CI: 0.57-0.92), which was maintained with extended follow up (median follow-up time: 56.3 months; HR, 0.73; 95% CI: 0.59-0.90). Patients with the ILC subtype, who account for 10-15% of all breast cancer patients, show unique clinicopathological characteristics. This exploratory analysis examined the PFS and OS in a subgroup of MONALEESA-3 patients with ILC, including those receiving tx in the 1L setting. Methods: Postmenopausal patients with HR+/HER2− ABC were randomized 2:1 to receive RIB + FUL or PBO + FUL in 1L and 2L settings. PFS and OS were evaluated by log-rank test and Cox proportional hazards model stratified per interactive response technology by lung/liver metastasis status and previous endocrine therapy. PFS and OS were summarized using Kaplan-Meier methods. The data cutoff for this analysis was January 11, 2023. Results: Among all MONALEESA-3 patients (N=726), 120 (16.5%) had the ILC subtype, 77 of whom were treated with RIB + FUL and 43 with PBO + FUL. Additionally, of the 354 MONALEESA-3 patients receiving 1L tx, 56 (15.8%) had the ILC subtype, with 38 treated with RIB + FUL and 18 with PBO + FUL. Patient demographics and disease characteristics were balanced between the two tx arms in this subgroup. Patients had a median age of 63 y (RIB arm: 64 y; PBO arm: 63 y); 17.5% of patients had de novo ABC (RIB arm: 18.2%; PBO arm, 16.3%), and 81.7% patients had experienced relapse >12 months after completion of (neo)adjuvant therapy (RIB arm: 80.5%; PBO arm, 83.7%); 83.3% of patients had bone metastasis (RIB arm: 81.8%; PBO arm, 86.0%), and 45.0% had visceral metastasis (RIB arm: 48.1%; PBO arm: 39.5%). The baseline characteristics were similar among those with ILC receiving 1L tx and remained balanced between the two tx arms. Patients with ILC receiving RIB + FUL showed a longer PFS (median: 20.5 months) than those receiving PBO + FUL (median: 9.4 months, HR: 0.56; 95% CI: 0.37-0.86). The median PFS was further prolonged in patients with ILC receiving 1L tx with RIB + FUL (median: 26.3 months) over PBO + FUL (median: 18.1 months; HR: 0.78; 95% CI: 0.39-1.59). Additionally, patients with ILC receiving RIB + FUL had a longer OS (median: 51.2 months) than those receiving PBO + FUL (median: 30.8 months, HR: 0.62; 95% CI: 0.39-0.98); the median OS in patients with ILC receiving 1L tx with RIB + ET vs PBO + FUL was 59.6 months vs 40.0 months (HR: 0.54; 95% CI: 0.25-1.19). Rates of adverse events (AEs) including neutropenia, alanine aminotransferase elevation, and aspartate aminotransferase elevation in RIB + FUL-treated patients in the ILC subgroup (and in the 1L tx ILC subgroup) were comparable to those observed in the overall MONALEESA-3 population. Conclusions: Similar to the overall MONALEESA-3 population, RIB + FUL tx demonstrated an improvement in PFS and OS over PBO + FUL in patients with ILC, including among those treated in the 1L setting. The efficacy benefit of RIB observed in patients with HR+/HER2- ABC with the ILC subtype, including those receiving 1L tx, further supports the use of RIB in this patient population.
Presentation numberPS1-10-28
Phase 1/2a trial of new generation PARP1-selective inhibitor saruparib + next generation selective ER degrader (SERD) camizestrant in patients (pts) with advanced/relapsed ER+/HER2-negative or low (HER2−) breast cancer (PETRA Module 6)
Timothy A Yap, University of Texas MD Anderson Cancer Center, Houston, TX
T. A. Yap1, K. Yonemori2, W. Li3, S. Kitano4, F. Lynce5, A. Sharp6, A. Falcón7, J. Garcia-Corbacho8, R. D. Baird9, J. Balmaña10, S. Nanda11, I. Song12, K. Sugibayashi13, L. Womersley14, M. Squatrito15, E. Gangl16, T. Milenkova17, S. J. Luen18; 1Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Medical Oncology, National Cancer Centre Hospital, Tokyo, JAPAN, 3Oncology Department, Cancer Center, First Affiliated Hospital, Jilin University, Jilin, CHINA, 4Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 5Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 6Drug Development Unit, Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, London, UNITED KINGDOM, 7Department of Medical Oncology, Hospital Universitario Virgen del Rocio, Seville, SPAIN, 8Department of Medical Oncology, Hospital Clínico Universitario Virgen de la Victoria, Malaga, SPAIN, 9Department of Oncology, Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UNITED KINGDOM, 10Department of Medical Oncology, Vall d’Hebron University Hospital, Barcelona, SPAIN, 11Early Global Development, AstraZeneca, Gaithersburg, MD, 12Early Global Development, AstraZeneca, Waltham, MA, 13Early Global Development, AstraZeneca, Cambridge, UNITED KINGDOM, 14Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 15Translational Medicine, AstraZeneca, Cambridge, UNITED KINGDOM, 16Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology Safety Science, R&D, AstraZeneca, Waltham, MA, 17Early Oncology Clinical Development, AstraZeneca, Cambridge, UNITED KINGDOM, 18Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, AUSTRALIA.
Background: Saruparib (AZD5305) is a highly selective, potent inhibitor and trapper of PARP1. Camizestrant is an oral next generation SERD and complete ER antagonist. PETRA (NCT04644068) is a Phase 1/2a, modular, open label, multicenter study of saruparib as monotherapy or in combination with anticancer agents in pts with advanced solid malignancies. We present safety and preliminary efficacy of saruparib + camizestrant in pts with advanced/relapsed ER+/HER2- breast cancer. Methods: Pts were aged ≥18 years with histologically/cytologically confirmed breast adenocarcinoma, a locally-documented ER+ and HER2- tumor and an ECOG PS 0/1. Pts were required to have recurrence or progression on ≥1 line of endocrine therapy, and were allowed ≤2 prior lines of chemotherapy in the advanced setting and ≤1 prior PARP inhibitor. Pts could receive prior CDK4/6 inhibitors or fulvestrant, but not oral SERDs. Pts received 60 mg saruparib + 75 mg camizestrant daily orally. The primary objective was assessment of safety and tolerability; secondary objectives included pharmacokinetic (PK) assessments and preliminary antitumor activity. Results: At data cutoff (Mar 31, 2025), 38 pts had received ≥1 dose of the combination. Median age was 56 years; median prior lines of therapy was 3.5 (range, 1-13). BRCA1/BRCA2 mutation (m) and PALB2m were detected in 8 (21.1%) and 2 (5.3%) pts, respectively, based on local testing. Safety is summarized in the Table. Hematological toxicities included anemia and neutropenia; gastrointestinal toxicities included nausea and vomiting. PK data showed no evidence of drug interaction between saruparib and camizestrant. Saruparib exposure was comparable following single-dose saruparib versus saruparib at steady-state in combination with camizestrant, for AUC (42,219 vs 41,703 h*ng/mL) and Cmax (2,834 vs 3,250 ng/mL). Eight of 35 evaluable pts achieved confirmed partial responses (by RECIST v1.1) for an objective response rate (ORR) of 22.9%; median duration of response was 5.5 months. In addition, 12 pts (34.3%) achieved stable disease for a disease control rate of 57.1%. An ORR of 33.3% was reported in 9 pts with measurable disease and BRCA1/BRCA2m or PALB2m. The median progression-free survival was 4.4 months (80% CI, 2.0-5.6) overall and 6.1 months (80% CI, 4.4-7.3) in pts with mutations. Conclusion: Saruparib + camizestrant was well tolerated with no new safety signals versus prior monotherapy studies of each drug. PK of saruparib in combination with camizestrant was consistent with monotherapy data. Preliminary efficacy of the combination was promising and compared favorably with camizestrant monotherapy. This combination is being tested in a randomized Phase 3 trial in pts with ER+/HER2- and BRCA1m/BRCA2m or PALB2m (EvoPAR-Breast01; NCT06380751).
| Safety, n (%) | Total (N=38) |
| Any AE | 38 (100) |
| Any SAE | 5 (13.2) |
| Any AE Grade ≥3 | 15 (39.5) |
| Any AE leading to interruption of: Saruparib Camizestrant | 15 (39.5) 11 (28.9) |
| Any AE leading to reduction of: Saruparib Camizestrant | 6 (15.8) NA |
| Any AE leading to discontinuation of: Saruparib Camizestrant | 1 (2.6) 2 (5.3) |
| Most common TEAEs, any grade / Grade ≥3: Anemia* Nausea Neutropenia* Thrombocytopenia* Fatigue and asthenia* Vomiting | 19 (50.0) / 8 (21.1) 18 (47.4) / 1 (2.6) 11 (28.9) / 3 (7.9) 10 (26.3) / 0 9 (23.7) / 0 8 (21.1) / 1 (2.6) |
| *Grouped terms. AE, adverse event; NA, not applicable; SAE, serious adverse event; TEAEs, treatment-emergent adverse events |
Presentation numberPS1-10-29
Complete tumor response during first-line endocrine therapy (ET) plus Cyclin Dependent Kinase 4/6 inhibitors (CDK4/6i) predicts excellent clinical outcomes in patients with Hormone Receptor-positive, Human Epidermal growth factor Receptor 2-negative advanced Breast Cancer (HR+/HER2- aBC)
Claudio Vernieri, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
C. Vernieri1, R. Caputo2, M. Dieci3, P. Vigneri4, M. Giuliano5, G. Curigliano6, A. Toss7, A. Botticelli8, R. Pedersini9, S. Cinieri10, M. Lambertini11, B. Tagliaferri12, G. Rizzo13, M. Sirico14, M. Giordano15, L. Gerratana16, I. Meattini17, A. Fabi18, M. Piras19, A. Zambelli20, F. Pantano21, A. Gennari22, N. La Verde23, D. Sartori24, O. Garrone25, D. Generali26, F. Jacobs1, F. De Braud1, G. Pruneri27, L. Provenzano1, PALMARES-2 study group; 1Medical Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 2Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, ITALY, 3Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, ITALY, 4Department Clinical and Experimental Medicine, University of Catania, Catania, ITALY, 5Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, Naples, Naples, ITALY, 6Division of Early Drug Development for Innovative Therapy, IRCCS European Institute of Oncology, Milan, ITALY, 7Division of Medical Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, ITALY, 8Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, ITALY, 9Medical Oncology Department, ASST Spedali Civili of Brescia, Brescia, ITALY, 10U.O.C. Oncologia medica, Presidio Ospedaliero “Antonio Perrino”, Brindisi, ITALY, 11Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genoa, ITALY, 12Medical Oncology 1, ICS Maugeri-IRCCS, Pavia, ITALY, 13Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, ITALY, 14Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, Meldola, ITALY, 15SC Oncologia, ASST Lariana, Como, ITALY, 16Breast Medical Oncology, CRO Aviano National Cancer Institute, Aviano, ITALY, 17Unità di Radioterapia Oncologica, Azienda Ospedaliero Universitaria Careggi, Florence, ITALY, 18UOSD Medicina di Precisione in Senologia, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Rome, ITALY, 19Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, ITALY, 20Oncology Department, Papa Giovanni XXIII Hospital, Bergamo, ITALY, 21Medical Oncology, Università Campus Bio-Medico di Roma, Rome, ITALY, 22Department Translational Medicine, University of Piemonte Orientale, Novara, ITALY, 23Department of Oncology, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, ITALY, 24Oncology Unit, AULSS3 Veneziana, Mirano, ITALY, 25SC Oncologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milan, ITALY, 26SC Oncologia Multidisciplinare, ASST Cremona, Cremona, ITALY, 27Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY.
Background: The CDK4/6i palbociclib (P), ribociclib (R), and abemaciclib (A) combined with ET are the mainstay first-line therapy for patients (pts) with Hormone Receptor-positive, HR+/HER2- aBC. The impact of best tumor responses on long-term clinical outcomes has never been explored in this setting. Methods: PALMARES-2 (NCT06805812) is the largest, real-world (rw) study designed to investigate the effectiveness of first-line ET+CDK4/6i in pts with HR+/HER2- aBC treated in 25 Italian Institutions. Here, we investigated the impact of different types of radiologically and clinically assessed best tumor responses – complete response (CR), partial response (PR), stable disease (SD) or progressive disease (PD) – on patient rw progression-free survival (rwPFS), time-to-chemotherapy (rwTTC), or overall survival (rwOS). Multivariable Cox regression models were used to adjust the association between the type of best tumor response and clinical outcomes for 16 clinically-relevant covariates. Logistic regression was used to identify variables predicting CR. Results: Of 3933 pts included in this analysis, 487 pts (12.4%) achieved CR, while 1620 (41.2%), 1535 (39.0%) and 292 (7.4%) pts achieved PR, SD and PD as best tumor response, respectively. Pts achieving CR had remarkably better rwPFS, rwTTC and rwOS when compared to pts achieving PR, SD or PD regardless of tumor endocrine sensitivity/resistance (Table). In the CR cohort, the three CDK4/6i were associated with similar rwPFS (R vs P: aHR 1.06, P =0.735; A vs P: aHR 0.93, P =0.763). Higher tumor expression of estrogen receptor alpha (ERα), as well as the presence of node or soft tissue metastases were associated with higher CR probability (OR: 1.10 per 10% increase, P=0.014; OR: 2.04, P=0.001; OR: 1.77, P=0.010, respectively). Conversely, higher tumor burden (OR 0.51 for each additional metastatic site, P <0.001), older age (OR: 0.79 per 10 years increase, P<0.001), ECOG PS ≥1 (OR: 0.52, P<0.001), higher Ki-67 expression (OR: 0.92 per 10% increase, P=0.025) and endocrine-resistant disease (OR: 0.78, P=0.047) were associated with lower CR probability. Among pts achieving CR, early CDK4/6i discontinuation because of toxicities was not associated with worse rwPFS (P=0.185) or rwOS (P=0.440). Conclusion: Achieving CR during first-line ET+CDK4/6i therapy is associated with excellent clinical outcomes in pts with HR+/HER2- aBC. These findings can guide clinicians in personalizing the management of pts with CR during ET+CDK4/6i, including CDK4/6i dose reduction/discontinuation in case of severe toxicities, or tailoring radiological disease re-evaluations.
| Subgroup | Best Response (N; %) | Median rwPFS (months) | aHR CR vs. PR/SD/PD (95% CI), P val | Median rwTTC (months) | aHR CR vs. PR/SD/PD (95% CI), P val | Median rwOS (months) | aHR CR vs. PR/SD/PD (95% CI), P val | ||||||||
| Whole cohort | CR (487; 12.4%) | 64.0 | 0.36 (0.31-0.42), <0.001 | NR | 0.23 (0.19-0.29), <0.001 | NR | 0.26 (0.19-0.34), <0.001 | ||||||||
| PR/SD/PD (3447; 87.6%) | 22.1 | 32.2 | 61.3 | ||||||||||||
| Endocrine-sensitive | CR (338; 12.8%) | 67.8 | 0.39 (0.32-0.47), <0.001 | NR | 0.24 (0.19-0.32), <0.001 | NR | 0.30 (0.21-0.43), <0.001 | ||||||||
| PR/SD/PD (2293; 87.2%) | 29.0 | 40.4 | 68.0 | ||||||||||||
| Endocrine-resistant | CR (149; 11.4%) | 47.2 | 0.32 (95% CI 0.25 – 0.41), p = <0.001 | NR | 0.23 (0.17 – 0.31), <0.001 | NR | 0.21 (0.13 – 0.33), <0.001 | ||||||||
| PR/SD/PD (1153; 88.6%) | 13.7 | 21.5 | 46.6 |
Presentation numberPS1-10-30
Real-world treatment utilization and outcomes of trastuzumab deruxtecan (T-DXd) among patients with hormone receptor-positive (HR+) HER2-low metastatic breast cancer (mBC) in the United States
Adam Brufsky, UPMC Magee-Womens Hospital, Pittsburgh, PA
A. Brufsky1, P. A. Kaufman2, M. Inoue-Choi3, C. Lam4, C. Nordon5, E. John6, L. Luo7, H. Rugo8; 1Division of Hematology and Oncology, UPMC Magee-Womens Hospital, Pittsburgh, PA, 2Division of Hematology and Oncology, University of Vermont, Burlington, VT, 3Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 4US Oncology Medical Affairs, AstraZeneca, Gaithersburg, MD, 5Oncology Outcomes Research, AstraZeneca, Cambridge, UNITED KINGDOM, 6Global Medical Affairs Payer Biometrics, Statistics, AstraZeneca, Cambridge, UNITED KINGDOM, 7Oncology Data & Analytics, AstraZeneca, Gaithersburg, MD, 8Department of Breast Medical Oncology, City of Hope, Duarte, CA.
Introduction: T-DXd, a HER2-directed antibody-drug conjugate, was the first treatment approved for patients (pts) with HER2-low (immunohistochemistry [IHC] 1+, IHC 2+ / in situ hybridization-negative [ISH−]) mBC, who had received a prior line of chemotherapy, based on the results from the Phase 3 trial, DESTINY-Breast04. This study describes demographic, clinical, and disease characteristics as well as real-world treatment outcomes of patients with HER2-low mBC treated with T-DXd, stratified by line of therapy (LOT) since mBC diagnosis. Methods: This was a retrospective longitudinal observational cohort study using patient-level electronic health record-derived de-identified data from the nationwide Flatiron Health database. Pts aged ≥18 years with no prior cancer (excluding thyroid and non-melanoma skin cancer), de-novo or recurrent HER2-low mBC diagnosed after Jan 1, 2018, who had received T-DXd between Aug 5, 2022 (FDA approval date), and Jan 1, 2025 (data cutoff), were eligible. Demographic and clinical characteristics were assessed at T-DXd initiation (index date). Descriptive statistics were used to explore pt characteristics, and the Kaplan-Meier (KM) method was used to analyze real-world time to next treatment (rwTTNT) and real-world time to treatment discontinuation (rwTTD). The overall median follow-up time was calculated using the reverse KM method. This analysis focuses on patients with HR+ mBC. Results: 1232 pts were included in the analysis; the overall median age at index date was 64.0 years. T-DXd was initiated as the first-line (1L), 2L, 3L, and ≥4L therapy following mBC diagnosis for 7.3% (n=90), 17.5% (n=216), 24.0% (n=296), and 51.1% (n=630) of pts, respectively. The reason for initiating T-DXd in 1L after mBC diagnosis (n=90) was not available. Pts who received T-DXd in earlier LOTs were more likely to be treated in a community versus academic setting and have recurrent disease (Table). Overall median follow-up time was 14.0 months. Median rwTTNT (95% CI) following the initiation of T-DXd as 1L, 2L, 3L, and ≥4L was 7.4 (5.0, 11.8), 9.7 (8.3, 11.3), 7.9 (6.7, 9.3), and 7.4 (6.6, 8.2) months, respectively. Median rwTTD (95% CI) for 1L, 2L, 3L, and ≥4L was 7.1 (5.0, 11.8), 9.3 (8.3, 10.7), 7.8 (6.5, 9.2), and 7.1 (6.4, 7.8) months, respectively. Conclusion: Except for patients with HR+ HER2-low mBC who initiated T-DXd in 1L after mBC diagnosis, a trend was observed toward worse outcomes (as indicated by shorter rwTTNT and rwTTD) in pts who initiated T-DXd in later lines. Further study is warranted as these results suggest earlier initiation of T-DXd for eligible pts with HR+ HER2-low mBC (in accordance with approved labels) prolongs duration of disease control.
| 1L (n=90) | 2L (n=216) | 3L (n=296) | ≥4L (n=630) | |
| Median age at the start of T-DXd treatment, years (interquartile range) | 60.5 (51.0, 71.0) | 64.0 (53.0, 72.0) | 64.0 (55.0, 70.0) | 63.0 (55.0, 72.0) |
| Female, n (%) | 89 (98.9) | 211 (97.7) | 293 (99.0) | 620 (98.4) |
| Academic practice, n (%)*† | 5 (5.6) | 20 (9.3) | 39 (13.2) | 82 (13) |
| Community practice, n (%)*† | 82 (91.1) | 180 (83.3) | 233 (78.7) | 477 (75.7) |
| Academic/community practice, n (%)*† | 3 (3.3) | 16 (7.4) | 24 (8.1) | 71 (11.3) |
| Breast cancer diagnosed at Stage I–III, n (%) | 68 (75.6) | 146 (67.6) | 204 (68.9) | 385 (61.1) |
| Breast cancer diagnosed at Stage IV (de novo), n (%) | 13 (14.4) | 56 (25.9) | 74 (25.0) | 205 (32.5) |
| Metastasis site, n (%) | ||||
| Bone | 65 (72.2) | 163 (75.5) | 249 (84.1) | 541 (85.9) |
| Brain | 7 (7.8) | 20 (9.3) | 41 (13.9) | 75 (11.9) |
| Liver | 40 (44.4) | 118 (54.6) | 193 (65.2) | 394 (62.5) |
| Lung | 26 (28.9) | 71 (32.9) | 87 (29.4) | 210 (33.3) |
| Number of prior lines of chemotherapy in the metastatic setting, n (%) | ||||
| 0 | 90 (100.0) | 151 (69.9) | 135 (45.6) | 85 (13.5) |
| 1 | 0 | 65 (30.1) | 133 (44.9) | 301 (47.8) |
| 2 | 0 | 0 | 28 (9.5) | 159 (25.2) |
| ≥3 | 0 | 0 | 0 | 85 (13.5) |
| Number of prior lines of endocrine therapy in the metastatic setting, n (%) | ||||
| 0 | 90 (100.0) | 74 (34.3) | 37 (12.5) | 16 (2.5) |
| 1 | 0 | 142 (65.7) | 136 (45.9) | 116 (18.4) |
| 2 | 0 | 0 | 123 (41.6) | 226 (35.9) |
| ≥3 | 0 | 0 | 0 | 272 (43.2) |
| 12-month event-free rate for rwTTNT (95% CI) | 0.33 (0.23, 0.48) | 0.37 (0.30, 0.46) | 0.33 (0.27, 0.40) | 0.27 (0.23, 0.32) |
| 12-month event-free rate for rwTTD (95% CI) | 0.34 (0.23, 0.48) | 0.35 (0.27, 0.43) | 0.31 (0.25, 0.38) | 0.27 (0.23, 0.31) |
*A small number of missing values are not shown; †in the database, 75% of the data were from the community and 25% from academic settings
Presentation numberPS1-11-01
Impact of prior CDK 4/6 inhibitor treatment on the efficacy of everolimus and exemestane or fulvestrant in hormone receptor positive HER 2 negative (HR+/HER2-) metastatic breast cancer (MBC): a restrospective analysis of 144 patients.
Maria Otaño, Hospital Universitario Donostia- OSI Donostialdea, Donostia, Spain
M. Otaño, A. Paisan, C. Fernandez, A. Lahuerta, N. Ancizar, A. Urruticoechea, I. Alvarez Lopez; Medical Oncology, Hospital Universitario Donostia- OSI Donostialdea, Donostia, SPAIN.
INTRODUCTION HR+/HER2- MBC remains one of the leading causes of cancer related mortality in woman worldwide. Standard treatment includes endocrine therapy and targeted agents such as everolimus, an mTOR inhibitor. The impact of prior CKD4/6 inhibitor treatment on the efficacy of everolimus is not well defined. This study evaluates the effect of prior CDK4/6 exposure on survival outcomes in HR+HER2- MBC treated with this combination. OBJETIVES To compare progression-free survival (PFS) and overall survival (OS) according to prior CKD4/6 inhibitor exposure in patients treated with everolimus and exemestane or fulvestrant. METHODS Retrospective analysis of 144 women with HR+/HER2- MBC treated with the combination of everolimus plus exemestane or fulvestrant between October 2010 and October 2023. Two cohorts were compared: cohort 1 without previos CDK 4/6 inhibitor and cohort 2 with previous CKD4/6 inhibitor (palbociclib or ribociclib). The analysis was conducted on June 2025. RESULTS A total of 144 women were included. The median follow up was 31.3 month (2.23-103). 11 patient were on treatment on the time of analysis. Table 1 highlights baseline characteristic. Median PFS was 7.4 months in the overall population, 8.2 month in cohort 1 and 6.1 months in cohort 2. The adjusted HR for previous treatment lines was 1.14 for previous CKD4/6 treatment (p 0.474). The HR for progression in patient with previous CDK 4/6 was 1.7 (p=0.119) independent of previous line duration. Median OS from the start of everolimus was 37 months in the overall population, 40.7 month in cohort 1 and 35.9 months in cohort 2. The adjusted HR for previous treatment lines was 1.1 (p= 0.6) for previous CKD4/6 treatment. Everolimus was discontinued due to toxicity in 27% of patients; 33% required dose reduction. Main toxicitys included mucositis (44& any grade), pneumonitis( >G2 17%) and hyperglycemia (>G2 3%). Table 1
| Patient Characteristics | Overall population N= 144 | No prior CDK4/6 N= 51 | Prior CDK 4/6 n= 93 | ||||
| Age | 58.7 (27-80.8) | 59.3 (27-77) | 58.3 (34.3-80.8) | ||||
| Postmenopause | 111 (77%) | 42 (82%) | 69 (74%) | ||||
| Stage IV at diagnosis | 49 (34%) | 13 (25.4%) | 36 (38.7%) | ||||
| Previous endocrine lines on metastatic setting | |||||||
| 1 | 87 (53.4%) | 19 (37.2%) | 68 (73%) | ||||
| 2 | 41 (28.4%) | 22 (43.13%) | 19 (20.4%) | ||||
| 3 | 13 (9%) | 7 (13.7%) | 6 (6.6%) | ||||
| 4 | 3 (2%) | 3 (5.8%) | 0 (0%) | ||||
| Previous chemotherapy on metastatic setting | |||||||
| Yes | 39 (27%) | 22 (43.1%) | 17 (18.2%) | ||||
| No | 105 (72.9%) | 29 (56.8%) | 76 (81.7%) | ||||
| Last treatment line duration | 17.5 months(0.92-74.7) | 13.3 months (0.9-38.2) | 19.7 months (1.6-74.7) | ||||
| Everolimus concomitant endocrine treatment | |||||||
| Fulvestrant | 44 (30.5%) | 1 (2%) | 43 (46.2%) | ||||
| Exemestane | 100 (69.4%) | 50 (98%) | 50 (53.7%) |
CONCLUSION To our knowledge, this is the second largest reported series to date supporting the prior hypothesis that prior CDK4/6 inhibitor treatment is associated with shorter PFS and OS in HR+/HER2- MBC patients treated with everolimus and exemestane. These findings highlight the need to optimize post CDK4/6 treatment strategies. BIBLIOGRAPHY 1. Baselga J et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012 Feb 9;366(6):520-9.2.Mo H et al. Real-World Outcomes of Everolimus and Exemestane for the Treatment of Metastatic Hormone Receptor-Positive Breast Cancer in Patients Previously Treated With CDK4/6 Inhibitors. Clin Breast Cancer. 2022 Feb;22(2):143-148.
Presentation numberPS1-11-02
The ADELA study: a double-blind, placebo-controlled, randomized phase 3 trial of elacestrant + everolimus versus elacestrant + placebo in ER+/HER2- advanced breast cancer (ABC) patients with ESR1-mutated tumors progressing on endocrine therapy (ET) + CDK4/6i
Antonio Llombart-Cussac, Hospital Arnau de Vilanova; Translational Oncology Group, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU; Medica Scientia Innovation Research (MEDSIR), Valencia, Spain
A. Llombart-Cussac1, J. M. Pérez-García2, E. Lopez-Miranda3, C. Saavedra4, V. Carañana5, I. Blancas6, C. Hinojo7, A. Cortes-Salgado8, E. Galve-Calvo9, R. R. Zhang10, M. Sampayo-Cordero11, D. Alcalá-López11, J. Carvalho-Santos11, O. Boix11, A. Garrido12, C. H. Barrios13, G. Curigliano14, R. Bartsch15, A. Hardy-Bessard16, T. Wasserman17, J. Cortés18; 1Department of Medicine, Hospital Arnau de Vilanova; Translational Oncology Group, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU; Medica Scientia Innovation Research (MEDSIR), Valencia, SPAIN, 2Department of Medicine, International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 3Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 4Department of Medicine, IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana; Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, SPAIN, 5Department of Medicine, Hospital Arnau de Vilanova, Valencia, SPAIN, 6Medicine department, San Cecilio Clinic University Hospital; Medicine department, Granada University; Instituto de Investigación Biosanitaria de Granada (ibs.Granada), Granada, SPAIN, 7Department of Medicine, Hospital Universitario Marqués de Valdecilla – Instituto de Investigación Valdecilla (IDIVAL), Santander, SPAIN, 8Medical Oncology Department, Hospital Universitario Ramón y Cajal (IRYCIS), Madrid, SPAIN, 9Department of Medicine, Hospital Universitario de Basurto, Bilbao, SPAIN, 10Department of Business, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 11Scientific department, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 12Department of Trial, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 13Department of Medicine, Latin American Cooperative Oncology Group (LACOG) – Oncoclínicas Group, Porto Alegre, BRAZIL, 14Department of Medicine, European Institute of Oncology, IRCCS; University of Milano, Milan, ITALY, 15Department of Medicine 1, Medical University of Vienna, Division of Oncology, Vienna, AUSTRIA, 16Department of Medicine, Centre Armoricain d’Oncologie, CARIO-HPCA; UNICANCER, Plérin, FRANCE, 17Global Medical Affairs – Solid Tumors, Menarini Group, New York, NY, 18Department of Medicine, International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group; IOB Madrid; Hospital Beata María Ana; Universidad Europea de Madrid; Hospital Universitario Torrejón, Ribera Group; Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN.
Background: ET+CDK4/6i is the standard-of-care (SOC) in 1L ER+/HER2- ABC; however, tumors eventually develop resistance. Constitutive activation of the PI3K/AKT/mTOR pathway can contribute to endocrine resistance in breast cancer. ESR1 mutations are a common type of acquired resistance that emerges in 40-50% of patients in the metastatic setting after prolonged aromatase inhibitor exposure. There is an unmet need for novel therapeutic approaches to overcome resistance mechanisms and improve outcomes in patients with ER+/HER2- ABC with ESR1-mutated tumors progressing after ET+CDK4/6i. Elacestrant is a next-generation oral SERD that binds to ER-alpha, inducing its degradation. In the EMERALD study, single-agent elacestrant improved PFS vs SOC ET in patients with ESR1-mutated tumors (HR 0.55; 95% CI 0.39-0.77; P=0.0005) [Bidard 2022]. Differences were notable among patients who received prior ET+CDK4/6i ≥12 months; median PFS with elacestrant was 8.6 vs 1.9 months with SOC ET (HR 0.41; 95% CI 0.26-0.63) [Bardia 2024]. The crosstalk between the ER and PI3K/AKT/mTOR pathways provides a rationale for evaluating elacestrant+everolimus (a mTORC1 inhibitor). In the ELEVATE phase 1b trial (NCT05563220), the combination of elacestrant and everolimus demonstrated a median PFS of 8.3 months (95% CI, 3.9 to NR) in all patients (N=23) with ER+/HER2- ABC who progressed after ET+CDK4/6i, regardless of ESR1-mutation status (Rugo, ASCO 2025); elacestant 345 mg + everolimus 7.5 mg was identified as the RP2D [Rugo ESMO 2024]. Safety was consistent with the known profile of everolimus+SOC ET. The ADELA study compares elacestrant+everolimus vs elacestrant+placebo in ER+/HER2- ABC patients with ESR1-mutated tumors progressing on ET+CDK4/6i. Design and Methods: ADELA (NCT06382948) is an international, multicenter, double-blind, placebo-controlled phase 3 trial. Eligible patients are adults (≥18 years) with ER+/HER2- ABC and centrally confirmed ESR1 mutations, previously treated with 1-2 lines of ET for ABC, and evidence of disease progression on prior ET+CDK4/6i for ABC after ≥6 months. Patients receiving CDK4/6i-based adjuvant therapy are eligible provided that disease progression is confirmed after ≥12 months of treatment but <12 months following CDK4/6i completion. Other criteria include adequate organ function, ECOG PS 0-1 and no prior use of elacestrant or other SERDs, PROTAC, CERAN, or novel SERM, and/or PI3K/AKT/mTOR inhibitors. Exclusion criteria include prior chemotherapy for ABC and active uncontrolled/symptomatic brain metastases and/or leptomeningeal disease. Patients will be randomized 1:1 to 28-day cycles of oral elacestrant 345 mg + everolimus 7.5 mg QD or elacestrant 345 mg + placebo QD until disease progression or unacceptable toxicity. Patients will receive dexamethasone mouthwash during the first 8 weeks. Stratification factors are presence of visceral metastases (yes vs no) and duration of prior CDK4/6i therapy (≥12 vs <12 months). The primary objective is to evaluate PFS assessed by blinded independent review committee. Secondary endpoints include investigator-assessed PFS, OS, ORR, CBR, DoR, TTR, best percentage change in tumor burden, safety, and HRQoL. Status: Planned enrollment is 240 patients. Rrecruitment is ongoing across Spain, France, Greece, Italy, Germany, Austria, Czech Republic, United Kingdom, and Brazil.
Presentation numberPS1-11-03
Real-world treatment patterns and overall survival in patients with HR+/HER2- metastatic breast cancer treated with chemotherapy following endocrine therapy in the US
Tiffany A. Traina, Memorial Sloan Kettering Cancer Center, New York, NY
T. A. Traina1, C. Rossi2, M. Levesque-Leroux2, A. Tardif-Samson2, P. Gagnon-Sanschagrin2, A. Guérin2, S. Vlassak3, V. Guan4, J. Salcedo4; 1Department of Medicine, Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY, 2-, Analysis Group ULC, Montréal, QC, CANADA, 3Global Medical Affairs, BioNTech SE, Mainz, GERMANY, 4Health Economics & Outcomes Research, BioNTech US Inc, Cambridge, MA.
Background: Current therapeutic approaches for patients with HR+/HER2-low metastatic breast cancer (mBC) do not differ from patients with HR+/HER2- mBC, with systemic chemotherapy remaining the standard of care (SOC) following progression on multiple lines of endocrine therapy (ET)-based treatment or in cases of primary endocrine resistance. Given the scarcity of real-world data on treatment patterns and real-world overall survival (rwOS) in this population, further research is essential to inform decision making and better understand the effectiveness of SOC treatments. Methods: A cohort of adults (≥18 years old) treated with chemotherapy following ET for HR+/HER2- mBC in the United States (US) was identified retrospectively in the Komodo Research Database (1/1/2016-4/30/2024). A 12-month washout period was used to identify patients with newly diagnosed mBC based on the presence of ≥2 medical claims with diagnosis codes for the primary BC followed by ≥2 claims with diagnosis codes for secondary neoplasms, excluding breast, skin, or lymph nodes. HR+/HER2- statuses were inferred from treatments received based on expert’s input. As such, HER2 expression profile below the threshold for positivity was not further stratified. Patients were included if they initiated chemotherapy after ≥2 prior lines of ET-based treatment (with or without targeted agents) or within 6 months of starting first-line ET + CDK4/6i (primary endocrine resistance) in the mBC setting. Treatment sequences in the mBC setting were described. rwOS was assessed from chemotherapy initiation to follow-up end among patients who initiated chemotherapy between 1/1/2017-10/31/2022 (i.e., had ≥18 months of potential observation, based on expert clinical input) using the Kaplan-Meier method. Results: The study identified 1,371 patients receiving chemotherapy for HR+/HER2- mBC. Median age at chemotherapy initiation was 59 years, 98.0% were female, 71.3% were White, 12.6% were Black or African American, 9.2% were Hispanic or Latino, and 3.8% were Asian or Pacific Islander. Most patients were commercially insured (67.8%), followed by Medicare (24.4%) and Medicaid (7.8%). The most common comorbidities were liver disease (30.3%), diabetes (21.4%), cardiovascular disease (20.6%) and chronic obstructive pulmonary disease (15.8%). Median time from initiation of mBC treatment to follow-up end was 31.6 months. Among the 1,371 patients, 25.1% initiated chemotherapy after one line, 49.1% after two lines, and 25.8% after ≥3 lines of mBC treatment. Most common regimens at chemotherapy initiation were capecitabine-based (60.2%), paclitaxel-based (17.3%) and albumin-bound paclitaxel-based (10.4%). Most patients (53.6%) received a next line of therapy, 37.7% remained on initial chemotherapy, and 8.7% discontinued all treatments after chemotherapy initiation. The most common next regimens included additional chemotherapy (49.1%), ET + other targeted therapy (excluding CDK4/6i; 10.9%), and ET + CDK4/6i (4.1%). Among 996 patients with sufficient potential observation to assess rwOS (≥18 months), median rwOS was 18.9 months (95% confidence interval [CI]: 17.4, 20.0 months), and rwOS rates at 12, 24, and 36 months were 65.0% (95% CI: 61.9%, 67.8%), 40.5% (95% CI: 37.4%, 43.6%) and 24.1% (95% CI: 21.2%, 27.1%), respectively. Conclusions: Real-world outcomes for patients with HR+/HER2- mBC treated with chemotherapy following progression on multiple lines of ET-based treatment or with primary endocrine resistance are poor, with median rwOS of approximately 1.5 years. These results highlight a need for more effective therapies with tolerable safety profiles to improve outcomes among patients with HR+ and HER2- or HER2-low mBC expression profiles who progress on ET-based treatments.
Presentation numberPS1-11-04
Her2-low expression is associated with inferior progression-free survival in hr-positive metastatic breast cancer treated with CDK4/6 inhibitors
Seyda Gunduz, Koç University School of Medicine, Istanbul, Turkey
F. Kemik1, B. Koylu1, C. I. Kikili1, N. Demir1, B. Guney2, A. K. Guren3, E. Karanci4, D. Tunali1, S. Lacin1, D. Tural1, R. U. Gursu2, O. Kostek3, D. Kivrak Salim4, F. Selcukbiricik1, S. Gunduz1; 1Department of Medical Oncology, Koç University School of Medicine, Istanbul, TURKEY, 2Medical Oncology, Istanbul Training and Research Hospital, Istanbul, TURKEY, 3Department of Medical Oncology, Marmara University School of Medicine, Istanbul, TURKEY, 4Department of Medical Oncology, Antalya Training and Research Hospital, Antalya, TURKEY.
Background: CDK4/6 inhibitors have become a standard treatment option for patients with hormone receptor-positive (HR+), HER2-negative metastatic breast cancer. However, there remains a need for reliable biomarkers to predict treatment response. Recently, tumors classified as HER2-low have emerged as a distinct subgroup, exhibiting unique biological characteristics compared to traditional HER2-negative disease. This study aimed to investigate the impact of HER2 immunohistochemical expression levels on progression-free survival (PFS) in HR+ metastatic breast cancer patients treated with CDK4/6 inhibitors. Methods: This multicenter, retrospective study aimed to evaluate the impact of HER2 immunohistochemical (IHC) expression on PFS in patients with HR+, HER2-negative metastatic breast cancer treated with CDK4/6 inhibitors (ribociclib or palbociclib). Patients were classified as HER2-negative (IHC 0) or HER2-low (IHC 1+ or 2+/ISH−) based on IHC scores. The primary endpoint was PFS, defined as the time from initiation of CDK4/6 therapy to radiological or clinical progression or death. Survival outcomes were analyzed using the Kaplan-Meier method and compared with the log-rank test. Factors associated with PFS were further evaluated using univariate and multivariate Cox regression analyses. Results: A total of 344 patients were included in the study. The mean age was 58.7 ± 12.3 years, and 70.6% of the patients were postmenopausal. The median follow-up duration was 20.0 months (IQR: 11.4-34.4). The progression rate was 38.3% in HER2-negative patients (IHC 0) and 52.5% in HER2-low patients (IHC 1+ or 2+/ISH−). According to Kaplan-Meier analysis, the median progression-free survival (PFS) was 36.4 months (95% CI: 27.1-45.7) in the HER2-negative group and 20.4 months (95% CI: 8.5-32.2) in the HER2-low group (log-rank p = 0.028). In univariate analysis, shorter PFS was significantly associated with high tumor grade (G3), PR-negativity, HER2-low expression, presence of visceral metastasis, and involvement of multiple metastatic sites at treatment initiation. In multivariate analysis, HER2-low expression (p = 0.041), visceral metastasis (p = 0.001), and PR-negativity (p = 0.003) were identified as independent predictors of inferior PFS. Conclusion: In HR+ metastatic breast cancer patients treated with CDK4/6 inhibitors, HER2-low expression was associated with shorter progression-free survival compared to HER2-negative disease. These findings suggest that HER2-low tumors may represent a biologically distinct subgroup with prognostic implications. HER2 expression levels should be considered when assessing prognosis, and further prospective studies are warranted to validate these observations.
|
Variable |
Category | n (%) |
| Age Group | <65 / ≥65 | 231 (67.2) / 113 (32.8) |
| Menopausal Status | Premenopausal / Postmenopausal | 101 (29.4) / 242 (70.6) |
| ECOG Performance Status | 0 / 1 / ≥2 | 318 (92.4) / 22 (6.4) / 4 (1.2) |
| Comorbidities | Yes / No | 161 (46.8) / 183 (53.2) |
| Histological Subtype | IDC / ILC / Other and missing | 263 (76.4) / 47 (13.7) / 34 (9.9) |
| Histologic Grade | Grade 2 / Grade 3/ missing | 167 (48.6) / 70 (20.3) / 107 (31.1) |
| ER Expression | ≥90% / <90% | 262 (76.2) / 82 (23.8) |
| PR Status | Positive / Negative | 292 (84.9) / 52 (15.1) |
| HER2 IHC Score | 0 / 1+ / 2+ | 242 (70.3) / 56 (16.3) / 46 (13.4) |
| Ki-67 Proliferation Index | <20% / ≥20% / missing | 136 (39.5) / 191 (55.5) / 17 (4.9) |
| Number of Metastatic Sites | 1 / ≥2 / missing | 211 (61.3) / 101 (29.4) / 32 (9.3) |
| Visceral Metastasis | Yes / No | 207 (60.2) / 137 (39.8) |
| Type of Metastasis | De novo / Recurrent | 209 (60.8) / 135 (39.2) |
| CDK 4/6 Agent | Ribociclib / Palbociclib | 271 (78.8) / 73 (21.2) |
Presentation numberPS1-11-05
Phase II trial of pembrolizumab in combination with paclitaxel in the hormone receptor-positive metastatic breast cancer enriched with tumor mutational burden determined by whole exome sequencing: Korean Cancer Study Group trial (KCSG BR20-16)
Joohyuk Sohn, Yonsei University College of Medicine, Seoul, Korea, Republic of
J. Sohn1, Y. Moon2, J. Lee3, J. Kim4, K. Lee5, M. Ahn6, H. Ahn7, Y. Park8, S. Kim9, A. Kim9, E. Kim9, Y. Yang9, Y. Cha10, J. Byun11, K. Lee12, J. Kim13, H. Kim14, I. Woo15, S. Koh16, K. Lee17, M. Lee18, J. Jung19, Y. Chae20, G. Kim1, M. Kim1, K. Kim1; 1Division of Medical Oncology and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Division of Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center CHA University, Seongnam, KOREA, REPUBLIC OF, 3Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, KOREA, REPUBLIC OF, 4Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 5Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 6Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, KOREA, REPUBLIC OF, 7Division of Medical Oncology and Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, KOREA, REPUBLIC OF, 8Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF, 9Department of Biomedical Systems Informatics and Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 10Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 11Division of Medical Oncology, Department of Internal Medicine, College of Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea, Incheon, KOREA, REPUBLIC OF, 12Center for Breast Cancer, National Cancer Center, Goyang, KOREA, REPUBLIC OF, 13Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, KOREA, REPUBLIC OF, 14Division of Hematology and Oncology, Department of Internal Medicine, Soonchunhyang University Hospital, Cheonan, KOREA, REPUBLIC OF, 15Division of Medical Oncology, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, KOREA, REPUBLIC OF, 16Division of Hematology-Oncology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, KOREA, REPUBLIC OF, 17Department of Hematology and Oncology, Ewha Womans University Hospital, Seoul, KOREA, REPUBLIC OF, 18Department of Internal Medicine, Inha University School of Medicine, Incheon, KOREA, REPUBLIC OF, 19Department of Internal Medicine, Hallym University Medical Center, Dongtan Sacred Heart Hospital, Hwaseong, KOREA, REPUBLIC OF, 20Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF.
Background: In hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC), chemotherapy is considered for patients resistant to endocrine therapies including CDK4/6 inhibitors (CDK4/6i) or those with visceral crisis. Pembrolizumab, a PD-1 inhibitor, has not shown its efficacy in unselected HR+/HER2- MBC patient population. This study evaluated pembrolizumab in combination with weekly paclitaxel as first-line chemotherapy in HR+/HER2- MBC patients enriched with tumor mutational burden (TMB), assessed as an integral biomarker. Methods: In this multicenter, single-arm, phase II trial, patients with HR+/HER2- MBC considered resistant to endocrine therapy were prescreened to determine TMB. Fresh tissue was procured through tumor biopsy and was sent to central laboratory for whole-exome sequencing (WES). TMB was calculated as the number of high-confidence nonsynonymous somatic single nucleotide variants detected by MuTect2(GATK v3.5) from WES of tumor-normal pairs. Inclusion criteria for TMB counts were initially defined as ≥70 nonsynonymous mutations for the first 30 prescreened patients and subsequently redefined upper 20% counts with every 30 patients by that time. Prior chemotherapy in the metastatic setting was not allowed. Patients received pembrolizumab (200 mg every 3 weeks) plus paclitaxel (80 mg/m² on days 1, 8, and 15 of a 28-day cycle) until disease progression or unacceptable toxicity. The primary endpoint was investigator-assessed objective response rate (ORR) per RECIST v1.1. Secondary endpoints included progression-free survival (PFS), overall survival (OS), duration of response (DoR), and safety. Results: Between March 2022 and December 2024, 105 patients were prescreened across 18 centers. The trial was terminated early due to slow recruitment after enrollment of 23 patients. The median age was 59 years (range, 32-74). All patients had prior exposure to CDK4/6 inhibitors, and approximately half (n=11, 47.8%) of patients had received two lines of endocrine therapy before enrollment. The investigator-assessed ORR was 65.2% (95% CI, 42.7-83.6%). At a median follow-up of 13.5 months, median PFS (mPFS) was 7.5 months (95% CI, 6.0-11.6), and median DoR was 4.3 months (95% CI, 3.9-not reached). Patients who experienced any grade of immune-related adverse events (irAEs) (n=9) demonstrated significantly longer mPFS compared to those without irAEs (9.1 vs. 5.8 months; HR=0.25; 95% CI, 0.08-0.77; P=0.016). An increase in TMB was positively correlated with the extent of tumor reduction, suggesting that patients with higher TMB experienced more pronounced tumor shrinkage by RECIST (R=-0.42, P=0.048). Median overall survival (OS) was not reached at the time of data cutoff. In terms of adverse events (AEs), peripheral neuropathy was the most frequent non-hematologic AE (n=15, 65.2%), and hypothyroidism was the most common immune-related AE (n=3, 13%). One patient discontinued treatment due to grade 3 irAE. Conclusion: Integration of biopsy followed by TMB determination through bioinformatic process as a biomarker-integral trial was feasible. Pembrolizumab in combination with weekly paclitaxel demonstrated promising clinical activity and manageable safety as first-line chemotherapy in TMB-enriched HR+/HER2- MBC patients.
Presentation numberPS1-11-06
Impact of sarcopenia on efficacy and toxicity profile of CDK4/6 inhibitors in HR+/HER2- metastatic breast cancer patients: a real-world analysis
Denise Drittone, Sant’Andrea University Hospital, Rome, Italy
D. Drittone1, M. Zerunian2, M. Mariniello1, C. Lucci1, L. Esposito1, N. Spiezia1, M. Delle Chiaie1, F. Schipilliti1, G. Arrivi1, M. Francone2, F. Mazzuca1, S. Pisegna1; 1Medical Oncology, Sant’Andrea University Hospital, Rome, ITALY, 2Radiology Unit – Department Of Surgical-Medical Sciences and Translational Medicine, Sant’Andrea University Hospital, Rome, ITALY.
Background: CDK4/6 inhibitors (CDK4/6i) combined with endocrine therapy (ET) are standard first-line treatment in HR+/HER2- metastatic breast cancer (mBC). Sarcopenia is prevalent in cancer patients and linked to poorer outcomes and higher toxicity, but its role in CDK4/6i-treated patients remains under-investigated. This real-world retrospective study evaluates sarcopenia’s impact on survival outcomes (PFS/OS) and toxicity in mBC patients receiving CDK4/6i. Methods: Retrospective analysis of clinical and radiological data from patients (pts) with histologically confirmed HR+/HER2- stage IV mBC at Sant’Andrea University Hospital was performed. All pts had de novo metastatic or relapsed disease treated with first-line CDK4/6i plus ET. Sarcopenia was assessed using baseline CT scans with third lumbar vertebra (L3) as reference. Body composition analysis used AI-based automatic segmentation software (DeepCatch). Skeletal muscle index (SMI) cutoff <38.5 cm²/m² classified pts as sarcopenic. Statistical analyses included univariate/multivariate Cox proportional hazards models and Kaplan-Meier survival curves. Results: The cohort included 75 mBC pts (median age 58 years; 53 postmenopausal, 22 premenopausal). BMI distribution: 1 underweight, 51 normal weight, 14 overweight, 9 obese. 33 pts received palbociclib, 40 ribociclib, 2 abemaciclib. ET included aromatase inhibitors (n=41) or fulvestrant (n=34). Any grade toxicity occurred in 34 pts (45%), with G3/4 events in 23 cases (31%). Best response was distributed as following: 3 CR, 29 PR, 31 SD, 12 PD. Overall, 42 pts (56%) were sarcopenic. Seventeen pts (23%) had bone-only disease, 58 (77%) visceral metastases. No significant difference in surivival outcomes was observed: mPFS 27 vs 23 months , mOS 51 vs 47 months in sarcopenic vs non-sarcopenic pts (HR=0.79, p=0.47, HR=0.672, p=0.28), respectively. Clinically relevant sarcopenia prevalence difference was observed between ET types: 56% with fulvestrant vs 34% with aromatase inhibitors (p=0.05). Significant associations were identified between sarcopenia and severe toxicity (G3/4): 61% of patients with severe toxicity were sarcopenic vs 37% with mild-moderate toxicity (p=0.05). While BMI categories showed no significant correlation with sarcopenia (p=0.08), BMI as continuous variable demonstrated superior discriminatory ability, showing significant protective effect (OR=0.798, 95% CI: 0.668-0.955, p=0.01). Each BMI unit increase associated with 20% sarcopenia odds reduction, with sarcopenic patients presenting lower mean BMI (23.0 vs 25.8 kg/m², p=0.003). No significant associations were found between sarcopenia and CDK4/6i type, age, menopausal status, or disease location (p>0.05). Multivariate analysis confirmed BMI as continuous variable (p=0.01) and severe toxicity grading (p=0.046, OR=3.118) as independent sarcopenia predictors. Conclusions: In mBC patients treated with first-line CDK4/6i plus ET, BMI and high-grade toxicity emerged as independent sarcopenia predictors. However, sarcopenia did not represent a negative prognostic factor for PFS and OS in our analysis. The significant correlation between sarcopenia and severe toxicity suggests need for nutritional screening and closer monitoring of sarcopenic patients to optimize treatment tolerability. These findings support implementing body composition assessment in clinical practice for patients receiving CDK4/6i plus ET. Further investigation is warranted to validate and expand these results.
Presentation numberPS1-11-07
First-in-human, phase 1a/b study of GDC-4198 (RGT-419B), a next generation CDK4/2 inhibitor, in patients with hormone receptor positive HER2- locally advanced/metastatic breast cancer (LA/mBC) who progressed on prior CDK4/6 inhibitors
Seth A Wander, Massachusetts General Hospital, Harvard Medical School, Boston, MA
S. A. Wander1, R. Zuniga2, H. Han3, S. Sadeghi4, R. Shatsky5, K. Clifton6, M. Dhawan7, J. Xie8, R. Xin8, K. Kalinsky9; 1Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 2Medical Oncology & Hematology, New York Cancer & Blood Specialists, New York City, NY, 3Medical Oncology, Moffitt Cancer Center, Tampa, FL, 4Hematology Oncology, University of California at Los Angeles, Santa Monica, CA, 5Medical Oncology, University of California San Diego, La Jolla, CA, 6Oncology, Washington University School of Medicine, St. Louis, MO, 7Oncology, Genentech, Inc., South San Francisco, CA, 8Oncology & Immunology, Regor Pharmaceuticals Inc., Cambridge, MA, 9Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA.
Introduction: GDC-4198 (also known as RGT-419B) is a CDK4/2 inhibitor, acting as an oral, highly potent small-molecule inhibitor of CDK4 with substantial activity against CDK2. Here we present interim data from a phase 1, first-in-human, multicenter trial of GDC-4198 as a monotherapy or in combination with endocrine therapy (ET) in patients with hormone receptor positive (HR+) human epidermal growth factor receptor 2 negative (HER2-) LA/mBC. Methods: Eligible patients received treatment for LA/mBC including ET and CDK4/6i. GDC-4198 monotherapy was administered orally in continuous 28-day cycles across four cohorts (25, 75, 150 mg once daily [QD], and 150 mg twice daily [BID]) using a standard 3+3 dose escalation design to identify the maximum tolerated dose (MTD) and recommended dose for expansion (RDE). Patients in the combination therapy cohort received GDC-4198 150 mg BID + aromatase inhibitor (AI). The primary objective was to assess safety and tolerability; secondary objectives included exploratory efficacy. Results: As of May 2025, 32 patients (28 monotherapy; 4 in combination with AI) received GDC-4198. The median age was 62 years (range 49-80). The median number of previous lines of therapy was 3 (range 1, 7). All patients received prior CDK4/6i and ET (100%). The most common (occurring in ≥10% of patients) treatment-emergent adverse events (TEAEs; all causality) were nausea, diarrhea, vomiting, white blood cell count decrease, neutrophil count decrease, fatigue, and headache. The incidence of grade ≥3 treatment-related adverse events (TRAEs) was low (16%). No patients have discontinued GDC-4198 due to TRAEs. There were no treatment-related SAEs. The MTD has not been reached. In patients with measurable disease and at least 1 post-baseline tumor assessment (n=24), 4 patients had confirmed partial responses in the monotherapy cohorts (1 at 150 mg QD; 3 at 150 mg BID). Conclusions: GDC-4198, with potent and selective activity against CDK4 and CDK2, was well-tolerated both as a single agent and in combination with AI. GDC-4198 demonstrated preliminary evidence of efficacy in a post-CDK4/6i setting. Further dose escalation with GDC-4198 as a single agent is ongoing and updated data will be presented at the meeting.
Presentation numberPS1-11-08
Integrative modeling of multimodal real-world data for improved risk stratification of first-line CDK4/6 inhibitor outcomes in patients with estrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer
Francisco Sanchez-Vega, Memorial Sloan Kettering Cancer Center, New York, NY
F. Sanchez-Vega1, S. Nandakumar 1, G. Long2, W. Chatila1, S. Sreenivasan 3, M. Carey4, M. Keddar2, J. Davies5, M. Zatzman1, M. Miller2, E. de Bruin 6, S. Khosla2, N. Ceglia1, H. Lees1, F. Nagib1, Y. Gong1, M. Donoghue1, N. Schultz1, S. Shah1; 1-, Memorial Sloan Kettering Cancer Center, New York, NY, 2Clinical Data Sciences, AstraZeneca, Cambridge, UNITED KINGDOM, 3Clinical Data Sciences, AstraZeneca, Gaithersburg, MD, 4-, Tempus AI Inc, Chicago, IL, 5Oncology Data Science, AstraZeneca, Barcelona, SPAIN, 6Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM.
Background: CDK4/6 inhibitor in combination with endocrine therapy is a cornerstone of treatment for hormone receptor-positive metastatic breast cancer. We developed a machine learning model that used clinical, genomic, and transcriptomic features to stratify patients based on response to first-line CDK4/6 inhibitor treatment and identify predictors of response. Methods: Patients with ER-positive/HER2-negative metastatic breast cancer, biopsy samples from before or <30 days after initiation of first-line treatment, curated clinical outcome data in a breast dataset from the Tempus real-world multimodal database, and data from targeted DNA sequencing (Tempus xT assay) and whole transcriptome sequencing (Tempus xR assay) were included. Driver DNA alterations associated with breast cancer were extracted at the gene and pathway levels. Cell type proportions and cell states were decomposed from bulk RNA sequencing data using EcoTyper. Genomic, transcriptomic, and clinicopathological features (age, tissue site, de novo disease, stage, early recurrence) and treatment characteristics (CDK4/6 inhibitor, endocrine therapy partner, adjuvant treatment) were used as explanatory variables in an integrative risk stratification model that used the OncoCast algorithm to predict real-world progression-free survival (rwPFS) in patients treated with first-line CDK4/6 inhibitors. Risk-set adjustment was used to mitigate the effect of patients’ delayed entry into the cohort due to the timing of their Tempus sequencing tests. Results: Overall, 251 patients were included. Patients were stratified into two risk groups (high-risk [median rwPFS 11.2 months, 95% confidence interval [CI]: 7.2-17.6]; low-risk [median rwPFS 18 months, 95% CI: 14.9-not available]; log-rank p<0.001). Features associated with the high-risk group included higher rates of TP53 alterations, >4 metastatic sites, and high expression of genes involved in unfolded protein response, cell cycle, metabolic, proliferative, and androgen response pathways. Features associated with the low-risk group included high expression of genes involved in early estrogen response pathway. Conclusion: Our study shows how multimodal real-world data collected during routine care can provide valuable insights into the biology of response to CDK4/6 inhibitor in patients with metastatic breast cancer and help improve patient stratification.
Presentation numberPS1-11-09
Real-world prevalence of ESR1 mutations (ESR1m) among patients with estrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC) after first-line (1L) treatment with endocrine therapy (ET) and/or a cyclin dependent kinase 4/6 inhibitor (CDK4/6i)
David Chandiwana, Arvinas Operations, Inc., New Haven, CT
D. Chandiwana1, D. Benjumea2, K. Greco1, C. Grace Rose3, S. Stergiopoulos3, J. Liao4, M. Edwards1; 1Medical Affairs, Arvinas Operations, Inc., New Haven, CT, 2Evidence Strategy, Genesis Research Group, Hoboken, NJ, 3HV&E (HTA, Value & Evidence), Pfizer, Inc., New York, NY, 4HEOR, Guardant Health, Palo Alto, CA.
Background: Patients (pts) with ER+/HER2- MBC often develop ESR1m during or after 1L treatment with ET with or without a CDK4/6i. Reported ESR1m prevalence rates vary, ranging from 20% to 50% after ≥1 line of therapy in the metastatic setting. Here, we explore the real-world prevalence of ESR1m and co-occurring mutations after 1L ET and/or CDK4/6i among pts with MBC in the GuardantINFORM database. Methods: This was a retrospective cohort study of adults in the United States with ER+/HER2- MBC and a Guardant360 comprehensive genomic profiling blood test of cancer-related gene mutations in circulating tumor DNA (ctDNA) between Jan 1, 2014 and Aug 31, 2024. Eligible pts had received any of the following 1L treatments within 6 months of diagnosis of metastatic disease: ET monotherapy (aromatase inhibitor [AI], selective ER modulator [SERM], or selective ER degrader [SERD]), CDK4/6i monotherapy, or ET (AI/SERM and/or SERD) + CDK4/6i combination therapy. Pts must have subsequently discontinued 1L therapy and received second-line (2L) treatment with follow-up >3 months. Pts were considered ESR1 evaluable if they had an ESR1 test ≤90 days before initiating 2L therapy (to rule out false negatives) or had a positive ESR1m result at any time before starting 2L therapy. ESR1m positivity was defined as missense mutations in codons 310-547. The proportion of ESR1-evaluable pts who were ESR1m positive was stratified by the type and duration of 1L treatment. The incidence of co-occurring AKT1 E17K, PIK3CA, PTEN, CCND1, ERBB2, FGFR1, KRAS, MYC, NF1, and RB1 mutations was analyzed. Results: Of 8335 adults with MBC treated with 1L ET and/or a CDK4/6i who had a Guardant 360 test, 4790 went on to receive 2L treatment with sufficient follow-up, and 1511 were ESR1 evaluable. Of these, 686 pts (45%) were ESR1m-positive, with the highest rates of ESR1m positivity among pts who received 1L SERD monotherapy or SERD + AI/SERM, and those who received 1L treatment for 12 to <24 months (Table). Among pts with ESR1m, 68% (466/686) had ≥1 other co-occurring mutation at start of 2L therapy, most commonly in genes involved in the PI3K/AKT signaling pathway (AKT1 E17K, PIK3CA, or PTEN; 50% [341/686]). Conclusions: In this real-world analysis of the GuardantINFORM database, 45% of pts with ER+/HER2- MBC were ESR1m-positive following 1L treatment with ET and/or CDK4/6i. The ESR1m positivity rate remained >40% in most subgroups analyzed according to 1L therapy type and 1L treatment duration. Other mutations co-occurred in >65% of pts with ESR1m.
| 1L Treatment | ESR1m Positive, % (n/N) | ||
| All | 45 (686/1511) | ||
| ET monotherapy | |||
| AI/SERM | 38 (153/400) | ||
| SERD | 52 (97/185) | ||
| ET + CDK4/6i | |||
| AI/SERM + CDK4/6i | 48 (242/500) | ||
| SERD + CDK4/6i | 42 (65/156) | ||
| CDK4/6i monotherapy | 43 (55/127) | ||
| SERD + AI/SERM | 52 (26/50) | ||
| SERD + AI/SERM + CDK4/6i | 52 (48/93) | ||
| Duration of 1L treatment | |||
| <6 months | 34 (157/468) | ||
| 6 to <12 months | 48 (140/289) | ||
| 12 to <24 months | 55 (208/379) | ||
| 24 to <36 months | 52 (105/201) | ||
| ≥36 months | 44 (76/174) |
aPts were considered ESR1 evaluable if they had an ESR1 test ≤90 days before initiating 2L therapy (to rule out false negatives) or had a positive ESR1m result at any time before starting 2L therapy.
Presentation numberPS1-11-10
Real-world progression-free survival 2 (rwPFS2) and tumor response with CDK4/6is + aromatase inhibitor (AI) in patients (pts) with HR+/HER2- metastatic breast cancer (MBC)
Hope S Rugo, City of Hope Comprehensive Cancer Center, Duarte, CA
H. S. Rugo1, A. Brufsky2, R. M. Layman3, X. Liu4, B. Li5, L. McRoy4, A. B. Cohen6, M. Estevez7, P. Cottu8, M. Thill9, G. Curigliano10; 1Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, 2Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, 3Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4US Oncology Medical Affairs, Pfizer Inc., New York, NY, 5Global Biometrics & Data Management, Pfizer Inc., New York, NY, 6Research Oncology, Flatiron Health Inc., New York, NY, 7Research Sciences, Flatiron Health Inc., New York, NY, 8Department of Oncology, Université Paris Cité, Paris, FRANCE, 9Department of Gynecology and Gynecological Oncology, Agaplesion Markus Krankenhaus, Frankfurt, GERMANY, 10Department of Oncology and Hemato-Oncology, University of Milano, European Institute of Oncology, IRCCS, Milan, ITALY.
Background CDK4/6is, including palbociclib (PAL), ribociclib (RIB), and abemaciclib (ABE), plus endocrine therapy, have been the standard of care for first-line (1L) HR+/HER2- MBC treatment. There have been no head-to-head randomized clinical trial data comparing CDK4/6is. The Palbociclib Verifying Evidence of Real-world Impact Study (P-VERIFY) is the largest real-world comparative effectiveness study to date (N = 9146), suggesting no significant differences in overall survival (OS) and progression-free survival (PFS) between the 3 CDK4/6is plus AI. This P-VERIFY data analysis compared rwPFS2 and real-world tumor response (rwTR) between 1L PAL, RIB, and ABE plus AI for HR+/HER2- MBC. Methods P-VERIFY is a retrospective Flatiron Health Research Database analysis of pts with HR+/HER2- MBC who initiated 1L CDK4/6i + AI between Feb 2015 and Nov 2023. All 9146 pts in P-VERIFY were included for rwPFS2 analysis, while 8010 (87.6%) had at least one tumor response assessment and were eligible for rwTR. rwPFS2 was defined as the time from 1L CDK4/6i + AI initiation (index date) to disease progression on 2L treatment or death from any cause, whichever came first. rwTR was defined as the change in burden of disease over the course of 1L index treatment based on the treating clinician’s assessment of radiologic evidence. Pts were followed until death, last medical activity, or May 31, 2024 (data cut-off date). Stabilized inverse probability treatment weighting (sIPTW) was used to balance pt characteristics. KM curves and Cox regression were used to compare rwPFS2. Logistic regression was used to compare rwTR. Results After sIPTW, baseline demographics and clinical characteristics were generally balanced between treatment groups. Median rwPFS2 (95% CI) was 35.8 (34.6−37.5) months for PAL + AI, 41.7 (37.5−46.2) months for RIB + AI, and 35.6 (32.2−45.5) months for ABE + AI. There were no significant rwPFS2 differences between RIB + AI vs PAL + AI [HR (95% CI) = 0.99 (0.89−1.10)], ABE + AI vs PAL + AI [HR = 0.95 (0.85−1.07)], and ABE + AI vs RIB + AI [HR = 0.97 (0.83−1.13)] (all P > 0.05). The best overall rwTR rates were 55.5% for PAL+AI, 57.4% for RIB+AI, and 57.4% for ABE+AI; these did not significantly differ between the 3 groups (OR = 1.08 [0.95−1.23] for RIB + AI vs PAL + AI; OR = 1.08 [0.94−1.25] for ABE + AI vs PAL + AI; and OR = 1.00 [0.84−1.20] for ABE + AI vs RIB + AI). Clinical benefit rates were similar among PAL + AI (85.8%), RIB + AI (86.3%), and ABE + AI (86.5%). Conclusions This large real-world comparative analysis suggests that rwPFS2 and rwTR do not significantly differ between 1L RIB + AI, ABE + AI, and PAL + AI in pts with HR+/HER2− MBC. These findings align with previous P-VERIFY analyses, which also suggested no significant differences in OS and PFS among the 3 CDK4/6is for this patient group.
Presentation numberPS1-11-11
Outcomes in patients with PIK3CA-mutated breast cancer treated with metformin
Catherine Travaline, Pennsylvania State University Hershey Medical Center, Hershey, PA
C. Travaline1, R. Plagens2, A. Elliott3, G. Sledge Jr.3, J. Hundal4, J. P. Leone5, S. L. Graff6, M. Lustberg7, M. Vasekar8; 1Hematology/Oncology, Pennsylvania State University Hershey Medical Center, Hershey, PA, 2Medical Affairs, Caris Life Sciences, Irving, TX, 3Medical Affairs, Caris Life Sciences, Phoenix, AZ, 4Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 5Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 6Lifespan Cancer Institute, Brown University Health, Providence, RI, 7Smilow Cancer Hospital, Yale University, New Haven, CT, 8Medical Oncology, Pennsylvania State University Hershey College of Medicine, Hershey, PA.
Background: Phosphoinositide 3-kinase (PI3K) inhibitors, including alpelisib (ALP), are commonly used to treat metastatic hormone receptor-positive (HR+) breast cancer (BC) with PIK3CA mutations (PIK3CAmut). However, hyperglycemia (HG) is a frequent and challenging side-effect. Metformin (MET) is a first-line treatment (Tx) for HG and has anti-neoplastic properties. We retrospectively reviewed tumor samples and associated clinical outcomes from real-world patients (pts) with HR+/HER2- BC treated with ALP and MET, focusing on prior vs concurrent Tx. Methods: A retrospective review was performed using metastatic BC samples that underwent next-gen sequencing of DNA (592-gene/whole exome) and immunohistochemistry (IHC) at Caris Life Sciences (Phoenix, AZ). IHC (intensity/% cells stained) was used to identify HER2- (≤1+ or ≤10%, or 2+/>10% and CISH-null), HR+ (ER+ (≥1+/≥1%) or PR+ (≥1+/≥1%)) samples. Pathogenic/likely pathogenic PIK3CAmut were identified in 3922 samples, of which 315 pts received ALP but not MET (“ALP alone”) and 198 pts received ALP + MET in various sequences, including MET prior to (but not concurrent with) ALP (n = 41) and MET concurrent with (but not prior to) ALP (n = 86). Endpoints, inferred from claims data, include overall survival (OS, date from first ALP Tx to last contact) and time on treatment (ToT, first to last ALP Tx). Hazards ratios (HR) and p-values were calculated using Cox proportional hazards models and log-rank tests, respectively. ICD10 codes for HG and type 2 diabetes mellitus (T2DM) were used to identify incidence of co-morbidities. Outcomes are reported as: median months (m); HR [95% CI]; p-value. Results: Compared with ALP alone, MET concurrent with ALP was associated with both longer OS (25.1 vs 13.6 m; HR = 0.61 [0.44-0.84]; p = 0.003) and longer ToT (6.0 vs 3.0 m; HR = 0.69 [0.53-0.90]; p = 0.006). However, MET prior to ALP had similar OS and ToT as ALP alone. An intermediate ALP+MET subgroup, MET prior to and concurrent with ALP (n=71), was associated with longer OS compared to ALP alone (22.6 m; HR = 0.59 [0.40-0.87]; p = 0.008), but similar ToT. HG was reported for 94 (29.75%) ALP alone and 60 (69.77%) MET concurrent with ALP pts. Among these, MET concurrent with ALP had significantly longer OS compared to ALP alone (26.1 vs 12.9 m; HR = 0.51 [0.33-0.80]; p = 0.003), as well as longer but not statistically significant ToT (5.7 vs 3.1 m; HR = 0.71 [0.49-1.03]; p=0.071). T2DM was reported for 101 (31.96%) ALP alone and 55 (63.95%) MET concurrent with ALP pts. Among these pts, MET concurrent with ALP was again associated with longer OS compared to ALP alone (28.7 vs 13.5 m; HR = 0.58 [0.37-0.92]; p = 0.0196), as well as longer but not statistically significant ToT (6.0 vs 3.2 m; HR = 0.72 [0.50-1.06]; p = 0.096). Conversely, MET prior to ALP had similar OS as ALP alone (14.3 m; HR = 1.23 [0.74-2.07]; p = 0.424), along with shorter though not statistically significant ToT (1.6 m; HR = 1.37 [0.80-2.35]; p = 0.25). Conclusions: Our finding that MET, when given concurrently with but not prior to ALP, was associated with improved outcomes suggests this therapy combination could have clinical benefits. Further research is warranted to examine this impact as well as any adverse effect profiles.
Presentation numberPS1-11-12
Rac/cdc42 inhibitor MBQ-167 in the estrogen receptor positive breast cancer tumor microenvironment
Ailed Cruz Collazo, University of Puerto Rico Medical Sciences Campus, San Juan, PR
A. Cruz Collazo1, N. Zaragoza-Rodriguez2, H. Fogle2, A. Torres-Sanchez1, N. Grafals1, V. Negron3, V. Carlo4, R. Papa5, S. Dorta-Estremera6, H. Franco2, S. Dharmawardhane1; 1Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, PR, 2Division of Clinical and Translational Cancer Research, UPR Comprehensive Cancer Center, San Juan, PR, 3Surgery, University of Puerto Rico Medical Sciences Campus, San Juan, PR, 4Pathology, Auxilio Mutuo Hospital Cancer Center, San Juan, PR, 5Biology, University of Puerto Rico Rio Piedras, San Juan, PR, 6Microbiology, University of Puerto Rico Medical Sciences Campus, San Juan, PR.
MBQ-167 is a dual Rac and Cdc42 inhibitor with significant anti-tumor growth and metastasis blocking activities in metastatic breast cancer cell and mouse models. MBQ-167 reduces both Rac and Cdc42 activation by GTP exchange and consequently, decreases phosphorylation of the common Rac and Cdc42 downstream effector p21-activated kinase (PAK). MBQ-167 is currently in Phase 1 clinical trials in advanced breast cancer patients, and so far, has shown safety and superior bioavailability up to 120 mg/kg BID (NCT06075810). Immune cell phenotyping from mouse tissue (tumor and spleen) and plasma from mice with tumors established from TNBC tumors demonstrated significant changes in myeloid cells and T lymphocytes indicating immune modulatory anticancer effects of MBQ-167 in the TME via inhibition of Rac and Cdc42. Immunohistochemistry (IHC) of breast cancer patient tissues demonstrate that Rac and Cdc42 are expressed in basal, HER2+ and (or) ER/PR+ breast cancers regardless of grade or type. However, the levels of their active downstream effector PAK, identified via IHC using a phospho (P) PAK antibody, is elevated in invasive breast cancer. Since the majority of breast cancers (~70%) are ER+, the objective in this study is to determine the effect of MBQ-167 in ER+ breast cancer, using an ex vivo drug testing platform. ER+ breast cancer patient tissues (>2 cm lesion) were collected immediately following surgical resection. Tissue slices (5 mm) were incubated in 0-1000 nM MBQ-167 for 24 h in ex vivo culture and fixed for H&E staining and IHC for PAK or p-PAK. A set of treated tissue slices were also snap frozen in liquid N2 and subjected to single cell RNA Seq to determine the effect of MBQ-167 on cellular heterogeneity in the TME. Results (N=5) show that when ER+ tissue slices following vehicle or 1000 nM MBQ-167 treatment were fixed and subjected to IHC using antibodies to p-PAKT423 or PAK, p-PAK levels were significantly reduced by ~70% without affecting total PAK levels. From the frozen samples treated with vehicle or 1000 nM MBQ-167 for 24 hrs, a total of 10 libraries was created for single cell RNA seq for 5 ER+ tumors. These data allowed us to systematically annotate the changes in gene expression in response to MBQ-167 in the different cell types within these tumor specimens. In conclusion, MBQ-167 reduces active p-PAK levels in ER+ breast cancer, and p-PAK levels can be used to demonstrate target engagement for the Rac/Cdc42 inhibitor MBQ-167 in ER+ breast cancer. These results inform the dynamics of Rac/Cdc42 inhibitor therapy in breast cancer patients and forward Phase 2 trials for this novel therapeutic in all breast cancer patients regardless of the clinical subtype. This study was supported by the DoD/BCRP HT94252310166, BC220526 and HT94252410094, BC230070 (to SD), Women’s Health Supplement from NIH/NIGMS 5P20GM103475 (to SD, RP, and HF), and Susan Komen for the Cure Aspire supplement (to ACC).
Presentation numberPS1-11-13
Ai-driven chemotherapy decision support via genomic subgroup identification in fertility-conscious and low-risk er+/her2− invasive ductal breast carcinoma
Wanru Guo, University of Maryland, Baltimore/ UM Greenebaum Comprehensive Cancer Center, Baltimore, MD
W. Guo, C. Tatsuoka; Department of Biostatistics, University of Maryland, Baltimore/ UM Greenebaum Comprehensive Cancer Center, Baltimore, MD.
Background: Invasive ductal carcinoma is the most common subtype of breast cancer, with endocrine therapy the standard treatment for ER+ patients. However, certain clinical and genomic risk factors support adding chemotherapy. Among young women under 50, treatment decisions must balance survival benefit with fertility preservation and overtreatment avoidance. Similarly, patients with low Nottingham Prognostic Index (NPI) scores are generally spared chemotherapy, yet a subset may still benefit from combination treatment. In this study, we apply a two-stage machine learning framework to identify (1) a clinically meaningful subgroup of young ER+/HER2− patients likely to benefit from combined chemo-endocrine therapy despite fertility concerns, and (2) genomic subgroups associated with chemotherapy benefit among patients with low-risk NPI profiles.Methods: We analyzed a longitudinal cohort of 423 IDC patients who received adjuvant treatment post-surgery, with 5-year overall survival (OS) and recurrence-free survival (RFS) as outcomes. A two-stage Virtual Twins (VT) ML framework was applied to identify subgroups benefiting from combination therapy (hormonal therapy + chemotherapy ± radiation) versus hormonal therapy alone, focusing on patients <50 years and those with low NPI. In Stage 1, multiple classifiers – random forest, multilayer perceptron (MLP), gradient boosting (XGBoost), Super Learner, and Bayesian additive regression trees (BART)- were compared for overall survival (OS) and recurrence-free survival (RFS) prediction. The best-performing model was used to estimate individual treatment effects (ITEs) via counterfactuals. In Stage 2, regression trees were trained on the ITEs using the top 10 predictive features to identify subgroups with differing treatment sensitivity. Results: OS and RFS were modeled using 19 clinical and multi-omics features (transcriptomics, CNV, mutations, methylation) selected from high-dimensional inputs based on biological clustering in Stage 1 of VT, enabling ITE-based subgroup identification via regression trees in Stage 2. Firstly, in ER+/HER2− patients under age 50, those with the greatest survival benefit from combination therapy had high NPI (>5.0) and harbored amplifications at 11q13/14 (CCND1, PAK1, RSF1, EMSY), 8p12, 8q, and 20q—indicative of proliferative and endocrine-resistant biology. These tumors also exhibited TRG/TRA deletions and high CD8+ infiltration. In contrast, younger patients with genomically quiet tumors-defined by isolated 8p12 amplification, 1q gain, 16q loss, or modest 8q/20q gains-and Luminal A or Claudin-low subtypes showed minimal benefit from combination therapy. Secondly, in patients with low NPI (<4.0), those with the greatest benefit from combination therapy were young (70) with 1q gain and 16q loss showed limited benefit, consistent with quiet Luminal A-like tumors.Conclusion: Virtual twin modeling identified ER+/HER2− IDC subgroups- despite clinical low-risk or fertility -conscious status– that derive meaningful benefit from adjuvant combination therapy when harboring high-risk genomic features. These findings support genomics-informed escalation in patients typically spared chemotherapy, highlighting the value of precision oncology in tailoring adjuvant care.
Presentation numberPS1-11-14
Early (≤12 weeks) vs Delayed (≥12 weeks) Initiation of Adjuvant Endocrine Therapy After Surgery on Recurrence in HR+ Breast Cancer: A Real-World Global Cohort Study
Safa Afridi, SUNY Upstate Medical University, Syracuse, NY
S. Afridi1, K. Doshi2, Z. Shah3, S. Gandhi4; 1Department of Internal Medicine, SUNY Upstate Medical University, Syracuse, NY, 2Department of Hospice and Palliative Medicine, UT Health San Antonio, San Antonio, TX, 3Department of Hematology and Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 4Department of Hematology and Medical Oncology, Emory Winship Cancer Institute, Atlanta, GA.
Background: Timely initiation of adjuvant endocrine therapy (AET) is a quality metric in hormone receptor-positive (HR+) breast cancer, but real-world data on optimal timing post-surgery are limited. We evaluated recurrence outcomes among patients initiating AET early (≤12 weeks) vs delayed (≥12 weeks) after surgery. Methods: We conducted a retrospective cohort study using the TriNetX Global Collaborative Network, which includes de-identified electronic health records from 146 healthcare organizations across the world. Women aged ≥18 years with Stage I-II HR+/HER2-negative breast cancer who underwent surgery and initiated adjuvant endocrine therapy were eligible. Surgery included partial mastectomy (e.g., lumpectomy, quadrantectomy), mastectomy with axillary lymphadenectomy, and simple mastectomy. Two cohorts were defined: Early AET Group: AET initiated within ≤12 weeks post-surgery (n=5,793) and Delayed AET Group: AET initiated after ≥12 weeks post-surgery (n=9,982).The primary outcome was breast cancer recurrence, defined using ICD-10 codes C77 (secondary neoplasm of lymph nodes) and C79 (secondary neoplasm of other sites), measured from 1 to 5 years after AET initiation. A multivariable Cox proportional hazards model estimated adjusted hazard ratios (aHRs) for recurrence, adjusting for age, disease stage, receipt of chemotherapy, Ki-67 status, and comorbidities (diabetes, chronic kidney disease [CKD], and coronary artery disease [CAD]). Results: A total of 15,842 patients were included: 5,810 in the Early AET group and 10,032 in the Delayed AET group. In univariate analysis, early AET was associated with a significantly lower risk of recurrence (HR 0.891, 95% CI: 0.799-0.992; p = 0.035). After adjusting for covariates in a multivariable model, early AET initiation remained independently associated with reduced recurrence (adjusted HR 0.891). Other significant predictors of increased recurrence included stage 2 disease, chemotherapy, diabetes mellitus, and coronary artery disease. Age was inversely associated with recurrence. Ki-67 and CKD were not significantly associated with recurrence. Conclusions: Initiating adjuvant endocrine therapy within 12 weeks post-surgery is associated with a significant reduction in recurrence among HR+/HER2-negative breast cancer patients. These findings reinforce the adoption of early AET initiation as a key quality metric in breast cancer care.
| Covariate | Adjusted HR | 95% CI | p-value |
| Age | 0.981 | 0.977 – 0.986 | <0.001 |
| Stage 2 | 2.187 | 1.962 – 2.437 | <0.001 |
| Chemotherapy vs No Chemotherapy | 1.957 | 1.748 – 2.191 | <0.001 |
| Ki-67 | 0.989 | 0.872 – 1.121 | 0.861 |
| Diabetes Mellitus | 1.160 | 1.024 – 1.313 | 0.019 |
| Coronary Artery Disease (CAD) | 1.225 | 1.039 – 1.445 | 0.016 |
| Chronic Kidney Disease (CKD) | 1.181 | 0.962 – 1.448 | 0.111 |
Presentation numberPS1-11-16
Clinical and Molecular Landscape of ESR1 and PIK3CA Co-Mutated Hormone Receptor-Positive Metastatic Breast Cancer (HR+ MBC): Insights from 8,626 Patients Including Polyclonality and TP53 Alterations
Abirami Sivapiragasam, Medical University of South Carolina, Charleston, SC
A. Sivapiragasam1, A. Dugan2, S. Fragkogianni2, M. Ciampricotti2, E. Williams2; 1Hematology Oncology, Medical University of South Carolina, Charleston, SC, 2Medical Affairs, Tempus AI Inc, Chicago, IL.
Background Patients with HR+ MBC are treated with first-line endocrine therapy plus CDK4/6 inhibitors, but resistance often emerges through ESR1 or PIK3CA mutations (MUT). Co-mutations (co-MUT), present in ~10-15% of patients, remain poorly understood. Here, we compare molecular features and Real-world overall survival (rwOS) among patients with co-MUT vs. those with single or no mutations (no-MUT). Methods We used Tempus Lens (Tempus AI, Inc., Chicago, IL) to identify patients with a primary diagnosis of MBC who had Tempus xF or xT testing from the Tempus multi-modal database (N = 8,626). Patients with tumor purity of at least 30% were assessed and all pathogenic or likely pathogenic (P/LP) somatic SNVs/indels for PIK3CA, ESR1, and TP53 were extracted. Polyclonal ESR1 and PIK3CA were defined as more than one distinct P/LP somatic SNV/indel. Tumor mutational burden (TMB) (mt/mB) was analyzed by DNA-sequencing. rwOS analyses were run for all samples collected in the year preceding any line of therapy using risk-set adjusted Cox models with the start of therapy as the index date, the later date of sequencing and treatment start as the study entry time, and a maximum follow up of 5 years. Results Of the total cohort, 53% of patients had no-MUTs, 32% were PIK3CA-MUT , 9% were ESR1-MUT, and 6% were co-MUT. As shown in the table, age and hormone receptor status were significantly different between the groups, but not race. The co-MUT group exhibited a higher prevalence of bone metastases, while ESR1-only patients had increased liver and lung involvement. TP53 P/LP MUT were present in 33% (N=903) of the PIK3CA-only patients and 24% (N=131) of the co-MUT patients. High TMB rate was significantly different between all groups and was highest in the co-MUT and PIK3CA patients (15% vs 14%). Polyclonal changes in both genes were also more frequent in the co-MUT group. The most common ESR1 mutation was p.D538G (6.4%), while p.His1047Arg was the predominant PIK3CA mutation (14%, n=1,187). Liquid biopsy identified 40% of co-MUT, while tissue sequencing detected 61%. Co-MUT patients had significantly worse rwOS compared to no-MUT (HR=1.75, p<0.001), though evidence of non-proportionality was observed. Conclusions Herein, we characterized the largest real-world cohort of co-MUT HR+ MBC patients, identifying distinct clinical and molecular features such as enriched bone metastasis, higher TMB, and increased polyclonality. The lower overall frequency of ESR1 MUT and co-MUT likely reflects that only 25% of patients had late molecular testing, suggesting these mutations often arise after prolonged endocrine therapy. In this subgroup, the ESR1 MUT rate was 24% and the co-MUT rate was 15%, as expected. Limitations include the retrospective design; future studies will address these and examine PTEN/AKT alterations.
| T-Dxd (n=82) | Chemo (n=248) | Adjusted HR* (95% CI) | |||||
| Median OS (95% CI), mo | 21.0 (15.1, 25.6) | 21.0 (18.3, 27.7) | 1.16 (0.79, 1.69) | ||||
| Probability of overall survival at 6 mo (%, 95% CI) | 81.1 (70.2, 88.4) | 85.1 (79.4, 88.7) | |||||
| Probability of overall survival at 12 mo (%, 95% CI) | 72.9 (59.2, 80.9) | 68.4 (61.6, 74,3) | |||||
| Median TTNT (95% CI), mo | 10.1 (7.8, 13.1) | 6.0 (5.1, 6.5) | 0.45 (0.30, 0.66) | ||||
| Probability of continuing therapy at 6 mo (%, 95% CI) | 70.0 (57.9, 79.0) | 49.2 (42.3, 55.2) | |||||
| Probability of continuing therapy at 12 mo (%, 95% CI) | 39.4 (26.1, 50.1) | 19.7 (14.4, 25.2) |
Presentation numberPS1-11-18
Prescription Patterns and Treatment Persistence in Patients with Metastatic Breast Cancer on CDK4/6 Inhibitors
Claire Sathe, Columbia University Irving Medical Center, New York, NY
C. Sathe1, R. Raghunathan2, M. K. Accordino1, M. M. Caplan1, J. D. Wright3, D. L. Hershman1; 1Hematology Oncology, Columbia University Irving Medical Center, New York, NY, 2Irving Comprehensive Cancer Center Core, Columbia University Irving Medical Center, New York, NY, 3Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY.
Background: Since the FDA’s approval of palbociclib, the first cyclin dependent kinase 4/6 inhibitor (CD4/6i), in 2015, these agents have transformed the treatment landscape for breast cancer (BC). In particular, the combination of CDK4/6i with endocrine therapy forms the cornerstone of the first-line treatment of hormone receptor-positive (HR+)/HER2-negative (HER2-) metastatic BC (mBC). Choosing among the 3 approved CDK4/6i (palbociclib, ribociclib, and abemaciclib) remains challenging, as each has a distinct side-effect profile and there are no head-to-head trial data comparing their therapeutic efficacy. This study examines real-world prescription patterns and usage trends of CDK4/6i among patients with mBC, providing insight into evolving prescription preferences and the tolerability of these treatments. Methods: Utilizing the MarketScan health care claims database, we identified patients prescribed palbociclib, ribociclib, and/or abemaciclib from 2/2015 (post-FDA approval of palbociclib) until 12/2022 (prior to FDA approval of CDK4/6i for adjuvant use). We assessed changes in prescription rates for specific CDK4/6i over time and the frequency of switches between these agents. To evaluate treatment duration, we analyzed cohorts of patients who received their first CDK4/6i prescription in 2015, 2016, and 2017, tracking ongoing prescriptions over subsequent years. Descriptive statistics, including frequencies, percentages, means, and medians, were employed to summarize our findings. Results: A total of 13,514 patients were prescribed a CDK4/6i between 2/2015 and 12/2022 (98.2% female; 23.1% 70 yo; 18.6% on Medicare, 19.9% on Medicaid). The percentage of patients < 50 yo and patients covered by Medicaid prescribed CDK4/6i consistently increased over time, from 17.9% and 11.5% in 2015, to 31.8% and 22.6% in 2022, respectively (p 70 yo and patients covered by Medicare decreased over time, from 16.9% and 24.9% in 2015, to 9.8% and 12.5% in 2022, respectively (p<0.0001). In 2015-2016, all patients on CDK4/6i were prescribed palbocilcib. From 2017 to 2022, the proportion of patients on CDK4/6i prescribed ribociclib and abemaciclib consistently increased each year. By 2022, among 2,682 patients with CDK4/6i claims, 1,726 (64.4%) were prescribed abemaciclib, 508 (18.9%) were prescribed ribociclib, and 448 (16.7%) were prescribed palbociclib. Switching between CDK4/6i was infrequent, with only 854 patients (6.3%) having claims for multiple distinct CDK4/6i. The most common switch was from palbociclib to abemaciclib (62.8% of patients with claims for at least 2 CDK4/6i), followed by palbociclib to ribociclib (13.2%). Among patients with a first claim for CDK4/6i in 2015-2017, long-term persistence was noted, with 11.1% of patients starting treatment in 2015 continuing into 2018, 12.4% of those starting in 2016 continuing into 2019, and 10.8% of those starting in 2017 continuing into 2020. Conclusions: This real-world study of CDK4/6i prescription patterns following their FDA approval for mBC reveals an increasing trend in younger and lower-income patients on Medicaid receiving these agents, likely reflecting improved access and coverage. Oncologists showed a preference for newer agents such as ribociclib and abemaciclib post-approval, even prior to overall survival data becoming available. The rarity of switching between CDK4/6i suggests good tolerability. Over 10% of patients maintained long-term use of CDK4/6i into the third year after the year of their initial prescription.
Presentation numberPS1-11-19
Predictive value of Progesterone Receptor expression in Advanced Breast Cancer. Systematic Review and Meta-analysis
Andrea Gottlob Pérez, Morales Meseguer University Hospital, Murcia, Spain
A. Gottlob Pérez, N. Blaya Boluda, M. Guirao García, E. García Torralba, F. Ayala de la Peña; Medical Oncology, Morales Meseguer University Hospital, Murcia, SPAIN.
Introduction and objectives: Endocrine therapy (ET) is the standard treatment for patients with luminal (HR+/HER2−) metastatic breast cancer (MBC). However, the predictive value of progesterone receptor (PR) expression in this setting remains controversial. This study aimed to evaluate PR expression as a predictive biomarker of progression-free survival (PFS) in patients with luminal MBC treated with ET. Materials and methods: A systematic search was conducted in PubMed, MEDLINE, and EMBASE to identify clinical trials (CTs) that included patients with luminal MBC treated with ET (either as monotherapy or in combination) and reported PR status. Data were analyzed using STATA v.12. A meta-analysis of proportions was performed to estimate pooled PR prevalence, and weighted means and frequencies were calculated. PFS according to PR status was the primary outcome. Results: Following the screening of 1,144 studies, 46 clinical trials (published between 1997 and 2024) met the eligibility criteria. Trial characteristics: phase II (40%) or phase III (48.9%); randomized design (71.7%); evaluated endocrine therapy (ET) either as monotherapy with parenteral SERDs (34.8%), or in combination with CDK4/6 inhibitors (39.1%) or PIK3CA/AKT/mTOR pathway inhibitors (26.1%). None of the trials investigating oral SERDs (N=31) reported PR status and were excluded from the meta-analysis. Patient characteristics (N=16,921): predominantly women (99.8%); median age 63 years (range 23-98); postmenopausal (52.2%) or premenopausal (10.9%); majority from Western countries (80%); visceral metastases in 48.2%, and bone-only disease in 19.1%; prior lines of therapy: none (23.9%) or ≤1 (28.3%). Most tumors were ER-positive (93.0%) and PR-positive (72.4%). The median follow-up across trials was 18.5 months. The predictive value of PR expression for PFS was reported in 25% of ET-only trials, 50% of ET+CDK4/6 trials, and 25% of ET+PIK3CA/AKT/mTOR trials. Table 1 summarizes the main findings. A statistically significant benefit was observed in patients with both PR-positive and PR-negative tumors who received combination therapy, compared to those treated with endocrine monotherapy (either fulvestrant or aromatase inhibitors [AIs]). Among patients receiving ET alone, clinical benefit was observed exclusively in those with PR-positive tumors. Overall heterogeneity across and within subgroups was consistent. Conclusion: PR is an accessible and standardized biomarker that may have predictive relevance in ET-based treatment strategies. In ET monotherapy settings, clinical benefit was higher in patients with PR-positive tumors. In contrast, in combination regimens, outcomes were independent of PR status.
| Group and name of the study | Hazard Ratio PR+ | Confidence Interval 95% PR+ | Weight % PR+ | Hazard Ratio PR- | Confidence Interval 95% PR- | Weight % PR- | ||||||||||||||||||||||||||||||||||||
| Group 1: Endocrine Therapy (ET) | ||||||||||||||||||||||||||||||||||||||||||
| CONFIRM | 0.86 | 0.73-1.05 | 7.90 | 0.75 | 0.71-0.92 | 11.97 | ||||||||||||||||||||||||||||||||||||
| FALCON | 0.73 | 0.56-0.94 | 7.15 | 1.04 | 0.67-1.62 | 4.54 | ||||||||||||||||||||||||||||||||||||
| SoFEA1 | 0.85 | 0.66-1.10 | 6.44 | 1.30 | 0.80-2.10 | 2.89 | ||||||||||||||||||||||||||||||||||||
| SoFEA2 | 0.94 | 0.71-1.23 | 5.57 | 0.85 | 0.49-1.48 | 4.30 | ||||||||||||||||||||||||||||||||||||
| I2 | 0.83 | 0.74-0.93 | 27.06 | 0.85 | 0.65-1.05 | 23.71 | ||||||||||||||||||||||||||||||||||||
| Group 2: ET+ iCDK4/6 | ||||||||||||||||||||||||||||||||||||||||||
| MONALEESA 2 | 0.62 | 0.46-0.82 | 7.40 | 0.36 | 0.20-0.65 | 9.20 | ||||||||||||||||||||||||||||||||||||
| MONALEESA 7 | 0.57 | 0.45-91 | 6.21 | 0.44 | 0.26-0.77 | 8.48 | ||||||||||||||||||||||||||||||||||||
| MONARCH 2 | 0.59 | 0.46-0.74 | 8.41 | 0.51 | 0.33-0.80 | 8.96 | ||||||||||||||||||||||||||||||||||||
| MONARCH 3 | 0.61 | 0.44-0.84 | 6.91 | 0.43 | 0.24-0.76 | 8.36 | ||||||||||||||||||||||||||||||||||||
| PALOMA 3 | 0.46 | 0.32-0.66 | 7.65 | 0.46 | 0.28-0.77 | 8.72 | ||||||||||||||||||||||||||||||||||||
| DAWNA 1 | 0.46 | 0.33-0.66 | 7.78 | 0.35 | 0.18-0.69 | 8.48 | ||||||||||||||||||||||||||||||||||||
| DAWNA 2 | 0.55 | 0.40-0.76 | 7.40 | 0.32 | 0.16-0.66 | 8.60 | ||||||||||||||||||||||||||||||||||||
| I2 | 0.55 | 0.48-0.61 | 51.75 | 0.41 | 0.32-0.50 | 60.79 | ||||||||||||||||||||||||||||||||||||
| Group 3: ET+iPI3K/AKT/mTOR | ||||||||||||||||||||||||||||||||||||||||||
| BELLE 3 | 0.66 | 0.50-0.85 | 7.52 | 0.70 | 0.42-1.18 | 5.94 | ||||||||||||||||||||||||||||||||||||
| BOLERO 2 | 0.37 | 0.25-0.50 | 8.78 | 0.43 | 0.23-0.65 | 9.56 | ||||||||||||||||||||||||||||||||||||
| ENCORE301 | 0.66 | 0.43-1.02 | 4.90 | |||||||||||||||||||||||||||||||||||||||
| I2 | 0.55 | 0.32-0.77 | 21.20 | 0.52 | 0.27-0.76 | 15.50 |
Presentation numberPS1-11-20
Rna-based ESR1 and PGR gene expression provides a real-time readout of hormone receptor status in patients with breast cancer
Lajos Pusztai, Yale School of Medicine, New Haven, CT
L. Pusztai1, S. D. Sisoudiya2, R. B. Keller-Evans3, J. J. Pao2, J. S. Ross4, V. Sethunath5, R. S. Huang3, A. B. Schrock3, L. Poyser4, L. Dennis6, G. Frampton2, E. M. Ebot2, E. S. Sokol2, S. Sivakumar2, A. C. Annan7; 1Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Computational Discovery, Foundation Medicine Inc., Cambridge, MA, 3Clinical Development, Foundation Medicine Inc., Cambridge, MA, 4Medical Affairs, Foundation Medicine Inc., Cambridge, MA, 5Genomic Analysis and Reporting, Foundation Medicine Inc., Cambridge, MA, 6Research and Development, Foundation Medicine Inc., Cambridge, MA, 7University of Oklahoma Health Sciences Center, University of Oklahoma, Oklahoma City, OK.
Background: The hormone receptor (HR) status, traditionally determined by IHC-based expression levels of the estrogen receptor (ER; encoded by ESR1) and progesterone receptor (PR; encoded by PGR) in tumor tissues, is used for clinical decision-making in breast cancer. With the increase in IHC-based predictive biomarkers, there is a need for development of tissue conservation strategies. Further, receptor subtype switching occurs in ~20% of cases during progression, potentially impacting treatment decisions made based on IHC at initial diagnosis and thus requiring repeat testing. Multiplex RNA gene expression could serve as a screening or surrogate for protein expression by IHC especially when tissue availability is limited. Herein, we examined a cohort of patients with breast cancer profiled with a targeted RNA sequencing assay to evaluate the accuracy of ESR1 and PGR expression when compared to ER/PR IHC expression and assess the feasibility of utilizing gene expression for HR subtype inference. Methods: The study cohort comprised 497 patients with breast cancer who received RNA-based profiling during routine clinical care, capturing the expression of 1,517 genes (FoundationOne®RNA; a laboratory developed test by Foundation Medicine, Inc.). IHC-based ER/PR annotation from pathology reports was extracted and compared to ESR1 and PGR RNA expression. Almost 25% of the samples were held out while the remaining were split into training and test sets (70:30). For the model, transcripts per million (TPM) cutoffs for ESR1/PGR that maximized accuracy in the training set were selected (ESR1 TPM cutoff: 517.3; PGR TPM cutoff: 50.3). Performance of the model was assessed in the test and the held-out sample sets using accuracy, sensitivity, specificity, and F1 score (range 0 to 1). Results: Hormone receptor IHC and gene expression were found to be highly correlated. ER+ cases by IHC had 54-fold higher ESR1 expression compared to ER- cases (median, IQR 6,969.7 [2,846-11,742.9] TPM in ER+ vs 128 [78-246.8] TPM in ER-; P<0.0001). Similarly, PR+ cases had 30-fold higher expression of PGR compared to PR- cases (median, IQR 242.6 [62.2-616.3] TPM in PR+ vs 7.5 [3.7-15.6] TPM in PR-; P<0.0001). In the training dataset, a cutoff of 517.3 TPM for ESR1 and 50.3 TPM for PGR maximized accuracy for predicting HR status. In the held-out set, applying a model with these cutoffs resulted in a sensitivity of 94% and specificity of 88% for detecting HR+ IHC status. Accuracy was 92% with an F1 score of 0.94, suggesting this model can achieve both high precision and high recall. Of the cases with low ER IHC staining (between 1-9%) (n=4), the model predicted two samples as HR+ and the other two samples as HR-. The median ESR1 TPM of the two samples predicted to be HR+, despite the low ER IHC staining, was 1,466.9 TPM whereas the TPM of the two samples predicted to be HR- was 66.4. Conclusion: ESR1 and PGR RNA expression is highly correlated with ER/PR IHC status, and gene expression-based thresholding from RNA expression profiling could potentially be utilized to infer real-time HR status with high accuracy. Utilizing a multiplex RNA expression methodology as a screen or surrogate for predictive biomarkers such as ER/PR will become increasingly important with more IHC-based predictive biomarkers on the horizon. Further validation will be required to assess its utility in clinical decision making for breast oncologists.
Presentation numberPS1-11-21
Sequencing PIK3CA, AKT and mTOR Inhibitors in HR+/HER2- Metastatic Breast Cancer: A Real-World Retrospective Analysis
Khushboo Pal, UTSW, Dallas, TX
K. Pal, J. Thomas, N. Sadeghi, S. Kanjwal, H. McArthur, S. Reddy, G. Delgado Ramos, S. Syed, C. Ahn, N. Unni; Hematology Oncology, UTSW, Dallas, TX.
Background The PI3K/AKT/mTOR signaling pathway is a well-established therapeutic target in hormone receptor-positive, HER2-negative metastatic breast cancer (HR+/HER2- MBC). Approved agents—everolimus (mTOR inhibitor), alpelisib (PIK3CA inhibitor), and capivasertib (AKT inhibitor)—have demonstrated median progression-free survival (mPFS) ranging from 5.5 to 7.3 months in patients previously treated with CDK4/6 inhibitors. Most clinical trials leading to the approval of PIK3CA and AKT inhibitors excluded patients with prior exposure to PIK3CA, AKT, or mTOR inhibitors. Despite lack of prospective evidence, these targeted drugs are prescribed sequentially in real-world settings. Methods We identified 72 HR+/HER2- MBC patients with PI3K/AKT/PTEN pathway alterations treated with everolimus, alpelisib, or capivasertib between January 2019 and January 2025. Of these, 30 (42%) received ≥2 pathway inhibitors sequentially (Cohort A), 28 (39%) received alpelisib alone (Cohort B), and 14 (19%) received capivasertib alone (Cohort C). Clinical data including age, measurable vs. non-measurable disease, disease site, reason for discontinuation (progression vs. toxicity), PIK3CA mutation subtype (helical vs. kinase domain), ESR1 mutation status, and prior therapy lines were collected. mPFS was defined as time from treatment start to radiographic progression or death, with censoring at last follow-up. PFS1 and PFS2 were measured independently for patients in Cohort A, and PFS1+2 was calculated to reflect cumulative disease control. ANOVA, log-rank, univariate analysis, and McNemar tests were used for survival comparisons. Results The median age of our patient population was 63 years; 23% were Hispanic. Number of prior lines of therapy in the metastatic setting was similar in all cohorts. All patients had prior CDK4/6 inhibitor exposure. mPFS1+2 was 14.0 months in Cohort A (95% CI: 9-28), compared with 7.0 months in Cohort B (95% CI: 5-8) and 6.0 months in Cohort C (95% CI: 3-8), with a hazard ratio of 0.428 for patients who received ≥2 versus a single agent (p<0.0001, log-rank). PIK3CA helical versus kinase domain mutations (44% vs 56%) and ESR1 co-mutation status (present in 63%) were not significantly associated with PFS in any cohort. AKT/PTEN co-alterations (16%, 15%, 35% in Cohorts A, B, and C) showed no significant impact. In patients with measurable disease, PFS1+2 in Cohort A was 16.3 months vs. PFS of 7.2 months and 7.0 months in Cohorts B and C, respectively (p=0.003642, ANOVA). Among those with non-measurable disease, median PFS did not differ significantly across cohorts. Within Cohort A, no significant difference in combined PFS was noted between patients with measurable vs. non-measurable disease (18.3 vs. 21.8 months, p=0.66) or in patients with ESR1 mutations (p=0.265). Interestingly, no association was found between progression or toxicity on the first drug and outcomes on the second drug within Cohort A (p=0.366). Conclusions In this real-world, post-CDK4/6i cohort of HR+/HER2- MBC patients, sequential use of PI3K/AKT/mTOR pathway inhibitors was associated with significantly longer disease control compared to single-agent use. Notably, measurable disease status and mutation profiles (e.g., ESR1, PIK3CA subtype) did not predict benefit from sequential therapy. Furthermore, toxicity from one agent did not preclude benefit from subsequent agents. Our findings support the consideration of sequencing pathway inhibitors regardless of tolerance to the first agent. These results reinforce sequential PI3K/AKT/mTOR inhibition as a viable strategy in modern metastatic breast cancer care, though prospective studies are needed to validate this approach in pretreated populations.
Presentation numberPS1-11-22
Distinct Transcriptional and Immunosuppressive Microenvironment Signatures in PIK3CA- and ESR1-Mutant Hormone Receptor Positive (HR+)/HER2- Metastatic Breast Cancer (MBC)
Abirami Sivapiragasam, Medical University of South Carolina, Charleston, SC
A. Sivapiragasam1, A. Dugan2, S. Fragkogianni2, U. Jariwala2, E. Williams2, M. Ciampricotti2; 1Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, 2Medical Affairs, Tempus AI, Inc., Chicago, IL.
Background Mutations (mut) in ESR1 and PIK3CA are key drivers of endocrine resistance in HR+ MBC, with ESR1 mut typically acquired after aromatase inhibitor therapy and PIK3CA mut often present early and clonally. The co-occurrence of these muts and their effects on the tumor immune microenvironment (iTME) and transcriptome are not well understood. Here, we compare gene expression and immune cell infiltration in patients (pts) with PIK3CA– and/or ESR1-mut versus wild-type (WT) HR+/HER2- MBC. Methods We used Tempus Lens (Tempus AI, Inc., Chicago, IL) to identify pts with a primary diagnosis of MBC who had Tempus xT/xF and xR testing from the Tempus multi-modal database. Pts with tumor purity of at least 30% were assessed and all pathogenic or likely pathogenic somatic SNVs/indels for PIK3CA and ESR1 were extracted. Pts were stratified into groups based on the presence of PIK3CA-mut and/or ESR1-mut. RNA-seq data were normalized to correct for assay/batch effects, quantified as transcripts per million (TPM) for all protein-coding transcripts, and reported as log2(TPM + 1). Immune infiltration was quantified using quantiseq. Differential gene expression (DGE) analyses were performed using Wilcoxon rank-sum tests; p values were adjusted for multiple comparisons using the Benjamini-Hochberg false discovery rate method. Results The study cohort included N=4,583 pts where 48% were WT, 36% PIK3CA-mut, 10% ESR1-mut, and 6% co-mut. Median age at diagnosis was 57 years and, among those with available race data, 77% were White, 12% were Black, 4% were Asian. DGE analyses revealed differences in genes known to promote cancer development and tumor growth. Specifically, compared to WT, both ESR1-mut pts and co-mut pts had higher expression of TFF1, FGA, FGB, ALB, and SCGB2A2 (p<0.001). Lower SFRP2 expression was specific to ESR1-mut tumors (p<0.001); PIK3CA-mutated pts also had higher expression of SCGB2A2 (mammaglobin) (p<0.001). iTME analysis revealed that PIK3CA-mut and co-mut tumors exhibited significantly higher levels of immunosuppressive cells, specifically M2 macrophages and regulatory T cells (Tregs)( p<0.001). This immunosuppressive profile was most pronounced in PIK3CA-mut pts, whereas ESR1-only pts showed the lowest numerical enrichment. Conclusions PIK3CA-mutant and co-mutant HR+/HER2- MBCs share a distinct immunosuppressive microenvironment, primarily driven by PIK3CA mutation status, and exhibit unique transcriptional profiles. SFRP2 downregulation is specific to ESR1-mutant tumors, while robust SCGB2A2 upregulation in PIK3CA-mutant and co-mutant tumors highlights its potential as a diagnostic and therapeutic target. These findings underscore the importance of further research to clarify the clinical significance of these molecular programs and to inform the development of new treatment strategies.
| No-mut (N=2,203) | PIK3CA-only (N=1,668) | ESR1-only (N=436) | Co-mut (N=276) | P-value | |
| FF1 Expression | 6.7 (3.2, 9.3) | 7.9 (5.5, 9.7) | 8.3 (6.0, 10.1) | 9.3 (7.2, 10.8) | <0.001 |
| FGA Expression | 0.5 (0.4, 2.3) | 0.5 (0.4, 1.6) | 7.2 (0.5, 10.9) | 3.5 (0.5, 10.3) | <0.001 |
| FGB Expression | 0.9 (0.5, 4.4) | 0.9 (0.5, 3.3) | 8.3 (1.0, 11.7) | 5.1 (0.8, 11.0) | <0.001 |
| ALB Expression | 2.9 (1.8, 6.4) | 2.9 (1.8, 5.3) | 10.9 (2.9, 14.5) | 6.9 (2.3, 14.1) | <0.001 |
| SCGB2A2 Expression | 4.0 (0.7, 8.5) | 7.7 (3.0, 10.4) | 5.0 (0.9, 9.1) | 8.2 (4.1, 10.8) | <0.001 |
| SFRP2 Expression | 6.1 (2.6, 8.2) | 6.9 (3.7, 8.7) | 2.3 (0.4, 5.7) | 3.5 (0.8, 6.8) | <0.001 |
| % of M2 macrophages | 5.3 (3.9, 6.9) | 5.8 (4.53, 7.31) | 5.1 (3.53, 6.53) | 5.1 (4.0, 6.9) | <0.001 |
| % of M1 macrophages | 3.2 (2.1, 4.5) | 3.4 (2.4, 4.6) | 2.8 (2.0, 3.0) | 3.3 (2.4, 4.4) | <0.001 |
| % of Tregs | 2.6 (2.0, 3.8) | 2.8 (2.1, 3.8) | 2.2 (1.6, 3.0) | 2.3 (1.7, 3.1) | <0.001 |
Presentation numberPS1-11-23
Clinicogenomic Features of Recurrence After Adjuvant Abemaciclib in HR+/HER2- Early Breast Cancer
Jimmitti Teysir, Memorial Sloan Kettering Cancer Center, New York City, NY
J. Teysir, E. Kohilakis, D. Audi Blotta, Y. Sallah, S. Shen, C. Dang, K. Jhaveri; Breast Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY.
Background: Adjuvant abemaciclib combined with endocrine therapy is approved for patients with high-risk hormone receptor-positive (HR+), HER2-negative early breast cancer. However, recurrence still occurs in a subset of patients, and the clinical and molecular features associated with early relapse are not well characterized. Methods: We identified patients who were treated with adjuvant abemaciclib at Memorial Sloan Kettering (MSK) from April 2019 to February 2024 whose disease subsequently recurred. Clinicopathologic and tumor next-generation sequencing data (MSK-IMPACT) were abstracted from patient records. Changes in hormone receptor expression and genomic alterations between primary and recurrent samples were evaluated. Results: Among 334 patients treated with adjuvant abemaciclib, 18 (5.4%) developed recurrence. Baseline clinicopathologic and treatment characteristics are summarized in Table 1. Median time from surgery to abemaciclib initiation was 5.8 months (range 2.0-18.8), and from abemaciclib initiation to recurrence was 14.2 months (2.4-43.7). Median duration of abemaciclib therapy was 12.6 months (0.5-34.6); discontinuation was due to disease progression (14; 77.8%), toxicity (2; 11.1%), therapy completion (1; 5.6%), or early switch to olaparib (1; 5.6%). 16 patients (88.9%) had distant metastases and 2 (11.1%) had locoregional/chest wall recurrence. Median duration of first-line metastatic therapy was 4.0 months (1.5-9.0), including 5 patients still on therapy at the time of data abstraction. Among 17 cases with paired immunohistochemistry data, ER expression declined in 11 (64.7%), with a median absolute decrease of 65% (range 9-95%), including complete loss in 4 tumors (23.5%). PR declined in 15 (88.2%), with a median absolute decrease of 20% (range 1-99), and complete loss in 13 (76.5%). Patients with ≥50% ER loss (8, 47.1%) had a median time to abemaciclib recurrence of 13.1 months, compared to 15.0 months in those with <50% loss (3, 17.6%) and 16.8 months in those with no loss (6, 35.3%). HER2 IHC increased in 8 tumors (47.1%), decreased in 3 (17.6%), and was unchanged in 6 (35.3%). Germline testing was available in 16/18 patients; 4 (25%) had pathogenic variants (BRCA2, n=2; PALB2, n=1; RAD51D, n=1). Tumor NGS findings from matched primary-recurrence pairs (n=6) and unmatched recurrence-only samples (n=8) are summarized in Table 1. Conclusions: In this cohort, recurrence after adjuvant abemaciclib occurred in 5.4% of patients and was characterized by frequent hormone receptor loss and enrichment of alterations associated with endocrine and CDK4/6 inhibitor resistance, including RB1, PTEN, and MYC, as well as targetable alterations such as PIK3CA and BRCA2. These findings highlight the potential value of molecular profiling earlier in the treatment course to assess risk of relapse in high-risk early-stage disease. Updated data will be presented at the meeting.
| Characteristic | Value |
| Age, median (range) | 51 (32–67) |
| Menopausal status, n (%) | Premenopausal: 9 (50.0%), Postmenopausal: 9 (50.0%) |
| Clinical stage, n (%) | II: 6 (33.3%), III: 12 (66.7%) |
| Pathologic tumor stage (T), n (%) | T1: 6 (33.3%), T2: 4 (22.2%), T3: 8 (44.4%) |
| Pathologic nodal stage (N), n (%) | N1mi: 2 (11.1%), N1: 6 (33.3%), N2: 7 (38.9%), N3: 3 (16.7%) |
| Grade, n (%) | G1: 1 (5.6%), G2: 7 (38.9%), G3: 10 (55.6%) |
| Histology, n (%) | Ductal: 14 (77.8%), Lobular: 3 (16.7%), Mixed: 1 (5.6%) |
| Radiation, n (%) | 18 (100.0%) |
| Chemotherapy type, n (%) | Neoadjuvant: 14 (77.8%), Adjuvant: 4 (22.2%) |
| Anthracycline-based chemotherapy, n (%) | 16 (88.9%) |
| Matched tumor sequencing: alterations present in both primary and recurrent samples (n = 6 patients) | PIK3CA (2), TP53 (1), PTEN (1), BRAF (1), PALB2 (1), ARID1A (1) |
| Matched tumor sequencing: alterations acquired at recurrence (n = 6 patients) | BRCA2 (2), ERBB2 (2), ARID1A (1) |
| Unmatched tumor sequencing: alterations in recurrence only samples (n = 8 patients) | TP53 (4), PIK3CA (3), PTEN (2), RB1 (2), ESR1 (1), ARID1A (1), ATM (1), CCND1 amp (3), MYC amp (1), FGFR1 amp (1), MDM2 amp (1), CDKN2A/B deletion (3) |
| Median time to recurrence in BRCA-altered (germline or somatic), months (range) | 3.2 (2.4–43.7) |
| Median time to recurrence in PIK3CA-altered tumors, months (range) | 17.2 (10.8–34.6) |
Presentation numberPS1-11-25
Fgfr1 promotes brain metastases of er+ breast cancer through canonical and non-canonical mechanisms in young and aged hosts
Morgan S Fox, University of Colorado, Aurora, CO
M. S. Fox1, J. A. Jaramillo-Gómez1, R. Marquez Ortiz1, K. Alvarez-Eraso1, M. J. Contreras-Zárate1, S. Koliavas1, P. Kabos2, N. J. Serkova3, C. A. Sartorius1, E. A. Wellberg4, D. M. Cittelly1; 1Pathology, University of Colorado, Aurora, CO, 2Medical Oncology, University of Colorado, Aurora, CO, 3Radiology, Animal Imaging Shared Resource, University of Colorado, Aurora, CO, 4Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK.
Estrogen receptor positive (ER+) breast cancer (BC) represents nearly 50% of all BC brain metastasis (BCBM) but remains understudied. Most ER+ BCBMs occur in postmenopausal women due to age or prior endocrine therapies, yet most in vivo ER+ BC models are estrogen (E2)-dependent and studied in young hosts. Thus, the mechanisms underlying metastatic progression in the aged/E2-depleted brain tumor microenvironment (TME) remain unknown. FGFR1-amplification (amp), a well-known driver of ER+ BC endocrine therapy resistance, was the only genomic alteration associated with increased risk of late recurrence in post-menopausal women with ER+ early BC on aromatase inhibitors, and FGFR aberrations are significantly higher in BCBM patients compared to non-BM in metastatic BC. While FGFRamp is often associated with auto-activation, TME-dependent activation of FGFR1 has emerged as an important mechanism modulating its activity. Here, we tested the hypothesis that canonical and non-canonical activation of FGFR1 by astrocytes and neurons promotes FGFR1-dependent ER+ BCBM in E2-high/young and E2-low/aged hosts. Intracardiac injection of ER+ PDX-derived cell lines showed higher BM incidence in FGFR1amp lines, and brain colonization was E2-dependent in young mice and E2-independent in aged hosts. FGFR1 knockdown (KD) did not alter 2D proliferation but decreased their growth in organotypic brain slices and their ability to colonize the brain in E2-high/young and E2-low/aged mice. Spatial transcriptomics of ER+ BCBMs and surrounding glial cells showed that while late-stage ER+ BCBMs are transcriptionally similar, aging and E2-depletion reduced FGF2 expression and FGF/FGFR signaling in surrounding glial cells, suggesting that TME-mediated paracrine activation of FGFR1 may be distinct in young and aged hosts. Since astrocytes were the main FGF2 source in the brain, we tested how changes in their FGF2 levels impact ER+ BC growth. Young astrocytes more effectively activated FGFR1 in ER+ BC cells than aged astrocytes and promoted FGFR1-dependent proliferation and mammosphere initiation of ER+ cells, suggesting that paracrine FGF2-driven FGFR1 activation contributes to ER+ BCBM growth in young hosts, but less in aged hosts. In the absence of FGF2, NCAM1, a cell adhesion molecule expressed in the surface of neurons and astrocytes, activates FGFR1 and contributes to neural development and synaptic plasticity. We found that like neurons, NCAM1 activated FGFR1 kinase activity and downstream signaling in ER+ BC, and induced differential expression of neuronal gene signatures including neural cell adhesion. FGFR1 KD decreased ER+ BC cell migration along neurites in co-culture with primary neurons and decreased the formation of synaptic puncta. NCAM1 KD in neuron-like SH-SY5Y neuroblastoma cells decreased migration of FGFR1amp ER+ BC cells, suggesting that FGFR1/NCAM1 facilitates interaction of ER+ BC cells with neurons and contributes to early BCBM colonization. Surprisingly, FGFR1 phosphorylation was higher in young versus aged mice at early stages of brain colonization but absent in all late-stage ER+ BCBMs, suggesting that paracrine activation of FGFR1 and interactions with neurons and astrocytes are critical for early BM colonization. Consistently, an FDA-approved FGFR inhibitor blocked ER+ BCBM when administered from early stages of BM colonization in young mice but did not block metastatic progression at late stages or in aged mice, suggesting limited efficacy of FGFR inhibitors to block non-kinase-dependent FGFR1 functions in vivo. Together, these studies suggest a novel mechanism whereby canonical and non-canonical activation of FGFR1 promote brain metastatic colonization of ER+ BC in young and aged/E2-depleted hosts, and caution that FGFR1 inhibition may only be effective to prevent but not to treat BMs.
Presentation numberPS1-11-26
Esr1 mutations and use of the oral serd elacestrant in metastatic breast cancer patients in austria: results from the agmt_mbc-registry
Simon P Gampenrieder, Paracelsus Medical University Salzburg, Salzburg, Austria
S. P. Gampenrieder1, G. Rinnerthaler2, A. Pichler3, W. Herz4, R. Pusch5, C. Dormann5, C. Suppan2, M. Sandholzer6, T. Winder6, S. Heibl7, L. Scagnetti7, C. Schmitt8, A. F. Zabernigg9, D. Egle10, C. Hager11, P. Pichler12, F. Roitner13, J. Andel14, K. Strasser-Weippl15, R. Bartsch16, V. Castagnaviz1, M. Hubalek17, M. Knauer18, C. F. Singer19, R. Greil20; 1Department of Internal Medicine III with Haematology, Medical Oncology, Paracelsus Medical University Salzburg, Salzburg, AUSTRIA, 2Division of Oncology, Department for Internal Medicine, Medical University Graz, Graz, AUSTRIA, 3Internal Medicine – Department for Haemato-Oncology, LKH Hochsteiermark, Leoben, AUSTRIA, 4Department of Surgery, Breast Health Center, LKH Hochsteiermark, Leoben, AUSTRIA, 5Internal Medicine I, Ordensklinikum Linz Barmherzige Schwestern – Elisabethinen, Linz, AUSTRIA, 6Department of Internal Medicine II, Academic Teaching Hospital Feldkirch, Feldkirch, AUSTRIA, 7Department of Internal Medicine IV, Klinikum Wels-Grieskirchen GmbH, Wels, AUSTRIA, 8Department for haematology and internal oncology, Kepler University Hospital Linz, Linz, AUSTRIA, 9Department of Internal Medicine, County Hospital Kufstein, Kufstein, AUSTRIA, 10Department of Gynaecology, Medical University Innsbruck, Innsbruck, AUSTRIA, 11Breast Center, Breast Center Dornbirn, Dornbirn, AUSTRIA, 12Department for Internal Medicine 1, University Hospital St.Pölten, St. Pölten, AUSTRIA, 13Department of Internal Medicine II, Hospital Braunau, Braunau am Inn, AUSTRIA, 14Department of Internal Medicine II, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, AUSTRIA, 15Department of Medicine I, Clinic Ottakring, Vienna, AUSTRIA, 16Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, AUSTRIA, 17Department of Gynaecology, Breast Health Center Schwaz, Schwaz, AUSTRIA, 18Breast Center, Tumor and Breast Center Eastern Switzerland, St. Gallen, SWITZERLAND, 19Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, AUSTRIA, 20Austrian Group Medical Tumor Therapy, AGMT gemeinnützige GmbH, Vienna, AUSTRIA.
Background: Elacestrant is the first oral Selective Estrogen Receptor Degrader (SERD) to be approved in the EU as of September 2023. Since its approval, testing for mutations in the estrogen receptor 1 (ESR1) gene has become standard care for patients with hormone receptor (HR)-positive, HER2-negative metastatic breast cancer (MBC) in low- to intermediate risk disease. ESR1 mutations are a known mechanism of resistance to aromatase inhibitors (AIs), with their frequency significantly increasing after AI treatment. This study presents the first real-world data from the Austrian Study Group of Medical Tumor Therapy (AGMT) MBC Registry regarding ESR1 testing and elacestrant use. Patients and Methods: The AGMT MBC Registry is a nationwide, multicenter, ongoing retrospective and prospective registry for MBC patients in Austria. For this analysis, only patients with sufficient data quality were included. ESR1 mutation testing was performed using various techniques and test kits, following local practices. Results: As of March 11, 2025, the registry included 3,094 patients, of whom 2,956 were evaluable. Among these, 132 patients (4.5%) had documented ESR1 testing. A total of 59 tests were performed on 54 patients after the approval of elacestrant: 33 (55.9%) via liquid biopsy and 26 (44.1%) via tissue analysis; three tests in three patients had no or inconclusive results. The majority of patients had HR+/HER2- disease (n=48; 92.3%). A total of 14 patients (27.5%) had a positive ESR1 result (30.4% of all tests). Six distinct ESR1 mutations were identified: D538G, E380Q, Y537S, Y537N, M396V, and L536_D538>P. Notably, three patients (5.5%) had multiple ESR1 mutations. These combinations were: E380Q/Y537S/M396V, D538G/E380Q, and D538G/Y537N. Of the 14 patients with positive ESR1 test results, 7 patients (50.0%) initiated elacestrant treatment between the first and eleventh line of therapy, with five patients continuing treatment at the data cut-off. Updated outcome data will be presented at the meeting.Eleven, 8 and 9 patients with ESR1 mutations (n=14) had additional testing for PIK3CA, TP53 and for BRCA1/2, respectively. In 7 patients co-mutations in PIK3CA were found. Four of these patients were treated with a PI3-kinase inhibitor, and one received elacestrant in combination with alpelisib. Co-mutations with TP53 and BRCA2 were found in 3 and 1 patient, respectively. Conclusion: This real-world data highlights the clinical relevance of ESR1 mutations in HR+/HER2- metastatic breast cancer patients and underscores the potential impact of molecular-guided treatment. The association of ESR1 mutations with co-mutations in PIK3CA, TP53, and BRCA2 suggests the need for personalized treatment approaches. Further follow-up and outcome data will provide more insights into the long-term efficacy of elacestrant in this cohort.
Presentation numberPS1-11-27
Evaluation of steroid hormone receptor expression and clinical outcomes in ER-positive breast cancer bone metastases
Anthony M Rossi, University of Arizona, Tucson, AZ
A. M. Rossi1, R. N. Plagens2, A. Elliott2, E. Tapia2, G. Sledge2, A. Elias3, J. Richer4, S. Ehsani1, J. L. Funk1; 1Medicine, University of Arizona, Tucson, AZ, 2na, Caris Life Sciences, Irving, TX, 3Medicine, University of Colorado, Auora, CO, 4Pathology, University of Colorado, Aurora, CO.
Background: Estrogen receptor-positive (ER+) breast cancer (BC) has a high affinity for progression in bone, which is often the initial metastatic site. ER+ BC treatment responses may depend on the expression of additional steroid hormone receptors (SHRs), such as progesterone receptors (PR) or androgen receptors (AR). However, the predictive value of these SHRs in determining ER+ BC bone metastases (BMET) outcomes remains largely unexplored. The possible functional impact of PR and AR targeting on bone-specific ER+ BC progression is also not known. To address this knowledge gap, we evaluated the predictive associations of AR and PR expression with clinical outcomes for ER+ BMET. Methods: A retrospective analysis was conducted using BC BMET samples analyzed at Caris Life Sciences, with survival outcomes inferred from insurance claims. HER2-/ER+ BMET (n=1677) specimens were identified using immunohistochemistry (IHC) (HER2-: ≤1+ intensity or ≤10% cells stained, or 2+ & >10% with CISH-null reflex test; ER+: ≥1+ & ≥1%). These samples were then stratified into four groups by IHC based on ±AR and/or PR expression (PR+: ≥1+ & ≥1%; AR+: ≥1+ & ≥10%). Associations were examined between AR ± PR expression and 1) overall survival (OS, inferred from biopsy to last contact), including CDK4/6 inhibitor time-on-therapy (CDKi TOT); 2) pathogenic gene mutations (mut) known to alter BC survival (ESR1mut or PIK3CAmut); or 3) AR-regulated gene expression. Results: SHR expression was common in ER+ BMET, with 87.1% AR+ and 59.3% PR+. “Triple positive” (AR+/PR+/ER+) BMET were the largest group (53.7%), followed by PR-null AR+/ER+ BMET (33.2%). AR-null ER+ BMET were much less common, with 5.6% or 7.3% PR+ or PR-, respectively. Only 9.4% of PR+/ER+ BMET were AR-, while 38.2% of AR+/ER+ BMET were PR-. AR positivity was associated with the longest OS for ER+ BMET. A similar trend for improved OS was observed in AR-expressing ER- BMET (n=305), though not statistically significant. When stratifying ER+ BMET by the proportion of AR+ cells stained by IHC, having a higher proportion (≥ median) was associated with longer OS only for PR+/ER+ BMET (Cox proportional hazards ratio (HR)=0.70, p=0.003 for high vs low AR % cells stained). There was no significant difference in CDKi TOT across groups. However, OS was significantly and uniquely longer for AR+ (not AR-) ER+ BMET patients (pts) who received CDKi, which was highest when PR was co-expressed (HR=0.62, p<0.0001 for pts with vs without CDKi), as compared to PR- (HR=0.73, p=0.012). ESR1mut prevalence was similar in AR+ (17.0%) vs AR- (10.7%) ER+ BMET, with OS reduced in ESR1mut (vs wildtype, wt) AR+/ER+ BMET (HR=1.92, p<0.0001). PIK3CAmut were more prevalent in AR+ (51.5%) vs AR- (27.3%) ER+ BMET. However, there was no difference in OS between PIK3CAmut vs wt for any SHR ER+ BMET subtype. While not SHR-regulated, RNA expression of the SHR pioneer factor FOXA1 positively correlated with that of AR, PGR (PR), and ESR1 (ER) in ER+ BMET across all SHR subtypes. Several known AR-regulated genes were among the most highly expressed genes in AR+ vs AR- BMET, consistent with functional AR signaling. Notably, expression of PIP, HMGCS2, and CYP4Z1 was significantly higher in AR+ vs AR- BMET, regardless of ER status. Conclusions: Our findings suggest that AR expression may have utility as a predictive biomarker of improved survival and response to CKD4/6 inhibitors in those with HER2-/ER+ BMET, particularly when co-expressed with PR, with evidence of functional AR signaling in both ER+ and ER- AR-expressing BC BMET. Whether AR in this context is a biomarker or mediator of improved survival cannot be determined. Therapeutic consequences of AR targeting to reduce AR+ BC progression in bone, also an AR-regulated tissue, may hold promise, although whether with an agonist or antagonist remains an intriguing and yet unanswered question.
Presentation numberPS1-11-28
Prognostic impact of type and location of germline BRCA2 pathogenic or likely pathogenic variants in patients with HR+/HER2- advanced breast cancer: results from the multicenter real-world PAMBRACA study
Emma Zattarin, University of Modena and Reggio Emilia, Modena, Italy
E. Zattarin1, E. Tenedini1, A. Marra2, A. Palazzo3, G. Griguolo4, C. Vernieri5, J. Etessami2, L. Pontolillo3, G. Landa4, A. Daneri6, M. De Monte5, R. Cuoghi Costantini1, O. Ponzoni1, M. Razeti6, C. Sposetti7, E. Barbieri8, M. Manni1, L. Cortesi9, G. Curigliano5, E. Bria3, V. Guarneri4, M. Dominici1, M. Lambertini10, A. Toss1; 1Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, ITALY, 2Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, ITALY, 3Department of Medical Oncology, Comprehensive Cancer Center, Agostino Gemelli University Polyclinic (IRCCS), Rome, ITALY, 4Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, ITALY, 5Department of Oncology and Hematology-Oncology, University of Milan, Milan, ITALY, 6Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 7Medical Oncology Department, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, 8Department of Medical Oncology, Azienda Ospedaliero-Universitaria di Modena, Modena, ITALY, 9Department of Medical Oncology, University of Modena and Reggio Emilia, Reggio Emilia, ITALY, 10Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genova, ITALY.
Background: The type and location of germline likely pathogenic or pathogenic variants (LP/PVs) in the BRCA1 and BRCA2 genes influence the risk of developing ovarian and breast cancer (BC) and their prognosis. However, their clinical impact in patients (pts) with advanced BC (aBC) remains to be fully elucidated. Methods:The PAMBRACA study is a multicenter, hospital-based, retrospective cohort study including BRCA1 and BRCA2 LP/PV carriers with hormone receptor positive/HER2-negative (HR+/HER2-) aBC treated with endocrine therapy (ET) plus cyclin-dependent kinase 4/6 inhibitors (CDK4/6i). Some pts also received PARP inhibitors (PARPi) after progression on CDK4/6i plus ET. For this specific analysis, BRCA1 carriers were excluded due to limited sample size. We collected information on type [insertion-deletion mutation (INDEL), single nucleotide variant (SNV), or copy number variation (CNV)], coding consequences (frameshift, nonsense, splicing, or missense), truncating versus non-truncating status, and exon location]. OS was defined as the time between metastatic disease diagnosis and pt death. Pts without an event were censored at the date of last follow-up. To account for potential immortal time bias, observation times were left truncated at the time of BRCA testing. Moreover, association between BRCA LP/PV type/location and pt progression free survival (PFS) during CDK4/6i plus ET or subsequent PARPi was explored. Multivariate Cox regression modelling was used to assess the association between LP/PV type/location, clinically relevant variables and pt outcomes. Results: Seventy-two pts with BRCA2 LP/PV diagnosed with HR+/HER2- aBC between Jan 1998 and Dec 2023 in six Italian Institutions were included. All pts received CDK4/6i plus ET and 36 (50.0%) received PARPi in the advanced setting. Detailed mutation type and location information was available for 57 cases (79.2%). Regarding BRCA2 LP/PV types, 37 (64.9%) INDELs and 20 (35.1%) SNVs were identified. The coding consequences included 33 (57.9%) frameshift, 16 (28.1%) nonsense, 5 (8.8%) splicing and 3 (5.2%) missense LP/PVs. Protein impact analysis showed 49 (85.9%) truncating, 3 (5.3%) non-truncating, and 5 (8.8%) unclassifiable LP/PV. Regarding LP/PV location, 32 (56.1%) LP/PV were located in exon 11, while 25 (43.9%) in other exons. No significant differences in baseline clinicopathologic variables (including age at BC diagnosis, menopausal status, pathological stage at BC diagnosis, tumor grading, estrogen/progesterone receptor levels, Ki67 levels, HER2 expression, presence of de novo metastatic disease, metastatic sites involvement) and LP/PV type/location (INDEL vs. SNV, frameshift vs. nonsense vs. splicing vs. missense, truncating vs. not truncating, and exon location) were observed. 5-year OS rate was 51% [95% confidence interval (CI) 37-72]. No BRCA2 LP/PV type or location was associated with OS, even after adjusting for clinically relevant covariates. At multivariable analysis, a number of metastatic higher than 3 [adjusted hazard ratio (aHR) 14.2, 95% CI 2.9-69.4, p = 0.001] and post-menopausal status at BC diagnosis (aHR 2.7, 95% CI 1.0-7.3, p = 0.056) were independently associated with poorer OS. No significant association was observed between BRCA2 LP/PV type/location and PFS with CDK4/6i plus ET or PFS with PARPi. Conclusions: In this cohort of pts with HR+/HER2- aBC harboring germline BRCA2 LP/PV, neither the type nor the location of the LP/PV was associated with OS. Furthermore, the effectiveness of CDK4/6i and PARPi appeared to be independent of the BRCA2 LP/PV type or location. Larger, prospective studies are warranted to further clarify the clinical impact of BRCA LP/PV types and locations on BC phenotype and outcome.
Presentation numberPS1-11-29
Factors predicting the effictiveness of CDK4/6 inhibitor rechallenge following progression on first-line endocrine therapy combined with a CDK4/6 inhibitor in ER+ metastatic breast cancer in the French ESME Database
Thomas PAPAZYAN, ICO Nantes, Saint Herblain, France
T. PAPAZYAN1, A. Lusque2, A. Mailliez3, T. Bachelot4, T. Grinda5, A. Goncalves6, E. Brain7, A. Savoye8, M. Arnedos9, C. Levy10, I. Desmoulins11, T. De La Motte Rouge12, C. Pflumio13, M. Mouret-Reynier14, V. Massard15, M. Leheurteur16, L. Bosquet17, W. Jacot18, M. Campone1, J. Frenel1; 1Medical oncology, ICO Nantes, Saint Herblain, FRANCE, 2Biostatistics, Oncopole Claudius Regaud IUCT-O, Toulouse, FRANCE, 3Medical oncology, Centre Oscar Lambret, Lille, FRANCE, 4Medical oncology, Centre Leon Berard, Lyon, FRANCE, 5Medical oncology, Institut Gustave Roussy, Villejuif, FRANCE, 6Medical oncology, IPC, Marseille, FRANCE, 7Medical oncology, Institut Curie, Paris, FRANCE, 8Medical oncology, Institut Godinot, Reims, FRANCE, 9Medical oncology, Institut Bergonié, Bordeaux, FRANCE, 10Medical oncology, Centre Francois Baclesse, Caen, FRANCE, 11Medical oncology, Centre Georges Francois Leclerc, Dijon, FRANCE, 12Medical oncology, Centre Eugene Marquis, Rennes, FRANCE, 13Medical oncology, ICANS, Strasbourg, FRANCE, 14Medical oncology, Centre Jean Perrin, Clermont, FRANCE, 15Medical oncology, Institut de cancerologie de Lorraine- Alexis Vautrin, Vandœuvre-lès-Nancy, FRANCE, 16Medical oncology, Centre Henri Becquerel, Rouen, FRANCE, 17Health Data and Partnerships Department, Unicancer, Paris, FRANCE, 18Medical oncology, Institut de Cancerologie de Montpellier, Montpellier, FRANCE.
Introduction: CDK4/6 inhibitors (CDK4/6i) have transformed the therapeutic landscape of hormone receptors-positive (HR+)/HER2-negative (HER2-) metastatic breast cancer (MBC). However, the clinical benefit of rechallenging with a CDK4/6i, either by maintaining the same agent or switching to another, after progression on first-line (1L) combined endocrine treatment (ET) remains unclear. Our study aimed to assess factors predicting the real-world effectiveness of CDK4/6i rechallenge after progression on 1L ET, using data from the ESME cohort, a large nationwide database of patients with MBC. Methods: The ESME-MBC cohort is a national cohort collecting individual-level patient data from 18 French Comprehensive Cancer Centers (NCT03275311). For this study, we included all patients aged 18 years or older with newly diagnosed HR+/HER2- MBC who began 1L ET with a CDK4/6i between January 2013 and December 2023. From this subpopulation, patients who had discontinued CDK4/6i due to disease progression and were later re-treated with a CDK4/6i in subsequent lines of therapy were selected to assess real world outcome including progression-free survival (PFS) and overall survival (OS) following the second CDK4/6i exposure. Multivariable analyses were conducted using Cox proportional hazards model including line of therapy, age, tumor grade, de novo MBC status, visceral metastases, number of metastatic sites, use of different CDK4/6i, and duration of initial CDK4/6i. Results: Among 2,711 patients who progressed on 1L ET + CDK4/6i, 177 (4.4%) underwent a CDK4/6i rechallenge during their treatment course. The initial CDK4/6i was palbociclib, ribociclib, and abemaciclib in 76.8%, 17.0% and 6.2% of patients, respectively, ET being mostly (86.4%) an aromatase inhibitor. After a 1L CDK4/6i-based treatment exceeding 24 months in 32.2% of patients, rechallenge occurred in 2L or later lines (3L++) in 77 (43.5%) and 100 (56.5%), respectively. At rechallenge, the median age was 63 years (range 28-89), 68.9% patients were postmenopausal, and 61.6% had visceral metastases. In 80.2% of cases, a different CDK4/6i was used, mosly abemaciclib (54.8%), in combination with fulvestrant (61.6%).With a median follow-up of 22.7 months [95%CI: 18.4-26.2] post-rechallenge, the second CDK4/6i was discontinued in 121 patients (68.4%) mainly due to disease progression (82.6%), followed by toxicity (10.0%) and other reasons (7.4%). In the whole population, the median PFS at rechallenge was 6.2 months [95% CI: 4.6-7.1]. Rechallenge resulted in a PFS of 6.9 months [95%CI: 4.5-10.6] in the 2L setting and 5.7 months [95%CI: 3.2-6.7] in 3L++ setting. Using a different CDK4/6i results median PFS was 5.7 months [95%CI: 4.1-6.7] and 7.8 months [95%CI: 3.8-9.6] when the same CDK4/6i was used. Multivariable analysis showed a worse PFS for Rechallenge in third 3L++ was the only factor associated with PFS (adjusted HR: 1.47, 95%CI: 0.98-2.21, p-value=0.06). Conclusion: This large multicenter retrospective study provides real-world evidence that switching with a second CDK4/6i after progression is feasible and may offer clinical benefit in selected patients.
Presentation numberPS1-11-30
Implications of PIK3CA/HER2 Status and Age/Comorbidities on Clinical Investigators’ (CIs) Selection of First-Line Systemic Therapy for Hormone Receptor-Positive (HR+) Metastatic Breast Cancer (mBC)
Neil Love, Research To Practice, Key Biscayne, FL
K. H. Pang1, J. O’Shaughnessy2, V. Kaklamani3, G. Kelly1, T. Wallace1, T. Cruse1, K. Miller1, K. A. Ziel1, D. Paley1, N. Love1; 1Clinical Content Department, Research To Practice, Key Biscayne, FL, 2Breast Disease Committee, Baylor University Medical Center, Dallas, TX, 3Breast Oncology Program, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX.
BACKGROUND: The 2023 and 2024 San Antonio Breast Cancer Symposium presentations of the Phase III INAVO120 and PATINA trials, respectively, have added important considerations in the decision of first-line therapy for select patients with HR+, PIK3CA-mutant or HER2-overexpressing mBC. In addition, the lack of direct comparison among the 3 approved CDK4/6 inhibitors (CDKis) has made choosing treatment a challenge in many clinical situations. This initiative attempted to identify how various factors influence CIs’ chioce of first-line therapy for HR+ mBC. METHODS: In April 2025, 20 US-based and international CIs completed a case-based survey on their choice of initial systemic therapy for HR+ mBC. For each case, a number of clinical factors (PIK3CA and HER2 status, age, performance status, comorbidities, site/extent of metastases, and prior adjuvant therapy) were varied. A modest honorarium was offered. RESULTS: Ribociclib (R) was the most common CDKi recommended for PIK3CA-wildtype, HER2-negative tumors, including patients with symptomatic visceral and asymptomatic bone metastases (Table). For patients with PIK3CA-mutated tumors with disease relapse 2 years into adjuvant endocrine therapy (ET), respondents most commonly recommended inavolisib (I)/palbociclib (P)/ET, and of great interest, 7 of 20 CIs would recommend this triplet for a patient with de novo disease. For patients with HR+, HER2-positive tumors, P/ET in addition to trastuzumab/pertuzumab was the most common recommendation for postchemotherapy/anti-HER2 maintenance treatment. However, for patients with multiple comorbidities and polypharmacy or for those who are frail and age 80 and older, respondents often did not recommend a CDKi. When one was used, it was commonly P. CONCLUSION: The clinical practice trend of selecting R as the preferred initial CDKi because of its consistent overall survival benefit has been modified by data from INAVO120 and PATINA, in which the perceived better tolerability of P led to its combination with targeted therapies that significantly improved outcomes. New questions revolve around the role of the I/P/ET combination for patients without endocrine resistance (de novo) and the use of a CDKi (P) in the HER2 maintenance setting for patients treated with trastuzumab deruxtecan based on recent data from the DESTINY-Breast09 study. Finally, the documentation that many investigators withhold a CDKi for older patients with poor performance status and comorbidities raises the issue of how to optimally identify patients for whom the risk/benefit ratio of these life-prolonging and generally tolerable agents does not weigh in their favor. These issues will be addressed in future surveys.
| Clinical presentation |
Most frequent endocrine therapy in addition to AI or fulvestrant |
|
HER2–, PIK3CA–
Symptomatic visceral metastases* |
R = 16 (80%) |
|
HER2–, PIK3CA–
Asymptomatic bone metastases* |
R = 17 (85%) |
|
HER2–, PIK3CA+
Symptomatic visceral metastases* |
I + P = 19 (95%) |
|
HER2–, PIK3CA+
Asymptomatic bone metastases* |
I + P = 15 (75%) |
|
HER2+ (IHC 3+)
Treatment response to THP |
P** = 17 (85%) |
| * while receiving an adjuvant aromatase inhibitor (AI) | ** with maintenance trastuzumab/pertuzumab |
Presentation numberPS1-12-02
Prognostic role of the innate immune system circulating cytokines in advanced luminal breast cancer (BC) patients (pts) of the GEICAM/2018-03 NIKOLE study
Elena García-Martínez, Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria, IMIB; GEICAM Spanish Breast Cancer Group, Murcia, Spain
E. García-Martínez1, E. Navarro-Manzano2, J. Cejalvo-Andújar3, B. Álvarez-Abril4, I. Garrido-Cano5, E. García-Torralba6, C. Tebar-Sánchez7, P. De la Morena-Barrio8, M. Martínez-Martínez9, F. Ayala de la peña8, C. Tapia-Céspedes10, J. Herranz11, I. Romero-Camarero12, F. Rojo13, B. Bermejo9; 1Medical Oncology, Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria, IMIB; GEICAM Spanish Breast Cancer Group, Murcia, SPAIN, 2Medical Oncology, Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria, IMIB,Centro Regional de Hemodonación, Murcia, SPAIN, 3Medical Oncology, Hospital Clínico Universitario de Valencia, INVLIVA; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 4Medical Oncology, Hospital Universitario Morales Meseguer, Murcia, SPAIN, 5Environmental Epidemiology and Cancer Area, INCLIVA, Instituto de Investigación Biomédica, Valencia, SPAIN, 6Medical Oncology, Hospital Universitario Morales Meseguer, Instituto Murciano de Investigación Biosanitaria, IMIB; Department of Medicine, Medical School, University of Murcia; GEICAM Spanish Breast Cancer Group, Murcia, SPAIN, 7Medical Oncology, INCLIVA, Instituto de Investigación Biomédica, Valencia, SPAIN, 8Medical Oncology, Hospital Universitario Morales Meseguer; GEICAM Spanish Breast Cancer Group, Murcia, SPAIN, 9Medical Oncology, Hospital Clínico Universitario de Valencia; INCLIVA, Instituto de Investigación Biomédica; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 10Medical Oncology, Hospital Clínico Universitario de Valencia; INCLIVA, Instituto de Investigación Biomédica, Valencia, SPAIN, 11Statics department, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 12Translational, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 13Medical Oncology, Hospital Universitario Fundacion Jimenez Diaz; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: Luminal BC represents around 70% of BC cases. Primary or secondary resistance to endocrine therapy (ET) is a treatment limiting factor. Little is known about the potential relationship between ET response and IIS. The main aim of this study is to characterize the IIS based on levels of circulating cytokines in luminal advanced BC. We previously reported first data about this relation, we do now explore its potential predictive and prognostic value and role in hormone resistance development. Methods: NIKOLE is a multicenter, prospective, observational, no post-authorization study in luminal advanced HER2-negative BC pts assigned to two different hormone-resistance cohorts (CohA, hormone-sensitive or CohB, hormone-resistant). Expression levels of cytokines and/or rNKG2D ligands: IL15, Granzyme, MICA, MICB, Perforin, IFNγ, IL6, IL8, IL10, TNFα, and VEGFA, were measured by the MILLIPLEX® immunoassays HCYTA-60K and HCKP2-11K in serum from serial peripheral blood collected at baseline (pts and healthy controls), after 6 weeks, at radiological re-evaluation (3 months after ET initiation) and progressive disease (PD). Log2 expression cytokine values were compared between groups and correlated with outcome including Clinical Benefit (CB) and Progression Free Survival (PFS). Continuous variables were compared using Student´s t-test, and contingency tables were analyzed with Fisher’s exact test. Differences in Kaplan-Meier (KM) plots were evaluated using log-rank test, Cox regression models were fitted and Bonferroni correction applied to adjust for multiple comparisons. Results: NIKOLE enrolled 6 healthy controls and 31 pts with advanced luminal BC treated with first or second ET. 25 (78%) first treatment pts were only considered, clinical characteristics are summarized in the table. No differences in baseline cytokines levels were found between pts vs controls nor CohA vs CohB. We observed significant increased levels between baseline and 3 months of IL15 (p=0.006), Granzyme (p=0.027), MICA (p=0.009) and MICB (p=0.018) and decreased VEGFA (p=0.005) and IFNγ levels (p=0.005), changes that were independent of pts hormone sensitivity status at the start of treatment. We also found better CB in pts with lower levels at 3 months of IFNγ (p=0.04) and VEGFA (p=0.04). Finally, MICA and granzyme baseline levels had prognostic value: pts with lower levels (median value as cutoff) showed longer PFS (26.55 vs 10.91 months) (p=0.035) and 26.55 vs 15.15 months (p=0.03), respectively. Conclusions: Here we highlight the importance of the IIS in advanced Luminal BC pts and the role of circulating cytokines as potential response predictive and prognostic biomarkers. Our results warrant additional research to investigate the specific role of the cytokines in the hormone-resistant BC and hopefully help in better pts’ selection to new immune therapies.
| Cohorts | CohA (n=16) | CohB (n=9) |
| Age (years), mean | 59 | 62 |
| Post-Menopausal Status, n (%) | 11 (69) | 9 (100) |
| ECOG Performance Status, n (%) (0; 1; 2) | 11 (69); 5 (31); 0 | 7 (78); 0; 2 (22) |
| Metastasis location, n (%) (Visceral; Bone; LN) | 9 (56); 11 (69); 7 (44) | 6 (67); 6 (67); 2 (22) |
| Histology, n (%) (Ductal NOS; Lobular Classical; Lobular Variant) | 13 (81); 2 (13); 1 (6) | 9 (100); 0; 0 |
| Tumor subtype, n (%) (LumA; LumB (HER2-); Lum NOS (HER2-)) | 9 (56); 6(38); 1(6) | 2 (22); 7(78); 0 |
| Pts status, n (%) | ||
| Treatment on-going | 9 (56) | 1 (11) |
| Discontinued for progression | 6 (38) | 6 (67) |
| Discontinued for death | 1 (6) | 1 (11) |
| Discontinued for Abemaciclib toxicity | 0 | 1 (11) |
| Treatment* (HT + TT combination), n (%) | ||
| AI + CI | 14 (88) | 2 (22) |
| AI + TKI | 1 (6) | 0 |
| ASI + CI | 0 | 1 (11) |
| SERMS + CI | 1 (6) | 3 (33) |
| SERMS + TKI | 0 | 1 (11) |
| SERDS + CI | 0 | 2 (22) |
| Outcome variables, n (%) | ||
| Clinical Benefit Rate (yes, SD 24 weeks) | 10 (63) | 5 (56) |
| Progression (yes) | 6 (38) | 6 (67) |
|
*LN= lymph node HT=Hormonotherapy TT= Targeted Therapy AI= Aromatase Inhibitor CI= Cyclin Inhibitor TKI= Tyrosine Kinase Inhibitor ASI= Adrenal Steroids Inhibitor SERMS= Selective Estrogen Receptor Modulators SERDS= Selective Estrogen Receptor Degraders SD= stable disease
|
Presentation numberPS1-12-03
Aberrant exon skipping of ALDOA confers tamoxifen resistance in breast cancer
Ying Duan, Yangzhou University, Yangzhou, China
Y. Duan, S. Yu, R. Sang, S. Zhang, Y. Yang, C. Bi, H. Sun; Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, CHINA.
Breast cancer is the most commonly diagnosed malignancy worldwide, with over 2.6 million new cases reported annually. A substantial proportion of these cases are estrogen receptor-positive (ER+), representing the most prevalent molecular subtype. Standard treatment for ER+ breast cancer typically includes adjuvant endocrine therapy with tamoxifen (TAM), which functions by antagonizing estrogen receptor signaling to reduce the risk of disease recurrence following surgery. Despite its clinical efficacy, more than 20% of patients experience either primary or acquired resistance to TAM, ultimately contributing to treatment failure and increased mortality. Cancer-associated alternative splicing (AS) events give rise to aberrant transcript variants that contribute to uncontrolled cell proliferation and therapeutic resistance. Despite growing evidence linking AS to tumor progression, the specific splicing variants involved in TAM resistance in breast cancer remain largely undefined. In this study, we profiled the landscape of AS events in nine pairs of matched primary and relapsed breast tumors from patients treated with TAM-based therapy. Through integrative experimental validations including Ribo-qPCR, RNA pull-down, mass spectrometry, and dual-luciferase reporter assays, we identified a key ALDOA splice variant associated with TAM resistance. Specifically, the inclusion of ALDOA exon 7.2 enhances the translation efficiency of the transcript, resulting in increased ALDOA protein expression, mTOR pathway activity, and the promotion of TAM resistance in breast cancer cells. Moreover, the inclusion of exon 7.2 in ALDOA mRNA is mediated by MSI1 via direct interaction. Elevated inclusion of ALDOA exon 7.2 or expression of MSI1 is associated with an unfavorable prognosis in patients undergoing endocrine therapy. Notably, treatment with Aldometanib, an ALDOA inhibitor, effectively restrains the growth of TAM-resistant breast cancer cells in vitro and in vivo. In conclusion, we provides a promising therapeutic avenue targeting ALDOA to combat TAM resistance.
Presentation numberPS1-12-05
Liquid Biopsy-Based Molecular Profiling Using Guardant360 CDx at Progression on CDK4/6i+ET: Findings from the AGO-B CAPTOR Study
Volkmar Mueller, Hamburg-Eppendorf University Medical Center, Hamburg, Germany
V. Mueller1, H. Huebner2, P. Ziegler2, S. Uhrig2, S. Brucker3, V. Thewes4, L. L. Volmer3, A. Hartkopf3, T. Engler3, C. C. Hack2, I. Juhasz-Boess5, H. Kolberg6, D. Lueftner7, M. P. Lux8, M. Schmidt9, H. Tesch10, M. Thill11, M. Untch12, P. Wimberger13, S. Heublein14, I. Nel15, H. Neubauer16, B. Rack14, J. C. Radosa17, F. Taran18, B. Aktas15, N. Ditsch19, L. Haeberle2, I. Sava-Piroddi20, R. Weinhold20, C. Mann20, E. Belleville21, K. Lüdtke-Heckenkamp22, M. van Mackelenbergh23, R. Pihusch24, C. Schem25, K. Apel26, H. Wagner27, A. Schneeweiss4, T. Lotz28, W. Janni14, T. N. Fehm16, P. A. Fasching2, B. Lex29; 1Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, GERMANY, 2Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen University Hospital, Department of Gynecology and Obstetrics, Erlangen, GERMANY, 3Department of Obstetrics and Gynecology, University Hospital of Tuebingen, Tuebingen, GERMANY, 4National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, GERMANY, 5Department of Gynecology, Albert-Ludwigs-University Freiburg, Freiburg im Breisgau, GERMANY, 6Department of Gynecology and Obstetrics, Marienhospital Bottrop, Bottrop, GERMANY, 7Immanuel Hospital Märkische Schweiz & Immanuel Hospital Rüdersdorf, Medical University of Brandenburg, Rüdersdorf bei Berlin, GERMANY, 8Department of Obstetrics and Gynecology, St. Louise Hospital, Paderborn, St. Josefs-Hospital, Salzkotten, St. Vincenz Hospital, Paderborn, GERMANY, 9Department of Obstetrics and Women’s Health, University Medical Center Mainz, Johannes Gutenberg University, Mainz, GERMANY, 10Oncology Bethanien, Center for Hematology and Oncology, Frankfurt am Main, GERMANY, 11Department for Gynecology and Gynecologic Oncology, Agaplesion Markus Hospital, Frankfurt am Main, GERMANY, 12Department of Obstetrics and Gynecology, Helios Clinics Berlin-Buch, Berlin, GERMANY, 13Department of Obstetrics and Gynecology, Technical University Dresden, Nationales Centrum für Tumorerkrankungen (NCT/UCC), Dresden, GERMANY, 14Department of Gynecology and Obstetrics, Ulm University Hospital, Ulm, GERMANY, 15Clinic and Polyclinic for Gynecology, University of Leipzig, Department of Gynecology, Medical Center, Leipzig, GERMANY, 16Department of Obstetrics and Gynecology, Center for Integrated Oncology (CIO Aachen, Bonn, Cologne), University Hospital and Medical Faculty of the Heinrich-Heine University Duesseldorf, Düsseldorf, GERMANY, 17Department of Obstetrics, Gynecology and Reproductive Medicine, Saarland University Hospital, Homburg, GERMANY, 18Department of Gynecology and Gynecologic Oncology, University Clinic Cologne, Köln, GERMANY, 19Gynecology, Obstetrics and Senology, Faculty of Medicine, University of Augsburg, Breast Center, University Hospital Augsburg, CCC Augsburg, Germany, Bavarian Cancer Research Center (BZKF), Augsburg, GERMANY, 20Oncology, Novartis Pharma GmbH, Nürnberg, GERMANY, 21Clinical Research Oncology, ClinSol GmbH & Co KG, Würzburg, GERMANY, 22Center for Hematology and Oncology, Niels Stensen Clinics, Franziskus Hospital Harderberg / Georgsmarienhütte, Georgsmarienhütte, GERMANY, 23Campus Kiel, University Hospital Schleswig-Holstein, Kiel, GERMANY, 24Oncologicum Rosenheim, medicum Rosenheim MVZ, Rosenheim, GERMANY, 25Mammazentrum, Jerusalem Hamburg Hospital, Hamburg, GERMANY, 26Oncology, Dr. Apel Medizinische Versorgung GmbH, Erfurt, GERMANY, 27Hematology and Medical Oncology, Joint Oncology Practice at Fürth Medical Center, Fürth, GERMANY, 28Medical Unit, GUARDANT Health, Palo Alto, CA, 29Department of Gynecology and Obstetrics, Kulmbach Hospital, Kulmbach, GERMANY.
Background: CDK4/6 inhibitors in combination with endocrine therapy have become the standard first-line treatment for patients with hormone receptor-positive, HER2-negative (HR+/HER2-) advanced breast cancer. While ribociclib is supported by the most robust overall survival data across multiple phase III trials, mechanisms of resistance and biomarkers for treatment efficacy remain incompletely understood. Liquid biopsies, particularly of circulating tumor DNA (ctDNA) analysis, offer an opportunity to characterize tumor evolution and identify actionable genomic alterations at clinically relevant timepoints. The AGO-B “Comprehensive Analysis of Spatial, Temporal and Molecular Patterns of Ribociclib Efficacy and Resistance in Advanced Breast Cancer Patients” (CAPTOR) trial (NCT05452213) is a single-arm, open-label phase IV study of patients with advanced HR+/HER2- breast cancer who were treated with ribociclib and endocrine therapy. Methods: Patients with HR+/HER2- advanced breast cancer receiving first-line treatment with ribociclib plus endocrine therapy were prospectively enrolled in the CAPTOR trial. At time of progression or end of treatment (EoT) patient were eligible for a Guardant360 CDx test. The test was initiated at physicians’ discretion. Blood samples were collected using Streck cfDNA BCT tubes and shipped to the GUARDANT Health central laboratory. ctDNA was extracted and sequenced using the FDA-approved Guardant360 CDx assay, covering 74 cancer-related genes and detecting single nucleotide variants (SNVs), insertions/deletions (indels), gene fusions, and copy number alterations. Testing results were returned to the treating physician via the GUARDANT health portal or via PDF upload into the CAPTOR electronic case report form. Results: Guardant360 CDx testing was initiated for 37 patients between December 2024 and June 2025. Of those, two were cancelled due to shipment delays and four were not yet processed at time of interim analysis, resulting in 31 successful ctDNA GUARDANT tests. The median time till receipt of samples at the GUARDANT central laboratory was 3 days (range 1-8) and the median time till report of test results was 12 days (range 7-53). Thirty (97%) had at least one alteration in the analyzed genes. The most frequent genes affected were PIK3CA (N=15, 48%), TP52 (N=12, 39%) and ESR1 (N=10, 32%). Two patients (6%) had a BRCA1 and three (10%) a BRCA2 mutation. Of those, one patient each had a potentially germline mutation in BRCA1 (41.5% variant allele frequency [VAF]) or BRCA2 (47.59% VAF). The median VAF was 0.83 (range 0.02-47.59). For twenty-two (71%) patients at least one alteration was detected with an approved indication in breast cancer. Twelve (39%) had an indication for Alpelisib due to an activating mutation in PIK3CA, nine (29%) an indication for Elacestrant based on an activating mutation in ESR1, sixteen (52%) an indication for Capivasertib due to PIK3CA, AKT1 or PTEN mutation and one patient (3%) had a ERBB2 amplification resulting in an indication for an anti-HER2 treatment. Of those, with a treatment indication based on ctDNA test results, treatment information after progress or EoT was available from seventeen (77%) patients. Of those, seven (41%) received treatment in accordance with ctDNA test result (29% Capivasertib, 6% Trastuzumab, 6% Elacestrant). Discussion: These interim results demonstrate the feasibility and clinical utility of Guardant360-based ctDNA testing at progression or end of therapy within the CAPTOR trial. A high proportion of patients showed actionable alterations, and nearly half of those with available follow-up received targeted treatment in line with the ctDNA findings.
Presentation numberPS1-12-06
Ribo-age: age-stratified descriptive analysis of adverse drug reactions in patients with advanced breast cancer treated with ribociclib plus nonsteroidal aromatase inhibitors: preliminary results
Tamara Díaz-Redondo, Regional and Virgen de la Victoria University Hospitals, Málaga, Spain
T. Díaz-Redondo1, R. Sánchez-Bayona2, P. Zamora Auñon3, I. Garcia-Fructuoso4, A. Bujosa5, J. Sánchez6, J. Ponce Lorenzo7, A. Jiménez Ortega8, E. Marin Olivo8, A. Corbera8, A. López9; 1Medical Oncology, Intercentre Clinical Management Unit, Regional and Virgen de la Victoria University Hospitals, Málaga, SPAIN, 2Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 3Medical Oncology Department, La Paz University Hospital., Madrid, SPAIN, 4Medical Oncology Department, Hospital Clinic of Barcelona, Translational Genomics and Targeted therapies in Solid Tumors, IDIBAPS, Barcelona, SPAIN, 5Medical Oncology Department, Hospital Son Llàtzer, Palma de Mallorca, Balearic Islands, SPAIN, 6Medical Oncology Department, Puerta de Hierro-Majadahonda University Hospital, Madrid, SPAIN, 7Oncology Department, University General Hospital Dr. Balmis, Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, SPAIN, 8Medical Affairs, Novartis, Barcelona, SPAIN, 9Oncology Department, University Hospital of León, León, SPAIN.
Background: Ribociclib (RIB), a CDK4/6 inhibitor, in combination with endocrine therapy (ET), has demonstrated significant improvements in progression-free survival (PFS) and overall survival (OS) in patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative (HR+/HER2−) advanced breast cancer (ABC), as evidenced by the MONALEESA phase III trials. Despite these robust clinical trial outcomes, to our knowledge, real-world data on safety and tolerability of RIB + ET, in the Spanish healthcare setting, remain scarce. This study aims to address this gap by evaluating the safety profile of RIB in combination with a non-steroidal aromatase inhibitor (NSAI) as first-line (1L) therapy in both younger and older patients with HR+/HER2− ABC, based on routine clinical practice in Spain. In this report, we show a preliminary result from the first 147 patients. The final analysis will include at least 500 patients. Methods: This is a non-interventional, retrospective cohort study involving patients diagnosed with HR+/HER2− ABC who initiated 1L treatment with RIB + NSAI between January 2021 and August 2023. Data was collected exclusively from medical records across participating centers. Following the final patient inclusion date, a subsequent 12-month follow-up period (until August 2024) is established to ensure adequate exposure to RIB across all participants and to monitor the potential emergence of adverse events (AEs). If discontinuation occurs for any reason, the duration of exposure to RIB was measured. Results: The median follow-up was 23.8 months (range: 3.1 – 43.4), and the median time on treatment was 18.39 months (range: 0.5 – 43.4). Stratified by age, the PFS rate at 24 months was 63.3% (95% CI: 52.6 – 72.3) in patients aged ≤ 65 years and 65.5% (95% CI: 47.0 – 78.9) in those aged > 65 years. In general, the safety profile was consistent with previously reported data, with no new safety signals. From 141 patients reporting at least one AEs, 16 discontinued RIB due to AEs (10.9% of the whole population). The two most common AEs for both, safety and tolerability are summarized in Table 1. Conclusions: Preliminary real-world data suggest that RIB + NSAI is well tolerated in both younger and older patients with HR+/HER2− ABC in the Spanish healthcare setting. PFS outcomes appear consistent across age groups. Final results from the full set of at least 500 patients will provide more definitive insights into the safety profile and treatment outcomes.
Presentation numberPS1-12-07
Impact of TP53 mutations on Treatment Duration with CDK 4/6 Inhibitors and Immune Checkpoint Inhibitors in ER-positive/HER2-negative Breast Cancer: Insights from Real-World Data
Masanori Oshi, Roswell Park Comprehensive Cancer Center, Buffalo, NY
M. Oshi1, M. Sugimori2, K. Kawashima1, A. Yamada3, K. Narui4, T. Ishikawa5, K. Takabe1; 1Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Cancer Genome Medicine, Yokohama City University Medical Center, Yokohama, JAPAN, 3Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 4Breast and Thyroid Surgery, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 5Breast Surgery and Oncology, Tokyo Medical University, Tokyo, JAPAN.
Background: TP53 mutation is a key driver of breast cancer. However, its clinical impact remains unclear. This study investigates the relationship between TP53 mutations and response to CDK 4/6 inhibitors (CDK4/6i) and immune checkpoint inhibitors (ICI) in breast cancer using real-world data (RWD) from the C-CAT (Center for Cancer Genomics and Advanced Therapeutics) cohort, with particular focus on age-related differences. The duration of treatment was used as a surrogate of unresponsiveness to treatment and progression of disease during treatment. Material and Methods: We used METABRIC data for initial survival analysis and performed Gene Set Enrichment Analysis (GSEA) to identify pathway enriched in TP53-mutated ER-positive/HER2-negative breast cancers. We also investigated the relationship of TP53 mutations and durations of CDK4/6i and ICI treatments in the C-CAT cohort, a database that includes clinical and genomic data of metastatic breast cancer patients from Japan. To evaluate treatment resistance in metastatic breast cancer, we used treatment duration as a clinical endpoint. The CCAT cohort was stratified by age into four groups: Adolescent and Yong Adults (AYA: 15-39 y.o.), perimenopausal (40-54 y.o.), menopausal (55-64 y.o.), and old (over 65 y.o.). Results: TP53 mutations were significantly associated with poor prognosis in ER-positive/HER2-negative breast cancer in the METABRIC cohort. GESA revealed that TP53-mutated tumors were enriched with pro-cancerous gene sets, particularly those related with cell proliferation and immune response, while TP53-wild type tumors enriched estrogen response gene sets. These findings led to investigating the impact of TP53 mutations on durations of CDK4/6i and ICI treatments. In the CCAT cohort. TP53 mutation was not associated with the duration ICI treatment. On the other hand, TP53 mutation was significantly associated with shorter duration of CDK4/6i treatment in entire ER-positive/HER2-negative breast cancer, but not in the patients who received CDK4/6i starting from late treatment phase. This trend was consistent across both Abemaciclib and Palbociclib, suggesting that TP53 mutations similarly affect treatment duration in both drugs. Further, AYA patients had the shortest treatment duration compared to the other age groups, with an earlier initiation of CDK4/6i treatment. The prevalence of TP53 mutation decreased with increasing age. When analyzing the impact of TP53 mutations by age group, TP53 mutation was significantly associated with shorter treatment duration in the perimenopausal, menopausal, and old groups, while no significant difference was found in the AYA group. Conclusion: This study demonstrated that TP53 mutation is associated with shorter duration of CDK4/6i treatment, particularly in the early stages, with similar trends observed for Abemaciclib and Palbociclib. These findings highlight the need for personalized treatment strategies based on TP53 mutation status. While the impact of TP53 mutations on ICI treatment duration was not significant, it suggests that other factors may influence ICI efficacy. Additionally, age-specific therapeutic strategies should be considered, as the impact of TP53 mutations on treatment duration varies by age group.
Presentation numberPS1-12-08
Real-world ESR1 mutation (ESR1m) testing and positivity rates in patients with ER+, HER2- metastatic breast cancer (MBC)
Lindsay A Williams, Eli Lilly and Company, Indianapolis, IN
C. Liao1, N. Bansal2, J. John3, M. Bhave4, F. Bidard5, T. Marquart6, A. Chawla7, L. Williams8; 1Value Evidence Outcomes – Oncology, Eli Lilly and Company, Indianapolis, IN, 2Value Evidence Outcomes, Eli Lilly and Company, Indianapolis, IN, 3Virtual Medical Hub, Eli Lilly and Company, Indianapolis, IN, 4Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 5Department of Medical Oncology, Institut Curie, Paris, FRANCE, 6Oncology Medical Affairs, Eli Lilly and Company, Indianapolis, IN, 7Global Medical Affairs, Eli Lilly and Company, Indianapolis, IN, 8HEOR Oncology, Eli Lilly and Company, Indianapolis, IN.
Background: As the MBC treatment landscape evolves, understanding ESR1m testing patterns for ER+, HER2- MBC is essential to identify patients for targeted therapies. Despite existing guidelines recommending testing of patients with MBC at progression for ESR1m, real-world data on testing and mutation rates are limited. Therefore, we described ESR1m testing and positivity (+) rates in patients with ER+, HER2- MBC to identify opportunities for improved ESR1m test utilization. Methods: This retrospective, observational study included patients aged ≥18 years at ER+, HER2- MBC diagnosis (Dx) who initiated 1st line (1L) therapy with aromatase inhibitors (AI) or selective estrogen receptor degraders (SERDs) ± CDK4/6 inhibitors (CDK4/6i) from 1/1/20-3/31/25. Flatiron Health Research Database (FHRD), a de-identified US-based electronic health record database, was used to estimate ESR1m+ rates in windows defined as: A) initial early breast cancer (EBC) Dx date or 90 days before de novo MBC Dx to last patient contact, B) MBC Dx date to 1L initiation date+28 days, C) during line of therapy (LoT;start to end date), and D) at LoT initiation (90 days before to 28 days after LoT initiation). Results: Of all 12148 patients, 48% (n=5874) had ≥1 ESR1m test and most tested patients (n=3650/5874; 62%) were tested only once (mean:1.6 test/patient). Among all patients, testing rates were similar at each LoT initiation (25-29%). In exploratory analyses of all patients, post-2023 testing rates were often higher than pre-2023, especially at 1L (34 vs 20%) and 2L initiation (36 vs 26%). When considering patients with data available on ESR1m testing by sample type (n=2028), ~60% were in tissue only and ~40% were in blood only. Among a subgroup of patients with de novo MBC who received 1L AI±CDK4/6i, testing rates at 2L and 3L initiation were 27-28%. For the subgroup of patients with EBC who relapsed and developed MBC while on adjuvant AI±CDK4/6i or ≤12 months after completion, the testing rate was the highest during 1L (38%). Among all patients, ESR1m+ was 15% during 1L and increased to 37% at 3L initiation. Among patients with de novo MBC (received 1L AI±CDK4/6i), ESR1m+ was highest at 3L initiation (40%). Among the relapsed patients, ESR1m+ was 23% at 1L initiation, 31% at 2L initiation, and 33% at 3L initiation. Conclusion: Among all patients with MBC, 52% were never tested and only 34-36% were tested at 1L and 2L initiation from 2023 onward. Importantly, lower rates of ESR1m+ were observed in the real-world compared to clinical trials. This may be due to suboptimal test timing, poor patient identification for testing, and frequent use of tissue-based testing, which is less sensitive than the recommended blood-based ctDNA used in clinical trials. These results highlight an unmet need for increased ESR1m testing using blood-based ctDNA to optimize patient identification for targeted therapies in ER+,HER2- MBC.
Presentation numberPS1-12-09
Therapeutic Implications of Concurrent ESR1 and PI3K Pathway Mutations in HR+/HER2- Metastatic Breast Cancer
Kimberly Boldig, Henry Ford Health, Detroit, MI
K. Boldig1, S. Ghosh2, V. Dabak1; 1Hematology Oncology, Henry Ford Health, Detroit, MI, 2Public Health Sciences, Henry Ford Health, Detroit, MI.
Individualized cancer treatments have revolutionized oncologic care for patients. Mutations in the ESR1 gene confer resistance to endocrine therapies by enabling estrogen receptor (ER) activity independent of estrogen binding. Similarly, PIK3CA mutations activate the PI3k/AKT/mTOR signaling pathway, promoting cellular proliferation, signaling, and metabolism. This signaling can sustain ER transcription in the absence of estrogen, resulting in resistance to anti-estrogen therapies. Recent therapeutic advances targeting ESR1 and the PI3k pathway have demonstrated improved progression-free survival (PFS). The EMERALD trial showed that elacestrant significantly prolonged PFS in patients with advanced hormone positive breast cancer and ESR1 mutations. Likewise, alpelisib and capivasertib improved PFS in patients with PIK3CA-mutated, HR+, HER2- advanced breast cancer who had progressed on prior endocrine therapy. Little is known about the clinical significance of concurrent ESR1 and PIK3CA mutations in breast cancer patients. Both are associated with worse prognosis and resistance to endocrine therapy. We identified 58 patients at our institution with ESR1 mutations and 99 patients with PIK3CA mutations. Our dataset specifically analyzed 30 patients with concurrent mutations who were treated with ESR1 andPIK3CA inhibitors. In this cohort, the median PFS of 6.0 months (mo) (95% CI: 3.91 – 8.09), and the median overall survival (OS) of 22 mo (95% CI: 19.71-24.29). All patients were female, with a median age of 56 years (range: 32-74 years). Most had previously received a CKD4/6 inhibitor (91.7%). The cohort was predominantly non-Hispanic white (83.3%), with 4% black and 1% Hispanic. At the time of analysis, 76.7% had experienced disease progression or death. Patients with concurrent mutations who were treated with alpelisib had a median PFS of 14.50 mo (95% CI: 8.01-20.91) and a median OS of 24.56 mo (95% CI: 13.79-35.32). Patients treated with capivasertib had a median PFS of 6.79 mo (95% CI: 3.26-10.32) and all patients remain alive. Median PFS for those treated with elacestrant was 12.77 mo (95% CI: 7.19-18.34) and median OS was 19.32 mo (95% CI: 16.83-21.81). Patients who received both elacestrant and a PIK3CA inhibitor had a median PFS of 11.85 mo (95% CI: 5.79-17.91), median OS of 42.10 mo (95% CI: 30.26-53.94). When patients received elacestrant first, followed by a PIK3CA inhibitor, median PFS was 5.92 mo (95% CI: 3.40-8.44), median OS was 18.90 mo (95% CI: 15.74-22.06). Alternatively, if a PIK3CA inhibitor was given first followed by elacestrant, median PFS was 14.01 mo (95% CI: 5.48-22.54), median OS was not reached. Patients with concurrent mutations who had received both medications were further analyzed based on location of metastasis. Those with only bone metastases had a PFS of 15.93 mo (95% CI: 7.66-24.16), OS of 44.20 mo (95% CI: 27.54-60.87). Those with visceral metastases had PFS of 8.39 mo (2.55-14.23), OS of 20.54 mo (95% CI:14.75-26.32). These findings align with results from the SOLAR-1 and CAPItello-291 trials. However, our data uniquely demonstrates that patients with concurrent ESR1 and PIK3CA mutations, who were treated initially with PIK3CA inhibitor followed by elacestrant, had longer PFS and OS. This sequencing may offer improved outcomes, as PI3K mutations may confer a worse prognosis than ESR1 mutations. Prior studies, including EMERALD and EMBER-3, have indicated shorter PFS in patients with PI3K mutations treated with oral SERDs compared to those without PI3K pathway alterations. These findings highlight the need for future studies comparing institutional data across larger cohorts and prospective trials to better define optimal sequencing strategies for this subset of patients.
Presentation numberPS1-12-10
Real-world effectiveness of CDK4/6 inhibitors in HR+/HER2- advanced breast cancer with bone marrow involvement: A case series from a single institution
William A Mantilla, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC)., Bogota, Colombia
W. A. Mantilla1, F. Canro2, M. A. Bravo1, P. Lopez3, D. Rubio3, H. Carranza1, S. X. Franco1; 1Breast Cancer Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC)., Bogota, COLOMBIA, 2Innpatient unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC)., Bogota, COLOMBIA, 3Pathology Unit, Keralty Group, Bogota, COLOMBIA.
Background: Bone marrow (BM) metastasis is a rare and challenging manifestation of advanced breast cancer (ABC), typically associated with high hematologic toxicity risk and poor outcomes. These patients are frequently excluded from clinical trials, limiting available evidence to guide treatment decisions. While chemotherapy is commonly recommended to achieve a rapid response, including hematologic recovery, CDK4/6 inhibitors (CDK4/6i) are increasingly used due to their efficacy in HR+/HER2- ABC. We aimed to assess the real-world (RW) efficacy of CDKis in this high-risk subgroup. Methods: We conducted a retrospective study of patients with HR+/HER2- ABC and BM involvement treated with CDK4/6i between 2022 and 2025 in a single institution located in Bogota, Colombia. Eligibility required histologic or smear-confirmed BM metastasis and receipt of at least one cycle of CDKi (palbociclib, ribociclib, or abemaciclib) in combination with endocrine therapy. Descriptive statistics were used to summarize clinicopathologic characteristics and hematologic outcomes. RW progression-free survival (rwPFS) was also described using the Kaplan-Meier method. Results: Fourteen patients were identified through retrospective chart review, two patients were excluded for being HER2-positive BC, two had HR positive BC but did not receive CDK4/6i, and one died before completion of the first treatment month with CDK4/6i. Nine patients were included (median age: 67 years). All were post menopausal with confirmed BM metastasis; De novo disease was present in 44.5% of the cases, and 8 Patients were ECOG <2; 55.5% had both bone and visceral disease. CDKis used included ribociclib (n=5), palbociclib (n=3), and abemaciclib (n=1). Hematologic abnormalities at baseline included anemia (66.6%), neutropenia (44.4%), and thrombocytopenia (22.2%). A hematologic response was observed in 44.4% at 6 months, with hemoglobin recovery being the most frequent and clinically relevant outcome. Median rwPFS was 32.9 months, combining progression and censoring by last follow-up. Dose was infrequent, with only one patient requiring dose adjustment. Conclusions: CDK4/6 inhibitors appear to be a feasible and effective treatment option in patients with HR+/HER2- ABC and BM involvement. Despite cytopenias traditionally being considered a contraindication for CDKi, a notable proportion of patients achieved hematologic recovery and clinical benefit. rwPFS was comparable to pivotal trials and similar reported cohorts. These findings support the cautious use of CDKis in this underserved population, and emphasize the value of RW data in informing care for this poor prognosis group.
Presentation numberPS1-12-11
Giredestrant immobilizes the Estrogen Receptor to potently suppress ER-active breast cancers
Ciara Metcalfe, Genentech, South San Francisco, CA
J. Guan1, W. Zhou1, J. Liang2, A. Collier1, P. Perez-Moreno3, M. Hafner4, C. Metcalfe1; 1Discovery Oncology, Genentech, South San Francisco, CA, 2Translational Medicine, Genentech, South San Francisco, CA, 3Product Development Oncology, Genentech, South San Francisco, CA, 4Computational Science, Genentech, South San Francisco, CA.
Estrogen Receptor α (ER) is one of the most successfully prosecuted therapeutic targets in oncology, with multiple ER-targeted therapies approved for the treatment of breast cancer. Understanding the limitations of approved agents re-energized the breast cancer community towards the development of next-generation endocrine therapies to further benefit patients. In particular, orally bioavailable selective ER antagonists and degraders (SERDs) were designed to overcome exposure limitations of fulvestrant and achieve deep and sustained ER inhibition, to both treat and delay resistance. Results from randomized clinical trials in women with pre-treated advanced metastatic breast cancer (mBC) support potential superiority of latest-generation SERDs in patients harboring ESR1-mutant tumor cells, while the gains for patients with no mutations detected appears more limited. It was hypothesized that ER degraders with a distinct mechanism of action i.e. proteolysis-targeting chimeras (PROTACs) that are engineered to directly recruit the CRBN E3 ligase to ER, would drive more robust depletion of ER protein and thus extend benefit to ER wildtype tumors. Recent data from the VERITAC-2 trial do not support that hypothesis, likewise demonstrating a more prominent advance for patients harboring ESR1 mutations versus those with no mutations detected. We evaluated the activity of investigational and approved endocrine therapies in 22 ER-expressing breast cancer models in vitro, and observed a broad range of anti-proliferative activity spanning complete arrest to no measurable impact. Leveraging a previously curated ER activity signature we discovered that a key determinant of SERD, in particular giredestrant, sensitivity is the extent of ER activation prior to treatment. Most critically, differentiation of endocrine therapies with distinct modes of action occurs most clearly in cell lines with high ER activity; ER activity-low cell lines are less sensitive to endocrine therapies and fail to discriminate between agents. This finding is important given the extensive heterogeneity in ER activity in 2/3L mBC, observed by leveraging the same ER activity signature in biopsies from patients participating in giredestrant trials. We next explored differentiating attributes of SERDs and PROTACs. Transcription factors, including ER, move dynamically within the nucleus to engage DNA and trigger gene expression. In prior studies, we proposed that a major mechanism through which SERDs achieve full ER antagonism is via profound reduction of ER mobility. We show that giredestrant, like other full ER antagonists, immobilizes ER, while ER PROTACs do not; we thus uncouple ER immobilization and ER degradation as distinct pharmacological features. PROTACs can promote greater degradation of ER protein than is achieved by giredestrant, however, increased ER depletion does not result in superior anti-proliferative potential. We propose that ER immobilization by SERDs is a key pharmacological attribute driving ER inhibition even in the absence of ER elimination. Thus, sub-maximal ER degradation by SERDs likely does not, in this context, represent a limitation to be overcome by direct E3 ligase recruitment. Rather, the limited gains observed for novel SERDs in 2/3L ESR1 wildtype mBC is more likely a function of tumor biology, whereby those tumors tend to be ER activity-low, a context in which differentiation of endocrine therapies is challenging. Advances delivered by latest-generation SERDs may be more apparent where ER activity is more consistently high; a number of ongoing clinical trials, including those in earlier treatment lines, will test this hypothesis.
Presentation numberPS1-12-12
Comparative transcriptomic and proteomic analysis of lasofoxifene and fulvestrant, in an ER-mutated metastatic breast cancer explant model, identifies potential molecular mechanisms underlying differential treatment efficacy
Muriel Laine, University Of Chicago, Chicago, IL
M. Laine1, D. A. Tieri1, C. Zhu2, I. Raman2, G. Chen2, B. S. Komm3, G. L. Greene1; 1BenMay, University Of Chicago, Chicago, IL, 2Microarray Core, UTSW, Dallas, TX, 3-, Sermonix Pharmaceutical, Columbus, OH.
Background: Metastatic breast cancer (mBC) is one of the leading causes of death among women. Mutations in estrogen receptor alpha (ERα) are acquired as the disease becomes resistant to current therapies, leading to tumor cell metastasis to distant organs. A combination of lasofoxifene (Las) and the CDK4/6 inhibitor abemaciclib has shown promising results in the ELAINE 2 trial, with significantly increased progression-free survival and good tolerability in mBC patients with activating ERα mutations who have progressed on endocrine/CDK4/6 therapy, including fulvestrant. In mouse studies, Las, as well as the Las plus palbociclib (Pal) combination, resulted in a significant reduction in primary tumor burden and metastasis compared to fulvestrant (Ful) and Ful plus Pal treatment.Methods: In this study, we used a mBC xenograft model with the ERα Y537S mutation (MCF7 ERα Y537S mutant) and GeoMx-DSP technology to investigate differences in transcriptional and protein expression levels between Las, Ful, and their combinations with Pal. We also examined differences between ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC).Results: As expected, our results show that, at both the transcriptome and protein levels, treatment with Las or Ful yields many similar outcomes. The addition of Pal shifts gene and protein expression in the same direction as Pal alone. While Las, like Ful, modulates the transcription of several genes involved in tumor growth and dissemination, the differences lie mainly in the intensity or significance of these alterations across several genes or proteins that lead to differences reflected in pathway analysis. Gene pathway analysis (GSEA/GOBP) using PanCK+ tumor cell selection suggests that Las, compared to vehicle, activates more immune pathways, while Ful activates more apoptosis pathways. Transcriptome analysis suggests that Ful is more involved in metabolic pathways, whereas Las treatment is more involved in actin filament modification and RNA/DNA modification pathways. Notably, PCNA protein expression is selectively repressed by Las in IDC compared to Ful. Conclusion: Our data reveal the complexity of Las vs Ful treatment outcomes in the MCF-7 Y537S mBC explant model and provide potential insights into the molecular mechanisms underlying the differential efficacy of Las and its combination with Pal in a metastatic breast cancer model with an activating ERα mutation.
Presentation numberPS1-12-13
Real-world rate of switching and reasons for switching among CDK4/6i drugs among first-line metastatic HR+/HER2- breast cancer patients
Sowjanya Reganti, Cancer Care Specialists, Reno, NV
S. Reganti1, R. Choksi2, V. Gorantla2, F. Kudrik3, D. Patt4, S. Reddy1, S. Rosenfeld5, D. Parris6, M. Wang6, A. Rui6, M. Gart6, C. Wall6, B. Wang6, P. Varughese6, J. Donegan6, L. Morere6, R. Geller6, J. Scott6, R. Mahtani7; 1Medical Oncology, Cancer Care Specialists, Reno, NV, 2Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 4Medical Oncology, Texas Oncology, Austin, TX, 5Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 6PrecisionQ, IntegraConnect, West Palm Beach, FL, 7Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: The combination of CDK4/6 inhibitor (CDK4/6i) (abemaciclib [abema], palbociclib [palbo], or ribociclib [ribo]) and endocrine therapy (ET) is standard-of-care for patients (pts) with hormone receptor-positive, HER2-negative metastatic breast cancer (HR+/HER2- mBC). These agents demonstrate similar improvements in progression-free-survival, but only ribo has shown consistent overall survival (OS) benefit across multiple phase III trials. This clinical benefit, as well as toxicity profiles, comorbid conditions, physician familiarity, and pt-specific factors, may influence initial selection and switching patterns in clinical practice. This study characterizes switching patterns in pts initiating first-line (1L) CDK4/6i + ET. Methods: We conducted a retrospective cohort study using the IntegraConnect PrecisionQ de-identified electronic health record (EHR) database, which includes data on >3 million cancer pts treated across >500 US care sites. Pts with HR+/HER2− mBC who initiated 1L treatment with abema, palbo, or ribo between 02/26/2018 and 05/16/2025 were included. Switching was defined as discontinuation of the initial CDK4/6i followed by initiation of another CDK4/6i. Data on whether the accompanying ET was changed at the time of the CDK4/6i switch were not assessed. Pts (n=179) were selected for a subset analysis based on the availability of curated discontinuation reasons (toxicity, disease progression, personal/financial), which were manually abstracted from clinical documentation. Pts were stratified by initial CDK4/6i, documented reason for switching, and time to switch (≤30, 31-90, 91-180, >180 days). Differences in time to switch by reason were assessed using Wilcoxon rank-sum testing. Results: Among 11,928 pts with HR+/HER2− mBC who initiated 1L treatment with CDK4/6i + ET, 7,499 used palbo, 2,371 used ribo, and 2,058 used abema. Of these pts, 577 pts (7.7%) switched from palbo, 307 (12.9%) from ribo, and 264 (12.8%) from abema to an alternative CDK4/6i. Toxicity was the most frequently documented reason for switching, reported in 91.8% of pts who switched from abema, 69.7% from ribo, and 49.5% from palbo. The median duration of therapy prior to switching for toxicity was longest for palbo (131 days), followed by abema (96 days) and ribo (95 days). Disease progression as a reason for switching was most commonly reported in pts initially treated with palbo (42.3%), followed by ribo (21.2%) and abema (6.1%). Pts who switched due to disease progression had a significantly longer time to switch than those who switched for other reasons (median 444 vs. 141 days; Wilcoxon P180 days for palbo (58.8%), whereas earlier switches (≤180 days) were more frequent for abema and ribo (~70%). Among those who switched from palbo, 64.1% switched to abema and 35.9% to ribo. Among pts who switched from ribo, 61.2% switched to palbo and 38.8% to abema. Of those switching from abema, 75.4% switched to palbo and 24.6% to ribo. Conclusion: In this real-world analysis of 1L CDK4/6i use in HR+/HER2- mBC, intra-class switching was common and most often driven by toxicity rather than disease progression. Time to switch varied by initial agent and reason for discontinuation with palbo users more frequently switching later and for progression, while earlier switches from abema and ribo were largely due to intolerance. This study’s retrospective design may introduce selection bias, and reasons for switching were not always documented. Comorbidities, tumor burden, prior treatments, and access to care were not captured, representing potential unmeasured confounders. Additionally, evolving data availability and drug approvals during the study period may have influenced treatment choices and switching patterns.
Presentation numberPS1-12-14
Tolerance and clinical outcomes in elderly patients with hormone receptor positive breast cancer receiving CDK 4/6 inhibitors at a rural comprehensive cancer center
Shahab Haghollahi, Dartmouth Health, Lebanon, NH
C. Lapp1, M. Afzal2, X. Xia3, T. MacKenzie3, H. Kuznia1, S. Haghollahi1; 1Internal Medicine, Dartmouth Health, Lebanon, NH, 2Hematology/Oncology, Dartmouth Health, Lebanon, NH, 3Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH.
Background Cyclin dependent kinase 4/6 inhibitors (CDK 4/6is) are approved by the FDA in patients with HR+ metastatic and locally advanced breast cancer. Studies have shown that elderly women have similar clinical effects with CDK 4/6i use compared to younger women, but they experience higher rates of dose reductions and delays. The objective of this study was to evaluate real-world outcomes of elderly patients treated with CDK 4/6is to help elucidate the unique considerations necessary for treatment in the geriatric population. Our cancer center primarily serves rural and socioeconomically diverse patients who often require significant distance to travel for care. To evaluate the impact of potential geographic and socioeconomic disadvantages on the tolerance to CDK 4/6is and the clinical outcomes, we are investigating the impact of national and state area deprivation index (ADI) in this study, as we postulate that higher ADI may correlate with poorer clinical outcomes and tolerance. Methods This retrospective chart review was approved by the Institutional Review Board. Data gathering and analysis was performed by the authors. A total of 64 patients with HR+ breast cancer treated with ET and CDK 4/6i therapy aged 65 and over at the time of diagnosis who received care at the Dartmouth Cancer Center over the last five years were included in this study. Tolerance was defined by an aggregate score that incorporated the number of side effects, number of dose interruptions, number of dose reductions, and hospitalizations. National and state ADI were calculated by the regional zip codes. Results The median age at diagnosis was 72 years. 25% (16) patients had locally advanced disease and 75% (48) had metastatic disease at time of treatment. In patients with metastatic disease, median progression-free survival (PFS) was 10.5 months and median overall survival (OS) was 16.5 months. Median number of CDK 4/6i dose interruptions and reductions were 1 and 0, respectively. Median duration of dose interruption was 2 weeks. Median state and national ADI scores were 6 and 47, respectively. Neither state nor national ADI scores were significantly associated with tolerance. BMI was not significantly associated with tolerance. COX proportional hazard models did not show significant difference in survival outcomes in patients based on ADI (state ADI >5 or <5), age (less than 70 years vs. greater than 70 years), CDK 4/6i drug (abemaciclib vs. ribociclib vs. palbociclib), or endocrine therapy (anastrozole vs. fulvestrant vs. letrozole). Patients with higher ADI had a 1.4% higher hazard of death compared to those with lower ADI, but the p value is not significant (HR= 0.986). Older patients had a 15.5% lower hazard of death compared to younger patients, but the p value is not significant (HR= 0.845). Discussion/Conclusion Age, ADI, and BMI did not influence tolerance or outcome measures in our patient population. Pooled data from pivotal trials showed median PFS for patients over age 70 on CDK 4/6is and ET was 33.1 months, compared to 27.3 months in younger patients. Our patients (age 65+) had much lower median PFS of 10.5 months. This may be due, in part, to less fit patients and the presence of underlying comorbid conditions compared to more selective populations in pivotal trials. It is also possible that rurality itself may affect outcome measures in a distinct way that is not captured through ADI.
Presentation numberPS1-12-15
Metabolic complete response on 18FDG-PET-CT is a key predictor of Progression-Free Survival in metastatic luminal breast cancer treated with CDK4/6 inhibitors (CDK4/6i).
Elizabeth dos Santos, AC Camargo Cancer Center, São Paulo, Brazil
N. Soldi1, V. Basílio1, C. Machado2, E. Carneiro1, F. Balint1, L. Gouveia1, L. Leite1, F. Makdissi3, S. Sanches1, A. Balieiro1, V. de Lima1, E. dos Santos1; 1Oncology, AC Camargo Cancer Center, São Paulo, BRAZIL, 2Economic sciences, Universidade Federal de Minas Gerais, Belo Horizonte, BRAZIL, 3Mastology, AC Camargo Cancer Center, São Paulo, BRAZIL.
Background: CDK4/6 inhibitors combined with endocrine therapy are the standard first-line treatment for HR+/HER2- metastatic breast cancer (MBC). However, predictive markers of treatment benefit remain limited. We hypothesized that 18F-FDG PET-CT may provide early prognostic insights in HR+/HER2- MBC. We evaluated the association between metabolic complete response (CMR) on PET-CT and progression-free survival (PFS) in patients treated with first-line CDK4/6 inhibitors and endocrine therapy. Methods: We conducted a retrospective cohort study including 95 patients with HR+/HER2- metastatic breast cancer (MBC) treated with first-line CDK4/6 inhibitors in combination with endocrine therapy between 2018 and 2023. All patients underwent serial 18F-FDG PET-CT scans to assess treatment response. Complete metabolic response (CMR) was defined as the complete absence of metabolic uptake on PET-CT at any point during first-line therapy, and patients were categorized as CMR or non-CMR. The primary endpoint was progression-free survival (PFS), and the secondary endpoint was overall survival (OS). Survival analyses were performed using Kaplan-Meier estimates and Cox proportional hazards regression models to assess associations between metabolic response and clinical outcomes. Results: Among 95 patients, 60% achieved CMR. Median PFS was 35.06 months in CMR versus 23.89 months in non-CMR patients (p = 0.0004). The incidence rate of progression or death was lower in CMR patients (0.017 events per month) compared to non-CMR (0.035 events per month). CMR was independently associated with prolonged PFS (HR = 0.42; 95% CI: 0.256-0.688; p = 0.001). For OS, CMR also predicted better outcomes (HR = 0.062; 95% CI: 0.014-0.272; p = 0.0011), with a 5-year OS rate of approximately 95% in CMR patients versus 30% in non-CMR patients. In multivariate analysis, CMR remained the only independent predictor of survival, regardless of prior endocrine therapy, metastatic burden, or hormone receptor levels. Conclusions: Metabolic complete response on PET-CT strongly correlates with improved PFS and OS in HR+/HER2- MBC treated with first-line CDK4/6 inhibitors. PET-CT may serve as a valuable non-invasive prognostic biomarker to guide early treatment decisions in clinical practice.
Presentation numberPS1-12-16
Chromatin Reader ZMYND8 Promotes Breast Cancer Bone Metastasis
Tianyue Qin, Icahn School of Medicine at Mount Sinai, New York, NY
T. Qin, I. Bado; Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.
Chromatin Reader ZMYND8 Promotes Breast Cancer Bone Metastasis Tianyue Qin1,2, Igor Bado1,3 1Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY2Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY3Division of Hematology/Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY Statement of Purpose: Breast cancer bone metastasis remains a major clinical challenge, with a five-year survival rate of only ~22% following distant recurrence. Treatment is hindered by therapeutic resistance, frequent metastasis relapse, and the emergence of secondary metastases, all of which contribute to poor patient outcomes. Understanding the molecular mechanisms driving these processes is critical. ZMYND8, a chromatin reader, has emerged as a key player in cancer biology. It is involved in histone modification recognition, DNA repair, and can also promote angiogenesis under hypoxic conditions. Importantly, ZMYND8 has been identified as a prognostic marker in breast and liver cancers. Our preliminary data show that ZMYND8 is significantly upregulated in bone metastases compared to naïve tumor cells in an MCF7 estrogen receptor-positive (ER+) breast cancer model. This suggests that ZMYND8 may play an active role in metastatic progression.To elucidate its function, we are employing both in vitro and in vivo ER+ breast cancer models. This research will advance our understanding of ZMYND8 in metastasis and potentially uncover new therapeutic targets. Methods: We developed ZMYND8 knockdown in MCF-7 cell lines expressing firefly luciferase. Cell proliferation of knockdown vs control cells was studied with two methods: IVIS imaging of bioluminescence and BrdU assay. Cell migration was studied using a transwell system. The number of cells that migrated was stained with crystal violet and counted. Wound healing assay was performed by scratching cells in culture and measuring area of the scratch after 96h. Immunofluorescence staining was performed on patient samples with ZMYND8 antibody and Pan-Keratin antibody. Tumor vs stomal cells were separated based on Pan-Keratin expression and ZMYND8 expression. ZMYND8 expression was quantified by fluorescence intensity in primary breast tumor vs bone metastasis and in bone tumor vs stroma. DNA damage was quantified based on γH2AX staining. ZMYND8 knockdown and control MCF-7 cells were injected to the intra-iliac artery of nude mice and tumor growth was measured with IVIS. Results: In vitro studies demonstrated that ZMYND8 significantly enhances the metastatic potential of ER+ breast cancer cells. Specifically, knocking down ZMYND8 in MCF7 cells decreased migration, proliferation, and wound healing capacity compared to control cells. In Immunofluorescence analysis on patient bone metastasis samples, ZMYND8 is more highly expressed in bone metastasis than primary breast tumor, and it is also more highly expressed in the tumor cells compared to stromal cells in the bones. ZMYND8 also co-localized with yH2AX staining, and their expression shows a linear correlation, indicating ZMYND8 is highly expressed at DNA damage sites. In vivo study showed less growth of ZMYND8 knockdown cells in mouse compared to control cells at 35 days, suggesting ZMYND8’s role in tumor progression. Conclusion: Collectively, these results implicate ZMYND8 as a critical regulator of breast cancer bone metastatic progression and highlight its potential as a therapeutic target in ER+ breast cancer. Ongoing and future research includes in vivo studies to understand ZMYND8’s role in bone metastasis progression, mechanistic studies to investigate its molecular activities, and its therapeutic implications.
Presentation numberPS1-12-17
Frequency and Prognostic Significance of ESR1 Mutations Post-CDK4/6 inhibitors in ER+/HER2− Metastatic Breast Cancer: Real-World Data from the GEICAM_RegistEM Study
Ángel Guerrero-Zotano, Instituto Valenciano de Oncología (IVO); GEICAM Spanish Breast Cancer Group, Valencia, Spain
Á. Guerrero-Zotano1, S. Antolín2, A. Tibau Martorell3, J. Cruz-Jurado4, R. Andrés5, S. Saura6, C. Rodríguez-Sánchez7, E. Galve8, C. Falo9, M. Margelí-Vila10, Á. Rodríguez-Lescure11, A. Miguel12, J. Chacón13, E. Adrover-Cebrian14, M. Corbellas -Aparicio15, M. Marin-Alcalá16, I. González-Maeso17, S. Varela-Ferreiro18, C. Arqueros19, A. Antón-Torres20, S. Servitja21, D. Moreno-Muñoz22, M. Merino23, A. Ballesteros-García24, M. Echarri-González25, J. Alonso-Romero26, R. Seijas Tamayo27, V. Iranzo-Cruz28, J. De la Haba-Rodríguez29, J. Guerra30, L. Palomar Abad31, J. Illarramendi Mañas32, A. Godoy Ortiz33, A. García-Palomo34, M. Ruíz-Borrego35, R. Villanueva36, S. Zazo37, Y. Jerez-Gilarranz38, C. Reboredo Rendo2, N. Ancizar Lizarraga39, I. Ceballos-Lenza40, L. Murillo Jaso41, L. Figuero42, J. Herranz43, N. Martín44, D. Fernández-García44, I. Álvarez López39, S. López-Tarruella45, F. Rojo Todo37; 1Medical oncology, Instituto Valenciano de Oncología (IVO); GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 2Medical oncology, Complejo Hospitalario Universitario A Coruña (CHUAC); GEICAM Spanish Breast Cancer Group, A Coruña, SPAIN, 3Medical oncology, Hospital Universitario Germans Trias i Pujol (ICO-Badalona); GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 4Medical oncology, Hospital Universitario de Canarias; GEICAM Spanish Breast Cancer Group, Santa Cruz de Tenerife, SPAIN, 5Medical oncology, Hospital Clinico Universitario Lozano Blesa; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 6Medical oncology, Complejo Hospitalario Universitario de Gran Canaria Dr. Negrin; GEICAM Spanish Breast Cancer Group, Las Palmas De Gran Canaria, SPAIN, 7Medical oncology, Hospital Clinico Universitario de Salamanca; GEICAM Spanish Breast Cancer Group, Salamanca, SPAIN, 8Medical oncology, Hospital Universitario de Basurto; GEICAM Spanish Breast Cancer Group, Bilbao, SPAIN, 9Medical oncology, ICO de Barcelona (Hospital Duran i Reynalds); GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 10Medical oncology, ICO Badalona- Hospital Universitario Germans Trias i Pujol; GEICAM Spanish Breast Cancer Group, Badalona, SPAIN, 11Medical oncology, Hospital General Universitario de Elche; GEICAM Spanish Breast Cancer Group, Elche, SPAIN, 12Medical oncology, ALTHAIA Xarxa asistencial de Manresa; GEICAM Spanish Breast Cancer Group, Manresa, SPAIN, 13Medical oncology, Hospital General Universitario de Toledo; GEICAM Spanish Breast Cancer Group, Toledo, SPAIN, 14Medical oncology, Complejo Hospitalario Universitario de Albacete; GEICAM Spanish Breast Cancer Group, Albacete, SPAIN, 15Medical oncology, Hospital Universitario Dr. Peset; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 16Medical oncology, Consorci Sanitari de Terrassa; GEICAM Spanish Breast Cancer Group, Terrasa, SPAIN, 17Medical oncology, Hospital Universitario Son Llatzer; GEICAM Spanish Breast Cancer Group, Palma de Mallorca, SPAIN, 18Medical oncology, Hospital Universitario Lucus Augusti; GEICAM Spanish Breast Cancer Group, Lugo, SPAIN, 19Medical oncology, Hospital de la Santa Creu i Sant Pau; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 20Medical oncology, Hospital Universitario Miguel Servet; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 21Medical oncology, Hospital del Mar; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 22Medical oncology, Hospital Universitario Fundacion Alcorcon; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 23Medical oncology, Hospital Universitario Infanta Sofia; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 24Medical oncology, Hospital Universitario de la Princesa; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 25Medical oncology, Hospital Universitario Severo Ochoa; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 26Medical oncology, Hospital Clinico Universitario Virgen de la Arrixaca; GEICAM Spanish Breast Cancer Group, El Palmar, SPAIN, 27Medical oncology, Hospital Nuestra Señora de Sonsoles; GEICAM Spanish Breast Cancer Group, Avila, SPAIN, 28Medical oncology, Consorcio Hospital General Universitario de Valencia; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 29Medical oncology, Hospital Universitario Reina Sofia Cordoba; GEICAM Spanish Breast Cancer Group, Córdoba, SPAIN, 30Medical oncology, Hospital Universitario de Fuenlabrada; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 31Medical oncology, Hospital de Sagunto; GEICAM Spanish Breast Cancer Group, Puerto de Sagunto, SPAIN, 32Medical oncology, Complejo Hospitalario de Navarra; GEICAM Spanish Breast Cancer Group, Pamplona, SPAIN, 33Medical oncology, Medical Oncology Unit, Virgen de la Victoria University Hospital, IBIMA; GEICAM Spanish Breast Cancer Group, Málaga, SPAIN, 34Medical oncology, Complejo Asistencial Universitario de Leon; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 35Medical oncology, Hospital Universitario Virgen del Rocio; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 36Medical oncology, H. de Sant Joan Despi Moises Broggi; GEICAM Spanish Breast Cancer Group, Sant Joan Despí, SPAIN, 37Medical oncology, Hospital Universitario Fundacion Jimenez Diaz; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 38Medical oncology, Hospital General Universitario Gregorio Marañon; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 39Medical oncology, Unidad de Cáncer de Guipúzcoa – Osakidetza; GEICAM Spanish Breast Cancer Group, San Sebastián, SPAIN, 40Medical oncology, Hospital Universitario de Canarias; GEICAM Spanish Breast Cancer Group, La Laguna, SPAIN, 41Medical oncology, Hospital Clínico Universitario Lozano Blesa; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 42Medical oncology, Hospital Clínico Universitario de Salamanca; GEICAM Spanish Breast Cancer Group, Salamanca, SPAIN, 43Statics department, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 44Translational, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 45Medical oncology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense Madrid; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: Endocrine resistance in ER+/HER2- metastatic breast cancer is frequently mediated by ESR1 mutations. These mutations typically arise after prolonged endocrine therapy (ET) exposure in the metastatic setting, particularly with aromatase inhibitors (AI). This study evaluates the real-world evidence incidence and prognostic impact of ESR1 mutations in Spanish patients (pts) with ER+/HER2- advanced breast cancer (ABC) progressing on 1st line CDK4/6 inhibitor (CDK 4/6i) + ET. Methods: The GEICAM/2014-03 RegistEM study (NCT02819882) is an ongoing non-interventional cohort that collects prospective data and biological samples from patients with ABC, across 38 sites in Spain. A subset of 125 pts with ER+/HER2- ABC and available plasma samples collected at progression after 1st line with CDK4/6i + ET (n=125) was analyzed using the Plasma-SeqSensei™ BC IVD kit (Sysmex Inostics) to identify and quantify ctDNA mutations in six BC-related genes, ESR1, AKT1, ERBB2, KRAS, PIK3CA and TP53. Median PFS and OS were estimated from the Kaplan-Meier survival curves. Hazard ratios with 95% confidence intervals were obtained from multivariable Cox regression and logistics regression models, adjusted by clinical confounders (age, first metastatic, visceral disease, prior sensitivity to ET). Results: ESR1 mutation rates after progression on 1st line CDK4/6i + ET were 41.6%. The incidence of mutations increased with longer duration of 1st line therapy and varied according to the ET partner. Among patients treated with fulvestrant (FULV), mutation frequencies were 15.8% for 24 months. In contrast, with aromatase inhibitors (AI), frequencies were 22.2%, 61.1%, and 65.2%, respectively. Overall, The most common ESR1 mutations were D538G (28%), Y537S (17%), Y537N (16%), and E380Q (11%), all more common with AI than with FULV. Specifically, D538G was found in 35% of AI-treated patients vs. 11% with FULV; Y537S in 21% vs. 8%; Y537N in 21% vs. 3%; and E380Q in 12% vs. 8%. The detection of ESR1 mutations after CDK4/6i + ET was significantly associated with shorter mPFS, and showed a consistent trend toward worse OS, particularly with polyclonal ESR1 mutations (Table 1). Co-occurrence of ESR1 with PIK3CA/AKT1 or TP53 mutations was in 16.8% and 14.4%, respectively. The negative impact of ESR1 mutations on PFS and OS was independent of co-mutations in PIK3CA/AKT1 or TP53. Conclusions: ESR1 mutations are frequent after progression on CDK4/6i + ET, particularly following >1 year of AI, and are associated with poorer outcomes, especially when polyclonal. Their prognostic impact is independent of co-mutations in PIK3CA/AKT1 and TP53 and varies according to specific ESR1 variants. These findings support the use of ESR1 mutation profiling to guide treatment decisions post-CDK4/6i.
| ESR1 mutation status and survival analysis after 1st line CDK4/6i+ET |
N |
Events (%) |
Median RW-PFS (months) |
aHR (95% CI) |
p-value (Wald test) |
| ESR1 mutation status | |||||
| NMD (ref.) | 73 | 66(90.4%) | 5.52 | 1 | |
| MD | 52 | 48(92.3%) | 4.39 | 1.59 (1.05-2.4) | 0.0277 |
| Monoclonal | 26 | 25(96.2%) | 6.77 | 1.23 (0.75-2.02) | 0.4036 |
| Polyclonal | 26 | 23(88.5%) | 3.47 | 2.34 (1.39-3.94) | 0.0014 |
| ESR1 Y537N status | |||||
| NMD (ref.) | 108 | 97(89.8%) | 5.92 | 1 | |
| MD | 17 | 17(100%) | 3.22 | 2.86 (1.64-5.01) | 0.0002 |
| ESR1 Y537S status | |||||
| NMD (ref.) | 104 | 94(90.4%) | 5.52 | 1 | |
| MD | 21 | 20(95.2%) | 3.96 | 1.71 (1.02-2.86) | 0.0408 |
| ESR1 D538G status | |||||
| NMD (ref.) | 92 | 85(92.4%) | 5.52 | 1 | |
| MD | 33 | 29(89.9%) | 3.75 | 1.49 (0.95-2.32) | 0.0793 |
| ESR1 E380Q status | |||||
| NMD (ref.) | 112 | 103(92%) | 5.19 | 1 | |
| MD | 13 | 11(84.6%) | 5.06 | 1.24 (0.65-2.35) | 0.5094 |
| N | Events (%) | Median OS (months) | aHR (95% CI) |
p-value (Wald test) |
|
| ESR1 mutation status |
|
|
|||
| NMD (ref.) | 73 | 56(76.7%) | 23.33 | 1 | |
| MD | 52 | 37(71.2%) | 16.85 | 1.36 (0.65-2.87) | 0.4142 |
| Monoclonal | 26 | 18(69.2%) | 24.66 | 0.86 (0.31-2.4) | 0.7783 |
| Polyclonal | 26 | 19(73.1%) | 15.47 | 1.86 (0.82-4.24) | 0.1401 |
| rwPFS (real word progression-free survival): Time from start of next treatment line to progression or death | OS (overall survival): Time from start of next treatment line to death | NMD: no mutation detected | MD: mutation detected | aHR: adjusted Hazard Ratio | CI: confidence interval |
Presentation numberPS1-12-18
Treatment with CDK4/6 inhibitors in elderly patients with HR positive/HER2 negative metastatic breast cancer: experience from a Spanish center
Mariana López Flores, Complejo Asistencial Universitario de León, Leon, Spain
M. López Flores1, M. Vázquez Diez1, J. A. Fong Gutiérrez1, E. Honrado Franco2, J. Sáez Hortelano3, S. Abella Álvarez1, M. Rodríguez García1, L. López González4, A. López González1; 1Medical Oncology, Complejo Asistencial Universitario de León, Leon, SPAIN, 2Pathology, Complejo Asistencial Universitario de León, Leon, SPAIN, 3Pharmacy, Complejo Asistencial Universitario de León, Leon, SPAIN, 4Radiology, Complejo Asistencial Universitario de León, Leon, SPAIN.
Introduction CDK4/6 inhibitors (CDK4/6i) combined with endocrine therapy (ET) are the standard first-line treatment for hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC). However, evidence in elderly patients remains limited. This study aimed to evaluate the efficacy and tolerability of CDK4/6i in elderly (≥70 years) versus younger (<70 years) patients with HR+/HER2- mBC. Methods We conducted a retrospective cohort study of patients treated with CDK4/6i at our center between 2014 and 2023. Clinicopathological data were extracted from electronic medical records. Survival outcomes and safety were assessed and stratified by age. Results A total of 169 patients were included: 116 (68.6%) <70 years (mean age 55.6) and 53 (31.4%) ≥70 years (mean age 76.2). Median follow-up was 44.9 (29.2-75.2) months. Approximately one-third of patients had de novo mBC in both groups. An ECOG performance status of 2 was observed in 13% of patients <70 years and 44% of those ≥70 years (p=0.001). Aromatase inhibitors were more commonly used in elderly patients, while fulvestrant was more frequent in younger ones (p=0.03). CDK4/6i were used as first-line in 60% of both groups. A significant difference in CDK4/6i choice was observed (p<0.001): palbociclib was the most frequently prescribed in ≥70 (63.5%), while <70 patients received all three agents equally. The number of cycles and grade 3-4 toxicities were similar; neutropenia was the most common severe toxicity (69%). Disease progression was the main cause of discontinuation (88%). Treatment was discontinued due to toxicity in 11% of <70 and 13% of ≥70 patients (p=NS). After progression, significantly fewer elderly patients received further treatment (92.6% vs 69.8%; p=0.001) Progression-free survival (PFS) was similar between groups (13.1 vs 11.1 months, p=0.536), and also among patients treated in first line (p=0.420). In multivariate analysis, de novo mBC was associated with shorter PFS [HR 1.60 (1.05-2.45); p=0.03], while first-line CDK4/6i use was linked to longer PFS [HR 0.58 (0.39-0.88); p=0.009]. Neither age nor CDK4/6i type affected PFS. Overall survival (OS) was shorter in elderly patients (35.9 vs 51.1 months, p=0.003). Receiving subsequent treatment was associated with improved OS [HR 0.33 (0.17-0.63); p=0.001]. Conclusions In this real world cohort, palbociclib was the most frequently prescribed CDK4/6i in elderly patients. Toxicity and PFS were comparable across age groups. However, elderly patients had shorter OS, potentially related to reduced access to subsequent treatments and age-related factors.
Presentation numberPS1-12-19
Disease natural history and care quality assessment of hormone receptor positive / human epidermal growth factor receptor 2 negative metastatic breast cancer (HR+/HER2- mBC) – an international multicentric study
François Duhoux, Cliniques universitaires Saint-Luc, Brussels, Belgium
S. Theophanous1, L. Revie2, O. Bouissou3, F. Duhoux4, P. Galgane Banduge5, A. Choudhury5, A. Collard6, W. Hai Deng3, G. Hall7, R. Foppen8, V. Gouthamchand5, I. Kaczmarczyk9, E. Krasniqi8, L. Licata10, A. Lobo Gomes11, L. Sanchez Gomez12, A. van Maanen6, G. Podevijn6, E. Ross3, J. Thonnard6, A. Thomas6, C. Twelves1, X. Fernandez13, C. Van Marcke4; 1LTHT, Leeds Teaching Hospital NHS Trust, Leeds, UNITED KINGDOM, 2EMEA, IQVIA Ltd, London, UNITED KINGDOM, 3OUH, Oslo University Hospital, Oslo, NORWAY, 4Medical Oncology, Cliniques universitaires Saint-Luc, Brussels, BELGIUM, 5Clinical Data Science, Maastricht University, Maastricht, NETHERLANDS, 6CUSL, Cliniques universitaires Saint-Luc, Brussels, BELGIUM, 7Medical Oncology, Leeds Teaching Hospital NHS Trust, Leeds, UNITED KINGDOM, 8IFO, IRCCS Ospedale Regina Elena, Rome, ITALY, 9Data Strategy, Access and Enablement (DSAE), IQVIA Ltd, London, UNITED KINGDOM, 10Medical Oncology, IRCCS Ospedale San Raffaele, Milan, ITALY, 11Uniklinik, RWTH Aachen, Aachen, GERMANY, 12Medical Data Works, Medical Data Works BV, Maastricht, NETHERLANDS, 13Commercial Research Management, DIGICORE, Brussels, BELGIUM.
Background Despite early detection and treatment, ~30% of early-stage HR+ HER2− breast cancer (BC) patients relapse. Rebiopsy at recurrence is recommended by international guidelines when feasible, as it informs optimal treatment selection. Molecular profiling of metastatic sites is essential to reassess HER2 status and detect actionable genomic alterations such as BRCA1/2, PIK3CA, and AKT1, guiding sequencing of therapies in the metastatic setting. The DIGICORE consortium enables detailed cancer phenotyping and quality of care assessment through automated near real-time data extraction. Here, we report real-world data from HR+ HER2- mBC patients treated at five European DIGICORE centres. Methods Cohorts of HR+ (ER ≥ 10%) HER2- mBC diagnosed between 2018 and 2023 were identified at each centre. Clinical data structured to the OMOP common data model and aligned to key oncological variables in the Minimal Essential Description of Cancer framework (DIGICORE) were automatically extracted. Data were analysed using the Vantage6 federation platform (IKNL, Netherlands) via Medical Data Works (Netherlands). Subcohorts were defined by disease history (de novo vs recurrent mBC) and HER2 status (HER2-low vs HER2-zero). Only anonymous, aggregate results were shared across centres, preserving privacy. Results A total of 1175 HR+HER2- mBC patients were identified. Due to ongoing curation, analyses focus on 529 patients with deep phenotypic data. Of these patients, 33.3% were de novo metastatic (60.2% HER2-low, 33.0% HER2-zero, and 6.8% undetailed HER2-) and 66.7% were a relapse of a previous early-stage BC (62.9% HER2-low, 36.0% HER2-zero, and 1.1% undetailed HER2-). Of the latter, 78.5% of patients were rebiopsied at relapse, of which 8.1% were HER2-positive at the early stage.Amongst patients who underwent genomic testing, 16.1% carried a germline BRCA1 or BRCA2 mutation, while 50%, 4.2%, and 4.2% carried a somatic PIK3CA, AKT1 or ERBB2 mutation, respectively. Among 320 patients with known metastatic sites, 32.5% had bone-only and 67.5% visceral metastases. ConclusionsThis federated network enables real-world analysis of phenotypic, genotypic, and care variables for patients with HR+ HER2- mBC. Improvement of data capture and curation, as well as addition of treatment patterns to the analysis, is ongoing. Notably, 8.1% of HER2- mBC are a relapse of HER2+ early BC, highlighting the need for rebiopsy. More than half of patients could benefit from therapy targeting an actionable gene mutation.
| De novo metastatic | Recurrent metastatic | ||
| Total patients (n) | 176 | 353 | |
| Age at index (median* – min, max) | 68 (25-93) | 63 (27-97) | |
| Disease stage at initial BC diagnosis (n, %) | I | N/A | 70** (20%) |
| II | N/A | 132 (37%) | |
| III | N/A | 100 (28%) | |
| IV | N/A | 6** (2%) | |
| Unknown/ Missing | N/A | 44 (12%) | |
| HER2 subclassification at metastasis diagnosis based on latest available assessment (n, %) | HER2 zero | 58 (33%) | 127 (36%) |
| HER2 low | 106 (60%) | 222 (63%) | |
| Unknown/ Not specified | 12 (7%) | 4 (1%) |
Presentation numberPS1-12-20
Circulating tumor DNA profiling of patients with early-line advanced luminal breast cancer
Marija Balic, University of Pittsburgh, Pittsburgh, PA
N. Dobrić1, S. O. Hasenleithner1, C. Suppan1, E. V. Klocker1, D. Hlauschek1, R. Graf2, C. Albertini3, D. Egle3, A. M. Starzer4, R. Bartsch4, G. Rinnerthaler1, P. J. Jost1, E. Heitzer2, N. Dandachi1, M. Balic5; 1Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, AUSTRIA, 2Institute of Human Genetics, Diagnostic, and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, AUSTRIA, 3Department of Gynecology, Breast Cancer Center Tirol, Medical University of Innsbruck, Innsbruck, AUSTRIA, 4Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, AUSTRIA, 5Division of Oncology, Hillmans Cancer Center, University of Pittsburgh, Pittsburgh, PA.
Background: Advances in the development of new endocrine treatment approaches for hormone receptor-positive (HR+)/HER2-negative (HER2-) advanced breast cancer (ABC) have significantly improved patient outcomes. However, a number of patients still have limited benefit from endocrine-based treatments, and even patients with prolonged progression-free survival (PFS) will progress at some point. To better understand the heterogeneity in treatment outcomes within the first line (1L), and between 1L and second line (2L), we performed circulating tumor DNA (ctDNA) profiling in a uniform multicenter cohort of patients with HR+/HER2- ABC in Austria. Samples were collected at defined timepoints prior to early lines of therapy. The goal of this study was to characterize ctDNA features associated with molecular and clinical heterogeneity in HR+/HER2- ABC.Methods: Patients with locally advanced or metastatic HR+/HER2– BC were included. ctDNA was analyzed using a 77-gene panel (AVENIO ctDNA Expanded Kit). Tumor fraction (TF) was estimated using two orthogonal approaches: untargeted aneuploidy assessment via mFAST-SeqS (reported as z-scores) and ichorCNA analysis of low-pass whole-genome sequencing (0.1x WGS) when available. The highest variant allele frequency (hVAF) per sample was used as an additional TF proxy. TF, hVAF, and somatic variant burden were compared between samples collected prior to 1L and 2L systemic therapy using mixed-effects models, and their associations with PFS were evaluated.Results: We analyzed 225 ctDNA samples from 184 patients with HR+/HER2– ABC (128 before start of 1L, and 76 before start of 2L), including 40 patients with paired samples. The overall TF was low (median z-score 2.49; range –0.5 to 208.3), with no significant difference between 1L and 2L samples by mFAST-SeqS (adjusted mean difference 6.33, 95%CI: -0.3-13.0; p = 0.061) or ichorCNA (adjusted mean difference -0.003, 95%CI: -0.06-0.07; p = 0.925). However, hVAF was significantly higher in 2L samples (adjusted mean difference 0.05, 95%CI: 0.01-0.09; p = 0.017). Somatic variants were detected in 84% 1L and 89% 2L samples, with pathogenic/likely pathogenic (P/LP) alterations in 63% and 70%, respectively. 2L samples harbored significantly more total (adjusted mean difference 2.58, 95%CI: 1.33-3.83; p = < 0.001) and subclonal (adjusted mean difference 2.1, 95%CI: 1.09-3.12; p = < 0.001) variants, including a higher P/LP ESR1 mutation frequency (30.2% vs. 7.8%). PI3K-pathway alterations (PIK3CA, AKT1, PTEN) were found in 41.4% 1L and 52.6% 2L samples. High TF (z-score ≥3) was associated with a shorter PFS in both 1L (17.9 vs. 50.6 months; p < 0.001) and 2L (4.6 vs. 7.3 months; p = 0.042) settings.Conclusions: Elevated ctDNA-derived TF prior to treatment was associated with significantly shorter PFS in advanced luminal BC across both 1L and 2L settings. While the spectrum and frequency of pathogenic and targetable alterations aligned with existing data, 2L samples showed higher TF (as measured by hVAF) and a greater burden of somatic variants, reflecting evolving molecular complexity. These findings underscore the utility of integrated ctDNA-based approaches for tumor burden assessment and molecular profiling. Further studies are warranted to elucidate the biological drivers of heterogeneity in early-line HR+/HER2– ABC and to optimize ctDNA-guided treatment strategies.
Presentation numberPS1-12-21
Association of weight changes compared to baseline with time on CDK 4/6 inhibitor in women with de novo Stage IV HR+HER2- breast cancer
Darya Kizub, MD Anderson Cancer Center, Houston, TX
D. Kizub1, S. Pasyar2, M. V. Phan3, J. Sukumar1, A. K. Arun1, A. Singareeka Raghavendra4; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Biostatistics, MD Anderson Cancer Center, Houston, TX, 3Pharmacy Quality-Regulatory, MD Anderson Cancer Center, Houston, TX, 4Breast Medical Oncology-Research, MD Anderson Cancer Center, Houston, TX.
Background and objectives: Overweight and obesity are associated with worse outcomes in breast cancer, while weight loss can delay the onset of endocrine therapy resistance in pre-clinical models. Weight gain is a common side-effect of endocrine therapy. We explored if weight loss delays resistance to endocrine therapy with CDK 4/6 inhibitors (CDK4/6i) in women with de novo hormone-receptor positive (HR+) /HER2- metastatic breast cancer. Methods: We analyzed patients with de novo HR+/HER2- metastatic breast cancer treated at MD Anderson Cancer Center with CDK4/6 inhibitors and endocrine therapy from January 2015 to December 2024 who experienced disease progression. Body weight and BMI were tracked at baseline (CDK4/6i initiation) and 3, 6, and 12 months from CDK4/6 inhibitor start. Mixed-effects models assessed associations between treatment duration and weight/BMI change. Cox proportional hazards models evaluated factors associated with overall survival (OS). Results: Among 488 patients who progressed on CDK 4/6i and endocrine therapy, palbociclib was the most used drug (86.3%). 80 (16.4%) experienced weight gain and 48 (9.8%) experienced weight loss at 12 months compared to baseline. Similar proportions were observed for change in BMI. Mixed-effects univariate and multivariate models showed that increases in absolute weight and BMI from baseline over time were associated with longer CDK4/6i therapy duration (p=0.010 and p=0.005, respectively) (Table 1). In univariate Cox analysis, longer CDK4/6i use was associated with improved OS (HR 0.50, 95% CI 0.44-0.57, p<0.001), while higher tumor grade and presence of BRCA mutations correlated with poorer OS. Conclusion: Contrary to our hypothesis, absolute weight and BMI increase from baseline was associated with longer CDK 4/6i use in de novo HR+/HER2- breast cancer. Weight trends and tumor biology may offer prognostic insight and warrant further study in prospective settings.
| Variable | Category | Estimate | p-value | 95% CI |
| Univariate regression, absolute weight change | ||||
| CDK 4/6i use duration | 0.48 | 0.008 | 0.13,0.83 | |
| Age | >50 vs <= 50 | 0.08 | .856 | -0.83,1.00 |
| Race | Asian vs white | -1.72 | 0.029 | -3.26,-0.17 |
| Black vs white | 0.66 | 0.403 | -0.88,2.19 | |
| Hispanic vs white | -0.11 | 0.875 | -1.51,1.28 | |
| PR primary | Positive vs negative | 0.04 | 0.952 | -1.13,1.20 |
| HER2 IHC | 1+ vs 0 | -0.09 | 0.860 | -1.20,1.00 |
| 2+ vs 0 | -0.30 | 0.649 | -1.62,1.00 | |
| Grade primary | II vs I | -0.74 | 0.413 | -2.51,1.03 |
| III vs I | -1.33 | 0.165 | -3.21,0.55 | |
| Lymphatic invasion | Positive vs negative | -0.01 | 0.992 | -1.17,1.16 |
| BRCA | BRCA2 vs BRCA negative | -1.15 | 0.362 | -3.64,1.33 |
| BRCA vs no germline testing | -0.003 | 0.994 | -0.93,0.92 | |
| Multivariate regression, absolute weight change | ||||
| CDK 4/6i use duration | 0.46 | 0.010 | 0.11,0.81 | |
| Race | Asian vs white | -1.59 | 0.43 | -3.13,-0.06 |
| Black vs white | 0.71 | 0.362 | -0.81,2.23 | |
| Hispanic vs white | -0.18 | 0.803 | -1.56,1.21 | |
| Multivariate regression, BMI change | ||||
| CDK 4/6i use duration | 0.19 | 0.005 | 0.06,033 | |
| Race | Asian vs White | -0.53 | 0.81 | -1.12,0.07 |
| Black vs white | 0.25 | 0.398 | -0.33,0.84 | |
| Hispanic vs white | 0.03 | 0.918 | -0.51-0.56 |
Presentation numberPS1-12-22
Exceptionally High Frequency of ESR1 Mutations in Pakistani Women with Endocrine Resistant Hormone Receptor positive Her2 neu negative Metastatic Breast Cancer
Yasmin Abdul Rashid, The Aga Khan University, Karachi, Pakistan
Y. A. Rashid1, S. Khan2, M. Anis2, R. Kumari2, R. Idress3, S. I. Azam4, I. Masroor5, A. K. Sattar6, A. Gauhar7, M. Rafiq8, A. A. Jabbar9; 1Oncology, The Aga Khan University, Karachi, PAKISTAN, 2Biological and Biomedical Sciences, The Aga Khan University, Karachi, PAKISTAN, 3Pathology & Laboratory Medicine, The Aga Khan University, Karachi, PAKISTAN, 4Community Health Sciences, The Aga Khan University, Karachi, PAKISTAN, 5Radiology, The Aga Khan University, Karachi, PAKISTAN, 6Surgery, The Aga Khan University, Karachi, PAKISTAN, 7Anaesthesiology, The Aga Khan University, Karachi, PAKISTAN, 8Institute of Biochemistry and Genetics Engineering, University of Sindh, Jamshoro, PAKISTAN, 9Oncology, Dr. Ziauddin Hospital, Karachi, PAKISTAN.
Exceptionally High Frequency of ESR1 Mutations in Pakistani Women with Endocrine-ResistantHormone Receptor Positive Her2 Negative Metastatic Breast CancerBackground:Aromatase inhibitor (AI) based hormone therapy combined with CDK4/6 inhibitors in hormonereceptor-positive (HR+), Her2-negative metastatic breast cancer (MBC) is considered the standard ofcare. Ligand-binding domain mutations in ESR1 gene is a well-established mechanism of resistancereported in these patients. The ESR1 gene mutation frequencies have been reported to be between15–50% in AI-treated cases. Limited data is available from South Asian population and to date, no studyhas reported the frequencies of ESR1 gene mutations in Pakistani cohort.Methods:We enrolled (n = 50) Pakistani women with endocrine-resistant HR+, Her2 – MBC to conduct aprospective study at Aga Khan University Hospital (AKUH), Karachi, Pakistan. Genomic DNA wasextracted from formalin-fixed paraffin-embedded (FFPE) biopsy blocks of these patients and allele-specific multiplex PCR assays were conducted to analyze the hotspot mutations (Y537S Y537C, Y537N,D538G, and E380Q) in ESR-1 gene. Clinicopathological data including age at diagnosis, metastaticpresentation (de novo vs recurrent), ER/PR via IHC, Her2 status, patient comorbidities, and priorchemotherapy were recorded. Data analysis and associations were evaluated using t, Fisher’s exact,Chi-square, Kolmogorov-Smirnov and Mann Whitney tests via SPSS v25 and GraphPad Prism 4software. A p-value of less than 5% was considered as significant.Results:Our data showed that median age at diagnosis was 48 years (23-75). Strong ER positivity was found tobe in 68%, intermediate 24% and only 8% patients had low ER positivity. PR-positivity was found in84% patients. Overall, 96% patients harbored the Y537S mutation, a frequency that is far exceedingthe global reports. This confers strong ligand-independent activation and profound endocrineresistance. Y537S prevalence was similar in recurrent vs de novo cases (26/28 vs 22/22; p=0.50). Thetwo Y537S-negative patients were significantly younger (mean age 30 vs 49 years; p<0.01). All patientswith comorbidities (n=30) carried Y537S (vs 18/20 without comorbidity; p=0.16). Her2-low status (IHC1+/2+) was found in 52% and was associated with more recurrent disease (62% vs 33%), thoughsurvival was similar. Prior chemotherapy (in 38/50 patients) correlated with recurrent disease (58% vs17%; p<0.01). Patients without prior history of receiving chemotherapy had a higher median numberof concurrent ESR1 gene mutations (3 vs 2), though not statistically significant.Conclusions:We report an exceptionally high ESR1 gene Y537S single nucleotide polymorphism in endocrine-resistant HR+, Her2– MBC Pakistani cohort. Thus, highlighting the importance of unique tumor biologyand clonal evolution. These mutations were enriched in patients with strong ER/PR expression andyounger age group. Our findings support routine ESR1 gene testing in our population, implying tailoredapproach and the need for region-specific therapeutic strategies.
Presentation numberPS1-12-23
Pi3k/akt/mtor inhibitors in hr+/her2- breast cancer: bridging the gap between evidence and community practice in toxicity management
Kia Jones, Medlive, Needham, MA
K. Jones1, T. Ackbarali1, K. Jhaveri2, T. Traina3; 1Hematology/Oncology Team, Medlive, Needham, MA, 2Section Head, Endocrine Therapy Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, 3Section Head, Triple Negative Breast Cancer Clinical Research Program, Memorial Sloan Kettering Cancer Center, Needham, NY.
BACKGROUND: PI3K/AKT/mTOR inhibitors represent a significant therapeutic advance for HR-positive, HER2-negative advanced breast cancer (ABC). However, real-world adoption in community settings remains limited. To understand clinician perceptions and challenges associated with managing treatment-related toxicities, a multi-phase CME initiative was developed to gather insights directly from community practitioners and deliver targeted education focused on adverse event (AE) management. METHODS: Phase 1 of the initiative consisted of a 90-minute virtual roundtable discussion featuring two academic and four community-based breast oncologists. Discussion topics included real-world barriers, specialist coordination, and strategies to individualize care. Qualitative analysis of the roundtable transcript was conducted to identify thematic insights and consensus strategies. These findings were synthesized and served as the foundation for phase two. Phase 2 was a 45-minute accredited activity focused on clinical evidence, AE mitigation, and patient-centered treatment planning. The content integrated roundtable-derived consensus and aligned with published evidence and guidelines on managing AEs in HR+/HER2- ABC. The activity launched on Medlive.com in October 2024 and will remain available for 1 year. Program outcomes were measured through pre-/post-activity assessments and post-activity evaluations. RESULTS: During the roundtable discussion (n=6), panelists identified hyperglycemia, rash, and diarrhea as the most challenging AEs associated with PI3K/AKT/mTOR inhibitor therapy. Hyperglycemia was rated by 100% of participants as the most difficult to manage, particularly in community settings where limited access to endocrinology was cited as a key barrier. Rash was identified by 83% of panelists as a common and distressing AE that frequently required dose interruption or discontinuation. Diarrhea was considered less severe but still impactful, with all panelists noting that early patient education and symptom recognition were essential to prevent treatment disruption. There was panel-wide consensus on the importance of multidisciplinary coordination to manage AEs effectively. However, 75% of the community-based panelists noted that limited access to dermatology, endocrinology, and/or pharmacy support often prevents optimal implementation of proven management strategies. Following the roundtable, 10,240 clinicians engaged with the education. Among CME participants, 83% completed the activity. Post-activity, 45% anticipated increased coordination with specialists to address toxicities, 34% reported greater intent to use PI3K/AKT/PTEN-pathway inhibitors in biomarker-selected patients, and 33% planned to adopt more rigorous biomarker testing protocols. Significant knowledge gains (p<.05) were observed in understanding of molecular mechanisms (+29%) and interpretation of key efficacy data from pivotal trials (+27%). CONCLUSIONS: The roundtable revealed critical barriers to integrating pathway-directed therapies in community settings, including gaps in specialist access, variability in supportive care infrastructure, and uncertainty around AE management. The educational component demonstrated measurable improvements in knowledge and intent to change practice, supporting the value of combining qualitative insights with structured learning to address implementation challenges. When grounded in real-world perspectives and reinforced by clinical data, education on targeted therapies can enhance clinician readiness to integrate these strategies into routine care and overcome barriers in community settings. Supported by an independent medical education grant from AstraZeneca.
Presentation numberPS1-12-24
Optimal sequencing after progression with CDK4/6 inhibitors in hormone receptor-positive metastatic breast cancer: a real-world analysis
Viviane Primo Basilio de Souza, AC Camargo Cancer Center, São Paulo, Brazil
V. Primo Basilio de Souza1, N. Soldi1, A. Cicilini1, T. Saruwatari1, L. De Mattos1, M. Camandaroba1, M. Tariki1, C. Machado2, S. Sanches1, A. Da Costa1, V. De Lima1, E. Dos Santos1; 1Clinical Oncology, AC Camargo Cancer Center, São Paulo, BRAZIL, 2Departamento de Medicina Preventiva e Social da Faculdade de Medicina da UFMG, UFMG, Belo Horizonte, BRAZIL.
Introduction: CDK4/6 inhibitors combined with endocrine therapy have transformed the management of hormone-positive/HER2-negative metastatic disease, consolidating their position as the standard of care in first-line therapy. However, there is still a significant gap in the literature regarding the best therapeutic strategy after progression and the prognostic factors that impact treatment sequencing in clinical practice.Methods: We conducted a retrospective study of patients with HR+/HER2- metastatic breast cancer treated with CDK4/6 inhibitors (palbociclib, ribociclib, or abemaciclib) between 2016 and 2025 at a referral cancer center. Clinical, pathological, and molecular variables were analyzed, focusing on progression-free survival after second-line therapy (PFS2), overall survival (OS), and progression-free survival after first-line therapy (PFS1). Statistical analyses included Kaplan-Meier curves, univariate and multivariate Cox models, and multiple imputation to address missing data.Results: Of the 377 patients, 177 experienced a second progression event, and 318 were included in the overall survival analysis. We observed that the type of second-line treatment significantly impacted PFS2. Strategies based on endocrine therapy, especially fulvestrant, demonstrated superior PFS2 compared to CDK4/6i rechallenge (median 11.3 vs. 5.4 months; HR 0.41, 95% CI 0.23-0.75, p = 0.004). Regimens containing capecitabine demonstrated inferior performance, with a median time of 6.9 months. In multivariate analysis with multiple imputation, factors such as visceral metastasis (HR 1.64, 95% CI 1.12-2.42, p = 0.01) and late use of CDK4/6i (HR 1.25, 95% CI 1.13-1.39, p < 0.001) predicted worse overall survival. Interestingly, early discontinuation of CDKi due to toxicity appeared as a protective factor (HR 0.41; p = 0.004), and 27% of patients maintained response without requiring second-line therapy at last follow-up, a hypothesis that raises discussions about early response and biological sensitivity. Conversely, 13% presented rapid progression leading to death before the initiation of second-line therapy. Regarding overall survival, a trend toward a negative impact of visceral metastasis (HR 1.64; p = 0.01) and elevated Ki-67 (HR 1.44; p = 0.01) was observed. In contrast, patients who received fulvestrant or everolimus after progression had better survival rates (median PFS2 of 12.1 months), outperforming chemotherapy-based regimens. The proportion of patients who did not receive second-line treatment included both individuals with rapidly fatal clinical progression (13%) and patients without documented progression at the end of follow-up (27%).Conclusion: This demonstrates that second-line therapeutic choices significantly impact post-progression outcomes. Endocrine strategies, particularly combinations of everolimus and fulvestrant, were associated with superior progression-free survival and overall survival compared with chemotherapy or CDK4/6 inhibitor reintroduction.
Presentation numberPS1-12-25
Institutional adherence to CDK4/6 inhibitor use in ER+/HER2- metastatic breast cancer: A retrospective cohort study
Mercy C Anyanwu, Icahn School of Medicine at Mount Sinai, New York, NY
M. C. Anyanwu1, N. Wagner2, H. Becerra3, W. Siddiqui1, I. Yao1, J. Catlett1, O. Tracy1, T. K. Esantsi1, M. Kier1; 1Dubin Breast Cancer, Icahn School of Medicine at Mount Sinai, New York, NY, 2Breast Center, Montefiore Einstein Comprehensive Cancer Center, New York, NY, 3Medical Oncology, Brookdale University Hospital and Medical Center, Brooklyn, NY, New York, NY.
Background: NCCN recommends the combination of CDK4/6 inhibitors (CDK4/6i) and endocrine therapy (ET) as the standard first line treatment in ER+/HER2- metastatic breast cancer (MBC). Despite CDK4/6i demonstrating significant progression-free survival in clinical trials, real-world adherence to this combination remains inconsistent. We characterize the factors that influenced the adherence to combination therapy in patients with ER+/HER2- MBC. Methods: We conducted a multi-center retrospective review of electronic health records of patients with ER+/HER2- MBC between years 2015 – 2022 and imputed data into a standardized REDCap database. Bivariate group comparisons were performed using chi-squared or ANOVA, as appropriate. Multivariate logistic regression was done to identify independent factors associated with adherence to combination therapy. Statistical analyses were performed using R. A p-value <0.05 was considered statistically significant. Results: 152 women with a median age of 64 years (IQR: 52.3-73.8) were included. 27.6% were Black/African American and 44.7% were White. The most prescribed CDK4/6i was palbociclib (65.2%), then abemaciclib (25.9%). ECOG performance status was < 2 in 49.3% of cases. 79.6% of patients had fewer than three metastatic sites. Adherence to combination therapy was seen in 112 patients (73.7%). Of the 40 patients who did not receive CDK4/6i, 75% had justification, mostly due to insurance (23.3%), patient preference (23.3%) and comorbidities (20%). No significant differences in combination therapy across race or ethnicity were found. Insurance influenced the type of CDK4/6i prescribed, with palbociclib being most frequently used among patients with mixed (61.9%), public (50.6%), and private (39.4%) insurance (p = 0.011). The multivariate analysis showed that patients with fewer than three metastatic sites were more likely to receive combination therapy (77.7%) vs those with three or more sites (58.1%), p = 0.027; OR= 3.41 (95% CI: 1.19-9.72), p= 0.022, independent of other variables analyzed. Conclusions: Patient with low burden disease (< 3 metastatic sites) were more likely to receive combination therapy with CDK4/6i and ET. These findings may be driven by the preferred use of chemotherapy in high burden disease/visceral crises as these patients were excluded from trials such as PALOMA-2 due to the need for rapid disease control.
Presentation numberPS1-12-26
Neurofibromin loss as a driver of bone metastasis in estrogen receptor-positive breast cancer.
Zifan Zhao, Baylor College of Medicine, Houston, TX
Z. Zhao1, Z. Zheng1, J. T. Lei1, M. J. Baik1, Y. Wu1, L. K. Somes1, O. A. Harb1, J. Wang1, U. Rasaily2, O. A. Chang1, H. Wang3, I. Bado4, Z. Gugala5, M. Anurag1, B. Zhang1, A. Sreekumar2, V. Hoyos1, X. H. Zhang1, E. C. Chang1; 1Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 2Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 3Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Bufflo, NY, 4Icahn School of Medicine, Mount Sinai, New York, NY, 5The Department of Orthopaedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX.
Approximately 80% of breast cancers (BCa) expresses estrogen receptor-α (ER), and ER+ BCa most frequently metastasizes to the bone. The driver of bone metastasis remains undefined. To address this un-met need, we focus on a key tumor suppressor, neurofibromin (NF1), which is both a GTPase-activating protein that represses RAS signaling and a direct transcriptional corepressor that represses ER. NF1 inactivation, often observed as somatic mutations, has been shown to be enriched in metastatic vs primary ER+ breast cancer. In our analysis, NF1 copy number loss (CNL) was identified in 62% of ER+ breast cancer patients in a cohort who subsequently developed metastases. Notably, NF1-CNL was especially prevalent in bone metastases as compared to metastases in other tissues/organs. To functionally assess the metastatic potentials of NF1 inactivation, luciferase-tagged human NF1KO ER+ BCa cells were orthotopically transplanted into mouse mammary fat pads. After primary tumor resection, bioluminescence imaging revealed that NF1KO cells disseminated more extensively than NF1⁺ controls, with marked enrichment in bone and skeletal tissues. Furthermore, when directly delivered into bone via intra-iliac artery injection, NF1KO ER+ BCa cells demonstrated significantly accelerated growth within the bone microenvironment. Bone metastasis in ER+ BCa is frequently accompanied by osteoclast (OC)-induced bone loss, which releases stored growth factors that fuel tumor progression, thus creating a vicious cycle. We analyzed previously published RNA-seq data and found that “ossification” and “bone-remodeling” are key functional categories enriched among genes whose expression is affected by NF1-depletion in ER+ BCa cells. Bone abnormalities are common in neurofibromatosis type 1 patients caused by germline loss of NF1. It is possible that bone remodeling is a core function controlled by NF1 but hijacked by cancer cells. To test this, two ER+ BCa cell lines were each co-cultured with the OC progenitor cell line RAW264.7 in the presence of RNAKL. OC formation, assessed by TRAP staining, microscopy, and qPCR, was more efficiently induced by NF1-depleted cells than by controls. To identify factors mediating this effect, we performed mass-spectrometry on the culture medium from the co-cultures, and RNA-seq on mRNA extracted from the cells. Both analyses revealed upregulation of ICAM1, a molecule that facilitates fusion of pre-OC and promotes OC formation. Pharmacological inhibition of ICAM1 confirmed its functional necessity for OC induction in this setting, as expected. Immune suppression is a critical driver of tumor progression. When co-cultured with T-cells derived from human PMBCs, NF1KO ER+ BCa cells more efficiently suppressed proliferation and IFN-γ production than NF1+ cells. We further developed a CAR-T cell line that exhibits cytotoxic effect on HER2-expressing cells, such as MCF7 cells. NF1KO MCF7 cells were more resistant to killing by these CAR-T cells, while concurrently induce apoptosis in these CAR-T cells. Collectively, these data support the model whereby NF1 loss in ER+ BCa cells induce OC formation and T cell suppression, increasing tumor burden in the bone. Proper targeting of RAS and ER pathways for treatments may help mitigate bone metastasis in NF1-deficient ER+ BCa.
Presentation numberPS1-12-27
Trends and Barriers in Biomarker Testing and Treatment Patterns in HR+/HER2- Metastatic Breast Cancer
Ibrahim M Abbass, Genentech, South San Francisco, CA
I. M. Abbass1, D. Lebovitch2, N. Jain3, T. Stricker3, S. Nunnery4, A. Pregenzer1, E. Behan1, R. Wang1, L. Chen1, P. Dhillon1, Y. Liu2, T. Szado1, E. Yu1, S. Ogale1; 1HEOR Oncology, Genentech, South San Francisco, CA, 2HEOR Oncology, Genesis Research Group, Hoboken, NJ, 3Oncology, OneOncology, Nashville, TN, 4Oncology, Tennesse Oncology, Nashville, TN.
Background Targeted therapies have transformed treatment for HR+/HER2- metastatic breast cancer (mBC), but optimal use depends on timely biomarker testing and matched therapy selection. This study evaluated national trends in PIK3CA, PTEN, AKT1 and ESR1 biomarker testing and treatment patterns among U.S. patients (pts) in community oncology clinics and examined factors associated with timely biomarker testing. Methods We conducted a retrospective cohort study of adult pts (≥18 years) with HR+/HER2- mBC treated between 01/01/2018 and 01/31/2025, using de-identified, electronic health record-derived data from the United States Flatiron Health Network. Eligible pts received at least one line of therapy at U.S. community oncology practices. Biomarker testing and treatment patterns across the first three lines of therapy (1L-3L) were analyzed descriptively. Logistic regression model evaluated associations between timely testing and baseline demographic, socioeconomic (SES), and clinical characteristics. Timely testing was defined as completing biomarker testing with results available before initiating a line of therapy for which an approved targeted treatment was available. Results Among 6,197 eligible pts who received 1L therapy, 3,550 progressed to 2L and 2,045 to 3L during the study period. Biomarker testing rates increased over time prior to 1L (from 4% in 2018 to 35% in 2024), 2L (from 9% to 69%), and 3L (from 13% to 83%). CDK4/6 inhibitors were the predominant 1L treatment regardless of biomarker status. In 2L and 3L, treatment patterns became more fragmented. Among pts with known PIK3CA mutations, the use of targeted approved therapies was 32% in 2L and 36% in 3L in 2019, compared to 27% and 29% in 2024. Among pts with ESR1+, elacestrant use rose from 22% in 2023 to 29% in 2024 in 2L and remained steady at 26-27% in 3L. In AKT1/PTEN+ pts, capivasertib use was 6% in 2L and 11% in 3L in 2024. Among pts with no test, or with negative or inconclusive biomarker results, 1% in 1L, 9% in 2L and 17% in 3L received Antibody Drug Conjugates in 2024. Treatment choices for dual-mutant (ESR1+ with either PIK3CA, AKT1 or PTEN) pts remained highly variable, with no dominant regimen: 27% received targeted therapies in 2L versus 34% in 3L. Pts were less likely to receive timely biomarker testing if they were older, lived in lower-SES areas, were treated at smaller practices, or had a worse ECOG score. However, timely testing became more common in more recent years. Conclusions Despite the availability of targeted therapies, major gaps persist in both biomarker testing and the uptake of these treatments. In 2024, nearly one-third of pts remained untested before 2L, and only 35% were tested prior to initiating 1L, limiting timely access to newly approved options. Even when actionable mutations were identified, over two-thirds of eligible pts did not receive matched therapies. Timely testing was less common in smaller practices and lower-SES settings, highlighting systemic barriers to precision oncology. To close these gaps, coordinated, equity-focused strategies are needed to improve timely testing and ensure appropriate use of targeted treatments across all pts populations. Study Number: SL46196 and PL-03044. Financial Statement: This study was sponsored by Genentech, Inc., a member of the Roche Group.
Presentation numberPS1-12-28
Real-world outcomes of first line palbociclib combined with endocrine treatment in HR+/HER2- metastatic breast cancer: 5-year data from the French ESME cohort
Thomas Grinda, Gustave Roussy, Villejuif, France
T. Grinda1, S. Eltaief2, E. Brain3, T. Bachelot4, V. Diéras5, F. Dalenc6, V. Massard7, M. Decrop8, A. Coumert8, M. Arnedos9, A. Mailliez10, M. Carton11, L. Bosquet12, J. Frenel13, W. Jacot14; 1Medicine Department, Gustave Roussy, Villejuif, FRANCE, 2biostatistics, unicancer, Paris, FRANCE, 3Medicine Department, Institut Curie, Villejuif, FRANCE, 4Medicine Department, Léon Bérard, Lyon, FRANCE, 5Medicine Department, Eugène Marquis, Rennes, FRANCE, 6Medicine Department, Oncopole Claudius Régaud IUCT-O, Toulouse, FRANCE, 7Medicine Department, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy, FRANCE, 8Women’s Cancer, Pfizer Oncology France, Paris, FRANCE, 9Medicine Department, Institut Bergonié, Bordeaux, FRANCE, 10Medicine Department, Centre Oscar Lambret, Lille, FRANCE, 11Biostatistics, Clinical research and Epidemiology Unit, , Paris, France., Institut Curie, Villejuif, FRANCE, 12Medicine Department, Unicancer, Paris, FRANCE, 13Medicine Department, Institut de Cancérologie de l’Ouest – Centre René Gauducheau, Saint-Herblain, FRANCE, 14Medicine Department, Institut régional du Cancer de Montpellier, Montpellier, FRANCE.
Background: CDK4/6 inhibitors (CDK4/6i) are the standard of carein first line (1L) treatment for HR+/HER2- metastatic breast cancer (MBC) patients.This study aimed to characterize patient profiles and outcomes in the FrenchESME real-world (RW) cohort for patients in academic centers receiving 1L palbociclibcombined with endocrine therapy (ET).Material and Methods: The ESME MBC cohort is a nationalregistry collecting individual-level patient data from 18 French ComprehensiveCancer Centers (NCT03275311). This analysis included all patients aged ≥18years with newly diagnosed HR+/HER2- MBC who initiated in 1L palbociclib within90 days after ET initiation, between January 2018 and December 2023 outsidea clinical trial. Overall survival (OS) and time to next treatment(TTNT) were assessed using Kaplan-Meier estimations. Multivariable modelling(Cox-regression) was undertaken to identify independent prognostic factors forthese endpoints.Results: Among 3,587patients treated in 1L by CDK4/6i combined with ET for HR+/HER2- MBC, 2,267 patientsinitiated palbociclib, including 340 (15.1%) premenopausal women and 7 (0.3%)men. Median age at MBC diagnosis was 69 years (range 29-95). Visceralmetastases were present in 38.0% of cases, 37.8% of patients had bone-onlydisease, and 53.1%, 22.5% and 24.4% of patients presented 1, 2 and ≥3 metastaticsites, respectively. De novo MBC accounted for 33.1% of patients. Forrelapsed MBC, treatment-free intervals (TFI) were ≤12 months in 22.5% and >12months in 31.7% of the whole population (missing data in 12.7%). Palbociclibwas combined with an aromatase inhibitor (AI) in 1,651 (72.8%) patients, mostlyin ET-sensitive disease (1,297/1,651 of de novo or TFI >12 months) andwith fulvestrant in 449 (19.8%) patients, mainly for ET-resistant disease (323/449of TFI ≤12 months). Atthe time of the data cut-off, the median follow-up was 54.9 months [95% CI 52.9-56.8];median OS was 49.9 months [47.4-52.3] and median TTNT was 21.3 months [19.8-22.3]in the overall population. These results varied by different subgroups (table1). Interestingly, multivariable Cox models showed age and TFI (and not ETpartner) as only independent prognostic factors for OS. Additional analysesaccording to the double stratification ET partner and endocrine sensitivitywill be presented at the conference.
| Table 1. OS and TTNT in selected subgroups (N=2,267) | |||||||
| Subgroup | Patients, n (%) | mOS [95%CI] | mTTNT [95%CI] | ||||
| Age | |||||||
| <65 | 807 (35.6) | 52.9 [49.3 – 56.5] | 17.4 [16.1 – 19.9] | ||||
| 65-74 | 794 (35.0) | 53.4 [49.5 – 59.1] | 22.8 [20.7 – 26.0] | ||||
| ≥75 | 666 (29.4) | 41.4 [36.9 – 45.2] | 23.0 [20.5 – 25.2] | ||||
| Nb of metastatic sites | |||||||
| 1 | 1204 (53.1) | 57.6 [54.5 – 62.5] | 26.5 [24.4 – 28.9] | ||||
| 2 | 510 (22.5) | 46.6 [41.8 – 51.5] | 18.0 [16.3 – 20.2] | ||||
| >=3 | 553 (24.4) | 36.6 [33.3 – 40.0] | 15.6 [13.8 – 17.1] | ||||
| Type of disease | |||||||
| Visceral | 862 (38.0) | 41.5 [37.3 – 44.7] | 16.3 [14.7 – 17.9] | ||||
| Bone-only | 857 (37.8) | 59.6 [54.8 – 64.0] | 26.3 [24.3 – 28.9] | ||||
| Endocrine sensitivity | |||||||
| De novo MBC | 750 (33.1) | 52.5 [48.2 – 58.9] | 24.1 [21.8 – 26.7] | ||||
| TFI ≤ 12 months | 511 (22.5) | 36.2 [31.9 – 40.2] | 11.9 [10.8 – 12.9] | ||||
| TFI > 12 months | 718 (31.7) | 56.5 [52.9 – 61.5] | 25.4 [23.4 – 28.7] | ||||
| ET partner | |||||||
| Fulvestrant | 449 (19.8) | 37.4 [34.1 – 41.2] | 12.8 [11.9 – 14.9] | ||||
| IA | 1651 (72.8) | 54.0 [51.0 – 57.0] | 23.9 [22.0 – 25.4] | ||||
| MBC: metastatic breast cancer; NA: not applicable; NR: not reached; OS: overall survival; TFI: treatment-free interval; TTNT: time to next treatment | |||||||
Conclusions: In this academic treatment setting, HR+/HER2- patientstreated with palbociclib + ET demonstrate similar outcomes overall and across subgroupfor TTNT and OS as seen in other RW and randomized studies. These results underscorethe impact of some characteristics on effectiveness outcomes, i.e. age, numberof metastatic sites, type of disease and TFI.
Presentation numberPS1-12-29
Real-world outcomes of second-line therapies in HR+/HER2- metastatic breast cancer after progression on CDK4/6 inhibitor
Alexandre PEINOIT, Institut de Cancérologie de l’Ouest, Saint-Herblain, France
A. PEINOIT1, F. LE BORGNE1, J. FRENEL1, L. MATHIOT1, F. BOCQUET1, A. PATSOURIS2, M. ROBERT1, E. BULTOT2, J. QUINTIN1, V. SIMMET2, M. CAMPONE1; 1Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 2Medical Oncology, Institut de Cancérologie de l’Ouest, Angers, FRANCE.
Introduction: Metastatic HR+/HER2- breast cancer (MBC) remains incurable. Following progression on CDK4/6 inhibitors and endocrine therapy, treatment options include further lines of endocrine-based regimens alone or in combination with targeted therapies such as PI3K inhibitors or everolimus, and chemotherapy. To date, no reliable predictive markers for response to second-line (2L) therapy have been established.Methods: We performed a single center cohort study of all patients with HR+/HER2- MBC who initiated 2L therapy following progression on first-line (1L) treatment with CDK4/6 inhibitors and endocrine therapy. The primary objective was to describe progression-free survival (PFS) and overall survival (OS) according to the type of 2L treatment and to identify prognostic factors associated with PFS and OS according to the type of therapy. Outcomes were assessed using the Kaplan-Meier method alongside multivariable cause-specific Cox regression models, which included factors such as the type of 2L treatment, duration of 1L CDK4/6 inhibitors therapy, metastatic sites, and HER2 expression. A prognostic model was developed for patients treated in the 2L and used to provide corrected comparisons of different treatments.Results: Between January 1, 2018, and December 31, 2023, 281 women were included. Median age was 67 years and 80% were postmenopausal. 30% had de novo MBC and 20% had bone-only metastasis. Additionally, 78% of patients had an ECOG Performance Status (PS) of 0 or 1. In the 2L, 149 patients were treated with chemotherapy alone or combined with maintenance endocrine therapy, 84 received everolimus plus endocrine therapy, and 48 were given other treatment. Median follow-up was 26.1 months (95% CI: 22.7 – 29.2). For patients treated with everolimus, the median PFS and OS were 4.9 months (95% CI: 3.9–6.5) and 23.4 months (95% CI: 19.4–36.7), respectively. In comparison, patients treated with chemotherapy had a median PFS of 4.4 months (95% CI: 3.4–5.7) and a median OS of 14.9 months (95% CI: 10.7–18.7) in the overall population. Among patients who received 1L CDK4/6 inhibitors for more than 12 months, those treated with everolimus had a median PFS of 4.5 months (95% CI: 3.7–6.5) and OS of 27.8 months (95% CI: 20.0–41.4). In contrast, patients treated with chemotherapy in this subgroup experienced had a median PFS of 6.7 months (95% CI: 4.6–8.9) and OS of 18.7 months (95% CI: 15.3–27.6). The multivariable Cox model incorporated ECOG-PS score, metastatic sites, 2L treatment category, duration of CDK4/6 inhibitors in 1L and HER2 expression. None of the interaction terms tested between the 2L treatment and the duration of CDK4/6 in 1L, the metastatic site and the HER2 expression were significant. We observed a significant increase in the risk of death for patients treated with chemotherapy alone versus everolimus HR = 1.80, (95% CI: 1.07-3.04), p = 0.0269. This risk also increases for patients with an ECOG-PS score > 2 HR = 5.20, (95% CI: 2.71-9.96), p <0.0001, patients with 12 months or less of CDK4/6 inhibitors in 1L HR = 1.59, (95% CI: 1.07-2.35), p = 0.0215 and patients with brain or liver metastasis compared to bone only metastasis HR = 2.58, (95% CI: 0.35-.94), p = 0.0042.Conclusion: These preliminary results suggest everolimus with endocrine therapy is an option for patients with HR+/HER2- metastatic breast cancer in 2L who received more than 12 months of CDK4/6 inhibitors in 1L, have an ECOG-PS score < 2, and present with bone-only metastasis. Chemotherapy may be considered otherwise. This database enabled the development of a prognostic model that identifies different patient profiles and helps clinicians choose the best treatment option. Prospective studies must be conducted to confirm the prognostic model.
Presentation numberPS1-12-30
Real-world outcomes of first-line cdk4/6 inhibitor use in hr+/her2− metastatic breast cancer: a single-institution experience
Fatima T FAQIHI, King Fahad Medical City, Riyadh, Saudi Arabia
F. T. FAQIHI, M. F. ALDAWOUD, N. ALGAZLAN, A. A. ALMAZYAD, A. R. ALTAMIMI, M. RUDAINEE, M. M. ALHARBI, A. A. ALWOHAIBI, A. K. ALTWAIRGI; Medial Oncology, King Fahad Medical City, Riyadh, SAUDI ARABIA.
Title:Real-World Outcomes of First-Line CDK4/6 Inhibitor Use in HR+/HER2− Metastatic Breast Cancer: A Single-Institution Experience Background:CDK4/6 inhibitors are a standard first-line treatment for HR+/HER2− metastatic breast cancer (MBC). However, real-world data are needed to confirm their effectiveness outside clinical trial settings. This study evaluates progression-free survival (PFS) and identifies predictors of early progression in patients who received first-line CDK4/6 inhibitors at a tertiary cancer center. Methods:We conducted a retrospective analysis of 82 patients with HR+/HER2− MBC treated at King Fahad Medical City, Riyadh, between January 2021 and December 2023. Patients were categorized based on receipt of first-line CDK4/6 inhibitors (n=59) vs non-CDK therapies (n=22). PFS was estimated using Kaplan-Meier methods. Subgroup analyses and logistic regression were performed to identify predictors of short PFS (<12 months) among CDK4/6-treated patients. Results:Among the 82 patients, median age was 50 years (range: 30-78), 93% were female, 70% had de novo MBC, and 44% had visceral metastases. Visceral crisis was observed in 7.3% of the cohort.Of the 59 patients treated with first-line CDK4/6 inhibitors (33 palbociclib, 26 abemaciclib). Visceral metastases were present in 61%, while 32% had bone-only disease. AI was the endocrine partner in 71%, and fulvestrant in 29%. Median PFS was 22.2 months (range: 1.0-55.4), with longer PFS in de novo vs recurrent (24.3 vs 16.4 mo) and bone-only vs visceral metastases (27.9 vs 18.5 mo). No significant PFS difference was observed between palbociclib and abemaciclib.Multivariable analysis identified recurrent presentation and visceral metastases as predictors of shorter PFS. To date, death events occurred in only 11 of 82 patients (13.4%), precluding meaningful OS analysis. Conclusion:First-line CDK4/6 inhibitors offer substantial PFS benefit in HR+/HER2− MBC. No difference in outcome was observed between palbociclib and abemaciclib. OS data remain immature due to limited follow-up and low event rate. Recurrent presentation and visceral disease were associated with shorter PFS and may warrant closer monitoring in clinical practice.
Presentation numberPS1-08-14
Prognostic impact of estrogen receptor expression levels in HR-positive, HER2-negative metastatic breast cancer treated with CDK4/6 inhibitors and endocrine therapy
Yeon Gil Jeong, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea, Republic of
Y. Jeong1, J. Lee2, K. Shin2, S. Bae3, S. Ahn3, J. Jeong3, Y. Cha4, J. Kim1; 1Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, KOREA, REPUBLIC OF, 3Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 4Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF.
Background: Cyclin-Dependent Kinase 4 and 6 (CDK4/6) inhibitors in combined with endocrine therapy represent the standard first-line treatment for hormone receptor (HR)-positive/Human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (MBC). Recent studies suggest that the level of estrogen receptor (ER) expression may influence treatment response. This study aims to evaluate the association between ER expression levels and clinical outcomes in this population. Methods: A retrospective study was conducted on 437 HR-positive, HER2-negative MBC patients who received CDK4/6 inhibitors (palbociclib, ribociclib, or abemaciclib) in combination with endocrine therapy at Gangnam Severance Hospital and Seoul St. Mary’s Hospital in Seoul, Korea, between 2017 and 2024. Among them, 276 patients with available ER expression level data who received this regimen as first-line palliative therapy were included in this study. ER expression was assessed using tumor tissue obtained from primary sites in de novo cases and from recurrent sites in relapsed cases. ER expression levels were stratified as high (Allred score 5-8) or low (Allred score 3-4). Progression-free survival (PFS) and overall survival (OS) were analyzed using Kaplan-Meier method and compared with the log-rank test. Results: Median follow-up was 33.7 months (range, 2.0–100.7), and median patient age was 54.3 years (range 28.0–91.6). Among the 276 patients, 195 (70.6%) received palbociclib, 70 (25.4%) received ribociclib, and 11 (4.0%) received abemaciclib. Based on the Allred score, 267 (96.7%) were classified into the high ER group and 9 (3.3%) into the low ER group. In addition, 120 (43.5%) had de novo stage IV disease and 156 (56.5%) had recurrent stage IV disease, of whom 82 (52.6%) were classified as endocrine-resistant and 74 (47.4%) as endocrine-sensitive. Patients with high ER expression had significantly longer PFS than those with low ER group (median PFS: 34.3 vs. 6.3 months; 95% CI: 27.6–42.0 vs 5.2–N/A; p=0.0014). Median OS was not reached in the high ER group, whereas it was 35.7 months in the low ER group (95% CI: 72.7–N/A vs 8.2–N/A; p < 0.0001). Conclusion: High ER expression was associated with improved clinical outcomes in patients with HR-positive/HER2-negative MBC treated with CDK4/6 inhibitors and endocrine therapy. Patients with low ER expression, on the other hand, may derive less benefit from CDK4/6 inhibitors-based treatment, and consideration of alternative approaches such as chemotherapy or emerging targeted therapies may be warranted in this subgroup. Although the difference was statistically significant (p<0.05), these results should be interpreted with caution due to the small sample size in the low ER group. Further studies with larger cohorts and longer follow-up are warranted to validate these findings.
Presentation numberPS1-08-13
A retrospective study to assess real world rates of dose modification, therapy discontinuation and treatment outcomes in patients receiving the oral CDK4/6 inhibitors Ribociclib and Palbociclib.
Clara Steele, Bon Secours Hospital Cork, Cork, Ireland
C. Steele1, J. Shaw2, D. O’Connor1, K. Dunne1, D. O’Mahony1, C. Murphy1; 1Department of Medical Oncology, Bon Secours Hospital Cork, Cork, IRELAND, 2School of Medicine, University College Cork, Cork, IRELAND.
Introduction: The oral CDK4/6 inhibitors palbociclib and ribociclib have demonstrated improved outcomes for hormone receptor positive (HR+), HER2-negative advanced breast cancer when combined with endocrine therapy in both the first- and second-line settings. In the first line setting the randomized phase Ill PALOMA-2 (palbociclib) and MONALEESA-2 (ribociclib) trials demonstrated improved progression free survival (PFS) while MONALEESA-2 also showed an improvement in overall survival rates. We aimed to examine the rates of treatment modification and cessation among patients at our cancer center receiving these agents for metastatic or unresectable advanced HR+/HER2- breast cancer.Methods: We conducted a single center retrospective review of patients treated with CDK4/6 inhibitors between 01 June 2014 and 01 June 2024. Patients who were initiated on endocrine therapy plus palbociclib or ribociclib for metastatic or unresectable advanced HR+/HER2- breast cancer within the above time frame were included. The primary endpoints were dose reductions, dose delays, and treatment cessation within the first six cycles, the secondary endpoint was PFS. Chi-squared tests were used to compare treatment modification rates between the groups, and PFS was assessed using Kaplan-Meier survival analysis. Statistical analysis was conducted using GraphPad PRISM Software.Results: A total of 84 patients were included in this study: 43 received palbociclib and 41 received ribociclib. Overall dose reduction rates were 34.8% (n=15/43) for the palbociclib group and 58.5% (n=27/41) for the ribociclib group, there was a statistically significant difference between the two groups (χ² 8.052, p=0.0045). 10/15 (66%) of patients requiring dose reductions of palbociclib had this reduction within the first six cycles, and three of these patients (n=3/10) had an additional dose reduction during the same period. 22/27 (82%) of patients requiring ribociclib dose reduction had this reduction within the first six cycles, and four of these patients (n=4/22) had an additional dose reduction during the same timeframe. Overall dose delays were 39.5% (n=17/43) for palbociclib and 58.5% (n=27/41) for ribociclib with statistical significance observed between the two groups (χ² 5.828, p=0.0158). Most of the treatment delays were in the first six cycles in both groups: 88.2% (n=15/17) of palbociclib delays and 96.3% (n=26/27) of ribociclib delays. In the first six cycles of treatment, neutropenia was the most common cause cited in both groups for dose reductions (palbociclib [n=5/10], ribociclib [n=17/22]) and dose delays (palbociclib [n=8/17], ribociclib [n=17/22]). Treatment discontinuation rates prior to completion of six cycles were 24.4% (n=10/41) on palbociclib and 19.5% (n=8/41) on ribociclib (χ² 0.175, p=0.676). We assessed PFS among patients receiving CDK4/6 inhibitors with an aromatase inhibitor and there was no statistical difference in PFS observed.Conclusion: The use of ribociclib in our cohort was associated with more treatment delays, dose reductions and subsequent dose reductions when compared with palbociclib. The reasons for dose reduction were in keeping with published adverse events i.e. neutropenia with both agents and hepatotoxicity with ribociclib. Survival analysis among the subset of patients receiving CDK4/6 inhibitors with an aromatase inhibitor showed no statistically significant difference in PFS. Numerical trends towards improved event free survival favored ribociclib, although this was not statistically significant, and a larger dataset may provide more information.
Presentation numberPS1-08-12
Rewiring Treatment Pathways: How Molecular Profiling After CDK4/6 Inhibitor (CDK4/6i) Failure Shapes Future Therapy in Luminal HER2-Negative Breast Cancer
Letícia Coelho de Mattos, AC Camargo Cancer Center, São Paulo, Brazil
L. Coelho de Mattos, A. Cicilini, N. Soldi, V. Basilio, M. Godner, L. Leite, A. Ribeiro, R. Souza, N. Pandolfi, M. Tavares, F. Pinto, A. Diniz, M. de Brot, F. Makdissi, S. Sanches, V. Lima, E. Santana dos Santos; Clinical Oncology, AC Camargo Cancer Center, São Paulo, BRAZIL.
Introduction: The incorporation of CDK4/6 inhibitors has significantly improved outcomes in patients with Luminal HER2-negative metastatic breast cancer. Nonetheless, resistance invariably emerges, limiting long-term efficacy and challenging subsequent treatment decisions. The therapy of choice after progression to cyclin inhibitor is not clearly defined, therefore molecular profiling may offer a valuable opportunity to uncover resistance mechanisms and identify actionable genomic alterations. This study aimed to characterize molecular alterations of luminal breast cancers treated with CDK4/6i in the real world setting, with the goal of guiding future therapy selection and optimizing treatment sequencing. Methods: Tumor samples were obtained from patients with HR+/HER2− breast cancer treated with CDK 4/6i, either from metastatic lesions collected at progression or from primary tumors prior to systemic therapy initiation. Following histopathologic evaluation and manual microdissection, DNA was extracted from FFPE tumor slides and sequenced using a targeted NGS panel (Somatic GS Focus Panel). The panel interrogates SNVs and Indels across 25 cancer-relevant genes (AKT1, ALK, ATM, BRAF, BRCA1, BRCA2, EGFR, ERBB2, ESR1, FGFR3, IDH1, IDH2, KIT, KRAS, MET, NRAS, PALB2, PDGFRA, PIK3CA, POLE, PTEN, RET, and TP53) and detects high-level copy number alterations in PTEN, EGFR and ERBB2. Variants were interpreted by molecular pathologists within a certified molecular diagnostics workflow. Results: Of the 72 patients selected, 16 (22%) were excluded due to insufficient DNA quality obtained from tumor samples or due to technical sequencing failure, resulting in 56 successfully sequenced cases. Among these, 30/56 (54%) presented a mutation in one of the genes analyzed. Pathway-level analysis revealed mutations in the PI3K/AKT/mTOR signaling in 21 (38%) cases (15 PIK3CA, 2 AKT1, 4 PTEN). Also related to resistance to endocrine therapy, ESR1 mutations were present in 3 tumors (5%). We also observed alterations related to homologous recombination deficiency in 5 cases (10%), including 1 BRCA1mut, 2 BRCA2mut, 2 ATMmut. Additionally, TP53 mutations were detected in 6 cases (8%) and ERBB2 mutations in 5 samples (7%). These genomic profiles reflect distinct therapeutic resistance mechanisms and highlight targetable vulnerabilities that may indicate the value of using agents like SERDs, PI3K inhibitors, or therapies targeting DNA repair deficiencies. Conclusion: Post-progression molecular profiling following CDK4/6 inhibitor failure reveals a diverse spectrum of clinically relevant alterations in luminal HER2-negative breast cancer. The predominance of PIK3CA, TP53 and ESR1 mutations, along with alterations in canonical signaling pathways, supports the integration of genomic data into post-CDK4/6 therapeutic planning. These findings reinforce the importance of a precision oncology approach to optimize treatment sequencing and personalize care for patients with HR+/HER2− metastatic breast cancer. The high failure rate (22%) of tissue testing underscores the need for more effective methods to obtain this valuable information, such as liquid biopsies.
Presentation numberPS1-08-11
Palbociclib in real clinical practic: Results of a single-center observational study
Aleksandr Sultanbaev, Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Bashkortostan, Ufa, Russian Federation
A. Sultanbaev1, I. Kolyadina2, K. Menshikov1, S. Musin1, S. Nadezda1; 1Department of Chemotherapy, Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Bashkortostan, Ufa, RUSSIAN FEDERATION, 2Department of Chemotherapy, Russian Medical Academy of Continuous Professional Education, Moscow, RUSSIAN FEDERATION.
Goals: Analyze of palbociclib using for treating patients with HR+/HER2- advanced breast cancer (mBC) at the Republican Clinical Oncology Dispensary. Methods: Data from 323 patients were analyzed. Our study examined the effectiveness of combination endocrine therapy with palbociclib in patients with metastatic HR+ HER2- breast cancer. Data on the clinicopathological characteristics of patients and disease progression during palbociclib administration were obtained by reviewing clinical data from patient records and radiological/pathological examination reports. Results: The median age of patients included in the study was 62 years. When assessing the antitumor response, a partial response was recorded in 54 patients (16.7%), stabilization in 212 patients (65.6%), and progression in 57 patients (17.8%). The median progression-free survival was 13 months. Grade 3 adverse events were noted in 23 patients: neutropenia in 21.7% of cases, hepatotoxicity in 47.8% of cases, cardiotoxicity in 17.4% of cases, and coagulopathy in 4.3%. No patient discontinued therapy due to adverse events. The best treatment results were achieved by patients who used the combination of palbociclib with an endocrine partner as the first line of treatment for advanced stage. The assessment of overall survival in our study showed slightly worse results than in the registration studies. The predicted median was 31 months, whereas in PALOMA-2 and -3 these indicators were 34-37 months; the 20-month survival rate in the PALOMA-3 study was also at the level of 60%, but in PALOMA-2 this indicator reached 80%. The effect of the depth of response to therapy and the time to subsequent progression was statistically significant; patients with stabilization of the tumor process on therapy with palbociclib had a higher risk of progression than those with a partial response. No cases of complete response were reported, and the frequency of disease control was 82.4%, which turned out to be quite close to the results of the PALOMA-2 study – 84.9% and higher than the results of therapy in PALOMA-3 – 66.6%. Conclusions: This analysis of real-world data on the use of palbociclib in real-world clinical practice confirms the RCT data on the efficacy and safety of using CDK4/6 inhibitors for the treatment of patients with HR+ HER2- mBC.
Presentation numberPS1-08-10
Adherence to Guidelines Recommended Germline Testing and Mutational Status of BRCA1/2 Genes and Beyond as a Predictor of Response to CDK4/6 Inhibition in Advanced Breast Cancer
Natalija Dedić Plavetić, University Hospital Centre Zagreb, Zagreb, Croatia
E. Paponja1, K. Čular2, K. Kanceljak2, K. Bilić3, M. Križić2, M. Matec2, M. Popović2, I. Rako4, N. Dedić Plavetić2; 1University of Zagreb, School of Medicine, Zagreb, CROATIA, 2Department of Medical Oncology, University Hospital Centre Zagreb, Zagreb, CROATIA, 3Department of Plastic Surgery, University Hospital Centre Zagreb, Zagreb, CROATIA, 4Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, CROATIA.
Objective: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) combined with endocrine therapy are the standard first-line treatment for hormone receptor-positive/HER2-negative advanced breast cancer (HR+ HER2- ABC). Germline BRCA1/2 mutation (gBRCAm) has been associated with poorer treatment outcomes in this setting, but the predictive value of other germline breast cancer susceptibility gene mutations (beyond-BRCAm) remains unexplored. Methods: We aimed to compare time to treatment failure (TTF) in gBRCAm and beyond-BRCAm vs. patients without confirmed germline mutations treated with CDK4/6i at the Department of Oncology of the University Hospital Centre Zagreb from 2018 to 2023. Clinical data were collected retrospectively and prospectively depending of the timing of germline testing. TTF, defined as discontinuation due to progression, toxicity, or patient preference, was analyzed using Kaplan-Meier estimates, and group comparisons were assessed with the log-rank test. Results: Among 389 patients treated with CDK4/6i at our institution in advanced setting, only 49 of them (12.6%) underwent germline genetic testing. Of these, 9 (18.4%) were gBRCAm (2 BRCA1, 4 BRCA2, 3 PALB2), 7 (14.3%) were beyond-BRCA (ATM, CHEK2, FANCI, FANCC, AIP, LZTR1, and MYC), and the remaining 33 (67.3%) were without confirmed germline mutations. Treatment failure occurred in 35 out of 49 patients. The overall median TTF was 20 months (95% CI, 16-29), with a mean TTF of 31.3 months (95% CI, 23.3-39.3). In the gBRCAm cohort (n=9), treatment failure occurred in 8 patients. Median TTF was 9 months (95% CI, 6-NA), and mean TTF was 19.9 months (95% CI, 6.0-33.8). In the non-gBRCAm cohort (n=40), treatment failure occurred in 27 patients. Median TTF was 23 months (95% CI, 18-56), and mean TTF was 34.6 months (95% CI, 25.4-43.8). TTF was significantly shorter in gBRCAm subgroup (log-rank χ² = 4.7, p = 0.03). In the beyond-BRCA subgroup (n=7), treatment failure occurred in 5 patients. Median TTF was 17 months (95% CI, 10-NA), and mean TTF was 35.6 months (95% CI, 13.9-57.3). In the group without confirmed germline mutations (n=33), treatment failure occurred in 22 patients. Median TTF was 24 months (95% CI, 18-NA), and mean TTF was 35.2 months (95% CI, 24.9-45.5). Log-rank comparison for the three subgroups yielded χ² = 4.7, p = 0.09. Conclusion: In this single-institution cohort, gBRCAm patients demonstrated significantly shorter TTF when treated with CDK4/6i combined with endocrine therapy, suggesting outcomes consistent with results of previous studies in this specific patient population. Patients with beyond-BRCAm showed TTF outcomes comparable to those without confirmed germline mutations, although interpretation is limited by the small sample size. Notably, only 12.6% of patients underwent germline genetic testing, highlighting a need to adhere to BRCA1/2 testing guideline recommendations for ABC to optimize individualized treatment strategies in HR+/HER2- ABC. Expanded genetic testing and further studies are warranted to clarify the predictive value and underlying mechanisms of resistance for both gBRCAm and beyond-BRCAm.
Presentation numberPS1-09-03
Let’s talk about sexuality: SEXual SATisfaction questionnaire in patients with Metastatic breast cancer (SEXSAT-M)
Josefina Cruz Jurado, Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
J. Cruz Jurado1, J. Chacón López-Muñíz2, S. Antolín Novoa3, F. Henao Carrasco4, B. Cantos Sánchez de Ibargüen5, S. Estalella Mendoza6, A. Santaballa Bertrán7, C. Salvador-Coloma8, Y. Fernández Pérez9, M. Muñoz Mateu10, C. Rodríguez Sánchez11, M. Ruiz Díaz12, B. Martín Calero1, M. Ramos Aránguez2, N. Gallego Pena4, B. Núñez García5, J. Baena-Cañada13, M. Rivero Silva14, C. Pagés Rivero14, J. Salvador Bofill15; 1Medical Oncology Department, Hospital Universitario de Canarias, Santa Cruz de Tenerife, SPAIN, 2Medical Oncology Department, Hospital General Universitario de Toledo, Toledo, SPAIN, 3Medical Oncology Department, Complejo Hospitalario Universitario A Coruña, A Coruña, SPAIN, 4Medical Oncology Department, Hospital Universitario Virgen de la Macarena, Sevilla, SPAIN, 5Medical Oncology Department, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, SPAIN, 6Medical Oncology Department, Hospital Universitario Puerta del Mar, Cádiz, SPAIN, 7Medical Oncology Department, Hospital Universitari i Politècnic La Fe, Valencia, SPAIN, 8Medical Oncology Department, Hospital Lluís Alcanyís de Xàtiva, Valencia, SPAIN, 9Medical Oncology Department, Hospital Universitario Central de Asturias, Oviedo, SPAIN, 10Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, SPAIN, 11Medical Oncology Department, Hospital Universitario de Salamanca, Salamanca, SPAIN, 12School of Psychology, Universidad Autónoma de Madrid, Madrid, SPAIN, 13Medical Oncology Department. Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Cádiz, SPAIN, 14Medical, Oncology, Pfizer – Spain, Madrid, SPAIN, 15Medical Oncology Department, Hospital Universitario Virgen del Rocío, Sevilla, SPAIN.
Background: Breast cancer (BC) treatments produce changes in patients that can interfere with their identity, attractiveness, self-esteem, social relationships, and sexual functioning. Data suggest that patients with metastatic BC (MBC) report significant sexual concerns and would like support in coping with treatment-related sexual concerns. A Spanish study, developed and validated the “Sexual Satisfaction Questionnaire” (SEXSAT-Q) that is capable of measuring sexual satisfaction in early BC. It fails to cover critical aspects of MBC. For this reason, a group of Spanish hospitals has adapted and validated a new version with the objective of measuring sexual satisfaction in MBC (SEXSAT-M study). Methods: First, a focus group was conducted with 6 patients to assess concept relevance and extract contents. A sample of 22 patients was recruited at a single center to pilot the first version for language comprehension. A sample of 223 patients was recruited at 12 oncology hospitals in Spain to validate the new questionnaire. A retest sample (58 patients) was used to check time stability. Patient satisfaction is structured in four related dimensions: self-image, psychological state (depression-energy), sexual activity, and medical care. The sexual activity dimension explores content such as level of sexual activity, interest in sexual activity, discomfort, libido, and satisfaction with sexual activity. The self-administered questionnaire includes 18 items plus 3 bolt-on items from the QLQ-BR23 questionnaire referring to sexual activity in the last month. Questions are scored using a 5-point Likert scale: from 0 (No, not at all) to 4 (Yes, a lot). To assess the psychometric properties feasibility, reliability, content, construct and concurrent validity were used. Results: Focus group participants corroborated the need to assess the impact of the disease on sexual life. The pilot sample (mean age: 51.3 years, 60.9% postmenopausal) was able to answer all items with little or no effort, and 77% answered the complete questionnaire, yielding a mean response time of 7.4 minutes. One item had to be reverted for better understanding. Regarding the validation sample (mean age: 48.7 years, 34.1% postmenopausal): Of the 219 usable cases, 58% responded to all questions, while the completion rate rises to 87.7% when the 3 bolt-on questions are not considered. Question 21 (Answer this question only if you were sexually active in the last month: To what extent did you enjoy sex?) has the higher blank response rate (33.3%). A patient’s score can be interpreted based on the centile in the distribution occupied by the direct score obtained. Dimension (average) scores resulted to arrange in three distinguishable levels: Libido (38.8) and Sexual Activity (42.8) located at the lowest level, Pain (51.4), Psychological State (52.5) and Medical Care (57.2) at an intermediate level, and self-image (70.3) at the highest level. Self-image shows a significantly higher average than all other dimensions (p<0.001), libido and sexual activity show a significantly lower average than all other dimensions (p0.05). Conclusions: The SEXSAT-M questionnaire was very well accepted and easy to complete by almost all patients, proving good face value and acceptance. Taking into account the results of this study, this new tool can help assess sexual satisfaction in the MBC population with the aim of improving sexual health at this stage of the disease. Our instrument gives the opportunity to expose a very sensitive content which MBC women have mentioned to be an important part of their lives, and which is seldom explored in regular practice.
Presentation numberPS1-09-04
A study of curative approach treatment along with metastasis-directed SBRT in bone-only oligometastatic breast cancer patients
Abhilash Dagar, All India Institute of Medical Sciences, New Delhi, DELHI, India
A. Dagar1, A. Ghosh2, A. Singhal3, A. Amritt1, J. Sharma4, A. Kumar5, A. Sharma1, A. Batra5, P. Patil1, S. Saini1, A. Mishra4, S. Deo4, A. Gogia5, D. Sharma1, S. Mallick1; 1Radiation Oncology, All India Institute of Medical Sciences, New Delhi, DELHI, INDIA, 2Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, INDIA, 3Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, DELHI, INDIA, 4Surgical Oncology, All India Institute of Medical Sciences, New Delhi, DELHI, INDIA, 5Medical Oncology, All India Institute of Medical Sciences, New Delhi, DELHI, INDIA.
A study of curative approach treatment along with metastasis-directed SBRT in bone-only oligometastatic breast cancer AbstractPurpose and Objectives: A combination of metastasis-directed ablative therapy and curative intent treatment for OMBC is an active area of research. We designed this prospective study to include a homogenous population of oligometastatic breast cancer patients with bone-only metastases. Materials and Methods: Patients with 1-5 bone only metastasis were included and received SBRT to the metastatic site followed by curative intent anthracycline and taxane based chemotherapy followed by surgical excision of the primary tumor. Patients with spinal metastasis received 30 Gy in 3 fractions over 3 days and non-spinal metastasis received 24Gy in 2 fractions over 2 days. Response assessment was done using PERCIST criteria. Results: Between June 2021 and April 2024, a total of 49 patients with a total of 84 metastatic sites were enrolled. After a median follow-up of 19.4 months (1.1-44.4 months), median PFS was 18.8 months (1.1-44.4 months) 95% CI 26.512-36.349, while the 1 and 2-year PFS were 81.63% and 71.43%, respectively. Local control at metastatic sites was 92.4% at first follow-up, and 97.9% had a complete metabolic response at last follow-up. Multivariate analysis for PFS showed patients who completed NACT (p-value: 0.05) and underwent surgical excision of the primary (p-value: 0.02) had significantly improved PFS, and hormone receptor positivity reached near significance (p-value: 0.07). Median OS for the entire cohort was 19.4 months (1.56-44.4 months), while the 1 and 2-year OS were 89% and 74%, respectively. Multivariate Analysis for OS revealed significantly improved OS for patients completing NACT (p-value:0.01), receiving LRRT (p-value:0.038), and presence of vertebral metastasis (p-value: 0.038). With SBRT, 96.4% of patients showed a decrease in pain medication step and requirement. The median time to pain response was 2.3 months (1.0-4.7 months). No Grade 3-4 toxicities were documented till last follow-up. Pain flare was the most common toxicity observed in 10.7% of the patients. Conclusion: Oligometastatic breast cancer is a very heterogeneous group of patients. Our study shows excellent outcomes in treatment-naïve, bone-only metastatic patients, emphasizing the need for patient selection.
Presentation numberPS1-09-06
Is there misdiagnosis and/or late diagnosis of metastatic ILC? A quantitative and qualitative patient-reported analysis.
Morgan E Cody, University of Pittsburgh, Pittsburgh, PA
M. E. Cody1, M. Balic2, R. Chang3, T. Cushing4, C. Desmedt5, C. Donnelly4, J. Foldi6, S. Freeney7, R. C. Jankowitz8, J. Levine4, L. Petitti4, N. Ryan4, K. Spencer9, G. Tseng10, C. Turner9, S. Oesterreich6, A. V. Lee6; 1School of Medicine, University of Pittsburgh, Pittsburgh, PA, 2UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, 3Biostatistics, University of Pittsburgh, Pittsburgh, PA, 4Non-profit organization, Lobular Breast Cancer Alliance, Delaware, DE, 5Laboratory for Translational Breast Cancer Research, VIB-KU Leuven Center for Cancer Biology, Leuven, BELGIUM, 6Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, 7Organization, Lobular Ireland, Ireland, IRELAND, 8Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 9Organization, Lobular Breast Cancer UK, Manchester, UNITED KINGDOM, 10Biostatistics and Computational & Systems Biology, University of Pittsburgh School of Public Health, Pittsburgh, PA.
Background Invasive lobular breast cancer (ILC) is the second most diagnosed type of breast cancer (BC) in women, after invasive breast cancer of no special type. The linear-file infiltration and diffuse growth hinder detection by conventional imaging. Metastatic ILC (mILC) often shows poor glucose uptake and is less sensitive to FDG-PET imaging. ILC metastasizes to unusual sites—peritoneum, GI tract, ovaries, urinary tract, leptomeninges, and orbit—which may go unrecognized after a long disease-free interval. Some metastatic sites cause nondescript symptoms, like GI pain, with published case reports of mILC misdiagnosed as gastric cancer. These atypical BC metastatic sites may lead to late and/or misdiagnosis, thereby delaying effective treatments. Objective Given the limitations in accurate and timely diagnosis of mILC, we developed a patient survey to investigate the patient-reported prevalence of delayed diagnosis or misdiagnosis of mILC and their impact on treatment outcomes. Methods A 45 question survey was developed and piloted with breast cancer researchers, clinical oncologists, and patient advocates. An IRB-approved survey was administered to patients with mILC via social media, advocacy organizations, and conferences to assess patients’ history, symptoms, diagnosis, and the impact of misdiagnosis or delays on treatment. Analyses including data QC and visualization were conducted in R using descriptive statistics. Incomplete or inconsistent responses were excluded, and summary statistics were stratified by four common mILC sites to highlight subgroup differences. Results 520 patient surveys were completed, with 450 patients diagnosed with ILC, and 321 diagnosed with mILC. Those with mILC, 33.3% (n=107) were diagnosed de novo at initial presentation. Of the patients diagnosed with mILC, 32.1% (n=103) presented with other medical conditions at diagnosis. Misdiagnosis was reported by 26.2% (n=84) of patients with mILC, and 31% (n=26) had ≥2 misdiagnoses before receiving an accurate one. The top 5 misdiagnoses (52% of all misdiagnoses) were benign breast condition (18%), bone related condition (18%), another type of BC (6%), diagnostic delay (7%), and primary GI cancer (3%). 44.5% of patients waited ≥1 year for an accurate diagnosis. The most reported contributors to delayed or misdiagnosis were inconclusive imaging, lack of ILC knowledge by providers, and initial misdiagnosis. Of the 321 mILC patients, 138 (42.9%) reported experiencing symptoms prior to diagnosis, the most common being back pain (16.5%), fatigue/malaise (14.9%), GI symptoms (11.8%), bloating (8.4%), and weight loss (8.1%). 47 patients described their misdiagnoses, with a total of 72 misdiagnoses: 23 GI, 19 GU, 15 neurological, and 15 hematological. This includes patients who reported >1 misdiagnosis of the same type, e.g., 17 patients had GU misdiagnosis, but the total number of GU misdiagnoses was 19. 38% were treated for their misdiagnosis, mostly with musculoskeletal, pain, or GI therapies; 6 patients received unnecessary cancer treatments. Diagnostic work-up for most patients included CT, biopsy, MRI, labs, and US. Although 40% of patients indicated they had a mammogram at the time of their initial misdiagnosis, ILC was detected in only 20.5% of these cases, detecting only 5 of 20 de novo mILC cases. Patients reported additional diagnostic testing within 1-3 mos. of their initial mammogram, including biopsy, US, and MRI. 47.6% of patients were in active BC surveillance at the time of their mILC diagnosis; however, no difference was seen in time to diagnosis vs those not under surveillance. Conclusion Our survey results underscore the urgent need to improve diagnostic strategies for mILC. Addressing delays and diagnostic errors in mILC is critical to optimizing treatment strategies and improving patient outcomes.
Presentation numberPS1-09-07
Central nervous system disease in patients with metaplastic breast cancer receiving antibody drug conjugates: a multi-institutional study
Sarah Premji, Mayo Clinic, Rochester, MN
S. Premji1, S. Jacob2, A. LeVee3, S. Marion2, S. Fisch4, L. Huppert4, J. Mortimer3, D. Schmolze3, A. Nayak2, A. DeMichele2, P. Shah2, R. Leon-Ferre1, D. Idossa1; 1Department of Medical Oncology, Mayo Clinic, Rochester, MN, 2Department of Medical Oncology, University of Pennsylvania, Philadelphia, PA, 3Department of Medical Oncology, City of Hope, Duarte, CA, 4Department of Medical Oncology, University of California – San Francisco, San Francisco, CA.
Introduction: Metaplastic breast cancer (MpBC) represents a rare subtype, constituting <1% of all breast cancer cases. It is pathologically characterized by epithelial and mesenchymal cellular components exhibiting a basal molecular pattern, hormone receptor and HER2-negativity, and high grade. Clinically, MpBC exhibits an aggressive course with early disease progression and poor survival. Antibody-drug conjugates (ADCs) have been shown to penetrate the central nervous system (CNS); however, little is known about the response of ADCs in MpBC, particularly in patients with CNS disease. Here we aimed to describe features of CNS disease in a subset of patients (pts) who received an ADC for MpBC. Methods: We conducted a multi-institutional retrospective observational study of pts with advanced MpBC treated with ADCs at 4 NCI-designated cancer centers (abstract #A-345). This analysis includes a subset of this cohort with CNS metastases, with the aim of examining the incidence, characteristics, and treatment patterns. Clinical outcomes were evaluated for pts with information available, including CNS median progression-free survival (mPFS; defined from initiation of systemic therapy to time of further documented CNS progression or death) and median real world PFS (rwPFS; defined from time of treatment initiation to documented disease progression or death). Results: We identified 48 pts with MpBC who received an ADC. Among them, we identified 15 (31%) who developed CNS metastases. Of these, the majority (n=11, 73%) had triple-negative breast cancer, and 4 (27%) had HER2-positive breast cancer. The median age was 50 years (32-75 years). The median interval from the initial MpBC diagnosis to CNS metastasis was 13 months (0-54 months). Excluding those with de novo CNS disease, the median interval from time of metastatic diagnosis to development of CNS metastasis [mets] was 3 months (0-22 months). 4 (27%) pts had de-novo metastatic disease, of which 2 (50%) presented with CNS disease. 4 (27%) pts had screening CNS imaging prior to their diagnosis of CNS metastatic disease. At the time of CNS disease diagnosis, 9 (60%) pts had oligometastatic intraparenchymal CNS disease (defined as 5 CNS mets) CNS disease, and 1 (6%) had leptomeningeal and intraparenchymal disease. Most pts 14(93%) with CNS disease were symptomatic at time of diagnosis. One pt underwent resection, 9 (60%) underwent stereotactic radiosurgery (SRS), 6 (40%) underwent whole brain radiation therapy (WBRT), and 1 pt received intrathecal chemotherapy with methotrexate. The median number of lines in the metastatic setting prior to documented CNS disease was 1 (range 0-3). The majority 13 (87%) underwent a change in systemic therapy at time of CNS disease diagnosis, with 60% changing to an ADC at the time of CNS diagnosis or progression. Among pts with progression of CNS disease warranting change in systemic therapy, the median time from first CNS event to further CNS progression was 6.5 months. Median CNS PFS was 3.5 months (range 1 -74 months) and median rwPFS was 4.4 months. For those patients who used ADCs after diagnosis of metastatic CNS diagnosis (12/15), median rwPFS was 4 months. Conclusions: Within this CNS sub-cohort study, we found that 31% of pts with MpBC developed CNS metastasis. The majority of pts had neurological symptoms at diagnosis, but most pts did not undergo routing screening for CNS disease. Given the high rate of CNS mets within a short timeframe from diagnosis with poor outcomes, CNS screening should be considered. The extremely short median CNS PFS and low median rwPFS of this cohort highlight the guarded prognosis associated with MpBC. Given the small sample size of this cohort, larger studies are warranted to validate these findings.
Presentation numberPS1-09-08
Impact of locoregional treatment after 6 months of palbociclib + letrozole combination therapy in de novo er+, her2- metastatic breast cancer patients: first results of the prospective palatine trial
Claire Bonneau, Institut Curie, Saint Cloud, France
C. Bonneau1, P. Cottu2, F. Berger3, R. Sabatier4, I. Desmoulins5, R. Nguefack6, C. Jouannaud7, X. Durando8, P. Augereau9, G. Emile10, E. Rassy11, T. Bachelot12, C. Boaziz13, S. Guillermet14, R. Le Scodan15, S. Mijonnet16, C. Vissac-Sabatier16, A. Beddok17; 1Surgical Oncology, Institut Curie, Saint Cloud, FRANCE, 2Medical Oncology, Institut Curie, Paris, FRANCE, 3Biometry Unit, Institut Curie, Saint Cloud, FRANCE, 4Medical Oncology, Institut Paoli-Calmettes, Marseille, FRANCE, 5Medical Oncology, Centre Georges François Leclerc, Dijon, FRANCE, 6Medical Oncology, Centre Hospitalier René Dubos, Pontoise, FRANCE, 7Medical Oncology, Institut Jean Godinot, Reims, FRANCE, 8Medical Oncology, Centre Jean Perrin, Clermont-Ferrand, FRANCE, 9Medical Oncology, ICO Site Paul Papin, Angers, FRANCE, 10Medical Oncology, Centre François Baclesse, Caen, FRANCE, 11Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 12Medical Oncology, Centre Léon Bérard, Lyon, FRANCE, 13Medical Oncology, GCS RISSA – Institut de cancérologie Paris Nord, Sarcelles, FRANCE, 14Medical Oncology, Centre Eugène Marquis, Rennes, FRANCE, 15Medical Oncology, Hôpital privé à Saint-Grégoire, Saint-Grégoire, FRANCE, 16R&D, Unicancer, Le Kremlin-Bicêtre, FRANCE, 17Department of Radiation Oncology, Institut Jean Godinot, Reims, FRANCE.
Background: De novo metastatic breast cancer (mBC) accounts for approximately 30% of ER+/HER2- mBC. Treatment relies on systemic therapy. Retrospective and prospective trials have attempted to demonstrate the benefit of locoregional treatment on overall survival (OS) with mixed results. However, all these trials were before the CDK4/6 inhibitors era. The primary objective of the present trial was to evaluate the overall survival (OS) rate at 24 months in ER+/HER2- de novo mBC patients receiving palbociclib plus letrozole combination with a planned locoregional therapy after 6 months of systemic treatment Material and methods: We conducted a prospective multicentric, single arm, phase II trial, including women from 23 centers with de novo ER+/HER2- mBC. Patients with advanced, symptomatic, visceral spread, at risk of short-term, life-threatening complications and visceral crisis, were excluded. All patients received palbociclib + letrozole for 24-26 weeks, followed by a locoregional treatment of the investigator’s choice: surgery and/or radiation therapy (RT). Palbociclib and letrozole were continued until progression. Assuming that a 24-month overall survival (OS) rate of 88% or lower is not clinically meaningful, and aiming for 93% power to detect a 24-month OS rate of 94% or higher, a total of 191 patients are required (one-sided alpha of 7%). Results: From 2019-10-28 to 2022-08-01, 209 patients were screened, 199 were evaluable and 191 patients were analyzed in ITT. Median age was 56 years (range 26-86) and 115 patients (59%) were post-menopausal. For the 183 patients with unilateral BC, the median tumoral size was 37.5 mm (range <1-120), 91 (85.8%) had local lymph node involvement and tumors were mostly grade II (N=132; 74.2%) or III (N=29; 16.3%) with a mean Ki67 at 27%. 190 (95.5%) patients had 3 or fewer metastasis sites with 37 (18.6%) having bone-only metastases. All patients received at least one cycle of palbociclib and letrozole with a median duration of 22 months (IQR 10.6-31.8). Then, 112 patients (56.6%) had surgery and RT, 15 (7.6%) had surgery only, 26 (13.1%) had RT only and 45 (22.7%) didn’t receive any locoregional treatment. After a median follow-up of 24.6 months (range 2-48.9), 42 patients died. The 2-year OS rate estimated by Kaplan-Meier in the ITT population was 85.2% (CI95%: 80.2 – 90.5), below the lower limit of 91.1% calculated by the Fleming method. We observed 93 recurrence events with a 2-year progression free survival at 58.1% (IC95%: 51.4 – 65.5), including 17 patients (8.5%) with a local recurrence. Overall, 186 (93.5%) patients presented at least one adverse event (AE) linked to either systemic or locoregional treatment. Common AE of palbociclib-letrozole were neutropenia (64.3%) including 41.7% grade (gr) 3 and 5.5% gr 4, fatigue (61.3%) mainly gr 1 (46.7%), gastrointestinal disorders (51.8%) of which 36.2% gr1, 13.6% gr 2 and 2% gr 3, and hot flushes (32.2%) mainly gr 1(22.6%). AEs reported with RT were skin disorders (27.6%) of which 17.6% gr 1 and 7% gr 2, and 8% gr 1 fatigue. Surgery induced 7.5% gr 1, 4 % gr 2 and 0.5% gr 3 AEs, notably 3% gr 1 breast pain and 2.5% gr 1 lymphocele. Conclusion: In patients with de novo ER-positive/HER2-negative metastatic breast cancer, locoregional treatment after 6 months of palbociclib + letrozole is feasible, well tolerated, and provides excellent local control, but was not associated with the expected 24-month OS. These results suggest that the decision to perform locoregional therapy should be individualized, considering metastatic burden, toxicity profile, patient preferences and risk of local progression that could threaten patients’ QoL. Financial disclosures: Sponsor: Unicancer Clinical trial identification: NCT03870919 Collaborators: Pfizer
Presentation numberPS1-09-09
Prognostic significance of chemotherapy-related amenorrhea in premenopausal women with de novo metastatic breast cancer
Chongxi Ren, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, China
C. Ren1, J. Sun2, L. Kong1, H. Yu1, F. Wang1, H. Qi1; 1General Surgery, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, CHINA, 2Breast Surgery, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, CHINA.
Prognostic significance of chemotherapy-related amenorrhea in premenopausal women with de novo metastatic breast cancer Chongxi Ren,Jianna Sun,Lingjun Kong,Hongjun Yu,Wang fang,Hongxia QiDepartment of General Surgery, Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cang Zhou City, China. Background: Breast cancer is the most commonly diagnosed tumor in women. Women with de novo metastatic breast cancer (dnMBC), as a heterogeneous group, especially in young patients, represent a peculiar population with respect to tumor biology, prognosis, clinical management and survivorship issues. Currently, systemic chemotherapy remains the cornerstone of treatment for this disease. Chemotherapy-related amenorrhea (CRA) is a common complication observed in premenopausal women with breast cancer, and the incidence of CRA ranges from 15% to 94% in patients with breast cancer after receiving chemotherapy. However, up to now, its role in dnMBC remains unknown. Aim: To to investigate the clinical risk factors associated with CRA and to assess its impact on the prognosis of premenopausal women with dnMBC. Settings and design: This study was designed as a retrospective cohort study and was approved by the Ethics Committee of Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine (NO. 2017-AF29-058). Methods: Using hospital-based database, we identified women with dnMBC diagnosed between March 2010 and December 2017, collected demographic data, tumor characteristics, histopathological reports, treatment types and courses (including records of chemotherapy response and menstrual effects), and survival data. A retrospective analysis was conducted on the associated factors and prognostic effects of CRA in 291 out of 518 premenopausal patients who received continuous adjuvant chemotherapy (ACT regimen) or chemotherapy with AC regimen. Premenopausal status was defined as last normal menses within the 6 weeks preceding initiation of chemotherapy; CRA as cessation of menses for at least 3 months not later than 3 months from the end of chemotherapy. The primary outcome was overall survival (OS). It was estimated by Kaplan-Meier method and log rank test. Univariable and multivariable analyses were performed using the COX proportional hazard model to identify statistically significant prognostic factors. All statistical analyses were performed using Stata 18.0. Results: A total of 291 premenopausal patients with de novo metastatic breast cancer were included in this study. The mean patient age was 45.1 years (SD 7.8). 29.1% of the patients are over 50 years old, and there are 10 patients who are 56 years old. The majority of the patients had histopathological diagnoses of invasive ductal carcinoma (96.7%). The mean age of patients was higher for patients who had clinical amenorrhea compared to those who did not have it: 48.7 (SD 5.1) vs. 36.9 (SD 6.7) years, with a significant statistical difference (Diff:-11.7;95%CI [-13.3 to -10.1];p<0.001]). Compared with women who had not experienced amenorrhea, the OS of patients with CRA was longer, with a significant statistical difference (hazard ratios: 0.68; 95% CI[0.51-0.90];p=0.006). The prognostic value of CRA was independent of age, hormone receptor status, chemotherapy regimen, and chemotherapy cycles. Conclusions: This cohort study indicates that in premenopausal women with dnMBC, the condition of CRA is common, and it has a favorable prognosis compared to patients without amenorrhea. Further large cohort studies are necessary to confirm these results. Keywords: De novo metastatic breast cancer; Chemotherapy; Amenorrhea; Prognosis
Presentation numberPS1-09-10
Effect of Primary Breast Surgery on Prognosis in Breast Cancer Patients with Isolated Bone Metastases
Izzet Dogan, Acibadem Healthcare Group, Istanbul, Turkey
I. Dogan1, A. Azizy2, S. Yücel3, I. D. Subası4, M. Bozkurt5, O. Dülgeroğlu6, A. Arican5, I. Yıldız5, C. Uras7; 1Medical Oncology, Acibadem Healthcare Group, Istanbul, TURKEY, 2Medical Oncology, Istanbul University Institute of Oncology, Istanbul, TURKEY, 3Radiation Oncology, Acibadem Research Institute of Senology, Istanbul, TURKEY, 4Breast Radiology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 5Medical Oncology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 6Breast Surgery, Acibadem University Atakent Hospital, Istanbul, TURKEY, 7Breast Surgery, Acibadem Research Institute of Senology, Istanbul, TURKEY.
Background: Breast cancer patients presenting with isolated bone metastases represent a distinct subgroup with relatively favorable prognosis compared to those with visceral metastases. The role of primary breast surgery in this population remains controversial, with limited consensus on its impact on survival outcomes. Clarifying the prognostic effect of surgical intervention on the primary tumor may guide optimal treatment strategies. In this study, we evaluated the impact of primary breast surgery on survival prognosis and other prognostic factors in patients with isolated bone metastases. Method: This retrospective study analyzed data from the Surveillance, Epidemiology, and End Results (SEER) database, including patients with isolated bone metastatic breast cancer, between 2000 and 2021. Relevant demographic and clinicopathological variables were extracted to assess their association with overall survival. Survival analyses were conducted using Kaplan-Meier estimates, and independent prognostic factors were identified through multivariate Cox proportional hazards modeling. Results: A total of 6,500 patients were identified who met the study criteria. 3398 (52.3%) of the patients were under the age of 65. Breast cancer subtype rates were determined as follows: HR+/HER2- (77.2%), HR+/HER2+ (12.8%), HR-/HER2- (6.5%), and HR-/HER2+ (3.5%). All patients had at least one bone metastasis, and there were no other metastases, including distant lymph node metastasis. Surgery was performed in 1513 patients (23.4%). The number of patients who received breast radiotherapy was 124 (1.9%), and the rate of patients who received chemotherapy was 62.8%. The 5-year survival rate was found to be statistically significantly higher in the patient group who underwent surgery (59.5% versus 38.6%) (p<0.001). Other factors were evaluated in terms of prognosis, including age (p < 0.001), gender (p = 0.184), race (p < 0.001), origin (p = 0.331), histopathological subtype (p < 0.001), radiotherapy (p < 0.001), and chemotherapy (p < 0.001). Multivariate analysis confirmed that breast surgery had a statistically significant effect on overall survival (p < 0.001, HR: 0.40). Conclucions: Isolated bone metastatic breast cancer is a special subtype in disease presentation. In recent years, new drugs that improve survival compared to chemotherapy have been introduced in breast cancer, and primary breast surgery should also be considered in selected patients with isolated bone metastatic breast cancer.
Presentation numberPS1-09-11
Comparison of Clinicopathological Features and Prognoses in Male and Female Patients with Denovo Metastatic Breast Cancer
Izzet Dogan, Acibadem Healthcare Group, Istanbul, Turkey
I. Dogan1, A. Azizy2, I. D. Subası3, I. Yıldız4, A. Arican4, S. Yücel5, O. Dülgeroğlu6, M. Bozkurt4, C. Uras7; 1Medical Oncology, Acibadem Healthcare Group, Istanbul, TURKEY, 2Medical Oncology, Istanbul University Institute of Oncology, Istanbul, TURKEY, 3Breast Radiology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 4Medical Oncology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 5Radiation Oncology, Acibadem Research Institute of Senology, Istanbul, TURKEY, 6Breast Surgery, Acibadem University Atakent Hospital, Istanbul, TURKEY, 7Breast Surgery, Acibadem Research Institute of Senology, Istanbul, TURKEY.
Background: De novo metastatic breast cancer (MBC) presents distinct clinical challenges and accounts for a small proportion of all breast cancer cases. While extensive data exist for female patients, the characteristics and outcomes of male patients with de novo MBC remain poorly defined. Comparing clinicopathological features and prognoses between male and female patients may help improve personalized management strategies for this rare subgroup. In this study, we compared the clinicopathological features and prognoses of male and female patients diagnosed with de novo metastatic breast cancer. Method: This retrospective study utilized data from the Surveillance, Epidemiology, and End Results (SEER) database, including male and female patients diagnosed with de novo metastatic breast cancer, between 2000 and 2021. Demographic and clinicopathological variables were collected to compare baseline characteristics. Survival outcomes were assessed using Kaplan-Meier analysis, and multivariate Cox regression was performed to identify independent prognostic factors. Statistical analyses were conducted using SPSS version 30.0. Results: A total of 60,590 patients met the study criteria. Of these patients, 713 (1.2%) were male. 56.1% were under 65 years of age. Pathological subtypes were HR+/HER2- (61.2%), HR+/HER2+ (15.9%), HR-/HER2- (14.3%), and HR-/HER2+ (8.6%). All patients had de novo metastatic disease. Metastatic sites were bone (64.2%), lung (30%), distant lymph node (27%), liver (24.2%), brain (7.3%), and other (18%). At the time of diagnosis, statistically significant differences were detected in patient groups in terms of age (p<0.001), marital status (p<0.001), origin (p<0.001), race (p<0.001), histologic type (p<0.001), breast cancer subtype (p<0.001), liver metastasis (p<0.001), lung metastasis (p<0.001) 5-year survival of de novo metastatic female breast cancer patients was found to be better than male patients (31.4% vs. 25.6%, p=0.036) Conclusions: Male breast cancer is quite rare. The clinicopathological features and metastatic spread of male and female patients at diagnosis differ. Female patients were found to have a better survival rate.
Presentation numberPS1-09-12
Trop2 expression and therapeutic opportunities in inflammatory breast cancer
Shayla Murray, MD Anderson Cancer Center, Houston, TX
S. Murray1, A. Nasrazadani1, C. Yam1, A. Alexander1, O. Baranov2, D. Goncharova2, F. Paradiso2, M. Hensley2, The MDACC Inflammatory Breast Cancer Team, W. A. Woodward3, R. Layman1, S. Saleem1, V. Valero1, M. C. Stauder3, A. Lucci4, S. X. Sun4, G. J. Whitman5, M. Patel5, H. Le-Petross5, Y. Lu5, A. Marx1, C. Yajima1, M. Kai1, L. Villarreal1, H. Lopez1, B. Lim1; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2BostonGene, BostonGene Corporation, Waltham, MA, 3Breast Radiation Oncology, MD Anderson Cancer Center, Houston, TX, 4Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, 5Breast Imaging, MD Anderson Cancer Center, Houston, TX.
Background: Inflammatory breast cancer (IBC) is a rare and aggressive subtype with poor outcomes, particularly in patients with residual disease post-neoadjuvant therapy. Antibody-drug conjugates (ADCs), such as sacituzumab govitecan (SG), have shown efficacy in hard-to-treat subtypes like TNBC and HR+/HER2- disease. This study aims to establish the consistent expression of TACSTD2 (TROP2) in IBC and evaluate its potential as a therapeutic target and biomarker to improve outcomes in aggressive IBC subtypes. Methods: Both IBC and non-IBC samples were evaluated using BostonGene’s comprehensive transcriptomic analysis platform, which includes RNA-seq data processed through an internally validated pipeline. Morgan Welch IBC Program at MD Anderson Cancer Center Registry and other related protocols were utilized to recruit patients. TROP2 expression levels were quantified and categorized as high, medium, or low. A total of 71 IBC samples from this cohort then were compared with 631 non-IBC samples, including non-IBC of no special type (NST), non-IBC TNBC, and PAM50 subtype-matched non-IBC (basal-like, luminal, and HER2-enriched). Univariate analyses were conducted to assess differences in TROP2 expression across these groups. To evaluate clinical relevance, progression-free survival (PFS) was analyzed in a subset of patients (n=11) treated with SG. Results: There was no statistically significant difference in TROP2 expression between IBC and non-IBC samples when compared as whole group. In subtype-matched comparisons, no statistically significant differences were observed, although 15 of 22 basal-like IBC samples had TROP2 expression above the median of basal-like non-IBC. TROP2 expression did not differ significantly between HR+ and TNBC subtypes (p = 0.6657), and while TNBC patients had numerically longer PFS (mean: 1048.4 days) compared to HR+ patients (906.8 days), the difference was not statistically significant. PFS while treated with SG was longest in high expressors (mean: 1142.0 days), followed by medium (994.7 days) and low expressors (983.0 days); however, the correlation coefficient between TROP2 expression and PFS was negligible (r = 0.084). Conclusion: Our study supports further investigation of the role of TROP2 in IBC and its potential in therapeutic development for this rare, aggressive subtype of breast cancer. Although no significant correlation was found between TROP2 expression and PFS among SG-treated patients, TROP2 alone may not serve as a reliable predictive biomarker in this population. Notably, this univariate analysis did not account for confounding variables such as age, race, histology, stage, or prior treatments. Future studies using multivariate models could better isolate the impact of transcriptomic TROP2 expression. Given the limited surface targets in TNBC and HR+/HER2− subtypes and the relatively consistent expression of TROP2, incorporating it into a multi-marker predictive model may enhance patient selection for TROP2-targeted therapies like SG. Acknowledgement: The transcriptomic analyses were performed via collaboration with BostonGene and MW IBC program. Morgan Welch IBC Research and Clinic Program is funded by the State of Texas Rare and Aggressive Breast Cancer Grant.
Presentation numberPS1-09-13
Development of Staging Guidelines for Metastatic Breast Cancer: Which method is best?
Jennifer K Plichta, Duke University Medical Center, Durham, NC
J. K. Plichta1, V. Rey2, M. Blasingame1, S. Luo1, S. M. Thomas2; 1Department of Surgery, Duke University Medical Center, Durham, NC, 2Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC.
Background: Breast cancer staging is a foundational component for determining prognosis, and multiple methods have been used to create staging guidelines. However, it remains unclear which statistical approach is best. As such, we sought to compare 3 methods for developing staging guidelines, using de novo metastatic breast cancer (dnMBC) as an example. Methods: Patients with dnMBC in the National Cancer Database (2010-2022) were identified (N=48,832). Variables used for staging included clinical T-category, grade, ER, PR, HER2, histology, bone-only metastases, brain-only metastases, visceral metastases, and number of metastatic sites. Stage groups were assigned based on 3-year overall survival (OS) rates: stage group IVA >70%, IVB 50-70%, IVC 25-<50%, and IVD <25%. Statistical methods: (1) We conducted an analysis of all possible variable combinations (AC), yielding 643 unique characteristic profiles; OS was estimated via Kaplan-Meier, which was used to assign a subgroup (IVA-D). (2) Recursive partitioning analysis (RPA) grouped patients with similar OS based on these same disease characteristics; 3-year OS rates were estimated for each terminal node; subgroups (IVA-D) were assigned. (3) Bootstrapping was used; the RPA was repeated 1000 times (B-RPA); subgroups (IVA-D) were determined based on the subgroup each profile fell into most often. Overall, patients were categorized as stage IVA-D based on these 3 methods (AC, RPA, B-RPA), resulting in 3 stage group assignments. Differences in stage group assignments were classified as minor (1 group) or major (>1 group). Performance metrics (Akaike Information Criterion, AIC) were compared. Results: Application of the 3 methods yielded similar findings (Table), although the AC analysis had the best performance (lowest AIC), followed by the B-RPA and RPA. Patient-level Comparison (N=48,832). The same stage group (IVA-D) was assigned to 87.8% of patients by all 3 methods. Comparing B-RPA to AC, 10.6% of patients had a minor stage discrepancy and 0.4% had a major discrepancy. The RPA compared similarly to AC (10.9% minor, 0.4% major). Profile-level Comparison (N=643). Comparing B-RPA to AC, there was 60.7% agreement in stage assignments, 31.4% minor discrepancies, and 6.8% major discrepancies. Comparing RPA to AC was similar (60.8% agreement, 30.8% minor, 7.3% major). Of the 160 most common profiles, there was 85% agreement for both B-RPA and RPA. Of the 160 least common characteristic profiles, there was only 36.3% and 36.9% agreement for the B-RPA and RPA, respectively. Conclusions: Although the AC analysis had the best performance, there was a high level of agreement in stage assignment at the patient level. Potentially due to smaller patient samples in some profile subgroups, there was a moderate degree of discrepancies at the profile level, although most were minor. These findings will help inform future staging guideline development.
| Stage IVA | Stage IVB | Stage IVC | Stage IVD | |
|
Number of profiles (N=643) |
||||
| AC | 66 | 173 | 166 | 231 |
| RPA | 32 | 212 | 204 | 195 |
| B-RPA | 18 | 229 | 210 | 186 |
|
Number of patients (N=48,832) |
||||
| AC | 3878 | 24316 | 14006 | 6606 |
| RPA | 4025 | 24724 | 13656 | 6427 |
| B-RPA | 3587 | 25353 | 13637 | 6255 |
|
Unadjusted 3-year OS rates |
||||
| AC | 75.7% | 61.9% | 38.8% | 14.3% |
| RPA | 72.9% | 61.3% | 38.6% | 15.8% |
| B-RPA | 73.4% | 61.4% | 38.2% | 15.4% |
|
Hazards Ratios from multivariable analysis* |
||||
| AC | Ref | 1.603 | 2.711 | 5.454 |
| RPA | Ref | 1.575 | 2.627 | 5.034 |
| B-RPA | Ref | 1.589 | 2.679 | 5.190 |
| *Multivariable analysis included stage group (IVA-D), age group (<50, 50+), sex, race/ethnicity, chemotherapy receipt, endocrine therapy receipt, immunotherapy receipt, radiation receipt, and surgery receipt. |
Presentation numberPS1-09-14
Evaluating clinical and molecular correlates of Inflammatory Breast Cancer using a scoring system-based chart review
Isaac S Chan, UT Southwestern Medical Center, Dallas, TX
C. M. Atallah1, A. Varner1, C. J. Kang1, P. Gowda1, K. Anderson1, D. Tovar1, R. Morey1, C. Hauer2, R. Sridharan2, S. Huang2, K. Lei2, S. M. Reddy2, H. L. McArthur2, J. Maués3, C. Hodgdon3, B. Zhang4, X. Zhang5, B. Lim6, I. S. Chan2; 1UT Southwestern Medical School, UT Southwestern Medical Center, Dallas, TX, 2Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 3Founder, GRASP Cancer, Baltimore, MD, 4Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 5Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 6Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer with a distinct clinical presentation and poor prognosis. Diagnostic ambiguity remains a challenge, and recent literature has proposed structured criteria to improve case classification. Methods: We conducted a retrospective chart review of 83 patients diagnosed with IBC and 83 patients with non-inflammatory breast cancer (non-IBC), seen at the University of Texas Southwestern Medical Center as part of the Dallas Metastatic Cancer Study. Each IBC case was evaluated using a structured scoring system adapted from the diagnostic guidelines proposed by Omarini et al. (2022) to assess the degree of conformity to established IBC features. Clinical and pathological characteristics were compared across groups using appropriate statistical tests. Results: Our findings reinforce several established associations in the literature. Compared to non-IBC patients, those with IBC exhibited significantly higher Ki-67 proliferation indices (1.42-fold increase, p<0.0001, Mann-Whitney U test), greater number of metastatic sites (2-fold increase, p<0.0001, Mann-Whitney U test), and higher clinical stage at diagnosis (p=0.04, Chi-square test). IBC cases were also associated with more aggressive disease biology as they were more likely to be ER negative (p=0.003, Fisher test) and PR negative (p=0.0184, Fisher test). Notably, we observed a novel pattern: IBC was significantly associated with specific metastatic sites, particularly the brain (4.5 times more likely in IBC patients, p<0.0001, Chi-square test) and lungs (2.08 times more likely in IBC, p=0.0028, Chi-square test), suggesting a potentially distinct metastatic tropism. Conclusions: This study supports the diagnostic utility of a structured scoring system for IBC and confirms key clinical and molecular trends observed in prior research. The novel association with specific metastatic sites warrants further investigation and may inform future efforts in IBC classification and management.
Presentation numberPS1-09-15
Prognostic Impact of BRCA Mutation Status on Outcomes with CDK4/6 Inhibitor Therapy in HR+/HER2- Metastatic Breast Cancer: A Real-World Cohort Study
Akshara Raghavendra, University of Texas MD Anderson Cancer Center, Houston, TX
A. Raghavendra, W. Qiao, A. Gutierrez, S. Damodaran, R. Layman, B. Arun; Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Germline BRCA mutations (gBRCA) are well-established predictors of therapeutic sensitivity in triple-negative breast cancer; however, their clinical implications in hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC) treated with CDK4/6 inhibitors (CDK4/6i) remain poorly defined. We evaluated the association of gBRCA status with treatment patterns and clinical outcomes in a large real-world cohort of patients receiving CDK4/6i-based therapy. Methods: This retrospective cohort study included 3,000 patients with HR+/HER2- mBC treated with CDK4/6i between January 2015 to December 2024. Patients were stratified into three groups: gBRCA-mutated (n=86), BRCA wild-type (n=913), and those without germline testing (n=2001). Demographics, tumor characteristics, treatment exposures, and outcomes were compared. Primary endpoints were progression-free survival (PFS) and overall survival (OS) from the initiation of CDK4/6i therapy. Kaplan-Meier analyses and univariate Cox proportional hazards models were used to assess survival outcomes by BRCA status. Results: Compared to BRCA wild-type patients, those with gBRCA mutations were younger at diagnosis (median age 44 vs. 52 years), more likely to have received platinum agents (24% vs. 9%) and PARP inhibitors (52% vs. 2%) in any treatment setting. No significant differences in ER or PR expression, HER2 IHC distribution, or site of progression were observed between groups. Median PFS was 11.3 months (95% CI: 7.4-15.6) in gBRCA patients, versus 20.7 months (95% CI: 17.6-23.1) in BRCA wild-type (WT) and 19.8 months (95% CI: 18.2-21.7) in untested patients (P<.001). Median OS from CDK4/6 initiation was significantly shorter in gBRCA– patients (33.7 months [95% CI: 28.9-64.9]) compared to BRCA WT (58.8 months [95% CI: 53.2-65.6]) and those without testing (45.7 months [95% CI: 43.8-48.9]) (P<.001). In univariate Cox models, gBRCA was independently associated with inferior PFS (HR=1.71, 95% CI: 1.34-2.17, P<.0001) and OS (HR=1.73, 95% CI: 1.27-2.35, P=.001). Additional factors associated with worse OS included platinum exposure (HR=2.03), visceral progression (HR=1.41), and receiving palbociclib compared to ribociclib (HR=1.89). Conclusions: In this large real-world cohort, gBRCA mutations were associated with significantly worse OS and PFS among patients with HR+/HER2- mBC treated with CDK4/6i. These findings suggest that gBRCA mutation status may have prognostic implications even in luminal disease and should be considered in treatment sequencing decisions. Prospective validation and exploration of optimal therapeutic combinations, including early use of PARP inhibitors or ADCs in this population, are warranted.
Presentation numberPS1-09-16
Optimal biological dose of eftilagimod alpha, a soluble LAG-3 protein, in metastatic breast cancer patients receiving weekly paclitaxel in AIPAC-003
Nuhad K Ibrahim, The University of Texas MD Anderson Cancer Center, Houston, TX
N. K Ibrahim1, I. Garcia Fructuoso2, F. Forget3, P. Chalasani4, P. Sánchez Rovira5, R. Poncin6, J. López7, S. Morales Murillo8, K. Papadamitriou9, S. Winckels10, C. Mueller10, F. Triebel11; 1Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Medical Oncology, Hospital Clinic de Barcelona, Barcelona, SPAIN, 3Medical oncology, Centre Hospitalier de l’Ardenne Vivalia, Libramont, BELGIUM, 4Division of Hematology and Oncology, George Washington University, Washington, WA, 5Medical Oncology Unit, Hospital Universitario de Jaén, Jaén, SPAIN, 6Department of Medical Oncology, Clinique Saint-Pierre Ottignies, Ottignies, BELGIUM, 7Department of Medical oncology, Hospital Universitario La Paz, Madrid, SPAIN, 8Medical oncology, Hospital Universitario Arnau de Vilanova, Lleida, SPAIN, 9Department of Medical Oncology, University hospital of Antwerp, Edegem, BELGIUM, 10Clinical Developement, Immutep GmbH, Berlin, GERMANY, 11Research & Development, Immutep S.A.S, Saint-Aubin, FRANCE.
Background: Eftilagimod alpha (efti) is an MHC class II agonist that mediates activation of antigen presenting cells (APC) like dendritic cells and monocytes, subsequently followed by T cell activation. Aligning with the FDA’s Project Optimus, we designed AIPAC-003 (NCT05747794), a randomized phase 2 trial testing efti plus paclitaxel (P) in metastatic breast cancer (mBC) patients (pts), to test 30 vs 90 mg efti to determine the optimal biological dose (OBD). Methods: Heavily pretreated female pts with HR+ and HER2-negative or HER2-low mBC resistant to endocrine-based therapy (ET) or with metastatic triple-negative breast cancer (mTNBC) not eligible to PD-(L)1-based therapy were enrolled. After an initial safety lead-in that tested a higher 90 mg dose of efti plus P, pts were randomized 1:1 to receive 30 mg or 90 mg efti plus P. Pts received P (80 mg/m2 IV on D1, 8 and 15) followed by efti (30 mg or 90 mg SC on D1 and 15) in a 4-week cycle up to 12 months. Imaging was done Q8W and assessed by the investigator per RECIST 1.1. Predefined dose-limiting toxicities (DLTs) were assessed by an IDMC. The primary objective was to define the OBD of efti. Results: 64 evaluable pts were randomized between May 2023-Sep 2024. 19% of pts had mTNBC. Most were previously treated with ET (89%), including CDK4/6 inhibitors (84%), and considered ET-resistant (81%). Pts had a median age of 58.5 years (range 34-85), and 64% had baseline ECOG 0. Safety data showed that adverse reactions leading to treatment discontinuation occurred in 13% of pts in the 30 mg arm vs 21% in the 90 mg arm (>70% related to paclitaxel toxicity), and two DLTs occurred, both hypersensitivity reactions in the 90 mg arm. A known safety signal of efti, local injection site reactions (LISRs) that were considered long-lasting (≥5 days) occurred in 19.4% of pts in the 30 mg arm and 33.3% of pts in the 90 mg arm. None of these LISRs led to treatment discontinuation. After 8 months minimum of follow-up, time to onset of response was 2.0 months (30 mg) versus 1.9 months (90 mg). Objective response rate (ORR) and disease control rate (DCR) are presented in Table 1. Conclusion: These data show that 90 mg efti is less tolerable and does not enhance efficacy. Therefore, the 30 mg SC dose of the APC activator, efti, previously administered in >500 pts with advanced or metastatic cancer in prior phase 1 and 2 studies, is considered the OBD, suitable for evaluation in larger pt populations.
| BOR1, n (%) |
30 mg efti + paclitaxel N=31 |
90 mg efti + paclitaxel N=33 |
| Complete Response | 0 | 0 |
| Partial Response | 13 (41.9) | 16 (48.5) |
| Stable Disease | 14 (45.2) | 10 (30.3) |
| Progression | 4 (12.9) | 7 (21.2) |
| ORR1, n (%) [95% CI] | 13 (41.9) [24.6-60.9] | 16 (48.5) [30.8-66.5] |
| DCR1, n (%) [95% CI] | 27 (87.1) [70.2-96.4] | 26 (78.8) [61.1-91.0] |
|
Abbreviations: BOR: best objective response; CI: confidence interval; DCR: disease control rate; ORR: objective response rate.
1 per RECIST 1.1 |
Presentation numberPS1-09-17
Phase 1 Clinical Testing of a First-in-Class Antisense Oligonucleotide Therapeutic against Advanced Solid Tumors of Multiple Tissue Origins
Zdravka Medarova, TransCode Therapeutics, Boston, MA
Z. Medarova1, S. Fu2, A. Varkaris3, M. McKean4, M. Barve5, A. Spira6, D. Vlock1, L. Fortin1, S. Dugan1; 1NA, TransCode Therapeutics, Boston, MA, 2NA, 1The University of Texas MD Anderson Cancer Center, Houston Texas, Houston, TX, 3NA, Termeer Center for Targeted Therapies, Massachusetts General Hospital, Boston, MA, Boston, MA, 4NA, START Center for Cancer Research, Salt Lake City, UT, Salt Lake City, UT, 5NA, Sarah Cannon research Institute at Mary Crowley, Dallas Texas, Dallas, TX, 6NA, NEXT Oncology Viginia, Fairfax, VA, Fairfax, VA.
Background: TransCode Therapeutics, Inc. is developing TTX-MC138, an antisense oligonucleotide (ASO) therapeutic targeting miR-10b, a critical driver of metastatic disease progression. Several in vivo preclinical studies with TTX-MC138 have successfully demonstrated its delivery to metastatic lesions, its ability to eliminate metastasis, and its capacity to elicit complete regression without recurrence. Thus, TTX-MC138 holds the potential to improve patient outcomes over currently approved and available treatment options. Methods: The first clinical study with TTX-MC138 was a first-in-human single-center, Phase 0, microdose study to demonstrate delivery of TTX-MC138-NODAGA-Cu64 to radiographically confirmed metastases in patients with advanced solid tumors. Preliminary analysis indicated accumulation of TTX-MC138 in metastatic lesions in a patient with metastatic breast cancer. Metabolite analysis demonstrated drug stability in circulation and a blood half-life of 18.7 hours. At a 100 microgram microdose, the drug showed robust PD activity in blood over the full-time course of the study. Clinical Study: NCT05908773.The second, ongoing clinical study with TTX-MC138 is a Phase 1/2 Multicenter, Open-Label, Dose-Escalation and Expansion Study of TTX-MC138 in Subjects with Advanced Solid Tumors. The study is employing a Bayesian Optimal Interval (BOIN) design to inform dose escalation among patients with previously treated advanced solid tumors. The BOIN design incorporates four dose levels: 0.8 mg/kg, 1.6 mg/kg, 3.2 mg/kg and 4.8 mg/kg. Patients receive a once monthly administration of TTX-MC138. Preliminary data review in 16 patients indicates no dose-limiting toxicities or serious adverse events. To date, all cohorts have enrolled. Preliminary analyses suggest that TTX-MC138 demonstrates a PK/PD profile consistent with preclinical results and results from TransCode’s Phase 0 clinical study. Specifically, results confirmed the Phase 0 observation that TTX-MC138 shows evidence of pharmacodynamic activity in the presence of high baseline expression of miR-10b (PCR), reaching a 66% inhibition at 24 hours after infusion, similar to that seen in the Phase 0 clinical study. Additionally, the level of TTX-MC138 increased with the increase in dose and remained consistent with repeated administration of TTX-MC138, suggesting a favorable pharmacokinetic profile. The process leading to use of TTX-MC138 in the clinic is critically dependent on the innate tropism of the TTX drug design engine to tumors and represents a first step towards developing effective nucleic-acid based therapeutics against cancer. Clinical Study: NCT06260774.
Presentation numberPS1-09-18
Efficacy of antibody drug conjugates in patients with advanced metaplastic breast cancer: a multi-institutional retrospective cohort study
Saya Jacob, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA
S. Jacob1, S. Premji2, S. Fisch3, A. LeVee4, S. Marion1, D. Schmolze5, J. Mortimer4, L. Huppert3, D. Idossa2, A. Nayak6, P. Wileyto7, A. DeMichele1, P. D. Shah1; 1Medicine, Hematology/Oncology, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA, 2Medicine, Hematology/Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 3Medicine, Hematology/Oncology, University of California, San Francisco, San Francisco, CA, 4Medicine, Hematology/Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, 5Pathology, City of Hope, Duarte, CA, 6Pathology, Hematology/Oncology, University of Pennsylvania Abramson Cancer Center, Philadelphia, PA, 7Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
Introduction: Metaplastic breast cancer (MpBC) is a rare, aggressive subtype of breast cancer that has limited evidence-based data to guide treatment. Historically, MpBC has had limited response to chemotherapy; however, the response of MpBC to antibody-drug conjugates (ADCs) remains poorly characterized. Methods: We performed a retrospective cohort study of patients (pts) with pathologic diagnosis of MpBC who received ADC therapy as part of standard of care in the metastatic setting across 4 NCI-designated cancer centers. The primary endpoints were objective response rate (ORR) at time of best response to ADC & real world PFS (rwPFS) of ADC compared to non-ADC therapy, defined as time from treatment start to time of progression or death. Results: We identified 48 pts with MpBC who received at least one ADC between 3/2016 and 10/2025. Median age was 54 years (range 31-77); all pts were biological females. Most pts had triple negative disease (n=39, 81%), 5 (10%) had HER2+ disease, 3 (6%) had hormone receptor positive disease & 17 (35%) were confirmed HER2-low (IHC 1+ or 2+, ISH nonamplified). The most common MpBC subtypes were with squamous differentiation (n=17, 35%), chondroid (n=11, 23%), matrix-producing (n=11, 23%) & spindle cell differentiation (n=10, 21%). Ten pts (21%) had de novo metastatic disease & 38 (79%) had recurrent disease. Most common metastatic sites were lung (n=33, 69%) & liver (21, 44%). 37/38 (97%) patients had curative intent anthracycline & taxane, with median disease-free interval of 16 months (range 2-241). 18 pts (38%) received curative intent immune checkpoint inhibitor. Most pts received ADC in the first (n=19, 40%) or second line (n=18, 38%) with Sacituzumab govitecan being the most common (n=38, 79%) ADC, followed by Trastuzumab deruxtecan (n=9, 19%). 7 pts (15%) received a second ADC. ORR at time of best response was 38% (n=18), with 4 pts experiencing complete response (1 patient who received ADC in 1L, 3 in the 2L) & 14 experiencing partial response. 3 pts (6%) experienced stable disease, & 27 pts (56%) experienced progressive disease as best response. Median rwPFS of pts receiving ADC in the first line was 2.3 months, compared to 4.3 months in those receiving non-ADC/chemotherapy (HR 1.34; 95% CI 0.72-2.49, p=0.89). Median rwPFS of pts receiving ADC in the second line was 2.6 months, compared to 2.3 months in those receiving non-ADC/chemotherapy (HR 0.56; 0.28-1.11; p=0.10). Toxicity resulted in dose reductions in 17 pts (35%), dose delays in 14 (29%), & ADC discontinuation in 2 (4%). Conclusion: In this multi-institutional cohort of pts with metastatic MpBC, ADC treatment yielded a 38% ORR, including four complete responses. However, rwPFS was poor overall & similar between ADC & non-ADC therapy. These findings highlight the need to develop new treatment strategies for MpBC, though a subset of pts may experience meaningful response to ADC.
| Objective response to ADC at time of best response | |||||||
| Overall | 18 of 48 (38%) | ||||||
| First line | 7 of 19 (37%) | ||||||
| Second line | 6 of 18 (33%) | ||||||
| Real world progression free survival in months | |||||||
| ADC | Non-ADC | HR (95% CI, p value) | |||||
| First line | 2.3 | 4.3 | 1.34(0.72-2.49, p=0.89) | ||||
| Second line | 2.6 | 2.3 | 0.56 (0.28-1.11, p=0.10) | ||||
Presentation numberPS1-09-19
Real world outcomes of sacituzumab govitecan in hormone receptor-positive and triple-negative breast cancer: Impact of prior therapy
Akshara Raghavendra, University of Texas MD Anderson Cancer Center, Houston, TX
A. Raghavendra, Z. Wang, R. Bssett Jr, S. Damodaran, C. Yam, V. Valero, P. Pohlmann, J. Litton, F. Meric-Bernstam, D. Tripathy, T. Adesoye; Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX.
Background: The antibody-drug conjugate sacituzumab govitecan (SG) is approved for patients with metastatic triple-negative breast cancer (mTNBC) and hormone receptor (HR)+/Human epidermal growth factor receptor 2 (HER2)- metastatic breast cancer (mBC). The impact of HER2-low status on treatment outcomes remains unclear. This study examines overall survival by subtype and prior therapies, explores HER2-low as a predictor of time to next treatment (TTNT), and evaluates TTNT as a potential surrogate for SG benefit.Methods: We identified 472 patients who received SG for HR+/HER2- mBC or mTNBC between 2020 and 2024. We compared overall survival (OS) by breast cancer subtype, treatment history, TTNT, and HER2-low status (1-2+ ISH neg) and investigated factors associated with OS.Results:The median age was 51 years (range, 24-85). Of the 472 patients, 292 (62%) had HER2-low status at any time and 184 (39%) had HER2-0 tumors. The median follow-up time was 9.1 months. Most (84%, n=396) received ≥1 prior therapy in the metastatic setting including chemotherapy (95%) while 34% (n=160) received ≥3 lines of therapy. Among HR+/HER2- mBC patients (n=118), 74% (n=87) received ≥3 lines of prior therapy. Among mTNBC patients (n=354), 38% (n=133) received prior ICI while 21% (n=73) received ≥3 lines of prior therapy. The median overall survival (mOS) duration was 15.3 months (95% CI: 13.5-17.5) for all patients, 12.1 months (95% CI: 8.6-14.9) for HR+/HER2- mBC patients, and 16.5 months (95% CI: 113.6-21.3) for mTNBC patients. Factors associated with worse OS were primary HR+ status (hazard ratio [HR], 1.31; 95% CI, 1.00-1.71; p=0.052), anthracycline therapying metastatic setting (HR, 1.51; 95% CI, 1.05-2.18; p=0.027), and ≥3 prior lines of treatment (HR, 1.49; 95% CI, 1.13-1.96; p=0.004). There were no significant differences in survival based on race, age, or prior ICI therapy. Among 223 patients who were evaluable for TTNT, median TTNT was 5.9 months (range, 0.8-47.8 months), and 49.3% had a TTNT ≥6 months. Patients with a TTNT ≥6 months were more likely than those with a TTNT <6 months to have received ADCs as a subsequent line of treatment (30% vs 14%; p<0.001). For the 223 patients evaluable for TTNT, the mOS was 16.4 months (95% CI, 13.7-20.3) for all patients, 19.9 months (95% CI, 10.2-not estimable) for HR+/HER2- mBC patients, and 16.4 months (95% CI, 13.6-20.3) for mTNBC patients, and HER2-low status at any time point was associated with better OS (HR, 0.68; 95% CI, 0.48-0.98; p=0.036).Conclusions: The survival outcomes in this real-world study were consistent with those reported for clinical trials. Among mTNBC patients, those who received fewer lines of prior chemotherapy had longer OS, and HER2-low status was significantly associated with OS. Prior ICI therapy did not significantly impact outcomes, but patients with >3 lines of prior therapy had worse outcomes, suggesting that earlier utilization of SG may be beneficial.
Presentation numberPS1-09-21
Outcomes of Immunotherapy in BRCA-Mutation Positive, Hormone-Receptor Positive, Breast Cancer: A Systematic Review
Hafiz Muhammad Hannan Javed, TidalHealth, Salisbury, MD
H. Javed1, F. Sial2, F. Ashraf3, M. Mushtaq4, M. Shahzad5; 1Internal Medicine, TidalHealth, Salisbury, MD, 2Medicine, King Edward Medical University, Lahore, PAKISTAN, 3Internal Medicine, Gujranwala Medical College, Gujranwala, PAKISTAN, 4Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, 5Hematology & Oncology, University of South Florida Morsani COM, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
Introduction:BRCA 1 and BRCA 2 are tumor suppressor genes, and mutations in these genes occur frequently in breast cancer (BC). BRCA-mutated (BRCA-m) cancers are often high grade, poorly differentiated, and can carry worse overall survival (OS). Immunotherapy in BC is currently mainly limited to triple-negative breast cancer, but several ongoing trials are investigating immunotherapy in BRCA-m, hormone-receptor-positive (HR+) breast cancer. We aim to explore the outcomes of these trials in this study. Methods:Following PRISMA guidelines, a comprehensive search of PubMed, Cochrane, Embase, Google Scholar, and ClinicalTrials.gov (inception to May 2025) was conducted using MeSH terms for “Hormone Receptor Positive”, “BRCA Mutated”, and “Immunotherapy.” After screening and excluding review articles, meta-analyses, studies without a patient population of interest, and those without relevant clinical outcomes, two studies were selected for inclusion and described systematically. Results:A total of two Phase II studies, involving 34 patients, were included in this systematic review. Mayer et al treated 18 patients with a combination of Niraparib and Dostarlimab in the neoadjuvant setting. The median age was 41.8 years. All patients were HR+, 72% (n = 13) were positive for a germline mutation in BRCA2 (gBRCA2), and 28% were positive for a mutation in gBRCA1. The percentages of patients with stages I, II, and III were 38.9%, 44.5%, and 16.7%, respectively. At the time of surgery, 19% (n = 3/16) had a pathological complete response (pCR), and 69% (11/16) had residual disease. The mean absolute increase in stromal tumor infiltrating lymphocytes (sTILs) in 12 evaluable patients was 11.9%. Commonly reported adverse events included rash (25.0%), elevated liver function tests (18.8%), diarrhea (12.5%), and hypertension (12.5%). Cortesi et al reported outcomes of Pembrolizumab and Carboplatin combination in 16 patients with gBRCA-m, HR+ m metastatic breast cancer. All patients were females with a median age of 50 years (range, 34-69 years), and the liver was the most common site of metastasis (n = 10, 62%). 38% (n = 6/16) had no prior treatment, 70% (n = 7/10) had previously received hormone therapy (HT), and 40% (n = 4/10) had received a combination of HT and CDK4/6 inhibitors. The overall response rate (ORR) was 47% (n = 7/15) with a disease control rate (DCR) of 87% (n = 13/15). DCR was 100% in patients in the first-line setting (n = 12). All patients reported treatment-emergent adverse events (TEAEs), <25% reporting grade 3 or higher TEAEs. Conclusion:The addition of immunotherapy to chemotherapy provides an additional therapeutic option to combat BRCA-m, HR+ breast cancer with an acceptable safety profile. However, current studies are limited by the small sample sizes and the short follow-up times.
Presentation numberPS1-09-22
Single-cell RNA Sequencing Reveals Intrathecal Treatment Sensitivity and Regulatory Mechanisms in Breast Cancer Leptomeningeal Metastasis
Jizhuo Gao, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
J. Gao, H. Li, S. Zhao, M. Li, L. Xu; Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, CHINA.
Background: Breast cancer leptomeningeal metastasis (BCLM) is a highly aggressive and rapidly progressive form of advanced metastasis associated with an extremely poor prognosis. Although intrathecal therapy represents a critical local treatment modality, significant inter-individual variability in treatment sensitivity exists, and its underlying mechanisms remain unclear. This study aims to comprehensively characterize the tumor microenvironment (TME) in the cerebrospinal fluid (CSF) of BCLM patients using single-cell RNA sequencing (scRNA-seq), and to identify key cellular subpopulations and molecular pathways associated with intrathecal treatment sensitivity. Methods: Cerebrospinal fluid samples from eight BCLM patients were collected at the Second Affiliated Hospital of Dalian Medical University and categorized into three groups: pre-treatment, post-treatment sensitive, and post-treatment resistant. Four normal CSF samples from the GEO database were included as controls. All samples were sequenced using the 10X Genomics platform, resulting in a total of 80,274 high-quality cells for downstream analysis after quality control. Results: Dimensionality reduction and clustering identified seven major immune cell types: T cells, monocytes/macrophages, neutrophils, dendritic cells, NK cells, plasma cells, and B cells, with significant differences in their distribution across treatment states. A substantial population of epithelial cells, including tumor cells, was also observed and further classified into seven functionally heterogeneous subpopulations. Copy number variation (CNV) analysis stratified epithelial cells into high- and low-malignancy subtypes, with the high-malignancy subtype exhibiting enhanced proliferative and migratory potential, otentially contributing to chemotherapy resistance. Some epithelial subsets were also present in normal samples, suggesting a non-malignant origin. Compared to controls, BCLM CSF showed a marked reduction in cytotoxic T cells and NK cells, along with an increase in immunosuppressive Tregs. Monocytes/macrophages were significantly enriched and displayed a bias toward M2 polarization. Downregulation of ribosomal protein genes was observed across multiple immune subsets, indicative of an immunosuppressive TME . Among immune populations, monocytes/macrophages exhibited the most distinct transcriptomic alterations across treatment groups and were divided into 12 functionally heterogeneous subclusters. Notably, the C3_S100A9⁺ subcluster was significantly enriched in the treatment-sensitive group, with high expression of inflammatory markers, ribosomal proteins, and regulatory lncRNAs. Functional enrichment analysis revealed its involvement in inflammatory cytokine activation, antigen uptake and presentation, cell adhesion, and multiple layers of immune signaling, indicating a highly activated immune phenotype. Pseudotime trajectory analysis positioned this subcluster at the terminal differentiation state along the treatment-sensitive trajectory. Cell-cell interaction analysis demonstrated enhanced ligand-receptor interactions between this subcluster and highly malignant tumor cells, implicating roles in adhesion, metabolism, and immune activation. These findings suggest that this subpopulation may contribute to intrathecal treatment response by reshaping the immune microenvironment. Conclusions: These findings highlight the potential of C3_S100A9⁺ monocyte-macrophage subcluster as both a predictive biomarker and a therapeutic target. Future studies will integrate transcription factor regulatory network analysis to further elucidate its underlying mechanisms, providing novel insights for precision therapy in BCLM.
Presentation numberPS1-09-23
Physicians’ Perception & Impact of Quality Improvement Intervention: AI-enabled HER2-low Metastatic Breast Cancer Patient Notification Program
Antonio Meo, Daiichi Sankyo, Inc, Basking Ridge, NJ
S. Mehta1, A. Meo1, A. Warner2, A. Hernandez2, A. Alajrash2, P. Kothiya2, L. Gordan2, M. Hussein2; 1US Medical Affairs, Daiichi Sankyo, Inc, Basking Ridge, NJ, 2Real-World Evidence, Florida Cancer Specialists & Research Institute, Lady Lake, FL.
Background: Trastuzumab deruxtecan (T-DXd) was initially approved (Aug 2022) for treatment (trt) of HER2-low (IHC 1+ or IHC 2+/ISH-) unresectable or metastatic breast cancer (mBC) who have received a prior chemotherapy (CT) in the mBC setting. At approval, physicians in the US faced barriers in identifying patients (pts) with HER2-low for appropriate management. Florida Cancer Specialists & Research Institute (FCS) implemented a quality initiative (QI) wherein a random sample of 365 pts with HER2-low mBC were identified from the EMR using artificial intelligence (AI), and on 10/24/2022, physicians received an email alert identifying pts with HER2-low and mentioning the newly approved T-DXd. A retrospective study was conducted to evaluate the impact of the AI-enabled HER2-low notification program. Methods: The FCS EMR data were utilized to identify – Cohort 1 (pts with HER2-low mBC receiving CT on 10/24/2021 who initiated next line from 10/25/2021‒10/23/2022) and Cohort 2 (receiving CT on 10/24/2022 and initiated next line from 10/25/2022‒10/23/2023). Trt patterns were assessed descriptively, and real-world progression free survival (rwPFS) was assessed using Kaplan-Meier curves for both cohorts. A sensitivity analysis was performed to assess immediate changes in trt utilization following intervention by calculating monthly T-DXd use per eligible pts. A survey was emailed to FCS physicians to evaluate their familiarity with HER2-low trt guidelines and their perceived value of such QI. Results: 77 and 69 pts in Cohorts 1 (median age: 63 yrs) and 2 (61 yrs), respectively, were included. Within 1 year, a significant increase in T-DXd utilization (p<.05) and decrease in chemotherapy use were observed between cohorts; T-DXd rates were slightly higher among pts in the notification program (Table). In sensitivity analysis, 1in 4 eligible pts utilized T-DXd in the intervention month/Oct 2022 whereas 3 in 7 utilized T-DXd in the following month/Nov 2022. There was a trend for improvement in population-level rwPFS from Cohort 1 to 2 (Table). All surveyed physicians (n=20) were very (35%) or somewhat familiar (65%) with current clinical guidelines for treating HER2-low mBC. A majority reported these types of notification programs as “very helpful” (65%) but recommended embedding such alerts into pt charts (vs emails) and tailoring to specifically target trt-eligible pts. Conclusions: QI such as AI-enabled notification program can increase awareness of trt guidelines and new indications. Real-world data metrics to assess impact of QI must be benchmarked for continuous enhancements of interventions design, maximize pt outreach, and ultimately improve pt-level health outcomes. No causal association can be inferred between trt patterns and intervention due to limited pre-intervention period and expected natural uptake of a new trt post-approval.
Presentation numberPS1-09-24
Efficacy and safety of Albumin-Bound Paclitaxel (SYHX2011) in Patients with Advanced Breast Cancer: A Multicenter, Randomized, Double-blind Phase III study
Cuizhi Geng, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
C. Geng1, Q. Zhang2, L. Zhang1, T. Sun3, F. Li4, B. Zhao5, G. Han6, Z. Tong7, H. Yang8, Y. Yin9, X. Kong10, Y. Wang11, J. Nie12, Y. Cheng13, Y. Zhang14, J. Luo15, C. Shan16, J. Yao17, S. Tan18, X. Ling19, H. Sun20, H. Li21, Y. Xin22, Y. Li22, D. Yin22, X. Luo22, M. Li22, H. Liu22; 1Department of Breast, The Fourth Hospital of Hebei Medical University, Shijiazhuang, CHINA, 2Department of Breast, Harbin Medical University Cancer Hospital, Harbin, CHINA, 3Department of Breast, Liaoning Cancer Hospital & Institute, Shenyang, CHINA, 4Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, CHINA, 5Department of Breast, Xinjiang Medical University Affiliated Cancer Hospital, Urumqi, CHINA, 6Department of Breast, Shanxi Cancer Hospital, Taiyuan, CHINA, 7Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA, 8Department of Medical Oncology, Affiliated Hospital of Hebei Univeristy, Baoding, CHINA, 9Department of Oncology, Jiangsu Province Hospital, Nanjing, CHINA, 10Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, CHINA, 11Department of Breast Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, CHINA, 12Department of Breast Surgery, Yunnan Cancer Hospital, Kunming, CHINA, 13Department of Medical Oncology, Jilin Cancer Hospital, Changchun, CHINA, 14Department of Medical Oncology, Beijing Hospital, Beijing, CHINA, 15Department of Breast Surgery, Sichuan Provinvial People’s Hospital, Chengdu, CHINA, 16Department of Breast, Affiliated Hospital of Jining Medical Univeristy, Jining, CHINA, 17Cancer Center, Union Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan, CHINA, 18Department of Oncology, Guizhou Provinvial People’s Hospital, Guiyang, CHINA, 19Department of Medical Oncology, The First Hospital of Lanzhou University, Lanzhou, CHINA, 20Department of Oncology, Jiamusi Tumor Tuberculosis Hospital, Jiamusi, CHINA, 21Department of Breast, Shandong Cancer Hospital, Jinan, CHINA, 22Clinical development division, CSPC Zhongqi Pharmaceutical Technology Co., Ltd, Shijiazhuang, CHINA.
Background: Paclitaxel is a highly active chemotherapy agent as the standard treatment for advanced breast cancer. Albumin-bound paclitaxel is SPARC mediated enhanced paclitaxel distribution in tumor by high binding affinity to albumin, with high incidence of rash after treatment. SYHX2011 is a novel albumin-bound paclitaxel, with most non-particular human albumin replaced with mannitol and sucrose. This study aimed to compare SYHX2011 and paclitaxel for injection (albumin-bound) (PAB) outcomes in the patients with breast cancer. Methods: This is a multicenter, randomized, double-blind, phase III trial (NCT05753865). Patients with histologically/cytologically confirmed unresectable locally advanced or metastatic breast cancer were randomized in a 1:1 ratio to SYHX2011 (260 mg/m2) or PAB (260 mg/m2) administered every 3 weeks by intravenous infusion. Randomization was stratified by previous taxanes use and rash occurrence (previous taxanes with rash or previous taxanes with no rash or no previous taxanes), as well as previous chemotherapy for the advanced disease (yes or no). The primary endpoint was objective response rate (ORR) assessed by independent review committee (IRC). Noninferiority was verified if the lower confidence bound of 95% confidence interval (CI) in rate ratio (RR) was higher than 0.75; and superiority would subsequently be evaluated and considered to be confirmed if it was higher than 1. Key secondary endpoint was incidence of rash during the first two administration cycles and the whole cycles. Results: 459 patients were enrolled, including 229 in the SYHX2011 group and 230 in the PAB group. The baseline demographic and disease characteristics were well balanced between two groups. The median (range) age was 56.0 years (27-84). 321 (69.9%) patients received previous treatment with taxanes, of which 317 (69.0%) without a history of rash during the treatment. 129 (28.1%) patients had previous chemotherapy for the advanced disease. The ORR assessed by IRC were 35.8% (95%CI 29.4-42.6) in the SYHX2011 group and 25.8% (95%CI 20.2-32.1) in the PAB group (RR=1.38, 95%CI 1.04-1.84; P=0.0124), indicating that SYHX2011 was noninferior to PAB. The superiority of SYHX2011 over PAB was also confirmed. The incidence of rash during the first two administration cycles in the SYHX2011 group was lower compared with those in the PAB group (13.6% vs. 34.3%, adjusted RR=0.40 [95%CI 0.275-0.577, P<0.0001]). Similar results on the incidence of rash during the whole cycles were also observed (16.2% vs. 42.6%, adjusted RR=0.38 [95%CI 0.275, 0.532], P<0.0001). Treatment-related adverse events (TRAEs) occurred in 98.2% of patients receiving SYHX2011, and 98.3% of patients receiving PAB. 111 (48.7%) patients in the SYHX2011 group and 101 (43.9%) patients in the PAB group experienced ≥grade 3 TRAEs. The most common TRAEs were decreased neutrophil count (15.8% vs. 11.3%), decreased white blood cell count (14.0% vs. 11.7%), decreased lymphocyte count (9.2% vs. 10.0%), and peripheral sensory neuropathy (10.5% vs. 10.4%). Conclusions: SYHX2011 demonstrated superior efficacy and significantly reduced the incidence of rash compared with PAB. SYHX2011 might be a more effective and safe treatment option for patients with advanced breast cancer.
Presentation numberPS1-09-25
Therapeutic Inhibition of the sEcad-PDIA4 Axis Suppresses Brain Metastasis in Inflammatory Breast Cancer
Xiaoding Hu, MD Anderson cancer center, houston, TX
X. Hu1, I. Longa Rizzo1, E. Schlee Villodre1, T. Phi1, K. Tesfamariam1, J. Song2, Y. Gong3, K. Savitri3, W. A. Woodward4, B. G. Debeb1; 1Breast Medical Oncology-Rsch,and Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, MD Anderson cancer center, houston, TX, 2Biostatistics, MD Anderson cancer center, houston, TX, 3Pathology,and Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, MD Anderson cancer center, houston, TX, 4Breast Radiation Oncology,and Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, MD Anderson cancer center, houston, TX.
Background: Brain metastasis is a frequent and lethal site of relapse in inflammatory breast cancer (IBC), a rare, aggressive, and highly metastatic breast cancer subtype. We identified soluble E-cadherin (sEcad), an 80-kDa extracellular fragment of full-length E-cadherin, as being significantly associated with increased risk of brain metastasis and decreased overall survival (OS) in patients with metastatic IBC. Functional studies further indicate that sEcad promotes brain metastasis progression in both HER2+ and triple-negative IBC models. However, the underlying mechanism remains poorly understood. Methods: Stable overexpression of sEcad in IBC cell lines [MDA-IBC3 (ER-/HER2+) and SUM149 (ER-/HER2-)] was achieved using lentiviral transduction. PDIA4 knockdown was performed in the sEcad-overexpressing cells via shRNA. To evaluate brain metastasis and survival, MDA-IBC3-sEcad cells were injected via tail vein and SUM149-sEcad cells via intracardiac route into SCID/Beige mice. Mice were treated with 35G8, a blood-brain barrier-permeable PDIA4 inhibitor, administered subcutaneously. In silico analysis was performed using multiple independent breast cancer patient datasets. Results: High serum sEcad levels in IBC patients correlated with poorer OS (=0.02) and earlier development of metastasis (p=0.006). Overexpression of sEcad in IBC cell lines enhanced cell proliferation, and colony formation, migration, invasion, and reduced cell death in vitro. Mice injected with sEcad-overexpressing SUM149 and MDA-IBC3 cells had significantly higher tumor growth rates compared to controls (SUM149: p=0.007; MDA-IBC3: p=0.006). Mechanistically, mass spectrometry and Bio-ID assays identified Protein Disulfide Isomerase Family A Member 4 (PDIA4) as a novel binding partner of sEcad, which was validated through co-immunoprecipitation. PDIA4 expression was markedly elevated in sEcad-overexpressing cells in vitro and in vivo. Knockdown of PDIA4 in sEcad-overexpressing cells significantly reduced migration, invasion and survival. In silico analysis shows that PDIA4 is highly expressed in aggressive ER-negative, basal-like, and HER2+ tumors (p < 0.0001), which have increased brain metastasis risk. PDIA4 levels are also higher in brain metastases than matched primary tumors (GSE125989, P=0.011). Treatment with the brain-permeable PDIA4 inhibitor 35G8 significantly decreased brain metastasis burden in SUM149-sEcad models, including a decrease in brain metastasis incidence (p = 0.05) and the number of brain metastatic lesions (p = 0.03).Conclusion: Our findings uncover a novel mechanism by which sEcad drives brain metastasis in IBC through its interaction with PDIA4. Inhibiting PDIA4 with the BBB-permeable compound 35G8 reduced brain metastatic burden, highlighting the sEcad-PDIA4 axis as a potential therapeutic target.
Presentation numberPS1-09-26
Phase II Trial of Anlotinib-Chemotherapy Combination in HER2-Negative Metastatic Breast Cancer: Therapeutic Efficacy and Proteomic Biomarker Profiling
Ting Xu, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
Y. Yuan, T. Xu, Q. Gu, L. Zhang, S. Li; Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, CHINA.
Background: The management of human epidermal growth factor receptor 2-negative metastatic breast cancer (HER2-negative MBC) in second-line or later settings remains challenging due to the absence of standardized treatment regimens and the limited therapeutic efficacy of chemotherapy. Anlotinib, a novel multi-target tyrosine kinase inhibitor, exerts its effects by inhibiting angiogenesis. This study prospectively evaluated the efficacy and safety of anlotinib combined with chemotherapy as a second-line or later therapeutic option for HER2-negative MBC. Methods: Between August 2022 and August 2023, we prospectively enrolled 33 HER2-negative MBC patients who had experienced treatment failure following at least one line of systemic therapy in the metastatic setting. All participants received anlotinib in combination with chemotherapy. The primary endpoint was median progression-free survival (mPFS), while secondary endpoints included overall survival (OS), objective response rate (ORR), clinical benefit rate (CBR), disease control rate (DCR), and safety. Exploratory analyses utilized the Olink Target 96 Immuno-Oncology Panel to identify proteomic predictors of therapeutic response from serum samples collected from participants. Results: The cohort consisted of patients with hormone receptor-positive (HR+) /HER2-negative breast cancer (N=23) and triple-negative breast cancer (TNBC) (N=10). After a median follow-up duration of 25.9 months (95% CI: 19.9-32.0), the overall mPFS was 8.3 months, and the median OS was 22.2 months. In the HR+ and TNBC subgroups, mPFS was 7.4 months versus 10.6months, respectively. The ORR was 33.3%, DCR reached 90.9% and CBR stood at 60.6%. The analysis of Olink revealed that five proteins, namely CSF-1, VEGF, IL-6, IL-10, and IL-12, exhibited statistically significant differences between the sensitive and non-sensitive groups (P < 0.05), and higher expression levels of these proteins experienced shorter PFS.Grade≥3 treatment- related adverse events were reported in 21.2% of patients including hypertriglyceridemia and neutropenia, with no treatment-related fatalities documented during the study period. Conclusion: Anlotinib combined with chemotherapy demonstrated promising efficacy and an acceptable safety profile for second-line or later treatment of patients with HER2-negative MBC. Baseline serum levels of VEGFA, CSF-1, IL-6, IL-10, and IL-12 identified through proteomic analysis show potential as predictive biomarkers for therapeutic response. These findings necessitate validation in larger randomized clinical trials.
Presentation numberPS1-09-27
Breast Cancer and Brain Metastases: A 13-Year Single-Centre Experience in a Diverse UK Population
Sunil Goyal, Leicetser Royal Infirmary, Leicester, United Kingdom
S. Goyal1, F. Mutamba1, M. Aamir1, A. Tucker1, E. Hurley2, K. Sampson1, B. Varadhan1, L. Balakrishnan1, S. Ahmed1, O. Ayodele1; 1Oncology, Leicetser Royal Infirmary, Leicester, UNITED KINGDOM, 2Science, LEDIDI, Oslo, NORWAY.
Background: Brain metastases (BM) affect approximately 30-35% of patients with metastatic breast cancer (MBC), significantly worsening prognosis and quality of life1. Although therapeutic options have expanded, real-world UK data describing the patterns of BM and their association with tumour subtypes and outcomes remain limited. Objectives: To characterise the clinicopathological features, treatment patterns, and survival outcomes of patients with breast cancer and BM at a single UK centre over a 13-year period, with a focus on breast cancer subtype distribution and associated prognoses. Methods: This was a retrospective study of 261 patients reviewed via the University Hospitals of Leicester NHS Trust brain metastasis registry on Ledidi between December 2011 and December 2024. Patients were included if imaging confirmed brain metastases; 14 were excluded due to incomplete or non-confirmatory imaging. Clinical data were extracted via manual review of electronic records. Subtypes were defined per ASCO/CAP guidelines (ER/HER2 status). Descriptive statistics were used to summarise baseline characteristics. Overall survival (OS) from BM diagnosis was calculated using medians and interquartile ranges (IQR), and statistical comparisons across subtypes were performed. Results: The final cohort included 247 patients. Median age was 57 years. Ethnic distribution: 81% White, 14% Asian, 3% Black. Most patients (83%) presented symptomatically; 88% had concurrent extracranial disease (74% visceral; 67% bone). Initial imaging: CT (53%), MRI (37%). Subtype breakdown: ER+/HER2- (40.5%), ER-/HER2- (27%), ER+/HER2+ (16%), ER-/HER2+ (12.7%). Median OS from BM diagnosis was 17 weeks (IQR 5-42). Survival varied significantly by subtype (p<0.01): ER-/HER2+ (40.5 weeks) ER+/HER2+ (31 weeks) ER+/HER2- (14 weeks) ER-/HER2- (9 weeks) Patients received a median of two systemic treatment lines (IQR 1-3; maximum 7). Whole-brain radiotherapy (WBRT) was the most common treatment (32%), followed by stereotactic radiosurgery (SRS) in 19.4%; treatment data were unavailable for 27.5%. Conclusions: In this large single-centre UK cohort, ER+/HER2- was the most frequent subtype, while ER-/HER2- was associated with the poorest prognosis. HER2+ subtypes showed superior outcomes, reflecting improved therapeutic options. These findings support the need for earlier brain surveillance in high-risk subtypes and underscore brain metastases as an area of unmet need in breast cancer management. References: 1. Akshara S. Raghavendra et al. Breast Cancer Brain Metastasis: A Comprehensive Review. JCO Oncol Pract 20, 1348-1359(2024). DOI:10.1200/OP.23.00794
Presentation numberPS1-09-28
Organ specific metastasis and survival across molecular subtypes in de novo stage IV breast cancer: A population based retrospective cohort study, 2010-2022.
Pragya Jain, Baptist Hospital of Southeast Texas, Beaumont, TX
P. Jain1, R. Patel1, N. Ganatra1, M. Patel2, A. Jamal1, S. Patel1, S. Modi3, T. Naqvi4; 1Internal medicine, Baptist Hospital of Southeast Texas, Beaumont, TX, 2Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, 3Medicine, Smt. NHL Municipal Medical College, Ahmedabad, INDIA, 4Hematology-Oncology, Baptist Hospital of Southeast Texas, Beaumont, TX.
Background : Understanding prognosis in de novo metastatic breast cancer (dnMBC) requires accounting for both where the disease first spreads and the tumour’s molecular subtype, yet recent population level studies that analyse these two determinants together are uncommon. Objectives : To determine, in a recent nationwide cohort, how the first metastatic organ and the tumour molecular subtype jointly influence breast cancer specific survival (BCSS) among women diagnosed with dnMBC between 2010 and 2022. Methods : Using the US SEER-17 Research database (diagnoses 2010-2022), we identified women presenting with dnMBC (N = 38,078). Molecular subtype was derived from estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2) status and classified as hormone receptor positive/HER2-negative (HR+/HER2-), HR+/HER2+, HR-/HER2+, or triple negative breast cancer (TNBC). The first metastatic organ was coded from SEER Combined Mets at Diagnosis (bone, brain, liver, lung, multiple, other/unspecified). BCSS was estimated with Kaplan Meier curves, and multivariable Cox models adjusted for age, race/ethnicity and grade generated adjusted hazard ratios (aHRs) with 95% confidence intervals (CIs). Reference categories were bone only (site) and HR-/HER2+ (subtype). Sensitivity analysis excluded unknown organ codes. Results : Spread pattern: Bone only metastasis predominated (59% overall; 73% in HR+/HER2-). Liver only, lung only and brain only spread accounted for 20%, 14% and 7%, respectively; 28% presented with ≥ 2 metastatic organs. TNBC showed the highest lung or multi-site involvement (≥ 35%). Survival: Five year BCSS differed markedly by site (log-rank p < 0.001): bone 31%, liver 12%, lung 14%, brain 5%, multiple 8%. Adjusted prognosis: Brain only metastasis carried the poorest outlook versus bone only (aHR 1.78, 95% CI 1.60-1.98), closely followed by multi organ dissemination (1.81, 1.76-1.87). Liver only disease conferred intermediate risk (1.33, 1.26-1.41), whereas lung only spread did not differ significantly from bone (1.03, 0.98-1.07; p = 0.30). Relative to HR-/HER2+ tumours, HR+/HER2+ showed the most favourable prognosis (0.75, 0.71-0.79); HR+/HER2- provided a modest benefit (0.88, 0.83-0.92); and TNBC more than doubled the risk of breast-cancer death (2.27, 2.14-2.40). All aHRs shifted by < 5% after excluding unknown sites. Conclusions : The first metastatic organ and tumour molecular subtype independently determine prognosis in dnMBC. Brain involvement and multi organ dissemination portend the worst survival, whereas lung only disease is comparable to bone only after adjustment. HR+/HER2+ tumours confer the most favourable subtype profile, while TNBC remains high risk. These findings support subtype guided imaging and site specific eligibility stratification in metastatic clinical trials.
Presentation numberPS1-09-29
A Rapid Digital Patient-Derived Organoid Guiding Therapy After Antibody Drug Conjugates (ADC)s In Patients with Metastatic Breast Cancer[YY1] [YY1]The size limit (including title and body) is 3,400 characters; this does not include spaces.
Yuan Yuan, Cedars-Sinai Medical Center, Lost Angeles, CA
Y. Yuan1, J. Bitar1, D. Lin1, J. Mota1, D. Marino1, K. Sargsyan1, M. Campbell1, M. Tighiouart1, Y. Choi1, H. Yu2, Z. Wang2, L. Vanderpool2, S. Kawakita2, F. Bustamante2, Z. Wang2, X. Shen2; 1Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Lost Angeles, CA, 2Precision Medicine, Terasaki Research Institute,, Woodland Hills, CA.
Background: Metastatic breast cancer (MBC) remains incurable despite recent incorporation of immune checkpoint inhibitors (ICI) and antibody drug conjugates (ADCs). Currently FDA-approved ADCs such as trastuzumab deruxtecan (T-DXd) or sacituzumab govitecan (SG) are routinely used in clinical practice, but data on optimal sequencing of these agents for individual patients are lacking. With the fast-paced clinical development of multiple ADCs in the MBC space, lack of trial data or clinical algorithm guiding subsequent therapy following initial ADC resistance, such as another ADC with a different molecular target or different cytotoxic payload, chemotherapy, or their combination with ICI, had emerged as a critical unmet need. The current study aims to develop a clinical diagnostic assay to guide optimal treatment for patients with MBC using digital patient organoid (DPO). The primary objective of this study is to assess the DPO platform’s ability to predict patient treatment response to therapy in a prospective clinical study in patients with MBC receiving or progressed on ADCs (T-DXd or SG), in order to develop a personalized tool to refine treatment strategies in the post ADC resistance setting. Methods: An institutional IRB was established for prospective collection of fresh tumor biopsies in patients with MBC undergoing treatment with either T-DXd or SG. Tumor biopsies were collected to assess DPO drug sensitivity to a dose range of T-DXd or SG and correlate these results with the progression free survival (PFS). DPO was generated immediately upon arrival. DPO was then subjected to ADCs (T-DXd, SG, Dato-DXd) or chemotherapy drug (Gemcitabine; Carboplatin; Eribulin) dosing experiments to determine drug sensitivities (AUC values calculated from CTG readouts) for downstream correlation analyses. Results: Between 09/2024-06/2025, 28 fresh tumor tissues were collected and processed. 11 samples had sufficient viable cells to pass QC for DPO establishment and were successfully assayed and 8 failed QC, giving a DPO establishment success rate of 57.9%. Organoids typically established and expanded within 7-14 days post-biopsy. Liver metastasis core needle biopsies posed the most challenges due to low initial live cell numbers. A total of 90 DPO assays were conducted for ADCs, payloads, and chemotherapy agents testing. Analysis revealed that prior clinical exposure and progression on an ADC correlated with reduced DPO sensitivity to the same agent, consistent with acquired resistance. The platform was capable of delineating between target- versus payload-based resistance. Notably, DPOs from heavily pretreated patients were often resistant to both ADCs but remained responsive to alternative agents, highlighting potential therapeutic opportunities. Conclusion: The DPO platform shows promise in predicting treatment responses and differentiating resistance mechanisms to ADCs in MBC. This approach provides a foundation for rational selection among different ADCs and chemotherapies, especially after acquired resistance to prior ADC treatment, in patients with advanced breast cancer.
Presentation numberPS1-09-30
Clinical and Genomic Associations of Atypical Metastases in Invasive Lobular Carcinoma (ILC)
Jane J Chen, New York Presbyterian Weill Cornell, New York, NY
J. J. Chen1, C. White2, Y. Chen2, F. Pareja3, A. Safonov4, P. Razavi4, C. Dang4, K. L. Jhaveri4, S. Shen4; 1Department of Medicine, New York Presbyterian Weill Cornell, New York, NY, 2Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 3Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 4Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: ILC exhibits a distinct pattern of metastatic spread compared to invasive carcinoma of no special type (NST), related to loss of E-cadherin function. Sites more commonly involved in ILC vs NST include the serosa, meninges, skin, and ovaries. However, the genomic and clinicopathological characteristics of patients at highest risk of developing these ‘atypical’ metastases remain unclear. Using a single-center cohort of patients with metastatic ILC, we sought to define features associated with atypical metastases and determine their impact on outcomes. Methods: Patients diagnosed from 2002-2022 with stage IV ILC were identified by histology on early-stage biopsy/pathology and/or CDH1 mutation on a metastatic site biopsy. Patients were required to have MSK-IMPACT somatic next generation sequencing and metastatic site data available. Metastatic sites were recorded based on imaging and/or biopsies at the time of metastatic diagnosis and were characterized as serosal (peritoneum, pleura, pericardium) vs non-serosal, and atypical (serosal, skin, non-liver gastrointestinal [GI], gynecologic [GYN], meninges, bone marrow, orbit, genitourinary [GU], endocrine) vs typical. Genomic analyses were performed on tissue biopsies obtained within 2 months of metastatic diagnosis. Pearson’s Chi-squared, Fisher’s exact, and Wilcoxon rank sum test were used to compare characteristics between groups. Overall survival (OS) was estimated using the Kaplan-Meier estimate and compared between groups using the log rank test in univariate analyses. Results: 651 patients were included. Median age at metastatic diagnosis was 61 years (IQR 54-68). 520 (86%) patients had hormone receptor-positive (HR+), 56 (9.5%) had HER2-positive, and 65 (11%) had triple negative (TN) disease. 261 (40%) had bone-only metastases. 50 (7.7%) had peritoneal, 31 (4.8%) had pleural, 48 (7.4%) had non-liver GI, 47 (7.2%) had GYN, and 3 (0.5%) had meningeal involvement. 75 (12%) patients had serosal and 194 (30%) had atypical site involvement. 139 (45%) patients had classic, 65 (21%) had pleomorphic, and 103 (34%) had other subtype ILC; subtype information was unknown for 344 patients. ILC subtype was not associated with serosal/atypical site involvement. More patients with serosal and/or atypical metastases were premenopausal at early-stage diagnosis than those without (61% vs 40%, p=0.005 and 51% vs 39%, p=0.013, respectively). More patients with serosal involvement had HR+ disease (94% vs 85%, p=0.028) and fewer had TN disease (2.7% vs 12%, p=0.019). Patients with atypical metastases were more likely to have progesterone receptor positive disease (59% vs 41%, p<0.001). ESR1 mutation was more common in those with serosal/atypical metastases (20% vs 7.5%, p=0.003 and 15% vs 6.6%, p=0.007, respectively), whereas ERBB2 mutation was less common (3.8% vs 14%, p=0.048 and 7.3% vs 15%, p=0.041, respectively). GATA3 mutation was more common in those with serosal metastases (13% vs 4.5%, p=0.019). Presence of serosal metastases was associated with worse OS (median 3.5 vs 4.5 years, p=0.003). OS did not significantly differ among patients with atypical vs typical metastases (4.1 vs 4.5 years, p=0.12). Conclusions: Serosal/atypical metastases were associated with early-stage premenopausal status, HR+ disease, and ESR1/GATA3 mutation. Serosal involvement at metastatic diagnosis was associated with worse OS which will be further examined in this cohort in multivariate analyses accounting for prior treatment as a variable, among others. Prevalence of serosal and/or atypical metastases may be underestimated given the limitations of traditional imaging modalities in ILC; confirmation of these findings in other cohorts is warranted.
Presentation numberPS1-08-16
Local interventions in oligometastatic breast cancer: systematic review and meta-analysis
Giuliano Tosello, Universidade do Oeste Paulista, Presidente Prudente, Brazil
C. Molina1, A. Dalama1, D. Silva1, G. Tosello1, M. Cruz2, A. Toledo1; 1Mestrado em Ciencias da Saude, Universidade do Oeste Paulista, Presidente Prudente, BRAZIL, 2Oncologia, Grupo Orizonti, Sao Paulo, BRAZIL.
Introduction: Breast cancer is the most common neoplasm among women worldwide. It is estimated that, in the 2023-2025 period, around 22,000 women will develop metastases, of which approximately 20% will have oligometastases. Some evidence suggests that local treatment with radiotherapy or surgery may prolong survival. However, the benefits of this approach are still not fully clear. Objective: To evaluate the effectiveness of local therapies, such as radiotherapy and surgery in combination with systemic therapy in the treatment of patients with oligometastatic breast cancer. Method: A systematic review was carried out with registration in the PROSPERO database (CRD42024525782), Medline via Ovid, EMBASE, Web of Science, Cochrane Library Databases, Scopus and Clinical Trials, until November 2024. RCTs were selected, involving women with oligometastatic breast cancer in systemic treatment associated with intervention, radiotherapy or surgery, compared to standard systemic treatment. The processes of study selection, data extraction, risk of bias analysis and level of evidence were carried out by independent and blind authors. For this, platforms such as Rayyan and a standardized Excel form were used. The methodological quality was analyzed by ROB-2 and the evidence was assessed with GRADE. For quantitative data analysis, the Review Manager – RevMan program (version 5.3) was used. Results: Among 717 studies, 2 randomized controlled trials were included in this review. When comparing the association of radiotherapy or surgery with systemic therapy, overall survival presents RR: 1.78 (CI: 0.26 – 12.17), 162 participants, high risk of bias, very low quality of evidence. For progression-free survival in this same comparison RR: 1.05 (CI: 0.76 – 1.46) 162 participants, high risk of bias, very low quality of evidence. Finally, in this same comparison for the secondary outcome, adverse effects, it was analyzed by subgroups, Grade 3 RR: 1.06 (CI: 0.52 – 2.13) 162 participants, high risk of bias, very low quality of evidence and the subgroup Grade 4 RR:0.42 (CI: 0.10 – 1.80) 162 participants, high risk of bias, very low quality of evidence. Conclusion: There are no benefits from the association of systemic therapy with radiotherapy or surgery in oligomestases compared to systemic therapy for the outcomes of overall survival, disease progression-free survival and adverse events.
Presentation numberPS1-08-17
Real-world outcomes comparing zoledronic acid and denosumab in patients with metastatic breast cancer and bone metastases: A propensity score matched analysis from a global federated health research network
Arshi Syal, Mayo Clinic, Jacksonville, FL
A. Syal1, Y. Arya1, C. Jones2, H. Mahadevia1, N. Batra1, A. Uriepero Palma3; 1Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 2Hematology and Oncology, UT Health San Antonio, San Antonio, TX, 3Internal Medicine, Jefferson Einstein Philadelphia Hospital, Philadelphia, PA.
Introduction: Patients with metastatic breast cancer frequently develop bone metastases and experience other skeletal-related events (SREs). These complications substantially impair quality of life and may negatively influence clinical outcomes. In this context, bisphosphonates and denosumab are the most commonly utilized therapeutic agents. We aim to study the real-world comparative data between bisphosphonates and denosumab in breast cancer patients with bony metastases along with evaluation of their toxicity profiles. Methods: A retrospective cohort study was conducted using the US Collaborative Network, TriNetX, covering January 2000 to December 2023, encompassing data from 105 global healthcare organizations. Female patients aged 18 and above with breast cancer metastatic to the bone were identified and then stratified into two groups based on treatment with zoledronic acid or pamidronate. The two groups were then propensity-matched based on age, sex, race, and comorbidities. We followed these patients for 5 years to assess outcomes, including mortality, osteoporotic fracture, hypercalcemia, pathological fracture, fall, and osteonecrosis. Results: We identified 7887 patients in the zoledronic acid cohort and 635 patients in the pamidronate cohort. After propensity matching, each cohort consisted of 625 patients with similar baseline characteristics. The average age was 66.4 years in the zoledronic acid cohort and 68.5 in the denosumab cohort. In the zoledronic acid cohort, the ethnicity distribution was 74.68 % White, 6.36% Hispanic, and 11.55% African American. In comparison, the denosumab group had an ethnicity distribution of 74.49% White, 10.31% African American, and 5.68% Hispanic. Our analysis found that within 5 years, patients with breast cancer metastatic to the bone who received zoledronic acid had a significantly lower risk of mortality (Risk difference: -3.49%, 95% Confidence Interval [CI]: -5.063 to -1.916), p-value <0.001) compared to denosumab. However, patients in the zoledronic acid arm were noted to have a higher risk of osteoporotic fracture (Risk difference: 0.817%, 95% CI: 0.232 to 1.402), p-value = 0.006), hypercalcemia [Hazard Ratio [HR]: 1.524, 95% CI: 1.407 to 1.65, p-value <0.001), and pathological fracture (HR: 1.501, 95% CI: 1.349 to 1.671), p-value =0.03). There was no significant difference in the occurrence of falls and osteonecrosis between the two subgroups. Conclusion: Our study revealed that patients with breast cancer with metastatic disease to the bone who received zoledronic acid had significantly lower rates of mortality but higher occurrence of osteoporotic fracture, hypercalcemia, and pathological fracture compared to those receiving denosumab. Additional longitudinal cohort studies are imperative to explore the outcomes between these agents and inform clinical practice guidelines in these patients.
Presentation numberPS1-08-18
The significance of HER2 Low status on clinical outcomes of breast cancer patients with metastatic hormone-receptor positive tumors treated in the CDK4/6 inhibitor era
Nava Siegelmann-Danieli, Maccabi Healthcare Services, Tel Aviv, Israel
S. Sharman Moser1, B. Horowitz2, R. Ginsburg2, G. Luria2, I. Livnat2, M. Hoshen1, S. Gazit1, N. Siegelmann-Danieli1; 1KSM Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, ISRAEL, 2Medical Affairs, AstraZeneca Israel, Kfar Saba, ISRAEL.
Background: HER2-low (IHC 1+ or 2+/ISH-) is a relatively new sub-type in breast cancer (BC) classification, describing tumors without HER2-oncogene overexpression but rather lower levels of HER2. We describe the characteristics and clinical outcomes of patients with hormone-receptor positive (HR+) metastatic BC (mBC) with HER2-low and HER2 IHC 0 tumors, treated with CDK4/6 inhibitors (CDK4/6i) prior to inclusion of trastuzumab-deruxtecan in the national formulary, in a 2.8-million-member Health Maintenance Organization (HMO). Methods: Patients aged >=18 years who initiated endocrine treatment with-CDK4/6i combination between 1st January 2018 to 31st December 2022 for mBC were identified in this retrospective cohort study with follow up until 31st December 2023. Patients were classified into HER2 IHC 0 and HER2-low, based on deidentified data extraction. Kaplan-Meier analysis was used to describe time-on-treatment (TOT, as a surrogate for progression-free-survival) and overall survival (OS). Results: A total of 1,067 patients were identified with a median age of 63 years, 60.7% were HER2-low, 31.4% were HER2 IHC 0 and 7.9% with missing data A total of 705 patients received first-line CDK4/6i with an aromatase inhibitor (AI) and 362 received CDK4/6i with fulvestrant (F). Median follow-up periods were 27.6 months and 23.0 months, respectively. For the group of patients receiving first-line CDK4/6i & AI, median TOT was 27.0 (95%CI: 23.5-34.4) and 22.6 (95%CI: 17.5,32.7) months for patients with HER2-low and HER2 IHC 0 respectively, and median OS was 67.0 (95%CI: 58.0, NR) months and 52.4 (95%CI: 47.4, NR) months, respectively (P=0.056). For the group of patients receiving CDK4/6i & F, median TOT was 11.6 months (95%CI: 10.0, 14.1), and median OS was 36.4 months (95%CI: 31.1, 40.3), and did not differ between groups. For all patients, a total of 523 patients (49%) received at least one subsequent line after CDK4/6i initial treatment and 346 (32.4%) continued to a further line, with TOT around 4 months. Conclusion: This study shows that HR+/HER2- metastatic breast cancer is a mixed population of IHC scores, with 60-65% of cases being HER2-low. The limited real-world TOT duration at post-CDK4/6i progression highlights a critical unmet need for new and more efficacious treatments for patients progressing on this modern endocrine-biologic regimen. In this real-world analysis, first-line CDK4/6i & AI treatment showed numerically higher OS by 14.6 months in HER2-low patients compared to HER2 IHC 0 tumors whereas in patients who received CDK4/6i&F at more advanced setting of their metastatic disease, OS did not differ by HER2 status. Those results stand in line with recent retrospective analysis of PALOMA-2 and PALOMA-3 trials, where HER2-low patients showed higher benefit from CDK4/6i in the 1st line setting compared with HER2 IHC 0, whereas in later treatment-lines adding CDK4/6i to endocrine therapy benefited all patients similarly. Additional studies are encouraged to support this retrospective observation.
Presentation numberPS1-08-19
Addressing real-world diagnostic and therapeutic gaps in HER2-low metastatic breast cancer: A quality improvement initiative in community oncology settings
Jane Meisel, Winship Cancer Institute of Emory University, Atlanta, GA
J. Meisel1, N. Sidiropoulos2, K. Valla3, M. S. Nawaz4, I. Dewald3, S. Dooyema3, J. Carter3, C. Heggen3, M. Kelly3; 1Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 2Pathology and Laboratory Medicine, University of Vermont Medical Center, Burlington, VT, 3Scientific Affairs, PRIME Education, New York, NY, 4Hematology and Medical Oncology, Hematology Oncology of Indiana, Indianapolis, IN.
Background: Accurate interpretation and reporting of HER2 testing results are essential for treatment selection in both HR+/HER2-negative and triple-negative metastatic breast cancer (mBC). The emergence of HER2-low and HER2-ultralow classifications has expanded therapeutic options, particularly with the availability of novel HER2-directed antibody-drug conjugates (ADCs) demonstrating clinical benefit in patients with low HER2 expression. However, consistent and accurate HER2 assessment remains a significant challenge, especially in community settings, and variability in pathology practices may contribute to missed therapeutic opportunities. This quality improvement (QI) initiative sought to address these challenges and advance evidence-based treatment planning for patients with HER2-low mBC in community oncology clinics by promoting sustainable changes in diagnostic and therapeutic practices. Methods: From 11/2024 to 4/2025, 91 healthcare professionals (HCPs) from 7 community oncology clinics completed baseline surveys assessing practice patterns, barriers, and facilitators related to HER2 testing, collaboration with pathologists, and treatment selection for patients with mBC. Clinics then participated in audit and feedback sessions to review site-specific data, identify root causes of practice gaps, and develop action plans. Representatives from each clinic later convened to review progress toward goals, address ongoing challenges, and refine action plans. Follow-up surveys were collected to assess sustained practice change. Results: Key challenges in identifying HER2-low mBC included interpretation of ambiguous or borderline HER2 results (51%), inconsistencies in testing and reporting (24%), and lack of familiarity with the clinical relevance of HER2-low (24%). Pathology reports infrequently included HER2-low (37%) or HER2-ultralow (17%) status. Additionally, 13% of HCPs noted their pathology reports only list IHC/ISH results that must be interpreted by oncology teams. Only 62% of HCPs found HER2 status to be easily accessible in their electronic medical record (EMR). Multidisciplinary tumor boards (MTBs) occurred at least monthly according to 53% of HCPs, yet pathologist participation was limited (55%). While 55% of HCPs were at least moderately comfortable (3/4/5 on 5-point Likert scale) interpreting and applying HER2 testing results to treatment decision-making, only 32% were very likely (5 on 5-point Likert scale) to recommend guideline-directed HER2-directed ADC therapy for a patient with HER2 IHC 1+ or 2+/ISH negative mBC. Top challenges cited regarding treatment selection included staying up to date with the latest approvals and guideline updates for patients with HER2-low disease (37%), and sequencing therapies for patients with other biomarker-directed treatment options (36%). These findings informed action plans to standardize reporting of HER2 results, improve multidisciplinary communication, integrate process improvements to MTBs, and improve HCP education on advances in HER2-low/-ultralow mBC. Follow-up surveys indicated improvements in HER2-low BC identification, communication with pathologists, review of prior pathology reports, and rebiopsy frequency to reassess HER2 status. Conclusions: This study identified gaps in HER2 reporting that hindered accurate identification of HER2-low mBC and selection of HER2-directed ADCs in community oncology practices. Future directions include a national survey of pathologists to identify barriers to consistent HER2-low reporting and to inform standardized workflows and communication strategies across oncology and pathology teams. Supporter: Supported by educational grants from Daiichi Sankyo, Inc. and AstraZeneca Pharmaceuticals.
Presentation numberPS1-08-20
Kdm6a loss affects epithelial-to-mesenchymal related gene expression and golgi apparatus morphology
Charli Worth, Baylor University, Waco, TX
C. Worth, A. Nambiar, J. Taube; Biology, Baylor University, Waco, TX.
Background: Epithelial-to-mesenchymal transition (EMT) is critical for development and is implicated in wound healing and cancer metastasis. Cellular plasticity, the ability of a cell to undergo EMT and its reversal mesenchymal-to-epithelial transition (MET), underlies tumor progression through metastasis and acquired chemotherapy resistance through dysregulated gene expression. The epigenetic basis of cancer cell plasticity is partially driven by histone modifying proteins including lysine (K)-specific demethylase 6A (KDM6A). KDM6A is a member of the COMPASS-like protein complex, and catalyzes the removal of methyl groups from H3K27me3, facilitating gene expression. Objectives: Previously we have shown that KDM6A is suppressed upon epithelial to mesenchymal transition (EMT), a form of cellular plasticity that facilitates invasion and dissemination of cancer cells. In this study, we sought to identify how KDM6A expression is regulated and how KDM6A expression and activity are required to maintain epithelial cellular identity. We also identify how KDM6A affects Golgi apparatus morphology as EMT progresses. Methods: To investigate the role of KDM6A in the context of EMT, KDM6A expression was knocked down and immunofluorescence and western blot were utilized to investigate its effects. KDM6A activity was measured using an enzymatic activity assay. Results: We observed that the KDM6A suppression or inhibition profoundly impacts cellular identity, leading to gain of mesenchymal and stemness properties. Moreover, KDM6A was determined to be regulated through sub-cellular localization leading to partial sequestration of COMPASS-like protein complex outside the nucleus, We conclude that KDM6A functions as a master regulator of epithelial cellular identity which is controlled by multi-factorial mechanisms including transcriptional suppression and altered protein localization.
Presentation numberPS1-08-22
Elevated Incidence of Malignant Hydronephrosis in Metastatic Invasive Lobular Carcinoma: A Single-Center Retrospective Study
Nihaal Reddy, UC San Diego Health, San Diego, CA
N. Reddy1, R. Shatsky2, K. Yeung2, A. Beck2, B. Larkin3, X. Zhang2; 1Department of Medicine, UC San Diego Health, San Diego, CA, 2Moores Cancer Center, UC San Diego Health, San Diego, CA, 3School of Medicine, UC San Diego Health, San Diego, CA.
Background: Invasive lobular carcinoma (ILC) accounts for 15% of breast cancer (BC) cases and is increasingly recognized for its unique biological characteristics. Metastatic lobular breast cancer (mILC) can have different organ tropism and patterns of infiltration than invasive ductal carcinoma (IDC), which are inadequately described in contemporary literature. Due to loss of e-cadherin, ILC often exhibits radiographically occult infiltrative growth patterns rather than the discrete masses as seen in IDC, limiting early detection and intervention, and putting patients at risk for rapid organ compromise. Hydronephrosis is one such dangerous manifestation of mILC caused by diffuse infiltration of the periureteral tissue and lymphatics that leads to encasement and constriction of the ureters, causing potential renal failure if not treated early. Incidence of malignant hydronephrosis (MH) in mILC is underreported. We describe the characteristics and treatment course of patients with mILC and MH who received care at our institution. Methods: We conducted a retrospective chart review of 82 patients with mILC treated at our institution. We collected data on patient demographics, disease characteristics, and interventions received. Results: Among 82 patients with mILC, 27 patients (32.9%) developed MH. Other common metastatic sites included bone (69.5%), peritoneum (34.1%), leptomeninges (28%), and liver (24.4%). On imaging, 10 of the 27 cases were classified as having mild hydronephrosis, while 8 were moderate and 9 were severe. 15 patients had bilateral hydronephrosis. 12 patients underwent ureteral stenting, and 6 underwent percutaneous nephrostomy tube placement. Of the 27 patients with MH, 11 retained normal renal function. 16 developed acute kidney injury (AKI) of any grade, which notably included 4 patients with only mild hydronephrosis. Of those 16 patients, 1 went hospice, 11 had grade 1-2 AKI with renal function recovery after urological intervention and 4 had grade 3 AKI as defined by anuria for at least 12 hours, rise in creatinine over 3x baseline, or recommendations for renal replacement therapy. Among the 4 patients with severe AKI, 2 recovered with urological intervention, and 2 transitioned to hospice. Conclusion: The observed rate of MH (32.9%) in our cohort notably exceeds previous reported estimates in both general BC populations (1.4%) and ILC-specific cohorts (11%-22%). MH can be challenging to diagnose and even mild cases can cause rapid renal impairment due to the infiltrative nature of mILC. With clinical attention, the majority of our patients had recovery and preservation of renal function. Given these high rates and the risk of renal injury often exceeding severity on imaging, clinicians should maintain a high level of suspicion and awareness as early recognition and prompt management are critical to minimize renal damage and improve outcomes.
| Patient Characteristics | Number (%) |
| Median Age (range) | 56.5 years (22 years – 77 years) |
|
ER+/HER2- Breast Cancer |
76 (92.7%) |
|
Triple Negative Breast Cancer |
5 (6.1%) |
| HER2+ Breast Cancer | 1 (1.2%) |
|
Bone Metastases |
57 (69.5%) |
|
Peritoneal Metastases |
28 (34.1%) |
|
Leptomeningeal Metastases |
23 (28%) |
|
Liver Metastases |
20 (24.4%) |
| Malignant Hydronephrosis (MH) | 27 (32.9%) |
|
Unilateral Hydronephrosis |
12 |
| Bilateral Hydronephrosis | 15 |
|
Mild Hydronephrosis |
10 |
|
8 |
|
| Severe Hydronephrosis | 9 |
|
No Urologic Intervention Received |
9 |
|
Ureteral Stenting |
12 |
| PCNs | 6 |
|
MH pts with no renal impairment |
11 |
|
MH pts with Grade 1-2 AKI |
11 |
| MH pts with Grade 3 AKI | 4 |
|
MH pts with recovered Renal Function after intervention |
13 |
|
MH pts who did not Recover Renal Function after intervention |
0 |
| Other (Transitioned to Hospice) | 3 |
Presentation numberPS1-01-13
Examining Antiemetic Management with Antibody-Drug Conjugates (ADCs) in Patients with Breast Cancer: Healthcare Provider (HCP) Insights from a SABCS Survey
Lee S. Schwartzberg, Renown Health, William N. Pennington Cancer Institute, Reno, NV
L. S. Schwartzberg1, L. Licata2, G. Bianchini2, Y. H. Park3, E. J. Roeland4, M. Massagrande5, F. Dato5, H. Iihara6, F. Scotte7, K. Jordan8, M. Aapro9, H. S. Rugo10; 1Renown Health, William N. Pennington Cancer Institute, Reno, NV, 2Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy, 3Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea, Republic of, 4Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 5Elma Research, Milan, Italy, 6Department of Pharmacy, Gifu University Hospital, Gifu, Japan, 7Interdisciplinary Cancer Course Division, Gustave Roussy Cancer Center, Villejuif, France, 8Department of Hematology, Oncology and Palliative care, Ernst von Bergmann Hospital, Potsdam, Germany, 9Genolier Cancer Center, Clinique de Genolier, Genolier, Italy, 10Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
Background: Antibody-drug conjugates (ADCs) such as trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG) have revolutionized breast cancer (BC) treatment, providing superior efficacy that allows for extended treatment durations. Nausea and vomiting (NV) are among the most frequent adverse effects associated with T-DXd and SG, with nausea rates of >70% for T-DXd and >60% for SG in BC clinical trials. According to antiemetic guidelines, patients receiving T-DXd and SG are at moderate-to-high risk for NV and should routinely receive prophylaxis with an NK1 receptor antagonist (RA)-containing regimen. With limited real-world data on antiemetic efficacy in the ADC setting, a survey of international healthcare providers (HCPs) was conducted to assess their experience. Methods: At SABCS 2024, HCPs completed a brief web-based survey to assess their experiences and perceptions of ADC emetogenicity and approaches to antiemetic prophylaxis. Target participants included HCPs familiar with ADCs who manage patients with BC. Eligible participants completed the survey on computers located in the exhibit hall. Results: Of 209 HCPs surveyed, 112 were eligible. Most were oncologists (69%) or hematologist/oncologists (21%) who spent 73% of time in patient care; nearly half were US-based (46%). Among all HCPs, 80% reported using ADCs in practice and/or in clinical trials; 99% had experience with T-DXd and 87% with SG. Among HCPs using T-DXd and SG, 78% and 83%, respectively, perceived them as either highly (32% and 28%) or moderately (46% and 55%) emetogenic. NV reported by HCPs in this survey was much lower than in the T-DXd and SG clinical trials; HCPs reported that 30%, 25% and 15% of their BC patients treated with T-DXd experience N and/or V during the acute (0-24 hours), delayed (days 2-5), and long-delayed (beyond day 5) phases, respectively. Similarly, the reported NV rates were 29%, 26% and 13%, respectively, for SG. HCPs reported that 88% of patients treated with any ADCs in an average month receive antiemetic prophylaxis; however, only 39% of HCPs report exclusively using a guideline-recommended NK1 RA regimen with T-DXd or SG. The majority of HCPs (94%) reported implementing some type of dose adjustments of ADCs due to NV. A third to nearly half of HCPs had at least sometimes implemented an ADC dose reduction or delay due to NV while a quarter reported interrupting or discontinuing ADC treatment (Table). More than half reported using rescue medication at least sometimes for delayed NV.
| Implemented Due to Nausea and/or Vomiting | ADC Dose Reduction | ADC Dose Delay | ADC Dose Interruption | ADC Discontinuation | Use of Rescue Medication in the Delayed Phase | ||||||
| Always | 2% | 0% | 0% | 0% | 1% | ||||||
| Often | 7% | 6% | 8% | 6% | 16% | ||||||
| Sometimes | 37% | 27% | 18% | 18% | 40% | ||||||
| Rarely | 37% | 40% | 42% | 31% | 34% | ||||||
| Never | 18% | 28% | 32% | 46% | 9% |
Conclusions: This survey highlights a gap between evidence and HCP perceptions regarding NV with T-DXd and SG, and reveals higher-than-expected dose reductions, likely reflecting suboptimal adherence to antiemetic guidelines. As ADCs gain prominence in BC treatment, these findings underscore the need for education and guideline-implementation to optimize NV prevention.
Presentation numberPS1-05-13
Cryotherapy combined with compression gloves and socks therapy reduce nab-paclitaxel-induced peripheral neuropathy in breast cancer patients undergoing neoadjuvant chemotherapy: a multicenter randomized controlled trial
Lize Wang, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
L. Wang1, Y. He1, J. Li1, X. Yu1, H. Sun1, J. Zhang1, B. Hua2, Z. Fan1; 1Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Breast Cancer Center, Peking University Cancer Hospital & Institute, Beijing, CHINA, 2Breast center, Department of General Surgery, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences, Beijing, CHINA.
Background: chemotherapy-induced peripheral neuropathy (CIPN) is a frequent and dose-limiting toxicity of nab-paclitaxel in the treatment of breast cancer. Effective strategies to mitigate CIPN remain limited. Cryotherapy and compression therapy have shown promise in reducing CIPN, but prospective randomized data are scarce. We investigated whether the combined use of cryotherapy and compression gloves/socks could alleviate nab-paclitaxel-induced neuropathy in patients receiving neoadjuvant chemotherapy. Methods: this multicenter, prospective, randomized controlled trial enrolled patients with stage ≥T2, hormone receptor-positive node-positive or triple-negative breast cancer (TNBC). All patients received dose-dense anthracycline-based chemotherapy (90-100 mg/m²) for 4 cycles followed by nab-paclitaxel (260 mg/m²) every 2 weeks for 4 cycles. Patients were randomized (1:1) to either standard care (control) or cryotherapy with compression gloves and socks worn during nab-paclitaxel infusion (intervention). Peripheral neuropathy was assessed by NCI-CTCAE grading, the Patient Neurotoxicity Questionnaire (PNQ), and the EORTC QLQ-CIPN20. The primary endpoint was the incidence of grade ≥3 neuropathy. Secondary endpoints included the incidence of grade ≥2 neuropathy, quality of life and pathological complete response (pCR) rate. (NCT05341141) Results: a total of 102 patients were enrolled. Three participants withdrew, and 99 were included in the final analysis-50 in the intervention group and 49 in the control group. The incidence of grade ≥3 neuropathy was significantly lower in the intervention group compared with the control group (4.1% vs. 18.8%, P =0.023). Similarly, grade ≥2 neuropathy occurred less frequently in the intervention group, indicating a consistent reduction in neurotoxicity (36.7% vs. 63.8%, P =0.008). Patients in the intervention group also reported significantly lower scores on CIPN20 assessments (P =0.008). According to PNQ, the incidence of grade D or E showed no statistically significant difference between groups (P = 0.475). Among all patients receiving biweekly nab-paclitaxel, the total pCR rate was 25.8%. In the TNBC subgroup, the total pCR rate reached 50.0%. The rate of treatment discontinuation or dose reduction due to adverse events was 20.4% in the control group versus 10.0% in the intervention group. No serious adverse events related to cryocompression were observed. Conclusions: cryotherapy combined with compression therapy significantly reduced the incidence and severity of nab-paclitaxel-induced CIPN in the neoadjuvant setting, while improving patient quality of life and treatment tolerability. Biweekly nab-paclitaxel yielded an acceptable pCR rate, especially in patients with triple-negative breast cancer. These findings support cryocompression as a safe, non-pharmacologic strategy to mitigate CIPN in breast cancer patients. Keywords: breast cancer, nab-paclitaxel, cryotherapy, compression therapy, peripheral neuropathy, neoadjuvant chemotherapy
Presentation numberPS1-05-15
Trends in the Utilization of Prophylactic Anti-Emetic Therapy among Patients Initiating trastuzumab deruxtecan for Metastatic Breast Cancer in the United States
Heather Moore, Duke Cancer Institute, Durham, NC
C. H. Shah1, C. Nordon2, E. Langford3, X. Zhao3, B. Adeyemi1, E. John2, A. M. Near3, H. Moore4; 1AstraZeneca, Gaithersburg, MD, 2AstraZeneca, Cambridge, UNITED KINGDOM, 3IQVIA, Durham, NC, 4Duke Cancer Institute, Durham, NC.
Background: Trastuzumab deruxtecan (T-DXd) is a HER2-directed antibody-drug conjugate approved for HER2+, HER2-low, and HER2-ultralow metastatic breast cancer (mBC) in the United States (US). In January 2023, T-DXd was re-classified as highly emetogenic by the US National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for antiemesis with a recommendation to use a triplet/quadruplet anti-emetic drug (AED) regimen as prophylaxis at treatment initiation to prevent therapy induced nausea and vomiting. However, real-world use of AED prophylaxis among patients initiating T-DXd and the characteristics of those who receive such therapy remains unclear. Methods: This retrospective study analyzed IQVIA PharMetrics Plus® health claims data between 1 January 2019 and 30 September 2024. Included were adult (≥18 years) patients with mBC who initiated T-DXd (index date) between 1 January 2020 and 1 April 2024, had 12 months of continuous data before index (baseline), and no other primary cancer at baseline. Due to a lack of biomarker information in claims data, treatment-based proxies were used to classify HER2 status. HER2+ status was deemed “plausible” if patients initiated T-DXd before August 2022 or had another HER2-targeted therapy prior to T-DXd. Alternatively, HER2-low status was assigned. The number of guideline-recommended AED classes initiated within 3 days before or on index date was used to categorize patients by prophylaxis group (none; mono; dual; triplet/quadruplet regimen). Patient characteristics were described using summary statistics as percentages for categorical variables and as means (or medians) for continuous variables. Results: Of 1,024 patients initiating T-DXd (mean [standard deviation] age: 59 [12] years; 56% HER2+), 31% had triplet/quadruplet, 51% had dual, 9% had mono, and 8% had no AED. Among patients with any AED prophylaxis use, 83% had 5-hydroxytryptamine type 3 antagonist (5-HT3a) + dexamethasone ± neurokinin 1 (NK1) receptor antagonist. From 1 January 2022 to 1 April 2024, the annual prevalence of triplet/quadruplet regimens increased from 19% to 70%, dual regimens decreased from 63% to 19%, mono regimens decreased from 10% to 4%, and no AED prophylaxis remained constant at 7%. Relative to patients with any AED regimen, those with no prophylaxis were older (mean age: 63 [12] years vs. 59 [12] years), used prior AEDs less frequently before the index prophylaxis window (77% vs. 90%), had a higher proportion with National Cancer Institute comorbidity index score >0 (48% vs 42%) and a lower proportion with brain metastasis (9% vs. 27%). No difference in patient characteristics was observed between patients with mono, dual, or triplet/quadruplet regimens. Triplet/quadruplet AED prophylaxis was used in 24% of the HER2+ subgroup (1 January 2020-1 April 2024) and 41% of the HER2-low subgroup (1 January 2022-1 April 2024). In the first 3 months of 2024, 63% of HER2+ patients and 74% of HER2-low patients were on triplet/quadruplet AED prophylaxis. Conclusion: While patients without AED prophylaxis were older, and had higher comorbidity burden, than patients with any prophylaxis, there were no differences in patient baseline characteristics among patients with mono, dual, or triplet/quadruplet AED prophylaxis. Guidelines are increasingly being followed in the US as demonstrated by the increased use of triplet/quadruplet AED prophylaxis among patients who initiated T-DXd after January 2023, with 5-HT3 antagonist+dexamethasone±NK1 receptor antagonist being the favored regimen. However, of 69 patients in the first 3 months of 2024, 7% of patients still had no AED prophylaxis, and 23% had mono/dual AED prophylaxis, indicating room for education for improvement of guideline implementation.
Presentation numberPS1-06-02
Breast MRI Biomarkers of Tumor Biology: Integrating Imaging with Pathology to Guide Clinical Care
Eduardo Carvalho Pessoa, UNESP, BOTUCATU, Brazil
E. C. Pessoa1, H. L. Couto2, C. K. C. PESSOA1, A. d. Pina1, H. D. VESPOLI1, D. R. DE PAULA1, B. D. A. FILHO1, E. A. p. NAHAS1; 1UNESP, BOTUCATU, Brazil, 2REDE MAMA, BELO HORIZONTE, Brazil
Background: Breast cancer exhibits substantial biological heterogeneity, and biopsy-based molecular profiling is limited by cost, sampling error, and inability to capture spatial variability. Breast magnetic resonance imaging (MRI) offers multiparametric assessment of tumor morphology, vascularity, and peritumoral tissue, but its role as a noninvasive biomarker of tumor biology remains underexplored. Methods: We retrospectively evaluated 340 women with invasive ductal carcinoma of no special type or invasive lobular carcinoma who underwent pretreatment 3T breast MRI. Imaging features were classified according to BI-RADS and correlated with histopathological and immunohistochemical markers, including tumor grade, estrogen receptor (ER), progesterone receptor (PR), HER2, Ki-67, and molecular subtype. Multivariable logistic regression was performed, with bootstrap validation and false discovery rate correction. Results: Larger tumors (>5 cm) were strongly associated with high grade (74.4%), HER2 positivity (26.8%), elevated Ki-67 (75.0%), and HER2-enriched subtype (14.6%). Paradoxically, round/oval shape and circumscribed margins—features often deemed benign—were predictive of ER/PR negativity, high Ki-67, and triple-negative subtype (OR 3.83, 95% CI 1.48-9.94, P=0.006). In contrast, irregular/spiculated margins correlated with Luminal A tumors and low-grade biology. T2 hyperintensity and peritumoral edema were among the strongest predictors of aggressiveness, independently associated with high grade, lymphovascular invasion, HER2 positivity, and Ki-67 ≥20% (P<0.001). Non-mass enhancement patterns were linked to HER2 positivity and intraductal spread, while rim enhancement and washout kinetics identified highly aggressive phenotypes, particularly triple-negative cancers. Multivariable models demonstrated good discrimination (AUC 0.68-0.82). Conclusions: Breast MRI provides robust imaging biomarkers that mirror tumor biology. Morphology, T2 signal, edema, enhancement patterns, and kinetic curves reflect proliferative capacity, receptor expression, HER2 status, and molecular subtype. By integrating imaging with pathology, MRI can enhance risk stratification, capture tumor heterogeneity beyond biopsy, and support precision treatment decisions. These findings highlight MRI as a powerful, noninvasive biomarker with direct translational potential in precision oncology.
Presentation numberPS1-08-05
First disclosure of efficacy and safety data for YL202/BNT326 (HER3 antibody-drug conjugate [ADC]) in advanced or metastatic HR+/HER2 null and HER2-low breast cancer: Phase 2 trial results
Jian Zhang, Phase I Unit, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
J. Zhang1, Y. Meng1, L. Cai2, T. Wu3, T. Sun4, H. Li5, Q. Ouyang6, F. Xu7, C. Trück8, M. Wenger8, R. Tibes8, N. Wang9, J. Bai9, S. Chin9; 1Phase I Unit, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, CHINA, 2The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, CHINA, 3Department of Oncology, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, CHINA, 4Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, CHINA, 5Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, CHINA, 6Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, CHINA, 7State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 8BioNTech SE, Mainz, GERMANY, 9MediLink Therapeutics (Suzhou) Co. Ltd., Suzhou, CHINA.
Background: YL202/BNT326 is an investigational ADC composed of an anti-HER3 IgG1 monoclonal antibody conjugated to ~8 molecules of a novel topoisomerase I inhibitor payload via a tripeptide linker. This phase 2 trial evaluates YL202/BNT326 monotherapy in patients (pts) with locally advanced or metastatic breast cancers (BC). Here, we report data from hormone receptor positive (HR+) BC patients with HER2 null (including HER2-ultralow) or HER2-low expression. Methods: In this Phase 2 trial (NCT06439771) conducted in China, pts with HR+ and HER2-null expression (HER2 no expression, HER2 ultralow expression [0 < IHC < 1+]) and HER2-low expression (IHC score of 1+, 2+/ISH-) who had received ≥1 prior CDK4/6 inhibitor, prior endocrine therapy, and 0-4 lines of prior chemotherapy were treated with 2.0, 2.5, or 3.0 mg/kg YL202/BNT326 Q3W. Primary endpoints are objective response rate (ORR), based on investigator assessment per RECIST 1.1, and recommended dose. Secondary endpoints include safety, disease control rate (DCR), duration of response, progression-free survival, and overall survival. Results: As of 1 September, 2025, 75 pts had received ≥1 dose of YL202/BNT326; 49 pts remain on treatment. Median age was 55.5 years, all pts were Asian, and the majority had ECOG PS 1 (73.3%). HER2 expression per local testing was HER2-low in 46 pts and HER2-null in 29 pts. Efficacy data (summarized in the table) is available for all 75 pts after a median follow-up of 5.5 months. More mature and additional data will be presented at the conference. Treatment-emergent adverse events occurred in 71 (94.7%) pts. Treatment-related adverse events (TRAEs) occurred in 70 pts (93.3%), with Grade ≥3 in 14 pts (18.7%). TRAEs led to dose reduction in 11 pts (14.7%). No dose discontinuation or deaths resulted from TRAEs. The most common TRAEs of any grade included decreased white blood cell count (57.3%), anemia (52%), decreased neutrophil count (50.7%), and nausea (34.7%). TRAEs of grade 3 or higher were less frequent, with decreased neutrophil count (8.0%), anemia (5.3%), and decreased white blood cell count (4.0%) being the most notable. Conclusions: The data suggest an encouraging clinical efficacy and manageable safety profile of YL202/BNT326 in BC pts with HR+ and HER2 null or HER2-low expression, with no treatment discontinuation due to TRAEs. This study continues to enroll pts with HR+ HER2-null/low BC and in addition is enrolling pts with triple-negative breast cancer. Furthermore, YL202/BNT326 is being evaluated in combination with pumitamig, an investigational anti-PD-L1 x VEGF-A bispecific antibody in pts with BC (NCT07070232).
| 2.0 mg/kg (N=37) | 2.5 mg/kg (N=35) | 3.0 mg/kg (N=3) | Total (N=75) | |
| Confirmed ORR by Investigator | ||||
| n (%) | 15 (40.5) | 13 (37.1) | 0 | 28 (37.3) |
| 95% CI | 24.8, 57.9 | 21.5, 55.1 | 0.0, 70.8 | 26.4, 49.3 |
| Unconfirmed ORR by Investigator | ||||
| n (%) | 17 (45.9) | 17 (48.6) | 0 | 34 (45.3) |
| 95% CI | 29.5, 63.1 | 31.4, 66.0 | 0.0, 70.8 | 33.8, 57.3 |
| Confirmed DCR by Investigator | ||||
| n (%) | 30 (81.1) | 33 (94.3) | 3 (100) | 66 (88.0) |
| 95% CI | 64.8, 92.0 | 80.8, 99.3 | 29.2, 100 | 78.4, 94.4 |
| CI = confidence interval, ORR = objective response rate, DCR = disease control rate |
Presentation numberPS1-08-06
Results of the phase 2 SUMIT-BC study, a randomized controlled trial of the cyclin-dependent kinase 7 inhibitor (CDK7i) samuraciclib with fulvestrant in advanced hormone receptor positive (HR+) breast cancer after a CDK4/6 inhibitor (CDK4/6i)
Sonia Pernas, Institut Catala d’Oncologia, Institut d’Investigació Biomèdica de Bellvitge, Barcelona, Spain
S. Pernas1, C. Karaçin2, C. A. González-Núñez3, S. Aksoy4, M. Bellet Ezquerra5, N. Karadurmuş6, O. Yazıcı7, M. Abu-Khalaf8, B. Bermejo9, M. González Cordero10, M. Gümüş11, C. Hinojo González12, R. Lozano Mejorada13, E. de la Mora Jiménez14, C. Zuloaga Fernandez del Valle15, Ç. Arslan16, S. Kurian17, M. Castro-Henriques18, D. Motola Kuba19, J. Pascual20, T. Pluard21, G. Szentmártoni22, F. Moreno23, G. Clack24, S. McIntosh24, R. Coombes25; 1Institut Catala d’Oncologia, Institut d’Investigació Biomèdica de Bellvitge, Barcelona, SPAIN, 2UHS Dr Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, TURKEY, 3Soltmed SMO, CDMX, MEXICO, 4Hacettepe University Cancer Institute, Ankara, TURKEY, 5Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 6SBU Gulhane Research and Training Hospital, Ankara, TURKEY, 7Gazi University Hospital, Ankara, TURKEY, 8Sidney Kimmel Comprehensive Cancer Center – Jefferson Health, Philadelphia, PA, 9Hospital Clínico Universitario de Valencia, Universidad de Valencia, Valencia, SPAIN, 10Hospital Universitario de Badajoz, Badajoz, SPAIN, 11Istanbul Medeniyet Universitesi, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, TURKEY, 12Hospital Universitario Marqués de Valdecilla, Santander, SPAIN, 13University Hospital of Salamanca, Salamanca, SPAIN, 14Actualidad basada en la Investigacion del Cancer, Guadalajara, MEXICO, 15Renati Innovation S.A.P.I de C.V., Jalisco, MEXICO, 16Medical Point Izmir Hastanesi, Izmir, TURKEY, 17MedStar Franklin Square Medical Center, Baltimore, MD, 18Hospital del Mar, Barcelona, SPAIN, 19CRYPTEX Investigacion Clinica S.A. de C.V., Mexico City, MEXICO, 20Hospital Vithas Málaga, Málaga, SPAIN, 21Saint Luke’s Hospital of Kansas City, Kansas City, MO, 22Semmelweis University, Budapest, HUNGARY, 23Hospital Clínico San Carlos, Madrid, SPAIN, 24Carrick Therapeutics, Dublin, IRELAND, 25Imperial College, London, UNITED KINGDOM.
Background: Samuraciclib (CT7001) is a small molecule, ATP competitive, selective oral inhibitor of the tumorigenic effects of CDK7. A phase I study in patients with HR+/HER2− advanced breast cancer (BC) who had previously been treated with a CDK4/6i demonstrated a favorable safety profile and clinical activity when combined with fulvestrant [Coombes et al. Nature Comms 2023]. This study showed that patients with no detectable TP53 mutation (TP53mt) in ctDNA at baseline and those without baseline liver metastases appeared to have better outcomes, a finding repeated in a second independent trial [Oliveira et al. ESMO BC 2025]. The phase 2 randomized SUMIT-BC (NCT05963984) study was designed to evaluate the efficacy, safety, pharmacokinetics (PK), and quality of Life (QoL) of samuraciclib combined with fulvestrant compared to fulvestrant monotherapy. Methods: Eligible patients were ≥18 years with histologically or cytologically confirmed ER+/HER2− locally advanced or metastatic BC not amenable to resection or radiotherapy of curative intent, had received an aromatase inhibitor (AI) combined with a CDK4/6i in the adjuvant or advanced setting, and had RECIST v1.1 evaluable disease. Baseline TP53 status was established using Guardant360 ctDNA analysis. Patients were randomized 1:1:1 to samuraciclib 360 mg QD + fulvestrant 500 mg IM, samuraciclib 240 mg QD + fulvestrant, or fulvestrant alone. Samuraciclib was administered as a new single oral tablet formulation. The primary endpoint was clinical benefit rate at 24 weeks. Secondary endpoints were tolerability, progression-free survival, overall response rate, duration of response, and PK of fulvestrant and samuraciclib. Results: 87 patients were screened and 59 were randomized to samuraciclib 360 mg + fulvestrant (n=20), samuraciclib 240 mg + fulvestrant (n=19), or fulvestrant alone (n=20). Data cut-off was September 2, 2025. Patient and disease characteristics and efficacy and safety results are shown in the table. No responses occurred in patients with detectable TP53mt. The most common AEs were gastrointestinal (GI). One patient treated with samuraciclib 240 mg discontinued due to GI AEs. Dose reduction permitted patients to stay on treatment long term. Samuraciclib single tablet exposure was consistent with that observed after dosing with the multiple capsules used in early development. Conclusions: Samuraciclib QD combined with fulvestrant demonstrated promising efficacy in ER+/HER2− locally advanced or metastatic BC previously treated using an AI with a CDK4/6i. The most common AEs were dose-related GI AEs. Pharmacokinetics of the new tablet formulation were acceptable. These data indicate that a phase 3 trial of samuraciclib with fulvestrant in a population with no TP53mt is warranted.
| Samuraciclib 360 mg + fulvestrant (n=20) | Samuraciclib 240 mg + fulvestrant (n=19) | Fulvestrant (n=20) | |
| Patient characteristics | |||
| Median age, years (range) | 61.5 (29-70) | 56.0 (40-72) | 59.0 (36-76) |
| ECOG performance status 0 (%) | 60.0 | 52.6 | 75.0 |
| Disease characteristics (%) | |||
| CIST measurable diseaseRE | 90.0 | 73.7 | 70.0 |
| No liver metastases | 75.0 | 78.9 | 80.0 |
| No TP53mt | 65.0 | 89.5 | 55.0 |
| Prior CDK4/6i ≥12 months | 60.0 | 78.9 | 85.0 |
| Clinical benefit rate (%) | 60.0 | 63.2 | 40.0 |
| Overall response rate (%) | 33.3 | 21.4 | 14.3 |
| Progression-free survival (months) | 7.8 | 8.5 | 5.6 |
| No liver metastases | 9.0 | 9.6 | 5.6 |
| No TP53mt | 14.5 | 9.6 | 6.8 |
| No TP53mt, prior CDK4/6i ≥12 months | 14.5 | 11.7 | 6.8 |
| Most common related AEs (%) | Any/Grade 3* | Any/Grade 3* | Any/Grade 3* |
| Diarrhea | 80.0/10.0 | 52.6/5.3 | 0/0 |
| Nausea | 75.0/15.0 | 36.8/0 | 15.0/0 |
| Vomiting | 60.0/5.0 | 31.6/0 | 5.0/0 |
| Samuraciclib dose reduction (%) | 50.0 | 31.6 | – |
| *No Grade 4 AEs reported for these Preferred Terms |
Presentation numberPS1-05-17
Impact of circuit based exercise in breast cancer survivorship
Kye Hee Cho, Yongin Severance Hospital, Yonsei University, Yongin, Korea, Republic of
K. Cho1, E. Han2, S. IM3; 1Yongin Severance Hospital, Yonsei University, Yongin, Korea, Republic of, 2National Jeju University Hospital, Jeju, Korea, Republic of, 3Severance Hospital, Yonsei University, Seoul, Korea, Republic of
Introduction: Adjuvant chemotherapy (AdjCTx) in breast cancer patients is associated with muscle weakness, fatigue, and functional decline that may persist into survivorship. While exercise is recognized as a supportive intervention, the role of circuit-based exercise (CBE) during active chemotherapy remains underexplored. This study investigated the feasibility and functional outcomes of a supervised CBE program in breast cancer patients with or without concurrent AdjCTx. Methods: This study included 40 postoperative breast cancer patients who completed a CBE program for at least two months between June 2020 and April 2023. Patients were stratified into AdjCTx (+) (n = 20) and AdjCTx (-) (n = 20) groups. The CBE protocol, delivered via the Milon® system, integrated aerobic and eccentric resistance training at 60% of one-repetition maximum. Functional outcomes included isokinetic knee extensor strength (peak torque), gait speed (10-meter walk test), balance (Berg Balance Scale), and mobility (Timed Up and Go). Body composition was assessed using bioelectrical impedance analysis. Within-and between-group changes were analyzed using repeated measures ANOVA and paired t-tests. Results: Both groups demonstrated significant improvements in knee extensor strength over time (P <0.05), with significant time effects (F = 10.75-12.34, P < 0.001). The AdjCTx (-) group demonstrated greater absolute gains, while the AdjCTx (+) group showed steady, progressive improvements without regression. BMI significantly decreased in both groups (P<0.001); changes in body fat percentage and ALMI were not statistically significant. Functional improvements in TUG and BBS were observed only in the AdjCTx (-) group (P<0.05), whereas the AdjCTx (+) group maintained baseline function. Gait speed improved significantly in both groups, with a greater increase in the AdjCTx (+) group (P=0.022), despite persistent strength deficits. Conclusions: CBE incorporating eccentric resistance is a feasible and well-tolerated during AdjCTx. It preserves lower-limb strength and functional capacity, highlighting its potential as an early, structured rehabilitation strategy to counteract treatment-related decline and support long-term survivorship in breast cancer patients.
Presentation numberPS1-05-29
Patient-reported reasons for discontinuing adjuvant endocrine therapy: a survey of early-stage breast cancer survivors
Tajana Silovski, Department of Oncology, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Zagreb, Croatia
A. Dugonjic Okrosa1, T. Silovski2, N. Dedic Plavetic2, D. Kifer1, A. Budisavljevic3, H. Silovski4, A. Seselja Perisin5, R. Kelemenic Drazin6, M. Skelin7, L. Jajac Brucic7, J. Jovic Zlatovic7, I. Mucalo1; 1Faculty of Pharmacy and Biochemistry, Zagreb, CROATIA, 2Department of Oncology, University Hospital Centre Zagreb, School of Medicine, University of Zagreb, Zagreb, CROATIA, 3Department of Medical Oncology and Hematology, General Hospital Pula, Pula, CROATIA, 4Department of Surgery, University Hospital Centre Zagreb, Zagreb, CROATIA, 5Department of Pharmacy, School of Medicine, University of Split, Split, CROATIA, 6Department of Hematology, Oncology and Clinical Immunology, General Hospital Varazdin, Varazdin, CROATIA, 7General Hospital of Sibenik-Knin County, Sibenik, CROATIA.
Background: Adjuvant endocrine therapy (AET) for early-stage breast cancer (eBC) profoundlyreduces the risk of recurrence¹, but the literature consistently reports high rates of discontinuation andnon-adherence2. This study aimed to identify the patient-reported reasons for discontinuing therapy. Methods: A self-administered, cross-sectional survey comprising open-ended questions wasconducted among female survivors of eBC who had been prescribed AET for a minimum of threemonths. Results: A total of 920 female survivors (median age 53 years) diagnosed with eBC were included inthe study. The median time since diagnosis was 2 years (IQR: 1.00-4.00), and 62% werepremenopausal at the time of their diagnosis.Among the 165 patients who discontinued or considered discontinuing AET without consulting theirphysician, the most frequently reported reasons were musculoskeletal pain (24.8%) and, as describedin responses to an open-ended question, unspecified severe adverse effects in general (18.8%). Thesewere followed by typical menopausal symptoms (15.5%), including hot flashes, night sweats, vaginaldryness, and osteoporosis and psychological effects (13.3%) such as depression, mood swings,memory problems, and irritability. In their open-ended responses, patients explained that despiteexperiencing significant adverse effects, they chose to continue AET, with fear of cancer recurrenceserving as the primary motivator. However, they also reported contemplating discontinuation due todiminished quality of life (QoL), emotional and cognitive challenges, and concerns about the long-term effects of the therapy.Discontinuations made in agreement with a physician (N=13) were primarily temporary and attributedto planned surgery (27.3%), switching AET (22.7%), gynaecological issues (20.4%), elevated liverenzymes (6.8%), or other adverse effects (22.7%). Conclusions: Although physicians rarely recommend discontinuing AET due to adverse effects, sucheffects remain the leading cause of patient-initiated treatment cessation. Effective side effectmanagement and personalised support are crucial for improving patients’ QoL and, in turn, promotingadherence to and persistence with AET. References: 1. Davies C, Pan H, Godwin J, Gray R, Arriagada R, Raina V, et al. Long-term effects ofcontinuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. The Lancet 2013;381:805-16. https://doi.org/10.1016/S0140-6736(12)61963-1. 2. Yussof I, Mohd Tahir NA, Hatah E, Mohamed Shah N. Factors influencing five-yearadherence to adjuvant endocrine therapy in breast cancer patients: A systematic review. Breast2022; 62:22-35. https://doi.org/ 10.1016/j.breast.2022.01.012
Presentation numberPS1-06-14
Identification of breast abnormalities using remote self-test Feminai – Breast Examination Kit™: a validation study
Karny Ilan, Sheba Medical Center, General Surgery Department, Tel Aviv, Israel
K. Ilan1, J. Mostafa1, T. Menes1, M. Sklair-Levy2; 1Sheba Medical Center, General Surgery Department, Tel Aviv, Israel, 2Sheba Medical Center, Breast Radiology Department, Tel Aviv, Israel
Identification of breast abnormalities using remote self-test Feminai – Breast Examination Kit™: a validation study Karny Ilan, MD, Jollanar Mostafa, MD, Prof. Tehillah Menes, Prof. Miri Sklair-LevyPresenting Author: Karny Ilan, MDDepartment: Breast Imaging, Merav Breast Health Center, Sheba Medical CenterEmail address: Karny10@gmail.comIntroduction and objectives Breast cancer remains the leading cause of cancer-related deaths among women globally, underscoring the importance of early detectionfor effective treatment. Traditional screening methods, including mammography, ultrasound, and MRI, are predominantly clinic-based and have notable limitations. Asthe healthcare landscape shifts towards remote care, innovative technologies are beingexplored to overcome these constraints. Advances in artificial intelligence are enhancing diagnostic precision, highlighting the need for accessible remote screeningsolutions. This study aims to identify and map breast pathologies using a wearable disposable breast patch which integrates infrared and bioimpedance sensors. Thus, addressing the need for more versatile and remote screening options.Methods This prospective, non-interventional, validation study evaluated 100 womenaged 25-75 undergoing breast cancer screening at the “Merav” Clinic, Tel Hashomer Hospital. Participants completed a medical questionnaire to assess their risk for breastcancer and were subsequently scanned with the FeminaiTM patch: a wearabledisposable breast patch incorporating thermometry and impedance sensors. Allparticipants underwent mammography or breast ultrasound, with several also receivingadditional breast MRI. A subset of 51 participants underwent biopsies. The data from thermal and impedance sensors, combined with questionnaire responses, wereanalysed using AI algorithms and compared against radiological interpretations(BIRADS scores) and biopsy results, enabling a comprehensive evaluation of thedevice’s efficacy in distinguishing between normal and abnormal breast conditions.Results A total of 100 women, with a mean age of 49, were examined using the Feminaibreast examination kit. Among the participants, 9 underwent ultrasound, 7 receivedMRI, and the remaining had mammograms. Of the cohort, 51 had a BI-RADS score of 3or higher, indicating abnormal findings, while 49 had a BI-RADS score of 1 or 2,indicating normal findings. Compared to imaging results, the device exhibited a sensitivity of 96%, specificity of 76%, and a negative predictive value (NPV) of 98%,accurately identifying all but one BIRADS 4 case, which biopsy results confirmed as normal tissue. 51 Biopsies were preformed for patients with BIRADS 4 and 5, 38 hadabnormal tissue findings (12 malignant, 26 benign). The device correctly identified all 38 cases with abnormal tissue findings. Notably, variations in infrared and impedance thresholds suggest potential for distinguishing between malignant and benign cases, ascompared to tissue biopsy results. Conclusions This validation study indicates that the Feminai breast examination kit achieved high sensitivity and NPV in this cohort. These promising results suggest the need for further validation to explore its potential as a valuable and cost-effective screening tool. The device’s performance suggests it could enhance early detection and improveaccessibility to breast cancer diagnostics, especially in remote and cost-sensitivesettings.
Presentation numberPS1-09-02
Intracranial activity of datopotamab deruxtecan (Dato-DXd) for patients with HER2-negative breast cancer and leptomeningeal disease (LMD): Results from Cohort C of the DATO-BASE phase 2 trial
Paolo Tarantino, Dana-Farber Cancer Institute, Boston, MA
P. Tarantino1, N. Zhou1, T. Li1, E. Kelly1, C. Strickland1, N. Chan2, M. DeMeo1, L. Anderson1, E. Pasternak1, C. Anders3, M. Ahluwalia4, R. Mahtani4, L. Hsu5, S. Ossing1, A. Waks1, C. Block1, A. Garrido-Castro1, S. Schumer1, I. Schlam1, D. Rimm2, N. Tayob1, S. M. Tolaney1, N. U. Lin1, S. Sammons1; 1Dana-Farber Cancer Institute, Boston, MA, 2Yale School of Medicine, New Haven, CT, 3Duke Cancer Center, Durham, NC, 4Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 5Brigham and Women’s Hospital, Boston, MA
Background: LMD represents a rare but devastating manifestation of metastatic breast cancer (MBC), with a significant unmet need for effective treatments. Datopotamab deruxtecan (Dato-DXd) is a TROP2-directed antibody-drug conjugate (ADC) that has demonstrated promising activity for HER2-negative MBC and clinical evidence of central nervous system (CNS) activity in patients with lung cancer. Methods: DATO-BASE (NCT06176261) is a multicenter, multicohort, single-arm, phase II trial enrolling patients with HER2-negative MBC and active CNS disease. Cohort C was an exploratory cohort enrolling patients with HER2-negative MBC and LMD, either newly diagnosed or progressive. Prior systemic therapy for LMD and prior ADCs were permitted. Patients received Dato-DXd 6 mg/kg IV every 3 weeks until progression, unacceptable toxicity, or withdrawal. Exploratory endpoints include overall survival (OS), radiographic and clinical response according to LANO (Leptomeningeal Assessment in Neuro-Oncology) criteria, intracranial progression-free survival (PFS), safety, quality of life, neurological function according to the NANO (Neurological Assessment in Neuro-Oncology) scale and biomarker analyses. Sections from archival tissue samples were stained for HER2 and TROP2 using the TROPLEX quantitative immunofluorescence assay, to obtain high-sensitivity (HS)-Trop2 and HS-HER2 expressions (attomoles/mm²). Results: A total of 10 patients with LMD were enrolled, of which 9 also had extracranial disease. Median age was 49 years (range 40 – 76), 8 patients had hormone receptor-positive disease and 2 had triple-negative disease; 60% had HER2-low disease. Eight patients had received prior local therapy for CNS disease (3 surgery, 2 surgery and stereotactic radiation, 3 whole-brain radiation). Patients had a median of 2.5 (range 0-6) prior lines of cytotoxic treatment, with 7 who had received prior ADCs (3 T-DXd, 2 SG, 2 both ADCs). Response rate by LANO criteria was 30% (n=3/10), with two partial responses in ADC-naive patients and one complete response in an ADC-pretreated patient (prior SG). With a median follow up of 8.1 months, the median intracranial PFS was 4.1 months (95% CI: 0.7 to not reached), and median OS was 4.7 months (95% CI: 0.7 to not reached). ADC-naïve patients (n=3) experienced numerically longer intracranial PFS (5.9 vs 1.4 months), and longer OS (13.3 vs 3.7 months) compared with ADC-pretreated patients. TROPLEX data was available for 8 patients, including two responders. All patients had HS-Trop2-high disease, whereas only 50% had HS-HER2-high disease. One LANO response each was observed in patients with HS-Trop2 > median and HS-Trop2 ≤ median, with the two groups showing a median intracranial PFS of 4.67 months vs 4.11 months, respectively. Similarly, one LANO response each was observed in patients with HS-HER2 > median and HS-HER2 ≤ median. Grade ≥2 treatment-related adverse events (TRAEs) occurred in 70% (n=7/10) of the patients, with the most common being stomatitis (40%, n=4), fatigue (30%, n=3) and nausea (30%, n=3); only one grade 4 TRAE was observed (dyspnea). No patient experienced interstitial lung disease nor grade 5 TRAEs. Out of 7 patients who had neurological symptoms at baseline according to the NANO score, improvement in at least one symptom was observed in 5 patients, with 1 patient showing stable symptoms and 1 patient missing follow up NANO assessment. Conclusions: In this exploratory cohort of DATO-BASE, Dato-DXd demonstrated promising intracranial activity in patients with HER2-negative MBC and LMD, including radiographic responses and improvement in neurological symptoms in most patients with baseline deficits. Outcomes appeared more favorable in ADC-naïve patients, although numbers were limited.
Presentation numberPS1-08-07
Final results from the Randomized Phase II study of Z-Endoxifen and Tamoxifen in Endocrine Refractory, Estrogen Receptor Positive, HER2 negative Metastatic Breast Cancer (Alliance A011203)
Matthew P Goetz, Mayo Clinic Department of Oncology, Rochester, MN
M. P. Goetz1, V. Suman2, J. Reid1, T. Dockter3, C. Strand3, W. F. Symmans4, J. Hawse5, A. Storniolo6, J. Doroshow7, J. M. Collins8, H. Streicher8, M. M. Ames1, A. Tarnower9, O. M. Hahn10, J. N. Ingle1, L. Carey11, A. Partridge12; 1Mayo Clinic Department of Oncology, Rochester, MN, 2Alliance Statistics and Data Management Center Mayo Clinic, Rochester, MN, 3Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, 4M.D. Anderson, Houston, TX, 5Mayo Clinic Department of Biochemistry and Molecular Biology, Rochester, MN, 6Indiana University, Indianapolis, IN, 7Division of Cancer Treatment and Diagnosis; National Cancer Institute, Bethesda, MD, 8Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 9University of New Mexico Cancer Center, Albuquerque, NM, 10Alliance Protocol Operations Office, University of Chicago, Chicago, IL, 11UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 12Dana-Farber/Partners CancerCare, Boston, MA
Background: In the treatment of ER+/HER2- metastatic breast cancer (MBC), randomized phase II/III trials testing selective estrogen receptor degraders (SERDs), proteolysis-targeting chimeras (PROTACs), complete estrogen receptor antagonists (CERANs) and selective ER modulators (SERMs) (mostly compared to fulvestrant) have demonstrated modest (1-2 months) improvements in progression-free survival (PFS), mainly limited to patients with ESR1-mutated MBC. In these studies, the median PFS (mPFS) ranges from 3 to 6 months, with shorter mPFS in patients with prior progression on CDK 4/6 inhibitor (CDK 4/6i). Here we report the final results of a randomized phase II study that compared Z-endoxifen (Endx) 80 mg/day with tamoxifen (Tam) 20 mg/day in postmenopausal women with estrogen receptor (ER)- positive/ HER2-negative MBC, and report outcomes according to ESR1, PIK3CA, PTEN and AKT alterations (Alliance A011203; NCT02311933). Methods: Postmenopausal women with unlimited prior endocrine regimens (including CDK 4/6i) and up to two chemotherapy regimens for MBC, with tumor biopsy confirmation of ER+ (>10% nuclear staining) and negative for HER2 amplification/overexpression were eligible. Prior adjuvant Tam was allowed if relapse was > 1 year following Tam completion. Patients randomized to Tam could cross over to Endx at progression. The primary end point was PFS analyzed by the interval censored data analytic approach. ctDNA was analyzed for ESR1 and PIK3CA/PTEN/AKT alterations using Guardant360 CDx 74-gene next-generation sequencing-based assay (Guardant Health). Results: Among 108 women who preregistered, 76 (Endx: 40; Tam: 36) were randomized and eligible. Patients were heavily pretreated; all patients had prior exposure to an aromatase inhibitor (AI), 50% had ≥ 2 AI based regimens in the metastatic setting and 50% in each arm had received fulvestrant. Approximately half on each arm received at least one chemotherapy regimen in the metastatic setting. Prior CDK 4/6i therapy was imbalanced (48% on Endx and 28% on Tam). Endx did not significantly prolong PFS compared to Tam (median PFS [mPFS] 4.3 vs 1.7 months; HR 0.72, p=0.168). Because arms were imbalanced according to prior CDK 4/6i exposure, we performed an unplanned analysis demonstrating that Endx improved PFS relative to Tam in CDK 4/6i-naïve patients (mPFS 8.3 vs 2.1 months; HR 0.39, p=0.0097) but not in patients treated with prior CDK 4/6i (mPFS 3.0 vs 1.5 months). In the subset of patients who consented for ctDNA testing (n=28), the mPFS for those with either an activating alteration in the PIK3CA/PTEN/AKT pathway or with an ESR1 mutation was similar as the overall trial. For the 24 patients who progressed on Tam and crossed-over to Endx, the median PFS was 3.5 months (range 1.5-15.0 months) with a 6-month clinical benefit rate of 33.3% (8/24; 90% CI: 17.8-52.1%).Conclusions: Endx did not meet the prespecified threshold for concluding that it significantly improved PFS relative to Tam in a cohort of heavily pretreated MBC. However, the observations of a > 6-month improvement in mPFS in CDK 4/6i-naïve patients, anti-tumor activity following crossover to Tam, and mPFS estimates similar to other novel endocrine agents in development all support the ongoing development of Endx. Support: U10CA180821, U10CA180882, U24CA196171, U10CA180820 (ECOG-ACRIN) and 5P50CA116201. Clinicaltrials.gov Id: NCT02311933. https://acknowledgments.alliancefound.org.
Presentation numberPS1-08-08
Monarche: subgroup analysis of adjuvant abemaciclib + endocrine therapy for hr+, her2-, high-risk early breast cancer by nodal status
Javier Cortes, International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, Spain
J. Cortes1, S. R. D. Johnston2, M. Martín3, S. M. Tolaney4, M. Goetz5, H. Rugo6, J. Sohn7, O. Ozyilkan8, M. Henriques Abreu9, M. Zaiss10, K. Maia Vianna11, H. Melgaard Nielsen12, A. Chan13, H. Kalofonos14, S. Morales15, B. San Antonio16, M. Munoz16, B. R Grimes16, S. Amdur16, P. Neven17; 1International Breast Cancer Center (IBCC), Quironsalud Group, Barcelona, SPAIN, 2Breast Unit, Royal Marsden Hospital, London, UNITED KINGDOM, 3Hospital General Universitario Gregorio Marañon, Universidad Complutense, CIBERONC, GEICAM, Madrid, SPAIN, 4Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, MA, 5Mayo Clinic, Rochester, MN, 6City of Hope Cancer Center, Duarte, CA, 7Yonsei Cancer Center, Seoul, KOREA, REPUBLIC OF, 8Başkent University, Ankara, TURKEY, 9Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), Porto, PORTUGAL, 10Praxis für interdisziplinäre Onkologie und Hämatologie, Freiburg, GERMANY, 11Centro Integrado de Oncologia de Curitiba (CIONC), Curitiba, BRAZIL, 12Department of Oncology, Aarhus University Hospital, Aarhus, DENMARK, 13Curtin Medical School, Faculty of Health Sciences, Perth, Western Australia, Perth, AUSTRALIA, 14Department of Medicine, Division of Oncology, University Hospital of Patras, Patras, GREECE, 15Hospital Universitari Arnau de Vilanova, Lleida, SPAIN, 16Eli Lilly and Company, Indianapolis, IN, 17Leuven Cancer Institute, Universitaire Ziekenhuizen (UZ), Leuven, BELGIUM.
The addition of two years (yrs) of adjuvant abemaciclib to ET resulted in statistically significant and clinically meaningful improvement in overall survival (OS) over ET in patients (pts) with HR+, HER2-, node-positive, high-risk EBC. At 7 yrs, abemaciclib+ET demonstrated sustained IDFS and DRFS benefits ([HR=0.73, 95% CI: 0.66, 0.82), and (HR=0.75, 95% CI: 0.66, 0.84)]). Higher nodal involvement (N2 or N3) is an independent prognostic factor for worse outcomes in EBC. However, previous monarchE data highlight that pts with limited nodal disease (1-3 axillary lymph nodes (ALN)) are at high recurrence risk when additional clinical risk factors are present, such as high-grade (G) tumor and/or large tumor size. Here, we show subgroup analyses by ALN.monarchE is a phase 3, open-label, randomized trial in pts with HR+, HER2-, node-positive, high-risk EBC. Pts were randomized (1:1) to receive ET for at least 5 yrs +/- abemaciclib for 2 yrs (treatment (tx) period). High-risk EBC was defined as either 1-3 ALN (N1) with G3 disease and/or tumor ≥5 cm or, ≥4 ALN (N2: 4-9, N3: ≥10) (Cohort 1 (C1)). A smaller group of pts were enrolled with N1, G≤2, tumor size <5 cm, and central Ki-67 ≥20% (C2). IDFS, DRFS and OS were assessed in C1 by ALN (N1, N2, N3) using the Kaplan-Meier (KM) method, and unstratified Cox proportional hazard model including interaction effect between tx arm and ALN. Of 5120 randomized pts in C1, 1761 (34.4%) had N1, 2223 (43.4%) had N2, and 1123 (21.9%) had N3 disease. At this analysis (data cut off:15Jul2025), in the ET arm, a comparable number of patients with N1 and N2 disease had IDFS events at 6 yrs (21.6% and 25.4%), while event no. in the N3 subgroup was notably higher (38.1%). A similar trend was observed in DRFS and OS (Table). With a median follow-up time of 76.8 months in C1, the addition of abemaciclib to ET reduced the risk of developing an IDFS event by 24.8% (N1), 31.5% (N2) and 27.4% (N3) compared to ET. The absolute improvements in IDFS rates at 6 yrs were 4.6% (N1), 6.9% (N2), and 8.1% (N3). Reductions in the risk of developing DRFS events were 25.2% (N1), 31.2% (N2), and 25.7% (N3), with corresponding 6yr absolute benefit of 3.8% (N1), 6.4% (N2), and 6.8% (N3). Abemaciclib+ET consistently reduced the risk of death compared to ET across ALN subgroups by 12.3% (N1), 15.5% (N2), and 26.8% (N3), which resulted in absolute improvements in OS of 1.4% (N1), 1.6% (N2), and 2.7% (N3). Baseline disease characteristics, IDFS and DRFS KM curves by ALN will be presented. In the ET arm, pts with N1 + ≥1 additional risk factor had recurrence and death risks comparable to pts with N2, while pts with N3 had poorer prognosis. The addition of adjuvant abemaciclib to ET led to a clinically meaningful reduction in the risk of recurrence and death regardless of nodal burden, confirming the consistent and sustained benefit of 2 yrs of tx with abemaciclib and supporting its use in eligible pts with node-positive high-risk EBC.
| Efficacy endpoints | C1 Subgroup by ALN |
Abemaciclib+ET Events/No. of pts |
ET Events/No. of pts |
6-year Rates, % |
HR | Interaction p |
| IDFS | N1 | 142/873 | 188/888 | 83.0 vs 78.4 | 0.75 (0.61, 0.94) | p = 0.82 |
| N2 | 197/1104 | 282/1119 | 81.5 vs 74.6 | 0.69 (0.57, 0.82) | ||
| N3 | 171/571 | 207/552 | 70.0 vs 61.9 | 0.73 (0.59, 0.89) | ||
| DRFS | N1 | 116/873 | 155/888 | 86.1 vs 82.3 | 0.75 (0.59, 0.95) | p = 0.85 |
| N2 | 171/1104 | 245/1119 | 84.1 vs 77.7 | 0.69 (0.57, 0.84) | ||
| N3 | 159/571 | 189/552 | 71.9 vs 65.1 | 0.74 (0.60, 0.92) | ||
| OS | N1 | 82/873 | 95/888 | 90.8 vs 89.4 | 0.88 (0.65, 1.18) | p = 0.63 |
| N2 | 110/1104 | 133/1119 | 90.4 vs 88.8 | 0.85 (0.66, 1.09) | ||
| N3 | 92/571 | 116/552 | 82.7 vs 80.0 | 0.73 (0.56, 0.96) |
Presentation numberPS1-08-09
Deep learning model for predicting hormonal therapy response using FES-PET in ER+ HER2− metastatic breast cancer
Mohyeldin S Abdelhalim, Alexandria Clinical Oncology Department, Alexandria University, Alexandria, Egypt
M. S. Abdelhalim1, A. M. Saad2, A. K. Morsi2, A. H. Algendy2, K. A. Belal1, A. S. Hussein2, A. M. ElDrieny3, S. W. Fouad1, Y. A. Elsaid4, O. Awad5, A. A. Ismail2, H. M. Hegazy1, B. El Sabaa6, S. H. Gamie7, A. A. Elsaid1; 1Alexandria Clinical Oncology Department, Alexandria University, Alexandria, EGYPT, 2Faculty of Medicine, Alexandria University, Alexandria, EGYPT, 3Department of Radiological Imaging, Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, EGYPT, 4Radiodiagnosis and Intervention Department, Alexandria University, Alexandria, EGYPT, 5Baylor College of Medicine, Houston, TX, 6Department of Anatomic Pathology, Faculty of Medicine, Alexandria, EGYPT, 7University of California San Diego, San Diego, CA.
Objective Endocrine therapy is the cornerstone of treatment for estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer, yet intrinsic and acquired resistance limit benefit. Reliable biomarkers are needed to identify likely responders. Conventional clinical factors (age, prior therapy, disease burden) offer modest predictive value. Molecular imaging with 16α-[18F]fluoroestradiol positron emission tomography (FES-PET) enables non-invasive, whole-body assessment of estrogen receptor availability and correlates with endocrine sensitivity. With advances in artificial intelligence, deep learning can integrate subtle imaging features with clinical variables for outcome prediction. This study evaluated whether FES-PET combined with simple clinical factors could predict endocrine therapy response using a deep learning framework. Materials and Methods We retrospectively analysed 60 patients treated at a private cancer center in Alexandria, Egypt, who underwent baseline FES-PET (18F-FES, Cerianna) prior to initiation of endocrine therapy. Imaging volumes were resampled to 128 by 128 by 128 voxels to standardise input. Clinical features included patient age, number of prior endocrine therapy lines, number of chemotherapy lines, and history of breast surgery (mastectomy versus breast-conserving surgery). All variables were normalised and concatenated with image-derived features. A three-dimensional ResNet-18 architecture, pre-initialised with weights from MedicalNet, was fine-tuned for classification. To mitigate class imbalance, focal loss was employed. The dataset was stratified into a training set (n = 48) and a test set (n = 12). Performance was evaluated after each epoch using accuracy, sensitivity, specificity, and area under the receiver operating characteristic curve (AUC). ResultsThe model converged rapidly, achieving optimal performance after only two epochs. At this point, it correctly classified 11 of 12 test cases, corresponding to a sensitivity of 100 percent for identifying responders and a specificity of 67 percent for non-responders. Overall classification accuracy reached 91.7 percent, with an estimated AUC of 0.83, reflecting strong discriminatory ability. Continued training beyond three epochs did not yield improvements, suggesting that transfer learning from MedicalNet enabled efficient adaptation to this small clinical dataset. Performance analysis demonstrated that integration of clinical features with FES-PET imaging contributed to balanced classification, highlighting the added value of combining routinely available patient variables with advanced image-derived features. ConclusionThis pilot study provides proof-of-concept that a deep learning framework integrating FES-PET imaging with simple clinical variables may predict endocrine therapy response with high accuracy in patients with ER+/HER2- metastatic breast cancer. The observation that robust performance was achieved after minimal fine-tuning suggests that transfer learning may be particularly advantageous in small, heterogeneous clinical datasets where conventional training would be limited by overfitting. These findings underscore the potential of artificial intelligence to unlock additional prognostic value from functional imaging while incorporating accessible clinical data. Nevertheless, the modest sample size and limited independent test cohort necessitate cautious interpretation, and external validation in larger, prospective, multi-institutional datasets will be critical. If confirmed, such models could be incorporated into clinical workflows to support treatment planning, sparing non-responders from ineffective therapy while enabling more personalised and precise care pathways
Presentation numberPS1-06-29
Linking PET to Pathology: FES SUVmax Aligns with ER IHC and Accurately Classifies ER at Reduced Cutoffs
Eric T Strand, Harvard Medical School, Boston, MA
E. T. Strand1, C. Ferguson2, E. M. Michaels3, A. A. Lokhandwala4, V. I. Bossuyt5, S. A. Wander3, S. G. Harrington2; 1Harvard Medical School, Boston, MA, 2Massachusetts General Hospital, Department of Radiology, Boston, MA, 3Mass General Brigham Cancer Institute, Boston, MA, 4Mass General Brigham Salem Hospital, Salem, MA, 5Massachusetts General Hospital, Department of Pathology, Boston, MA.
Background: Breast cancer is the second leading cause of death for women in the United States.1 Breast cancer subtyping and receptor expression analysis by immunohistochemistry (IHC) is critical in determining treatment strategy. 18F-16α-Fluoroestradiol (FES) positron emission tomography (PET) allows for noninvasive characterization of the estrogen receptor (ER) in patients with inaccessible biopsy sites, multiple lesions, or non-diagnostic pathology results. Though prior literature has shown FES-PET SUVmax thresholds of 1.5-2.0 provide accurate classification of ER positivity, such studies are subject to very small sample sizes and atypical analytical workflows.2,3,4 There remain limited data on the degree of FES uptake, measured by SUVmax, as a function of ER expression in biopsy samples. Methods: Via a retrospective single center review, we evaluated 98 breast cancer patients who underwent FES-PET-CT or FES-PET-MR from December 2023 to July 2025. We selected patients with biopsy ER IHC data within a +/-90-day window from FES-PET. Patients were excluded if surgical excision was performed on the biopsied site prior to FES-PET. ER IHC data was recorded, including % of cells staining (ER%) and ER staining intensity (1, 2, or 3+). SUVmax values of each biopsy site were analyzed via simple 3D ROI by a board-certified nuclear radiologist. Relationships between SUVmax and ER% were analyzed using Spearman rank correlation. SUVmax and ER staining intensity were compared using Kruskal-Wallis analysis. Receiver Operating Characteristic analysis determined SUVmax thresholds to predict ER IHC positive status (defined as ≥1% of cells with nuclear staining). Statistical analysis was carried out using Python (Pandas, Numpy, Scipy, Matplotlib, and Seaborn packages). Results: 32 patients with breast cancer and corresponding ER IHC who underwent FES-PET imaging were included, with a total of 34 biopsies performed within the cohort (22 before FES-PET, 12 after FES-PET). Biopsies included 13 (38.2%) breast, 7 (20.6%) locoregional, and 14 (41.2%) metastatic sites. Average time from biopsy to FES-PET was 13.8±34.6 days. Among 34 biopsies, 94.1% were ER IHC positive (average ER% 70.8±34.2%, 17 (50.0%) 3+). Three biopsies were ER low-positive on IHC (1-<10%). There was no significant relationship between ER% and corresponding SUVmax values (n = 34, r = 0.210, p = 0.233). There was a significant relationship between SUVmax and ER IHC intensity (n = 34, p = 0.044). Two positive biopsies (5.5%, 1+ and 95%, 3+) were negative on FES-PET performed after biopsy (42 and 61 days, respectively). Two positive biopsies (5.5%, 1-2+ and 100%, 3+) were negative on FES-PET performed prior to biopsy (9 and 49 days, respectively). ROC analysis determined SUVmax thresholds ≥1.4 accurately predict ER IHC positive status while retaining 100% specificity. An SUVmax threshold of ≥1.4 showed an AUC of 0.938, sensitivity of 87.5%, and overall accuracy of 88.24%. Six cases were not detected by the SUVmax ≥1.4 threshold and displayed an average ER% of 34.3±49.1 (two with no staining, two with 1+ staining, and two with 3+ staining). The remaining 28 cases that tested positive by SUVmax threshold displayed an average ER% of 78.6±25.1. Conclusion: Here we present one of the largest FES-PET IHC correlation studies to date. We show that FES-PET SUVmax magnitude is positively correlated with ER IHC staining intensity and that SUVmax thresholds lower than previously reported provide excellent ER classification in a heterogenous breast cancer cohort. Despite lower thresholds, FES-PET sensitivity and accuracy aligns with established values, which may provide more accurate classification and additional treatment options. In clinical practice, FES-PET will continue to provide critical adjunct information and may advance non-invasive receptor characterization strategies.
Presentation numberPS1-08-15
Dauntless-1: A Phase 2 Clinical Trial to Evaluate PMD-026, a First-in-Class Pan-RSK Inhibitor, Combined with Fulvestrant to Overcome Resistance to CDK4/6 Inhibitors in Advanced or Metastatic HR+/HER2- Breast Cancer
Sandra E Dunn, Phoenix Molecular Designs, Vancouver, BC, Canada
H. T. Khong1, M. Beeram2, R. Wesolowski3, H. S. Han4, N. J. Palaskas5, J. Margolis6, J. Peguero7, N. Sharp8, H. Rugo9, A. Jayanthan8, S. E. Dunn8, J. Leveque8, S. A. Wander10; 1Banner MD Anderson, Gilbert, AZ, 2START Center for Cancer Research, San Antonio, TX, 3The Ohio State University Comprehensive Cancer Center, Columbus, OH, 4H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 5Ronald Reagan UCLA Medical Center, Los Angeles, CA, 6Profound Research, Royal Oak, MI, 7Oncology Consultants, Houston, TX, 8Phoenix Molecular Designs, Vancouver, BC, Canada, 9City of Hope Duarte Cancer Center, Duarte, CA, 10Massachusetts General Hospital, Boston, MA
Background: Ribosomal s6 kinase (RSK) is situated at the convergence of the PI3K and ERK-MAPK pathways and plays a crucial role in cell growth and survival. It has been reported that up to 70% of breast tumors have a high expression of RSK, which is an indicator of poor prognosis in patients with breast cancer. RSK2 binds directly to estrogen receptor alpha (ERα) to promote tumor growth and the development of breast cancer in mice. RSK 1-4 have been linked to endocrine therapy (ET) and CDK4/6 inhibitor (CDK4/6i) resistance, which are common in patients with HR+/HER2- advanced or metastatic breast cancer (mBC). RSK drives resistance by promoting the G2/M phase of the cell cycle and bypassing G1/S control. PMD-026 is a first-in-class oral small molecule RSK inhibitor that halts G2/M progression and blocks growth in pre-clinical models of CDK4/6i resistance. In addition, PMD-026 inhibits the nuclear translocation of RSK2 (with no change to levels of nuclear ERα), reduces ERα transcription as a single agent equal to or better than fulvestrant or elacestrant, and synergizes with fulvestrant, oral selective estrogen receptor degraders (SERDs) or vepdegestrant to substantially inhibit tumor growth in CDK4/6i sensitive and resistant models. Combining PMD-026 and fulvestrant reduced ERα transcription by 84% as demonstrated by ELISA and yielded a 7000-fold improvement in the inhibition of tumor growth in soft agar assays. Design: Dauntless-1 is a Phase 2a study of PMD-026 plus fulvestrant in locally advanced or metastatic HR+/HER2 breast cancer patients with high RSK expression (>50%) previously treated with a CDK4/6i + ET combination (NCT04115306). Primary objectives are safety and progression-free survival. Secondary objectives are duration of response, overall response by RECIST V1.1, and overall survival. Exploratory objectives will evaluate these endpoints in the context of select mutations (ESR1, PIK3CA, TP53) and bone-only metastasis. Results: To date for the Dauntless-1 study, 30 patients have been screened for RSK2 expression (67% high, 13% intermediate, 20% low). All patients with high RSK2 expression experienced >12-month durability of response while on treatment with a CDK4/6i + ET regimen. The pharmacokinetics of 2 patients receiving PMD-026 at the initial starting dose of 200 mg Q12h were evaluated and compared to Phase 1 monotherapy results. The combination with fulvestrant increased the levels of PMD-026 with a median of 2.5 µM across 24 hr compared to 1 µM as a monotherapy. Higher than expected exposure with the combination relative to PMD-026 monotherapy was also associated with G3 rash and elevated transaminases, which resolved when PMD-026 was held. To date, PMD-026 at the new starting dose of 100 mg Q12h has been well tolerated with no rash or hepatotoxicity. Based on pharmacokinetic modelling, PMD-026 exposure at 100 mg Q12h is expected to cover the synergy range with fulvestrant. Conclusions: RSK2 is highly expressed in most front-line mBC patients receiving CDK4/6i + ET therapy. PMD-026 inhibits ER signalling and is highly synergistic with fulvestrant in preclinical studies, the translation of which into potentially meaningful clinical benefit is being explored in the Dauntless-1 study.
Presentation numberPS1-06-30
Predicting pathological response to neoadjuvant therapy in HER2-positive breast cancer by using <sup>68</sup>Ga-HER2 PET/CT: a prospective study
Yunjiang Liu, the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
Y. Liu, S. Gao, X. Zhao, X. Zhang; the Fourth Hospital of Hebei Medical University, Shijiazhuang, China
Background: To evaluate the utility of 68Ga-HER2 PET/CT in predicting pathological response to neoadjuvant therapy (NAT) in HER2-positive breast cancer. Methods: This prospective study included 45 newly diagnosed HER2-positive breast cancer patients requiring NAT. Patients underwent at least 4 cycles of NAT with trastuzumab plus pertuzumab or pyrrolizidine alkaloids. Post-NAT surgery was performed. Eligibility required age ≥18 years, stage II-IIIC HER2-positive breast cancer. Exclusion criteria included pregnancy or lactation, presence of severe comorbidities, inability to tolerate systemic therapy or surgery, and distant metastatic disease. 68Ga-HER2 PET/CT scans were obtained pre-NAT (PET1), at the end of cycle 2 (PET2), and pre-surgery (PET3). Standardized uptake values (SUVmax) were measured for the primary tumor and involved lymph nodes. The calculation of ∆SUVmax was performed according to the following formula: ∆SUVmax1 =(PET2 SUVmax-PET1 SUVmax)/PET1 SUVmax ×100% ∆SUVmax2 =(PET3 SUVmax-PET1 SUVmax)/PET1 SUVmax ×100% Pathological response following NAT was assessed using both the Miller & Payne system and the RCB system. Receiver operating characteristic (ROC) curves were generated to estimate diagnostic performance, and the area under the curve (AUC) was calculated. The Youden index was applied to identify the optimal cutoff value. Results: One patient was excluded from analysis due to the subsequent discovery of liver metastasis. 44 patients were included in the final analysis. 70.45% patients (31/44) achieved pathological complete response (pCR), while 29.55% were qualified non-pCR level. No significant differences in pCR rates were observed across age at diagnosis, menstrual status, clinical stage, ER expression, PR expression, Ki67 index, neoadjuvant regimen, type of breast surgery, or lymph node procedure. In contrast, HER2 status demonstrated a statistically significant association with pCR(P=0.025). Although no significant difference in primary tumor SUVmax values between the pCR and non-pCR groups at PET1 (3.76±1.97 vs 3.79±2.71, P=0.349), primary tumor SUVmax values at PET2 and PET3 were significantly lower in the pCR group compared to the non-pCR group (PET2: 0.45±0.71 vs 2.01±2.36, P=0.008; PET3: 0.12±0.39 vs 1.67±2.94, P=0.014). Optimal cutoff values for predicting pCR were: primary tumor SUVmax ≤1.15 at PET2 (AUC=0.738), ≤0.50 at PET3 (AUC=0.735); absolute ∆SUVmax≥61.14% at PET2 (AUC=0.742), and ≥71.63% at PET3 (AUC=0.743). Using the PET2 SUVmax and ∆SUVmax cutoff values, patients identified as NAT-sensitive at the end of cycle 2 had an 84.62% pCR rate, which was 14.17% higher compared to the overall pCR rate without the use of this imaging modality. Conclusion: Our data indicate that 68Ga-HER2 PET/CT assessment of early 68Ga-HER2 uptake changes may predict the NAT efficacy in patients with HER2-positive breast cancer, demonstrating a potential evaluation stratergy for early and late treatment response. Keywords: HER2-positive breast cancer, 68Ga-HER2, PET/CT, neoadjuvant therapy, pCR Clinical trial registration number: ChiCTR2400092565 (Chinese Clinical Trial Registry) Representative Images: The figure below presents sequential 68Ga-HER2 PET/CT scans of a HER2-positive breast cancer patient obtained pre-NAT, at the end of cycle 2, and pre-surgery.
Presentation numberPS1-08-24
A phase 1/2 trial of LY4064809 (STX-478), a pan-mutant-selective PI3Kα inhibitor in HR+, HER2- advanced breast cancer (ABC): Updated results from PIKALO-1
Komal Jhaveri, Medicine Department, Memorial Sloan Kettering Cancer Center, New York, NY
K. Jhaveri1, D. Juric2, A. Giordano3, A. Italiano4, G. Daniele5, J. Rodon6, A. Patnaik7, J. Bartolomé8, A. Elias9, P. Munster10, P. LoRusso11, A. Soyano12, M. de Miguel13, J. O’Shaughnessy14, R. Wesolowski15, G. Curigliano16, T. Hernández17, A. Lombard18, S. T. Estrem18, L. Du18, X. Xu18, A. Montero19; 1Medicine Department, Memorial Sloan Kettering Cancer Center, New York, NY, 2Termeer Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, Boston, MA, 3Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 4Early Phase Trials Unit, Institut Bergonié, Bordeaux, France, 5Phase 1 Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy, 6Drug Development Department, MD Anderson Cancer Center, Houston, TX, 7Medical Oncology, The START Center for Cancer Research, San Antonio, TX, 8Medical Oncology, Hospital Clínico San Carlos and IdISSC, Madrid, Spain, 9Medical Oncology, University of Colorado, Aurora, CO, 10Medical Oncology, University of California San Francisco, San Francisco, CA, 11Medical Oncology, Yale University, New Haven, CT, 12Breast Oncology Department, Moffitt Cancer Center, Tampa, FL, 13Centro Integral Oncologico Clara Campal, START Madrid – HM CIOCC, Madrid, Spain, 14Medical Oncology, Texas Oncology, Dallas, TX, 15Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 16Early Drug Development for Innovative Therapies Division, European Institute of Oncology, IRCCS and University of Milano, Milan, Italy, 17Medical Oncology, START Barcelona – HM Nou Delfos, Barcelona, Spain, 18Eli Lilly and Company, Indianapolis, IN, 19University Hospitals/Seidman Cancer Center, Case Western Reserve University Cleveland, Cleveland, OH
Background: PI3Kα inhibitors (PI3Kαi) have demonstrated clinical benefit in the ~40% of HR+, HER2- ABC with PIK3CA mutations (PIK3CAm). However, broad use is often constrained by: 1) toxicities mediated through wild-type PI3Kα inhibition, and 2) the exclusion of patients (pts) with diabetes (DM) or prediabetes (pDM). LY4064809 is an oral, allosteric, CNS-penetrant, pan-mutant-selective PI3Kαi with favorable safety, PK, and efficacy in pts with PIK3CAm HR+ HER2- ABC. Methods: In PIKALO-1 (NCT05768139), pts with PIK3CAm HR+ HER2- ABC received monotherapy (≥1 prior therapies, txs), LY4064809 plus fulvestrant (fulv, 1-2 prior txs), and LY4064809 plus fulv and CDK4/6 inhibitors (≤2 prior txs). Pts with pDM/DM were eligible; pts with uncontrolled DM (HbA1c ≥8% / FBG ≥140mg/dL) were excluded. Pts with prior PI3K/AKT/mTORi were excluded except for intolerance. Results: As of 7 Jul 2025, 121 pts with HR+, HER2- ABC were treated: 50 with LY4064809 (20-160 mg QD), 33 with LY4064809 (60-100 mg QD) + fulv, and 38 with LY4064809 (20-100 mg QD) + fulv + CDK4/6i (ribociclib, 9; palbociclib, 29). Median age was 62 (27-84) yrs, 49% had pDM/DM. Median prior txs was 2 (0-7) including: CDK4/6i (84%), SERD (51%), chemotherapy/ADC (31%/11%) and PI3Ka/AKT/mTORi (8%). Across all HR+, HER2- pts, all hyperglycemia events were low grade ([G]; 26% G1-G2, 37%/15% in pts with and without pDM/DM); no G≥3 events were reported. Other TEAEs ≥20% (any G/G≥3) were nausea (35%/<1%), fatigue (33%/5%), diarrhea (26%/<1%), neutropenia (25%/17%), and ALT/AST increased (24%/10%). Incidence of other common class-toxicities (rash [3%/0%] and stomatitis [5%/<1%]) were low. TEAEs led to dose reduction in 10 pts (8%) [ALT/AST increased (n=5 at 60, 80, and 160 mg), fatigue (n=3 at 60 and 100 mg), hyperglycemia (n=1 at 100 mg), dysesthesia (n=1 at 60 mg); all related], and discontinuation in 2 pts (2%) [AST increased (n=1 at 100 mg, related), intracranial hemorrhage (n=1 at 80 mg, unrelated)]. Throughout the study, LY4064809 exposures reached the target coverage for in vitro pAKT IC80 (from 20 mg) and in vivo efficacy (from 40 mg). No significant drug-drug interactions were noted with fulvestrant and palbociclib. PIK3CAm ctDNA was reduced at all dose levels across all txs. See table for efficacy. Updated safety, efficacy, biomarker, and PK data (data cutoff: Oct 2025) will be presented at the meeting. Conclusion: LY4064809, either alone or in combination with fulvestrant +/- CDK4/6 inhibitors, was well-tolerated, with notably lower incidence of PI3K inhibitor (PI3Ki)-class toxicities and no grade ≥3 hyperglycemia in HR+, HER2- pts with normal baseline glycemic control. Robust target coverage and promising antitumor activity were observed in heavily pre-treated pts with PIK3CAm HR+, HER2- ABC, highlighting LY4064809’s potential as a best-in-class mutant selective PI3Kα inhibitor. Table
| LY4064809 (n=44) | LY4064809 + fulvestrant (n=25) | LY4064809 + fulvestrant + CDK4/6ia (n=17) | |
| Median ToT (weeks) | 17 (4.1-77) | 18 (7.6-62) | 13 (8.4-38) |
| ORR | 18% (8) | 28% (7b) | 24% (4c) |
| DCR | 66% (29) | 88% (22b) | 82% (14c) |
| Efficacy evaluable pts had measurable disease and ≥1 post baseline response assessment or discontinued prior to the first post baseline response assessment. ORR includes PR and uPR; DCR includes PR, uPR, and SD. aIncludes palbociclib and ribociclib. bIncludes 2 uPR (ongoing, pending confirmation). cIncludes 4 uPR (ongoing, pending confirmation). Abbreviation: ToT, time on treatment. | |||
Presentation numberPS1-08-25
Equivalent survival benefits from first-line and second-line use of CDK4/6 inhibitors in HR+/HER2- advanced breast cancer: A computational analysis of phase 3 trials
Noah Schlachter, University of North Carolina at Chapel Hill, Chapel Hill, NC
N. Schlachter, L. A. Carey, A. Palmer; University of North Carolina at Chapel Hill, Chapel Hill, NC
Introduction: CDK4/6 inhibitors (CDK4/6i) are integral to managing HR+/HER2- advanced breast cancer (ABC), with approvals in both first-line (1L) and second-line (2L). The phase 3 SONIA trial found that 1L CDK4/6i therapy improved time to first progression (PFS1) but not overall survival (OS) or time to second progression (PFS2) compared to reserving CDK4/6i for 2L. Because most patients in SONIA received palbociclib, optimal timing remains uncertain for other CDK4/6i. We computationally analyzed phase 3 trial results to investigate whether CDK4/6i yield intrinsically greater PFS1, PFS2, or OS benefits in 1L versus 2L settings. Methods: We analyzed PFS and OS from pivotal phase 3 trials investigating palbociclib, abemaciclib, and ribociclib in HR+/HER2- ABC (Table). Using PFS distributions from 2L trials, we computed expected 1L PFS under the null hypothesis that CDK4/6i prolong time to progression, relative to endocrine therapy alone, by the same absolute durations at 1L or 2L. For OS, we investigated confounding from incomplete crossover in 1L trials, where a fraction of control arm patients did not receive CDK4/6i post-progression. We used 2L survival distributions to simulate OS with 100% crossover where all control patients receive CDK4/6i at 2L, testing the null hypothesis that CDK4/6i extend OS equally at 1L and 2L. For each CDK4/6i, we simulated trials with the same design as SONIA to compare expected PFS2, and time on therapy, for 1L vs 2L CDK4/6i. Results: Expected 1L PFS derived from 2L trial data did not significantly differ from observed 1L PFS for any CDK4/6i (P values ≥ 0.25, Cox proportional hazards), suggesting similar efficacy in delaying progression at 1L or 2L. For each CDK4/6i, simulated OS when all control arm patients cross-over to 2L CDK4/6i did not significantly differ from observed OS of 1L CDK4/6i (Table), suggesting that CDK4/6i prolongs OS similarly at 1L or 2L. For each CDK4/6i, simulated trials of 1L versus 2L CDK4/6i strategies did not predict significant differences in PFS2, but did predict longer time on CDK4/6i therapy at 1L, as observed in SONIA (Table). Conclusions: Analysis of PFS and OS results of phase 3 trials in 1L and 2L settings indicate that approved CDK4/6 inhibitors confer comparable durations of PFS2 and OS when added to endocrine therapy at either 1L or 2L in HR+/HER2- ABC. These results suggest that reserving CDK4/6i for 2L administration is a clinically valid approach without significant detriment to survival outcomes and may be preferable for many patients due to improved tolerability and affordability.
| SONIA results | Palbociclib | Abemaciclib | Ribociclib | |
| 1L trial | – | PALOMA-2 | MONARCH-3 | MONALEESA-2 |
| 2L trial | – | PALOMA-3 | MONARCH-2 | MONALEESA-3 |
| Crossover rate at 1L | 86% | 26.7% | 31.5% | 34.4% |
| Observed OS HR at 1L | 0.98 (P=0.83) | 0.95 (P=0.34) | 0.80 (P=0.066) | 0.76 (P=0.008) |
| Modelled OS HR at 1L with 100% crossover | – | 1.04 (P=0.55) | 0.93 (P=0.34) | 0.92 (P=0.27) |
| Observed PFS2 HR (1L vs 2L CDK4/6i) | 0.87 (P=0.10) | – | – | – |
| Modelled PFS2 HR (1L vs 2L CDK4/6i) | – | 0.87 (P=0.06) | 0.92 (P=0.25) | 0.92 (P=0.25) |
| Difference in time on CDK4/6i (1L-2L) (months) | 16.6 | 19.2 | 13.4 | 10.6 |
Presentation numberPS1-08-26
A phase 1/2, multi-part, open-label study of FWD1802, a novel oral selective estrogen receptor degrader (SERD), in patients with ER+/ HER2- locally advanced or metastatic breast cancer (BC)
Yanchun Meng, Fudan University Shanghai Cancer Center, Shanghai, China
Y. Meng1, F. Xu2, X. Qu3, Y. Shi4, H. Li5, Y. Sun5, X. Wang6, H. Liu7, S. Ding8, H. Hu9, H. Li10, J. Li11, M. Yan12, Y. Yin13, T. Sun14, W. Xie15, X. Chen16, J. Wang17, C. Zhu17, J. Zhang1; 1Fudan University Shanghai Cancer Center, Shanghai, CHINA, 2Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 3First Affiliated Hospital of China Medical University, Shenyang, CHINA, 4Tianjin Medical University Cancer Institute and Hospital, Tianjin, CHINA, 5Shandong Cancer Hospital, Jinan, CHINA, 6Sir Run Run Shaw Hospital,affiliated with Zhejiang University School of Medicine, Hangzhou, CHINA, 7Jilin Cancer Hospital, Changchun, CHINA, 8The Central Hospital of Yongzhou, Yongzhou, CHINA, 9Zhejiang Cancer Hospital, Hangzhou, CHINA, 10Sichuan Cancer Hospital, Chengdu, CHINA, 11Huizhou First Hospital, Huizhou, CHINA, 12Henan Cancer Hospital, Zhengzhou, CHINA, 13Jiangsu Province Hospital, Nanjing, CHINA, 14Liaoning Cancer Hospital & Institute, Shenyang, CHINA, 15Guangxi Medical University Cancer Hospital, Nanning, CHINA, 16The Second People’s Hospital of Yibin, Yibin, CHINA, 17Forward Pharmaceuticals, Wuhan, CHINA.
Background: About 2/3 of BC patients are ER+/HER2-. Endocrine therapy (ET) is the mainstay treatment, blocking the ER-driven signaling pathway to inhibit tumor growth despite rising resistance. FWD1802 is a novel oral SERD that has demonstrated potent anti-tumor effects in both wildtype and resistant ESR1-mutant xenograft tumors. Methods: Current study is an ongoing phase 1/2 single-agent trial. After a “3+3” dose escalation, and dose expansion in unselected patients in the Ph1 part, 2 doses were selected to enter the Ph2 part in ESR1 mutated (mESR1+) patients. The primary objective is to determine the recommended phase 2 dose and/or maximum tolerated dose (MTD). Secondary objectives include safety, tolerability, pharmacokinetics and anti-tumor efficacy. Key eligibilities include ≥1 prior endocrine therapy, ≤2 prior chemotherapies and ≤1 prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) in the advanced setting. FWD1802 is administered orally daily in 28-day cycles. A 7-day single dosing period prior to Cycle 1 is included in Part A. Results: As of 2025/7/3, 69 patients were treated with FWD1802: 25mg (n=1), 50mg (n=10), 75mg (n=23), 150mg (n=24), 300mg (n=7) and 450mg (n=4). 75mg and 150mg were selected in ph2 part. The median age was 56.4 years (range, 35 – 77), ECOG PS 0 (13%) and 1 (87%). Patients were heavily pretreated, receiving median 2 (1-7) lines of prior treatment in advanced setting, including 71% with prior CDK4/6i, 46% prior fulvestrant, and 52% prior chemotherapy. Visceral disease was present in 90% of patients, 55% with liver mets. Of note, in mESR1+ patients under ph2 doses, 97% had visceral disease and 65% with liver mets. Majority of treatment-emerged adverse events (TEAEs) were Grade 1-2, with 88 % of events Grade 1. TEAEs of ≥15% occurrence in total, 75mg and 150mg patients include: sinus bradycardia (63.8%, 52.2%, 66.7%), ALT increased (33.3%, 39.1%, 37.5%), AST increased (33.3%, 34.8%, 37.5%), hypertriglyceridaemia (33.3%, 43.5%, 33.3%), electrocardiogram QT prolonged (30.4%, 17.4%, 25.0%), hypercholesterolemia (24.6%, 17.4%, 25.0%), anaemia (24.6%, 26.1%, 20.8%), hyperglycemia (21.7%, 21.7%, 20.8%), white blood cell count decreased (18.8%, 21.7%, 20.8%), γ-GGT increased (15.9%, 8.7%, 16.7%), weight increased (15.9%, 17.4%, 16.7%) and photopsia (15.9%, 13.0%, 4.2%). No DLT was observed. Grade 3 TEAEs include: electrocardiogram QT prolonged (3%), hypocalcemia (3%), γ-GGT increased (1%), anaemia (1%), hypertension (1%), pneumonia (1%), femur fracture (1%), lymphocyte count decreased (1%) and hyponatremia (1%). Half-life of FWD1802 was 46.8~72 hours, and trend of linear PK was observed from 25mg to 450mg. Out of 60 response evaluable patients per RECIST 1.1 criteria, 10 (ORR 17%) confirmed PR were observed. In mESR1+ patients at Ph2 doses, 8 of 16 (50%) patients in 75mg reached PR (6 confirmed, 2 unconfirmed), and 3 of 11 (27%) patients in 150mg reached PR (2 confirmed, 1 unconfirmed). ctDNA analyses showed that response was associated with clearance of mESR1 ctDNA after dosing. Conclusions: FWD1802 showed great safety and tolerability with no DLT and TEAEs mostly of Grade 1-2. In late-line mESR1+ ET-resistant patients in ph2 doses, FWD1802 showed a superb 41% (11/27) ORR (unconfirmed). Considering linear PK property and comparable safety profile between 75mg and 150mg, and the overall highly promising ORR under ph2 doses, 150mg was selected for future monotherapy studies. The Ph2 expansion is still enrolling, and more monotherapy data will be updated once accumulated. (CTR20232130)
Presentation numberPS1-08-27
Randomized open-label multicenter phase 2 trial comparing first-line olaparib versus CDK4/6 inhibitor plus endocrine therapy in patients with gBRCAmut-associated HR+/HER2- advanced breast cancer [BR21-08, OPERA trial]
Kyong Hwa Park, Korea University College of Medicine, Seoul, Korea, Republic of
J. Kim1, J. Kim2, S.-H. Shin3, K. Lee4, J. Lee5, J. Sohn2, H. Kim6, K. Lee7, H. Won5, J. Jung8, K. Park1; 1Korea University College of Medicine, Seoul, Korea, Republic of, 2Yonsei University College of Medicine, Seoul, Korea, Republic of, 3Kosin university Gospel hospital, Busan, Korea, Republic of, 4National cancer center, Goyang, Korea, Republic of, 5Catholic University of Korea, Seoul, Korea, Republic of, 6University Cheonan Hospital, Cheonan, Korea, Republic of, 7Ewha Womans University Hospital, Seoul, Korea, Republic of, 8Hallym university Dongtan Sacred Heart Hospital, Dongtan, Korea, Republic of
Background: In HR+/HER2- advanced breast cancer (ABC), CDK4/6 inhibitors (CDK4/6i) plus endocrine therapy (ET) represent the current first-line standard. However, a subset of patients exhibit early resistance, and the impact of germline BRCA1/2 mutations (gBRCAmut) on treatment outcomes has not been fully evaluated. gBRCAmut tumors harbor homologous recombination deficiency and are potentially sensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. Although PARP inhibitors are established in gBRCAmut triple-negative BC, prospective data in gBRCAmut HR+/HER2- ABC are lacking. Methods: This open-label, multicenter, randomized phase II study (BR21-08, OPERA) enrolled patients with recurrent or metastatic HR+/HER2- ABC harboring germline BRCA1/2 mutations and no prior systemic therapy for advanced disease. Patients were randomized to olaparib (300 mg orally twice daily) or investigator’s choice of CDK4/6i plus ET. Upon progression, cross-over to the alternate regimen was allowed. The primary endpoint was progression-free survival on first-line therapy (PFS1). Based on an assumed HR of 0.375, 66 patients (33 per arm) were required to achieve 90% power with a two-sided alpha of 0.05. Results: As of September 2025, 10 patients were screened and 9 enrolled before early termination due to slow patient accrual. Median age was 50 years; 1 carried a gBRCA1 and 8 a gBRCA2 mutation. Six patients received CDK4/6i+ET (2 ribociclib, 2 abemaciclib, 2 palbociclib) and 3 received olaparib as first-line therapy. With median follow-up of 25.5 months, median PFS1 was 13.6 months in the control arm and 16.4 months in the olaparib arm. Two patients on abemaciclib remain on first-line therapy. Objective response rate (ORR) was 50% in the control arm (1 CR, 2 PR) and 33% in the olaparib arm (1 PR). Seven patients received second-line therapy: median PFS2 was 4.5 months for those receiving CDK4/6i+ET and 8.1 months for those receiving olaparib. The most common adverse event (AE) in the control arm during the 1st line therapy was neutropenia, with three grade ≥3 events, whereas nausea and anemia were most common in the olaparib arm, with no grade ≥3 events observed. Conclusion: In gBRCAmut HR+/HER2- ABC, first-line olaparib demonstrated numerically longer PFS and a favorable safety profile compared with CDK4/6i plus ET. These findings should be interpreted with caution given the small sample size.
Presentation numberPS1-08-28
The use of oral selective estrogen receptor degraders (SERDs) by community-based general medical oncologists (GMOs) in ER-positive (ER+), HER2-negative (HER2-neg) metastatic breast cancer (mBC): A clinician survey of practice patterns and practical challenges
Neil Love, Research To Practice, Key Biscayne, FL
K. H. Pang1, K. A. Ziel1, D. Paley1, S. A. Wander2, M. R. Lloyd3, N. Love1; 1Research To Practice, Key Biscayne, FL, 2Massachusetts General Hospital, Boston, MA, 3Beth Israel Deaconess Medical Center, Boston, MA
Background: The rapid pace of cancer research has created a challenging environment in which GMOs are constantly contending with the introduction of multiple novel agents and regimens often indicated for identical or overlapping patient populations. The 1/2023 approval of elacestrant (E) for patients with mBC and an ESR1 mutation (ESR1mut) whose disease progresses on endocrine therapy (ET) introduced new treatment considerations into a clinical situation which already had multiple approved therapeutic options. Since that time, several additional phase III reports — including 2 potentially practice-changing trials with novel SERD-based strategies (EMBER-3 and SERENA-6) — have added complexity to this space. This initiative sought to better understand GMOs’ familiarity with relevant research and their current and potential use of oral SERDs. Methods: In 9/2025, 50 US-based GMOs completed a case-based survey on the management of ER+ HER2-neg ESR1mut mBC. For each case, clinical factors (age, site/extent of metastases, symptom burden, duration of benefit with first-line treatment, PIK3CA mutations) were varied. The survey also asked GMOs to rate their level of familiarity with and interest in learning more about 12 phase III trials applicable to the management of patients with ER+ HER2-neg mBC. A modest honorarium was offered. Results: In general, GMOs self-reported familiarity with the 12 trials, with a mean overall rating of 3.7 out of 5. They were also very interested in learning more about these studies (mean rating 4.5). In terms of selection of therapy, for a 65-year-old patient with ESR1mut disease, progression 2.5 years after starting first-line CDK4/6 inhibitor (CDKi)/aromatase inhibitor (AI) and minimally symptomatic bone-only mets, 84% of GMOs recommend E. For the same patient with symptomatic visceral mets, 62% would deploy the SERD. For the same patient but with a coexisting PIK3CA mutation, E was preferred by 36% of GMOs in cases with bone mets and by 30% for symptomatic visceral disease. Other factors, including age (80 y) and shorter duration on CDKi/AI (10 mo), did not substantially affect treatment preferences across various situations. In terms of actual experience, GMOs reported using E a median of 4 times. Regarding select other oral SERDs (imlunestrant, camizestrant, giredestrant), 46% of GMOs believe data are not sufficient to determine comparative efficacy. However, 68% consider imlunestrant clinically equivalent whenever E is currently employed, and 72% would consider using it with abemaciclib in select situations. A majority (68%) of GMOs currently would utilize serial monitoring for ESR1mut during first-line treatment and early switching to a SERD (camizestrant) for patients with “molecular progression” ahead of overt clinical progression, as in SERENA-6. Conclusions: While GMOs are generally familiar with the key datasets, they are also highly motivated to learn more. GMOs have rapidly incorporated E into the management of progressive ER+ HER2-neg ESR1mut mBC. Similarities and differences in practice patterns were observed between GMOs and breast cancer research leaders (data available separately) for a number of clinical situations surveyed. In terms of novel SERDs, many GMOs consider the newly approved imlunestrant equivalent to E and would consider its use with abemaciclib in certain circumstances. Many GMOs would use camizestrant for patients found to have an ESR1mut without clinical disease progression, a perspective not shared by the majority of research leaders. This and other differences should be further studied to better understand what is driving the thoughts and recommendations of these different groups of clinicians.
Presentation numberPS1-03-05
Characterizing Nanomechanical Properties of Skin Tissue in Postmastectomy Implant Reconstruction
Papa Diogop Ndiaye, ARTIDIS AG, Basel, Switzerland
P. Ndiaye1, S. Nizzero2, M. Schaverien3, V. Hassid3, P. Shay3, J. Shuck3, C. Mark Warren3, J. Yu3, A. Ha3, G. Reece3, M. Roubaud3, A. Mericli3, M. Villa3, A. Francis,3, D. Adelman3, D. Baumann3, J. Christensen3, R. Dickey3, P. Hanwright3, S. Kapur3, J. Olenczak3, A. Sahin,3, E. Paredes3, J. Tilley1, A. Jizawi1, G. Srivastava1, S. Lasli1, T. Appenzeller1, M. Loparic1, M. Plodinec1, B. Smith3; 1ARTIDIS AG, Basel, Switzerland, 2ARTIDIS AG, Houston, TX, 3MDACC, Houston, TX
Introduction: Mastectomy remains a cornerstone of breast cancer treatment but often results in substantial disruption to body image and quality of life. Postmastectomy breast reconstruction (PMBR) offers physical and psychosocial restoration, yet failure rates, particularly following postmastectomy radiation therapy (PMRT), can exceed 20%. Known risk factors such as obesity, diabetes, smoking, and prior radiation increase complication rates, but no objective, individualized tool currently exists to guide patient selection or surgical planning. While patient-reported tools like the BREAST-Q provide retrospective insight into satisfaction and well-being, they do not predict outcomes or aid in preoperative decision-making. No validated nomogram or risk model broadly addresses implant failure in the setting of PMBR. Treatment-induced fibrosis alters tissue mechanics, suggesting that direct measurement of nanomechanical properties, such as stiffness, viscoelastic dissipation, and adhesion, could provide objective, biologically relevant markers of tissue quality and reconstructive risk. This study introduces dissipation; a quantifiable biomarker derived from high-resolution mechanical profiling and demonstrates its correlation with both subjective well-being and clinical outcomes. These findings offer an actionable, data-driven tool to stratify risk and optimize reconstructive planning, with immediate implications for clinical practice. Methods: This research is part of the EMPOWER study (NCT06584396), a single-center prospective trial at MD Anderson Cancer Center. Female patients undergoing preventive or curative mastectomy with tissue expander-based reconstruction were eligible. Since 2023, 130 patients have been enrolled, and 100 patients (131 samples) analyzed. At expander-to-implant exchange, surgeons collected skin and capsule specimens, which were trimmed and analyzed using the ARTIDIS ART-1 Nanomechanical Phenotype System, generating >10,000 data points per sample, including energy dissipation (aJ), stiffness (kPa), and adhesion (nN). Analyses assess associations between BREAST-Q Physical Well-Being scores (collected at 1 year post surgery) and the nanomechanical properties of resected tissues, combined with histopathological annotations. Results: Dissipation which refers to the ability of biological tissue to absorb and dissipate mechanical energy during deformation demonstrated strong correlations with subjective assessments from the BREAST-Q Physical Well-Being, particularly in parameters related to tissue softness, tightness, and discomfort. Dissipation values ranged from 150-800 aJ, with higher dissipation correlating with improved patient-reported softness and satisfaction of the breast hanging (p < 0.01) and lower values associated with fibrosis and dissatisfaction. Patients reporting chronic chest pain “all the time” exhibited dissipation peaks at 700 aJ, while those with no or occasional pain had peaks at 200-300 aJ. Conclusions: This is the first study to demonstrate that nanomechanical dissipation, a quantifiable measure of tissue viscoelasticity, correlates strongly with several measures of patient-reported physical chest wall properties. These early data support its potential as a biomarker to inform reconstructive planning and patient counseling in future workflows. Prospective correlation with clinical outcomes is ongoing, and further study is warranted to validate dissipation as a predictive tool in postmastectomy breast reconstruction.
Presentation numberPS1-10-08
Er riptacs drive tumor selective cell death in post-cdk4/6i resistance settings and demonstrate oral efficacy in cdx and pdx models
David E. Puleo, Halda Therapeutics, New Haven, CT
D. E. Puleo, M. A. Perry, M. B. Martin, C. D. Forbes, K. M. Jones, A. McGovern, A. Hundt, S. Fasciano, R. Stronk, M. P. King, A. Ward, K. Cooney-Walsh, K. Howard, C. Marshall, E. Attar, K. J. Eastman, K. Raina, K. J. Kayser-Bricker; Halda Therapeutics, New Haven, CT.
Background: Anti-estrogen therapies and CDK4/6 inhibitors (CDK4/6i) represent the backbone 1st line treatment for patients with ER+/HER2- metastatic breast cancer (MBC). However, patients ultimately progress on therapy due to heterogenous resistance mechanisms. Activating mutations in the estrogen receptor (ER) ligand-binding domain account for ~40-50% of this resistance. In this case, ER either fully (ER driven) or partially (ER co-driven) drives the disease. Next-generation therapies such as SERDs, PROTACs, and CERANs that degrade and/or antagonize ER are clinically active in the ER mutant setting but have weak or no activity in the ER wild-type (WT) setting where other heterogenous, non-ER mechanisms drive the disease (ER independent). Thus, there remains a significant unmet need to target ER co-driven and ER independent MBC. Regulated Induced Proximity Targeting Chimera (RIPTAC™) therapeutics are hetero-bifunctional small molecules that simultaneously bind a tumor-specific target protein (TP) and a pan-essential protein (EP). The TP:RIPTAC:EP ternary complex leads to selective inactivation of the EP only in target-containing cells. Breast cancer RIPTACs take advantage of ER expression to selectively inactivate BRD4 to induce breast cancer cell death. RIPTACs demonstrate potent cellular activity through BRD4 inactivation, regardless of ER driver status, and are expected to have broad activity across early- and late-line MBC. Methods: RIPTACs were tested for their ability to form a ternary complex between ER and BRD4. In vitro, pharmacodynamic response was assayed via qRT-PCR, and anti-proliferative activity in engineered and endogenous breast cancer cell lines was assessed. In vivo pharmacokinetic/pharmacodynamic analysis and tumor growth inhibition were assessed in both cell-line derived xenografts (CDXs) and patient-derived xenografts (PDXs). Agonist activity was assessed in the Ishikawa endometrial cell line in vitro and in juvenile rats in vivo. Results: RIPTACs form potent ternary complexes in vitro between BRD4 and both WT and multiple mutant forms of ER. Additionally, BRD4 pharmacodynamic response is observed only in ER-containing cells, resulting in nanomolar anti-proliferative activity in endocrine sensitive cell lines. RIPTACs cause significant tumor growth inhibition or regression in the endocrine sensitive MCF7 CDX model and in the ST941HI/PBR PDX model harboring ER Y537S. RIPTACs retain significant in vitro anti-proliferative activity in cell line models of post-CDK4/6i late-line resistance. Furthermore, breast cancer RIPTACs lack endometrial/uterine agonist activity, highlighting the modality’s therapeutic index and supporting the potential for clinical development across lines of BC treatment. Conclusions: These data demonstrate that RIPTACs are active across ER WT and ER mutant settings with a broad therapeutic index, thus providing an important potential treatment for patients with MBC. An ER-BRD4 RIPTAC is currently in IND-enabling studies with a Phase 1 FIH anticipated to start in 1H-2026.
Presentation numberPS1-05-05
At-home complete blood count monitoring: enhancing care pathways and improving patient experience
Toby Basey-Fisher, Entia, London, United Kingdom
S. Howell1, S. Taylor1, D. Ramanujam2, T. Basey-Fisher3, A. Brufsky4; 1The Christie NHS Foundation Trust, Manchester, United Kingdom, 2Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom, 3Entia, London, United Kingdom, 4UPMC, Pittsburgh, PA
Background: With myelosuppressive chemotherapy still extensively used and many antibody drug conjugates showing similar cytotoxic properties, complete blood counts (CBCs) continue to be a cornerstone of clinical decision making in cancer therapy. Historically, the need for in-clinic testing has presented logistical challenges for patients, reduced the efficiency of service delivery and delayed identification of complications such as febrile neutropenia, anemia and thrombocytopenia. To address this, the world’s first CBC analyzer has been developed for accessible use by cancer patients in the home (Liberty). The analyzer enables convenient patient self-testing from a single drop of capillary blood (16µL) through use of microfluidic consumables, centrifugation and fluorescent spectroscopy. The results are shared remotely to healthcare professional dashboards directly from the analyzer, providing the opportunity for more convenient and frequent CBC monitoring. Research Population and Methods: A cohort of breast cancer patients receiving systemic anti-cancer therapy (SACT) were selected for a qualitative sub-study from a larger multi-center, multi-indication comparator study evaluating at-home self-testing with Liberty versus standard of care venous blood testing at a health center. The primary endpoint was preference for at-home self-testing with Liberty versus standard of care venous blood testing at a health center. The secondary endpoint was healthcare resource utilisation across both monitoring pathways. Exploratory endpoints included patient experience of training, self-testing, ease of use and recommendation to other patients. Primary and exploratory endpoints were determined via questionnaires issued after at least two tests had been completed. Questionnaires asked participants their preference for self-testing compared to standard testing and to rate on a scale of 1-5, their experience of training, self-testing, acceptability of testing cadence, sense of control, perceived time savings, overall patient experience, likelihood to recommend Liberty to other patients and any additional comments. Results: Patients receiving a range of SACT (paclitaxel, ribociclib, abemaciclib, trastuzumab deruxtecan, sacituzumab govitecan) were recruited over 9 months to this sub-study (n=10). The mean score on participant preference for self-testing with Liberty versus the standard of care was 4.2/5 with 5 representing a strong preference for self-testing with Liberty (median 4.5). Self-testing with Liberty was shown to reduce the need for routine in-clinic blood tests by 96%. Scored out of a maximum of 5, the mean patient score for patient training was 5 (median: 5), experience of self-testing with Liberty 4.9 (median: 5), perceived sense of control 4 (median: 4.5), willingness to keep testing at the defined cadence 4.6 (median: 5), perceived time savings 4.6 (median: 5), overall experience 4.6 (median: 5) and likelihood to recommend Liberty to other patients was 4.9 (median: 5). No adverse events were reported. Conclusions: Liberty was the preferred pathway for blood monitoring and may help to reduce the significant burden faced by patients and healthcare systems for cancer treatment. Subsequent real-world usage of Liberty has demonstrated reductions of up to 91% of routine in-clinic blood tests and shown its utility in avoiding unnecessary emergency room attendance.
Presentation numberPS1-11-15
Current and future use of oral selective estrogen receptor degraders (SERDs) in the management of ER-positive (ER+), HER2-negative (HER2-neg) metastatic breast cancer (mBC): A survey of 20 breast cancer research leaders (RLs)
Neil Love, Research To Practice, Key Biscayne, FL
K. H. Pang1, K. A. Ziel1, D. Paley1, S. A. Wander2, M. R. Lloyd3, N. Love1; 1Research To Practice, Key Biscayne, FL, 2Massachusetts General Hospital, Boston, MA, 3Beth Israel Deaconess Medical Center, Boston, MA.
Background: The Jan 2023 approval of elacestrant (E) for patients with ER+ HER2-neg mBC and an ESR1 mutation (ESR1mut) with disease progression (PD) on endocrine therapy introduced new considerations for biomarker-guided decision-making. In the past year (y), 2 seminal phase III trials (EMBER-3, SERENA-6) reported potentially practice-changing findings with novel SERD-based strategies. We sought to determine the current and potential use of these agents. Methods: In Sep 2025, 20 US and international RLs completed a case-based survey on the management of ER+ HER2-neg ESR1mut mBC. For each case, a number of clinical factors (age, site/extent of metastases, symptom burden, duration of benefit with first-line treatment, PIK3CA mutations) were varied. A modest honorarium was offered. Results: All RLs employ ESR1mut testing, with 7 first doing so at metastatic diagnosis, 10 at PD on first-line treatment and the rest customizing their approach. 12 favor circulating tumor DNA and 8 have no preference between tissue and liquid ESR1 assays. For a 65-y-old patient with ESR1mut disease, PD 2.5 y after first-line CDK4/6 inhibitor (CDKi)/aromatase inhibitor (AI), and minimally symptomatic bone mets, 16 of 20 RLs recommend E. For the same patient with symptomatic visceral mets, only 4 would deploy the oral SERD. For the same patient but with coexisting PIK3CA mutations, E was preferred by 12 RLs in cases with bone-only mets but only by 3 for symptomatic visceral disease (Table). Other factors, including age and shorter duration on CDKi/AI, affected treatment preferences across various situations, with somewhat more RLs turning to E for an 80-y-old and fewer for a 65-y-old with PD after 10 mo of first-line treatment. Half of the RLs view select other SERDs as equivalent to E in efficacy and tolerability and 17 of 20 consider imlunestrant clinically equivalent whenever E is employed. All would use imlunestrant with abemaciclib in select situations. Regarding serial monitoring for ESR1mut during first-line treatment and early switching to a SERD (camizestrant) for patients with “molecular progression” (as in SERENA-6), 13 RLs believe more data are required but 7 endorse this approach now. Conclusions: RLs have rapidly incorporated E into the management of progressive ER+ HER2-neg ESR1mut mBC, but the decision to employ this agent appears to be heavily influenced by the global disease burden and the duration of benefit from first-line treatment. Most RLs consider the newly approved imlunestrant largely equivalent to E, and all would also consider its use in combination with abemaciclib in certain situations. Many fewer would use camizestrant for patients found to have an ESR1mut without clinical PD, but an important and significant minority believe this option should be available to select patients.
|
Clinical scenario |
Top 2 choices of systemic therapy |
|
65-year-old, minimally symptomatic bone metastases
ESR1 mutant, PIK3CA/AKT1/PTEN wild type (WT) |
Elacestrant (16/20) = 80%
Abemaciclib + fulvestrant (3/20) = 15% |
|
65-year-old, minimally symptomatic bone metastases
ESR1 mutant, PIK3CA mutant |
Elacestrant (12/20) = 60%
Capivasertib + fulvestrant (6/20) = 30% |
|
80-year-old, minimally symptomatic bone metastases
ESR1 mutant, PIK3CA/AKT1/PTEN WT |
Elacestrant (17/20) = 85%
Abemaciclib + fulvestrant (2/20) = 10% |
|
80-year-old, minimally symptomatic bone metastases
ESR1 mutant, PIK3CA mutant |
Elacestrant (15/20) = 75%
Capivasertib + fulvestrant (3/20) = 15% |
|
65-year-old, symptomatic visceral metastases
ESR1 mutant, PIK3CA/AKT1/PTEN WT |
Abemaciclib + fulvestrant (6/20) = 30%
Elacestrant (4/20) = 20% |
|
65-year-old, symptomatic visceral metastases
ESR1 mutant, PIK3CA mutant |
Capivasertib + fulvestrant (10/20) = 50%
Elacestrant (3/20) = 15% |
Presentation numberPS1-12-01
Brain fes pet standardized uptake value association with esr1 and pik3ca genomic alterations
Arman Sharbatdaran, New York Presbyterian – Weill Cornell Medicine, NY, NY
A. Sharbatdaran, L. Pontolillo, E. Elmoujarkach, S. Nehmeh, A. Brandmaier, R. S. Magge, R. Ramakrishna, S. C. Pannullo, J. P. S. Knisely, K. F. P. Beal, J. R. Osborne, M. Cristofanilli, J. Ivanidze; New York Presbyterian – Weill Cornell Medicine, NY, NY
Objectives: Breast cancer (BC) is the second most common cancer linked to brain metastases(BM), with 70% of patients having estrogen receptor-positive (ER+) disease. Brain metastases represent a major clinical challenge, second in frequency only to those arising from lung cancer.[F18]-Fluoroestradiol ([F18]-FES) PET is an FDA-approved molecular imaging tool that targets ER with high specificity (>90%) and allows whole-body assessment of ER expression. BrainFES PET has shown promise as an adjunct diagnostic tool in BC BM. Liquid biopsies, including circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes, are emerging non-invasive diagnostic and monitoring tools. Our purpose was to combine FES PET with liquidbiopsy in a pilot cohort to assess the potential of this multi-modal biomarker in evaluatingresponse to endocrine and radiotherapy in patients with ER+ BC BM.Methods: Following informed consent, patients with ER+ BC and known or suspected BM who underwent dedicated brain FES PET/CT and blood ctDNA analysis were included in this IRB-approved prospective study. Clinical and demographic data were collected via chart review. Dedicated dynamic brain PET/CT acquisition was performed from 0-90 minutes. FES PET was co-registered to post-gadolinium contrast 3D T1 MRI. Two 10-mL blood samples were collected and analyzed using the Guardant360 next-generation sequencing platform to detect genetic alterations. Codon variants of ESR1 and PIK3CA mutations were analyzed, and mutant allele frequency was assessed for personalized treatment. The average maximum standardized uptake values (SUV) in ESR1- and PIK3CA-positive and ESR1- and PIK3CA-negative groups were compared separately. Mann-Whitney and Spearman correlation tests were performed. Results: Nine patients with FES-positive lesions were stratified by PIK3CA and ESR1mutations. 5/9 (56%) were PIK3CA-positive, and 4/9 (44%) were ESR1-positive. The median FES PET SUV was 6.47 in the PIK3CA-positive and 3.20 in the PIK3CA-negative subgroup (p 0.015). The median FES PET SUV was 4.30 in the ESR1-positive and 3.30 in the ESR1-negative subgroup (p = 0.794). Strong correlation was found between FES PET SUV and mutation count, approaching statistical significance (r = 0.94, p = 0.06).Conclusions: Our preliminary analysis demonstrates higher FES PET SUV in patients with PIK3CA and ESR1 mutations, as well as a trend for greater SUV in patients with a greater number of mutations, noting limitations of small sample size. Integrating brain FES PET with liquid biopsy has the potential to optimize treatment response in patients with ER-positive BCBM. This multi-modal approach may enhance our understanding of therapy resistance mechanisms by integrating biologically targeted PET with molecular profiling to guide treatment decisions in patients with ER+ BC BM. References1. Barnholtz-Sloan JS, Sloan AE, Davis FG, et al. Incidence proportions of brain metastases in patients diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J Clin Oncol 2004;22:2865-2872 2. Lother D, Robert M, Elwood E, et al. Imaging in metastatic breast cancer, CT, PET/CT, MRI, WB-DWI, CCA: review and new perspectives. Cancer Imaging2023;23:53
Presentation numberPS1-12-04
Development and characterization of patient-derived xenografts of HR+/HER2- breast cancer following progression on CDK4/6 inhibitor-based therapy
Agnieszka Witkiewicz, Roswell Park Cancer Center, Buffalo, NY
A. Witkiewicz, S. Tzetzo, E. Schultz, E. Bailey, R. Kyne, T. O’Connor, E. Knudsen; Roswell Park Cancer Center, Buffalo, NY.
A limitation in the field of research around HR+/HER2- breast cancer is the lack of models that represent meaningful features of the disease. On the NCT04526587 clinical study, standard of care tumor tissues were collected to develop patient-derived xenografts (PDX) from patients that had progressed on CDK4/6 inhibitor-based therapy. As of September 2025 a total of 11 models have been developed. In general, there was a greater success in establishing models from solid tissue biopsies relative to liquid collections, such as ascites or effusion. The models expressed variable levels of the estrogen receptor and were molecularly characterized by whole-exome sequencing and gene expression analyses. The PDX models were highly represented by ESR1 mutations as well as PIK3CA and other common genetic lesions in metastatic HR+/HER2- breast cancer. All models fell into the Luminal B or HER2 subtypes of breast cancer based on absolute assignment of breast cancer intrinsic subtypes. The subtype of each model remained relatively static with passaging in NSG mice supplemented with estrogen pellets. Assessment of therapeutic sensitivity illustrated a general resistance to CDK4/6 inhibitors relative to established xenograft models (e.g. MCF7 xenografts). However, combination treatment with CDK4/6 and CDK2 inhibitors was highly effective in halting proliferation. Combined treatment resulted in potent arrested and generated in vivo gene expression changes indicative of therapy-induced senescence. In select cases, PDX tumors failed to proliferate following the cessation of treatment, supporting the potency of this therapeutic regimen. Pilot studies have used cell culture of select PDX models to define unique mechanisms of resistance and vulnerabilities. Together this work is providing new resources and insights upon which to combat therapeutic resistance in metastatic HR+/HER2- breast cancer.
Presentation numberPS1-09-01
Analysis of Prognostic Factors in HR+/HER2- Metastatic Breast Cancer with Long-Term Response to CDK4/6 Inhibitors
Thanh Nhu Nguyen, Hochiminh City Oncology Hospital, Hochiminh, Viet Nam
T. N. Nguyen1, D. T. H. Phan2, T. B. Diep1, H. D. Vo1, P. H. M. Vo1, Q. H. Nguyen1, T. T. T. Nguyen2, H. N. Vo1; 1Hochiminh City Oncology Hospital, Hochiminh, Viet Nam, 2Pham Ngoc Thach University of Medicine, Hochiminh, Viet Nam
Introduction: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) combined with endocrine therapy are the standard of care for hormone receptor-positive, human epidermal growth factor receptor 2-negative (HR+/HER2-) metastatic breast cancer (MBC). Although most patients eventually develop resistance, a subset achieves durable responses lasting at least 24 months with a single CDK4/6i. Identifying prognostic factors associated with this long-term benefit is essential for guiding clinical practice. Methods: We conducted a retrospective cross-sectional study of HR+/HER2- MBC patients treated with CDK4/6i plus endocrine therapy at Ho Chi Minh City Oncology Hospital between January 2021 and July 2023. Eligible patients were those who received continuous treatment with one CDK4/6i for ≥24 months. Progression-free survival (PFS) was estimated using Kaplan-Meier methods, and prognostic factors were evaluated by univariate and multivariate Cox proportional hazards regression models. Results: Of 236 patients treated with CDK4/6i, 80 met inclusion criteria by maintaining therapy with a single CDK4/6i for ≥24 months (30 with palbociclib [36.1%], 50 with ribociclib [63.9%]). Median follow-up was 31.4 months (range, 24.08-46.16). The median PFS for the cohort was not reached. Subgroup analysis demonstrated that patients achieving partial or complete response (PR/CR) did not reach median PFS, whereas those with stable disease (SD) had a median PFS of 37.65 months (95% CI, 34.11-41.19; p = 0.024). In multivariate analysis, both treatment response (PR/CR vs SD) and Ki-67 >30% were significantly associated with PFS. Conclusion: In HR+/HER2- MBC patients achieving long-term response (≥24 months) to a single CDK4/6i plus endocrine therapy, treatment response and high Ki-67 index (>30%) emerged as significant prognostic factors for PFS. These findings highlight the prognostic value of clinical response and proliferative activity in predicting durable benefit.
Presentation numberPS1-13-10
Elucidating Stemness-Associated Regulatory Pathways to Guide Personalized Treatment for Ductal Carcinoma in Situ
Fariba Behbod Behbod, University of Kansas Medical Center, Westwood hills, KS
F. B. Behbod1, E. Schueddig2, V. Kochat3, E. Arslan4, Y. Dallas2, P. Yang5, R. Madan2, Z. Y. Li3, T. Fields2, J. L. Wagner2, K. Larson2, C. Balanoff2, A. Aripoli2, A. Huppe2, O. Winblad2, J. Peterson2, M. Hill2, C. Smith2, C. Fabian2, E. J. Jeffers2, D. C. Koestler2, A. K. Godwin2, L. J. Kilgore2, M. T. Lewis6, S. A. Khan7, A. M. Thompson6, K. Rai1; 1University of Kansas Medical Center, Westwood hills, KS, 2University of Kansas Medical Center, Kansas City, KS, 3MD Anderson Cancer Center, Houston, TX, 4Takeda, Cambridge, MA, 5MD Anderson Cancer Center, Huston, TX, 6Baylor College of Medicine, Houston, TX, 7Northwestern University, Chicago, IL
Introduction: Ductal carcinoma in situ (DCIS), the most common form of non-invasive breast cancer, is considered a precursor to invasive ductal carcinoma (IDC). A critical gap remains in identifying which DCIS lesions are likely to progress to lethal and invasive disease and tailoring treatment accordingly. Methods: We evaluated the role of cellular plasticity and stem-like traits as drivers of DCIS progression. CytoTRACE, an algorithm that uses single-cell RNA sequencing (scRNA-seq), was utilized to evaluate cellular stemness (PMC7694873). Forty-three patient samples were subjected multi-omic technologies including scRNA-seq analysis of 17 patient-derived DCIS MIND models (8 progressed and 9 non-progressed), spatial transcriptomic analysis of 16 pure DCIS and DCIS with associated IDC (DCIS/IDC) and scATAC/RNA-seq analysis of 10 DCIS and DCIS/IDC samples. We utilized cell annotations published by Kumar and colleagues (PMC10168262). Spatial Graph Convolutional Network (SpaGCN) was used to map the location of stem-like cells. Ligand-receptor (LR) interaction analysis and transcription factor motif enrichment analysis were used to identify LR and regulatory networks within stem-like cells. Statistical tests including Mann Whitney U Test (MWU) was used to compare differences in stemness and a mixed-effect regression model (LMM) was used to assess tumor volume changes in control vs. treated animals. Results: Our preliminary data show that HER2-positive DCIS exhibits significantly higher enrichment in stem-like cells, which correlates with an increased risk of invasive and metastatic progression (MWU; p<0.0001). Analysis of the spatial transcriptomics dataset revealed a significant decrease in the proportion of Basal and Luminal secretory (LumSec) cell subclusters from benign to DCIS and IDC regions. This was mirrored by a significant increase in Luminal Hormone Responsive (LumHR) cells across the same progression (MWU; p<0.01), suggesting that LumHR cells are major contributors to DCIS, with their contribution significantly increasing with the transition to IDC. Consistent with this observation, CytoTRACE analysis confirmed that LumHR cells exhibited the highest stemness scores among all epithelial cell subclusters (MWU; p<0.01). ScRNA-seq and spatial transcriptomics further corroborated that DCIS with invasive potential displayed significantly higher stemness compared to DCIS with non-invasive potential (MWU; p<0.01). These included patient DCIS cells before they showed invasiveness in the MIND models. Therefore, stemness may serve as a signature of DCIS with a risk for future invasive potential. SpaGCN analysis identified a unique DCIS niche that was enriched with high-stem like cells. Further, LR analysis revealed interactions between CEACAM6 on high-stem-like LumHR cells with EGFR on myeloid and lymphatic cells. Finally, scATAC/RNA sequencing revealed an enrichment of FOXA1 transcription factor binding motifs in the cell clusters with the highest stemness scores in HER2+ DCIS. To target FOXA1, we employed bomedemstat, an inhibitor of lysine-specific demethylase 1 (LSD1). LSD1 inhibitors have been shown to target FOXA1 by blocking its demethylation and transcriptional activation (PMC7541538). In vitro, treatment of HER2+ SUM225 cells with bomedemstat decreased the expression of HER2, CEACAM6, and FOXA1. In vivo, bomedemstat showed a significant reduction in tumor volume over time in a HER2+ metastatic breast cancer PDX model (LMM; p<0.01). Conclusion: Cellular stemness may serve as a predictor of DCIS with a future risk for invasive progression. By targeting the key biological drivers of cellular stemness, our goal is to distinguish aggressive from indolent disease, ultimately reducing overtreatment and improving outcomes for patients with high-risk DCIS.
Presentation numberPS1-13-15
A new FDA approved docetaxel + albumin formulation for breast cancer treatment
Hirak Biswas, Syneos Health, Morrisville, NC
H. Biswas1, D. Desai2, Q. Sun3; 1Syneos Health, Morrisville, NC, 2Zydus Pharmaceuticals USA, Pennington, NJ, 3Zhuhai Beihai Biotech Co. Ltd., Zhuhai, China
Docetaxel was first approved as an anti-neoplastic agent in 1996 and is still used extensively for breast cancer treatment. Currently, available formulations for docetaxel use synthetic excipients, such as polysorbate 80, which are associated with hypersensitivity reactions. Here we describe a novel, polysorbate 80 free, docetaxel formulation with human albumin, BEIZRAY (BH009), which is FDA approved based on demonstrated bioequivalence to generic docetaxel. The bioequivalence study, NCT04889599, was a multicenter, randomized, open-label, 2-way crossover study, comparing the pharmacokinetics and safety of BH009 and docetaxel (Winthrop docetaxel injection, an authorized generic of Taxotere). 46 patients were randomized in the study and received single-dose of 75 mg/m2 BH009 or Taxotere IV over 60 min, with a 21-day washout between doses. The study demonstrated bioequivalence between BH009 and Taxotere based on PK parameters Cmax, AUC0-t and AUC 0-∞. Furthermore, BH009 demonstrated favorable grade 3/4 AE profiles than Taxotere. In conclusion, breast cancer patients now have a new docetaxel + human albumin formulation that is FDA approved via 505(b)(2) pathway, which may overcome the challenges associated with polysorbate 80 related hypersensitivity reactions.
Presentation numberPS1-13-08
Breast Cancer Risk Factors and Artificial Intelligence-derived Mammographic Risk Score in Black and White Women
Tuo Lan, Washington University School of Medicine, St. Louis, MO
T. Lan; Washington University School of Medicine, St. Louis, MO
Backgrounds Mammographic risk score (MRS), an artificial intelligence (AI)-derived metric from digital mammograms, captures breast tissue characteristics that may reflect cumulative exposures influencing breast cancer development. However, its relationship with well-established risk factors remains unclear. Methods This study included 9,623 cancer-free women from the Joanne Knight Breast Health Cohort who completed a baseline risk questionnaire and had MRS from screening mammograms. From 2008 to 2024, 468 breast cancer were diagnosed. We used linear regression to estimate β coefficients and 95% confidence intervals (CIs) for associations of MRS with lifestyle, reproductive, and previously validated risk factor models. We cross-classified MRS risk quintiles with risk factor models quintiles to compare differences in estimated incidence. Results MRS was positively associated with the Rosner-Colditz (β = 0.199, 95% CI: 0.138 to 0.260) and Tyrer-Cuzick scores (β = 0.072, 95% CI: 0.055 to 0.089). Being postmenopausal before age 50 (β age<50 post-menopause VS age<50 pre-menopause = -0.526, 95% CI: -0.646 to -0.406) were associated with lower MRS, whereas a history of breast biopsy (β yes VS no = 0.373, 95% CI: 0.303 to 0.443), postmenopausal BMI (β = 0.005 per 1kg/m2, 95% CI: 0.002 to 0.009), and breast density (β extremely dense~55% VS almost all fat~ 5% =0.374, 95% CI: 0.272 to 0.476) were associated with higher MRS (P <0.001 to 0.022). MRS improved risk factors models: breast cancer incidence increased by 53.2% per quintile of MRS within the Tyrer-Cuzick model and 44.9% within the Rosner-Colditz model (both P < 0.0001). Women reclassified as high risk by MRS were more often younger and Black women. Conclusions MRS captures biologically relevant imaging features shaped by reproductive and metabolic exposures and improves breast cancer risk prediction, particularly for younger and Black women. Incorporating MRS into clinical tools could enable earlier identification of high-risk women, guide risk-reduction strategies, and tailor screening guidelines.
Presentation numberPS1-13-17
Rna-based immune features associated with benefit from distinct microtubule inhibitor therapy for metastatic her2-negative breast cancer: a post hoc analysis of the calgb 40502 (alliance) phase iii randomized clinical trial
Elijah K Blige, The Ohio State University, Columbus, OH
E. K. Blige1, K. V. Ballman2, A. Michmerhuizen3, M. Vater1, L. A. Carey3, A. H. Partridge4, W. F. Symmans5, M. A. Watson6, C. M. Perou3, D. G. Stover1, H. S. Rugo7; 1The Ohio State University, Columbus, OH, 2Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, 3University of North Carolina, Chapel Hill, NC, 4Dana-Farber Cancer Institute, Boston, MA, 5MD Anderson Cancer Center, Houston, TX, 6Washington University School of Medicine, St. Louis, MO, 7University of California-San Francisco, San Francisco, CA
Background: Microtubule inhibitors remain a standard chemotherapy in the management of HER2-negative metastatic breast cancer (MBC). CALGB (Alliance) 40502 was a phase 3 randomized study involving 799 patients with MBC receiving first-line chemotherapy, to determine the optimal chemotherapeutic agent among paclitaxel, nab-paclitaxel, or ixabepilone (with or without bevacizumab). Secondary analysis suggested inferior PFS with nab-paclitaxel specifically among patients with hormone receptor positive (HR+)/HER2-negative breast cancer. Correlative analysis demonstrated significant association of stromal tumor infiltrating lymphocytes (sTILs) with improved progression-free (PFS) and overall survival (OS) in CALGB40502. We hypothesized that immune activation would be associated with differential benefit among distinct microtubule agents. To address this, we evaluated the sTILs and RNA-based immune features association with clinical outcomes, including evaluation of nab-paclitaxel versus paclitaxel.Methods: To limit heterogeneity, analyses focused on pre-treatment primary tumor breast samples with central review/pathologist-enumerated sTILs in accordance with International TILs Working Group methods and RNAseq (n = 280). PAM50 molecular subtypes were assigned based on transcriptomic features. RNAseq data were used to generate immune deconvolution estimates from 5 distinct algorithms (CIBERSORT, xCell, ABIS, ConsensusTME, ImmuCellAI). 1,018 curated breast cancer gene expression signatures GES) were derived from previously published studies. Associations between sTILs and GES were evaluated with sTILs being a categorical variable with the thresholds <5% (low) and (≥5%) high. Results: In the evaluable population, basal-like PAM50 subtype was enriched for high sTILs category (p=1e-4), as anticipated. T-cell and B-cell immune signatures and immune deconvolution estimates were among most highly associated with sTILs. In addition to the expected prognostic association of high T-cell GES, high B-cell-related IgG signature score was also significantly associated with improved overall survival (OS) among triple-negative breast cancer (TNBC) in CALGB 40502 (HR = 0.50,95% CI:0.30-0.86, log-rank p=0.01) but not in HR+/HER2-. In an exploratory analysis of young patients (age at study entry <50 years), high sTILs were associated with improved OS among TNBC patients but worse OS among those with HR+/HER2- MBC (HR = 2.19, 95% CI:1.10-4.36, log-rank p=0.022). High (above median) RNA-based Xcell total immune score was significantly associated with worse OS in both CALGB 40502 and a validation dataset of primary breast cancers, METABRIC (OS HR = 2.12, 95% CI:1.01-4.45, log-rank p=0.042). To further investigate the clinical trial finding of inferior PFS for nab-paclitaxel compared with paclitaxel in patients with HR+/HER2- MBC, there was a significant enrichment of T-cell signatures associated with poor outcome in patients receiving nab-paclitaxel (Fisher exact p<0.001).Conclusion: In this translational analysis of sTILs and RNAseq from a large phase III clinical trial, sTILs and immune signatures were not uniformly associated with better outcome. Rather, high sTILs or immune signatures were associated with worse OS in young patients with HR+/HER2- breast cancer and higher T-cell signatures were associated with inferior PFS with nab-paclitaxel among HR+/HER2- MBC patients. These data reinforce the importance of context/subtype-specific interrogation of the tumor-immune microenvironment.Support: U10CA180821, U10CA180882; https://acknowledgments.alliancefound.org. Bristol Myers Squibb (BMS); Clinicaltrials.gov Identifier: NCT00785291
Presentation numberPS1-13-26
Digital Self Service for Breast Cancer Screening: Early Experience with a Direct-to-Patient Program in a Federally Qualified Health Center
Erin Hultgren, Kintegra Health, Gastonia, NC
E. Hultgren1, D. Smith1, D. Santillan1, D. P. Miller2, L. Alexander1; 1Kintegra Health, Gastonia, NC, 2mPATH Health, Winston-Salem, NC
Background: Federally qualified health centers (FQHCs) serve lower income populations that have disproportionately low cancer screening rates; less than 50% of eligible women seen in FQHCs are up to date with mammograms. Additionally, many women at elevated breast cancer risk are unaware of their risk status, limiting access to genetic counseling and enhanced surveillance. Unfortunately, primary care providers report a lack of time to counsel women about screening and calculate individual risk scores. A digital health program delivered directly to patients outside the confines of a clinical visit could overcome many of these barriers. We evaluated a new “digital self-service” program for breast cancer screening in a pilot study in a FQHC in North Carolina. Methods: Each week, an automated query at the FQHC identified English or Spanish-speaking women aged 45 to 75 years who had no record of a screening mammogram in the prior 12 months and were scheduled for a primary care appointment in the next 9 to 16 days. The new digital health program, called mPATH-Breast, sent identified women an automated text message inviting them to determine their personal risk for breast cancer by clicking a hyperlink that launched the mPATH-Breast web app. The web app, available in English and Spanish, included a landing page informing women of the importance of breast cancer screening, allowed women to estimate their personal lifetime risk using the Gail model, confirmed they were due for routine screening, displayed a brief animated decision aid video, and allowed them to request either a mammogram or a referral to a high risk breast clinic as appropriate based on their risk score. mPATH-Breast then automatically transmitted mammogram and referral requests to the FQHC scheduling team and generated a one-page summary of each patient’s app use for upload into the electronic health record (EHR). Results: Between June 2025 and August 2025, the mPATH-Breast program sent text message invitations to 207 women, of which 193 (93%) were successfully delivered. Of the 193 women reached, 47 (24%) clicked the embedded hyperlink, and 17 of the 47 (36%) started the mPATH-Breast program. Fifteen of those 17 women (88%) completed the entire program. All 15 women (100%) elected to estimate their personal risk for breast cancer, with lifetime risks ranging from 3.2% – 9.8%. Nine of the 15 women (60%) reported having a mammogram in the last 12 months, and 4 women (27%) were confirmed due for average-risk mammography, of whom all 4 (100%) requested a mammogram. Conclusion: This early pilot demonstrates that a direct-to-patient digital program can effectively engage women in FQHC settings, where mammography uptake is otherwise low. Nearly all women who started the program completed it and chose to estimate their personal breast cancer risk; every woman found to be overdue requested a mammogram. Notably, more than half of women reported having a recent mammogram that was not documented in the EHR, underscoring the common challenges of capturing screening completed outside the health center and the importance of accurate record integration. Taken together, these results show that a direct-to-patient digital program can improve risk awareness, drive immediate action, and offer a scalable, patient-centered strategy to strengthen breast cancer prevention and early detection in underserved populations. Future research should examine strategies for increasing patient engagement with digital tools for screening.
Presentation numberPS1-13-18
Single Intratumoral Drug Injection Yields Complete Response (CR) in Metastatic Breast Cancer (MBC) Bone Lesions: Results from a Phase 2a Trial
Bryan S Margulies, Zetagen Therapeutics, Syracuse, NY
M. K. S. Heran1, B. S. Margulies2, J. C. Loy2, D. Villa1; 1University of British Columbia, Vancouver, BC, Canada, 2Zetagen Therapeutics, Syracuse, NY
Background: MBC bone defects occur in the axial skeleton in 70% of patients (pts), decreasing survival and quality of life (QoL). Pathologic fracture and spinal cord compression are skeletal-related events (SRE) that further reduce survival. MBC bone lesions create destructive lytic defects that produce debilitating pain often requiring opioid pain management, palliative radiation therapy, and surgery. Reducing SRE risk will improve QoL. Zeta-BC-003, is a first-in-class intratumoral injectable drug therapy that contains a specific concentration of a well-known small molecule, that works via a new molecular pathway. Zeta-BC-003 is comprised of a collagen-calcium phosphate biomaterial infused with N-allyl noroxymorphone resulting in CR in 6 consecutive pts in a Phase 2a trial, consistent with 2 pts previously treated via Compassionate Use (CU) with 2-yr follow-up (Palma et al. Pain Manag, 2023). Methods: ZGMBC (NCT05280067) is a non-randomized, open label Phase 2a trial currently at the University of British Columbia. Ten female pts were enrolled (8/23-3/25) with spinal MBC lytic bone lesions and a Spinal Instability Neoplastic Scale (SINS) score ≥3 and ≤9. Under sedation, 1 lesion per patient was treated with a single intratumoral injection of Zeta-BC-003, via an 18-gauge spinal needle and fluoroscopic guidance. Pts were followed with CT and MRI at discharge and days 84 & 180. Primary endpoints: SREs, Bone defect size change; Pain measured using the Numeric Rating Scale (NRS); and Postoperative prescription opioid use. Secondary endpoints: QoL via SF-12v2, SINS, and treatment response via the MD Anderson Criteria (MDAc) and RECIST v1.1. Results: Pts had a mean age of 52-yrs (range: 32.5 – 67.4) and 9 were pre/peri-menopausal. Six pts had HR+ tumors while 2 were HR+/HER2+, 1 was HER2+/HR-, and 1 was TNBC. Four pts had non-surgically accessible bone MBC (ribs, ilium, skull), 3 had liver metastases (mets), 1 had lung mets, and 3 had mediastinal lymph node mets. Five pts were given bisphosphonates. In the 6 pts that have completed the study there were no SAEs, AEs, or SREs. There were 6 treated lesions for the 6 pts, with one patient having 3 additional adjacent lesions for a total of 9. The mean bone defect volume decreased 87.9% (±8.5%) at day 180. Seven of the MBC bone defects were healed, resulting in normal bone morphology by imaging. All 6 pts had a CR treatment response via the MDAc, which is defined as ‘fill-in’ of the bone defect via CT + no active tumor (‘hot spots’) on MRI. A 24.2% increase in the Physical Component Summary (PCS) and a 12.1% increase in the Mental Component Summary (MCS) showed improved QoL (SF-12v2). Pain decreased 4.16% per the mean NRS. The Morphine Equivalent Dose (MED) was calculated for the 3 pts treated with opioids, which showed a decrease of 66.9%, 66.7%, and 33.3%. QoL improved with decreased MED, with PCS increasing for those pts 36.7%, 13.4%, and 58.4%. Decreased MED related to a lower MCS for 2 pts (2.4% & 6.9%) while the third increased 47.1%. The mean SINS increased 18.5%, from 4.5 to 3.7, indicating increased stability. Conclusion: There were no SREs and Zeta-BC-003 provided a CR (no active tumor) in all 6 pts for all 9 lesions, decreased MBC bone defect volume, increased stability (SINS), decreased pain (NRS & MED), and increased patient QoL. Zeta-BC-003 prevented SREs while 53% of MBC pts with bone mets reported an SRE (Parkes et al. Oncologist 2018). These data are consistent with the pts treated via CU and followed for 2-yrs, in which a CR was observed in all 7 treated MBC bone lesions coupled with decreased pain via the NRS (50%) and 76% less opioid drug use via the MED (day 0: 250-mg vs 2-yrs: 60-mg). These results offer the first non-palliative intratumoral treatment that ceases MBC lytic lesion activity, regrows bone, reduces SRE risk, while increasing QoL and possibly overall survival.
Presentation numberPS1-13-11
Personalized Whole-Genome-Based ctDNA Dynamics During Neoadjuvant Therapy Across Breast Cancer Subtypes: Early Insights From MONITOR-Breast
Julia Foldi, University of Pittsburgh Medical Center, Pittsburgh, PA
J. Foldi1, G. Hogan2, M. LaBella2, B. Mabey2, M. Balic1, D. Muzzey2, K. Johansen Taber2, J. Jasper2; 1University of Pittsburgh Medical Center, Pittsburgh, PA, 2Myriad Genetics, Salt Lake City, UT.
Introduction In patients with breast cancer who receive neoadjuvant treatment, therapy adjustments are primarily guided by predefined imaging and clinical assessments. Circulating tumor DNA (ctDNA)-based molecular residual disease (MRD) testing offers a real-time approach to evaluate treatment response and inform escalation or de-escalation strategies. Prior studies have generally examined binary MRD status at sparse timepoints, resulting in limited resolution and a lack of quantitative data on treatment response. The MONITOR-Breast study employs an ultrasensitive, whole-genome sequencing (WGS)-based assay with frequent ctDNA sampling across the neoadjuvant period. Here, we report initial results from an exploratory analysis of this multi-center, prospective study, and provide a quantitative, high-resolution dataset on ctDNA dynamics. Methods Patients with stage I-III breast cancer of any subtype who were planning to receive neoadjuvant chemotherapy were prospectively enrolled in MONITOR-Breast. WGS of core needle biopsy tissue from the primary tumor was used to design individualized capture panels with up to 1,000 tracking variants (Precise® MRD, Myriad Genetics). Plasma was collected before treatment initiation, prior to each neoadjuvant chemotherapy cycle, after completion of neoadjuvant therapy, up to 7 days before or 3 days after surgery, and approximately 3 weeks after surgery. Subtype and treatment-specific ctDNA trajectories were assessed relative to imaging and pathologic response (complete pathologic response (pCR) vs residual disease (RD)). Results At the time of analysis, 87 patients with available MRD results were enrolled. Subtypes included HR+/HER2- (n=21), HR+/HER2+ (n=22), HR-/HER2+ (n=10), and triple-negative breast cancer (TNBC, n=34). Clinical stage distribution was I (n=24), II (n=48), and III (n=15). Most patients presented with a single primary tumor (n=83); four had multiple primaries. Histological subtypes comprised invasive ductal carcinoma (n=80), invasive lobular carcinoma (n=6), and mucinous carcinoma (n=1). 734 plasma samples were analyzed, ranging from 1 to 15 timepoints per patient (median=10). Neoadjuvant treatments included chemotherapy (n=85), HER2-targeted therapy (n=33), and immunotherapy (n=37). Follow-up time ranged from 4 to 29 months (median=14). 44 patients had undergone surgery with documented pathologic response. Panels were successfully designed for 80 of 87 patients (92%). Baseline ctDNA, prior to any treatment initiation, was detected in 74 of 80 (92.5%) patients, with the following distribution of tumor fractions: >100 parts per million (PPM) (77.0%), 20-100 PPM (16.2%), and <20 PPM (6.8%). ctDNA levels showed a rapid decline across chemotherapy cycles; at cycle 2, fewer than half (45%) remained ctDNA+ (19% at <20 PPM), decreasing to 25% by cycle 4 (56% at 100 PPM and the rest at <20 PPM. All patients achieving pCR (n=22) were ctDNA- at surgery, while 28.6% (n=4) of patients with RD were ctDNA+ at surgery. Patients achieving pCR typically demonstrated early response with sustained ctDNA clearance, while persistent or re-emergent ctDNA was associated with RD. HER2+ and TNBC tumors demonstrated faster clearance compared to HR+ tumors. Conclusions Ultrasensitive, personalized ctDNA monitoring enables high-resolution tracking of neoadjuvant treatment response. Early ctDNA clearance was strongly associated with pCR, supporting its potential as a biomarker for treatment de-escalation, whereas persistent ctDNA positivity may identify patients at risk of RD. Together, these findings establish the feasibility and clinical relevance of integrating ultrasensitive ctDNA assays into real-time neoadjuvant treatment management.
Presentation numberPS1-13-19
Impact of Access to CGP-Guided Recommended Therapies on Overall Survival in Invasive Lobular and Ductal Carcinomas: Insights from theC-CAT Registry
Masaki Makita, tohoku university, sendai city, Japan
M. Makita; tohoku university, sendai city, Japan
Abstract Background/Purpose: The clinical benefit of accessing comprehensive genomic profiling (CGP)-guided recommended therapies in invasive lobular carcinoma (ILC) remains poorly defined.This study aimed to evaluate the impact of achieving CGP-guided recommended therapy on overall survival (OS) among breast cancer patients enrolled in the nationwide C-CAT registry. Methods: We retrospectively analyzed 7,363 breast cancer cases (IDC: 6,902; ILC: 461) registered in the C-CAT database between June 2019 and June 2025 who underwent CGP testing and provided consent for secondary use. Patients were stratified according to receipt of CGP-guided recommended therapy. Kaplan-Meier curves were generated and survival differences were tested by log-rank analysis. Results: In ILC, frequent alterations included CDH1 (309 cases), PIK3CA (278), and TP53 (182). Subtype distribution comprised HER2-positive (0.3%), luminal (73.7%), luminal/HER2 (2.2%), and triple-negative (17.7%). A total of 55 ILC patients (12%) achieved recommended therapy, most commonly with pembrolizumab monotherapy (n=11) or capivasertib-based regimens (n=10). These patients demonstrated significantly longer median OS compared with those not receiving recommended therapy (35 vs. 16 months; P=0.019). Conclusions: This large nationwide registry analysis demonstrates that access to CGP-guided recommended therapy is associated with a significant improvement in OS in ILC. Notably, despite limited availability of targetable alterations and trial opportunities in ILC, meaningful survival benefit was observed. These findings underscore the clinical importance of expanding access to CGP-driven recommended therapies for all breast cancer patients, and highlight the value of large-scale real-world evidence in rare subtypes such as ILC.
Presentation numberPS1-13-28
Integration of NIR-II Imaging and Molecular Profiling Identifies Subtypes of Metastatic Sentinel Lymph Nodes in Breast Cancer
Guo-Jun Zhang, The Breast Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Beijing University Cancer Hospital, Kunming, Yunnan, China
Y. zhu1, W. Chen1, J. chen1, L. zhang2, Z. Wu2, Z. Wei3, Q. Zhang3, X. Duan3, T. Guo1, S. Guan2, S. Song4, Z. Guo2, Y. Tang2, C. Yang4, C. Guo3, C. Peng3, G. Zhang2; 1School of Medicine,Xiamen University, Xiamen,Fujian, CHINA, 2The Breast Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Beijing University Cancer Hospital, Kunming, Yunnan, CHINA, 3Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, CHINA, 4Department of Pathology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Beijing University Cancer Hospital, Kunming, Yunnan, CHINA.
Introduction:Identification of metastatic sentinel lymph node (mSLN) is necessary for staging axillary and guiding following treatment in breast cancer patients. mSLNs exhibit significant heterogeneity in morphology and immune microenvironment, and might associate with the risk of recurrence/metastasis. Intraoperatively, frozen sections are utilized to assess SLN’s metastatic status, and to decide whether to perform axillary dissection or not. Previous studies primarily illustrated that near-infrared fluorescence (NIRF) imaging could visualize mSLNs in animal models. Thus, the purpose of this study is to investigate NIRF’s capability to stratify the subtypes of mSLNs, by combining with histopathological findings, and further to explore the mechanism underlying. Here, we report a TROP2-targeted second window of NIRF imaging approach for non-invasive, real-time identification of mSLNs subtypes. Materials and Methods:A novel NIR-II probe, ICG-SG, was synthesized by conjugating Sacituzumab Govitecan (SG) with ICG-Mal. In vivo NIRF imaging was applied to visualize mSLNs in mouse breast cancer model. In addition, mSLNs were stratified into distinct subtypes based on pathological features and mean fluorescence intensity (MFI). FFPE tissue sections from 345 invasive breast cancer patients were retrospectively analyzed, and histological subtypes of human mSLNs were assigned according to the classification established in the preclinical models. Moreover, three FFPE specimens per subtype underwent high-resolution spatial transcriptomics (Stereo-seq) and single-cell RNA sequencing (scRNA-seq) to map subtype-specific molecular features and mechanisms driving secondary lymph node metastasis. The association of subtypes with prognosis was also analyzed.Results and SummaryIn this study, we developed a TROP2-targeted probe ICG-SG for noninvasive, real-time detection of mSLNs in breast cancer. In both human- and mouse lines-derived models, we observed that ICG-SG exhibited significantly higher MFI than ICG-IgG in bilateral metastatic SLN models. In unilateral metastasis models with ICG-SG, mSLNs also showed markedly higher MFI than non-metastatic SLNs. Based on MFI and pathological features, mSLNs were classified into three subtypes: subcapsular (highest fluorescence), paracortical-medullary (intermediate, yet above normal), and replacement (negative fluorescence, below normal). Paracortical-medullary and replacement types had a higher risk of secondary LNs metastasis, which was also confirmed in a cohort of 345 patients, with odds 5.51- and 9.56-fold higher than subcapsular type, respectively. Whole-transcriptome analysis of representative mSLNs revealed distinct molecular and immune profiles across subtypes. Paracortical-medullary and replacement MSLNs exhibited aggressive tumor traits, impaired immune responses, and fibroblastic reticular cell-driven stromal remodeling, generating a pro-tumor microenvironment. In summary, mSLN subtypes intraoperatively and non-invasively determined predict non-sentinel lymph node metastasis and correlate closely with patient prognosis. The integrative imaging and molecular approach provides a framework for precise, real-time and noninvasive mSLN stratification and potential guidance for clinical decision-making.
Presentation numberPS1-13-24
Safety analysis of ASCENT-03, a phase 3 study of sacituzumab govitecan (SG) vs chemotherapy (chemo) for previously untreated advanced triple-negative breast cancer (TNBC) in patients (pts) who are not candidates for PD-(L)1 inhibitors (PD-[L]1i)
Sara Hurvitz, UW Medicine, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
S. Hurvitz1, A. Bardia2, S. M. Tolaney3, K. Punie4, C. Barrios5, A. Schneeweiss6, R. Hegg7, E. Tokunaga8, J. Sohn9, S. Kim10, S. Im11, Y. Park8, B. Rapoport12, D. Motola13, D. A. Yardley14, F. Dalenc15, M. Oliveira16, B. Pistilli17, G. Vidal18, S. Paluch-Shimon19, B. Xu20, T. Valdez21, D. Zhang21, S. Padman21, J. Cortés22; 1UW Medicine, Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 2David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, 3Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 4Oncology Center Antwerp, Ziekenhuis aan de Stroom, Antwerp, BELGIUM, 5Latin American Cooperative Oncology Group (LACOG), Porto Alegre, BRAZIL, 6Heidelberg University Hospital and German Cancer Research Center, Heidelberg, GERMANY, 7University of São Paulo, São Paulo, BRAZIL, 8National Hospital Organization Kyushu Cancer Center, Fukuoka, JAPAN, 9Yonsei Cancer Center, Seoul, KOREA, REPUBLIC OF, 10Asan Medical Center, University of Ulsan College of Medicine, Seoul, KOREA, REPUBLIC OF, 11Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, KOREA, REPUBLIC OF, 12The Medical Oncology Centre of Rosebank, Clinical and Translational Research Unit (CTRU), Faculty of Health Sciences, University of Pretoria, Pretoria, SOUTH AFRICA, 13Cryptex Investigación Clínica, Mexico City, MEXICO, 14Sarah Cannon Research Institute, Nashville, TN, 15Oncopole Claudius Regaud, IUCT-Oncopole, Toulouse, FRANCE, 16Vall d’Hebron University Hospital, Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, SPAIN, 17Gustave Roussy; IHU-National PRecISion Medicine Center in Oncology, Villejuif, FRANCE, 18West Cancer Center and Research Institute, Memphis, TN, 19Hadassah – Hebrew University Medical Center, Jerusalem District, ISRAEL, 20Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 21Gilead Sciences, Inc., Foster City, CA, 22International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Hospital Universitario Torrejón, Ribera Group,Universidad Europea de Madrid, Medica Scientia Innovation Research, Barcelona, SPAIN.
Background: In ASCENT-03 SG demonstrated statistically significant and clinically meaningful improvement in progression-free survival versus chemo for pts with previously untreated, locally advanced unresectable or metastatic TNBC who were not candidates for PD-(L)1i. We report the first in-depth safety analysis. Methods: Pts were randomized 1:1 to SG (10 mg/kg IV days 1 and 8 per 21-day cycle) or chemo (taxane or gemcitabine + carboplatin [G+C]) until disease progression or unacceptable toxicity. Exposure-adjusted incidence rates (EAIRs; number of pts with ≥ 1 specific treatment-emergent adverse event [TEAE] per pt-year of exposure) were measured in an exploratory analysis. Incidence, severity, time to onset, duration, and impact of supportive care measures for select TEAEs were assessed. Results: The safety set included 551 pts who received ≥1 dose of study drug (SG: 275; chemo: 276, 44% gemcitabine + carboplatin, 56% taxane). Pts received SG for median 8.3 months, taxane for 6.3 months, and G+C for 5.8 months. EAIRs, incidence, and time to onset/duration of specified TEAEs are shown in the Table. Grade ≥ 3 TEAEs occurred in 66% and 62% of pts treated with SG and chemo, respectively. The most common TEAEs with SG were neutropenia (68%), nausea (61%), and alopecia (55%) and with chemo were neutropenia (57%), anemia (50%), and fatigue (47%). Any-grade/grade ≥ 3 neutropenia was reported in 68%/45% of pts with SG and 57%/41% with chemo. EAIR of neutropenia was comparable between treatment groups. Neutropenia leading to dose reduction occurred in 20% of pts in both groups; 1 case in the SG group and 3 in the chemo group led to treatment discontinuation. Granulocyte-colony stimulating factor (G-CSF) was used at any time in 59% and 38% of patients and as primary prophylaxis in 20% and 10% pts in the SG and chemo groups, respectively. Any-grade/grade ≥ 3 diarrhea was reported in 54%/9% of pts with SG and 20%/1% with chemo. EAIR of diarrhea was higher in the SG group than the chemo group. Diarrhea leading to dose reduction occurred in 5% of pts in the SG group and 1% in the chemo group; 1 case in the SG group led to treatment discontinuation. Antidiarrheal treatment was given to 72% of pts who experienced diarrhea in the SG group and 44% in the chemo group. Conclusions: In pts with previously untreated advanced TNBC who were not candidates for PD-(L)1i, the SG group had a similar rate of grade ≥ 3 TEAEs and lower rates of TEAEs leading to dose reduction and discontinuation. Accounting for the longer duration of treatment with SG, EAIR of neutropenia was similar between treatment groups, while diarrhea remained higher with SG. It is important to manage these AEs proactively according to established guidelines. Taken together, the overall safety profile was consistent with the known safety profile for SG.
| EAIRs EAIR (95% CI) | SG (n = 275) | Chemo (n = 276) | Difference (95% CI) |
| Any TEAEs | 40.21 (35.58, 45.27) | 21.66 (19.14, 24.40) | 18.55 (13.17, 24.20) |
| Grade ≥ 3 TEAEs | 1.85 (1.59, 2.14) | 2.02 (1.73, 2.35) | −0.18 (−0.59, 0.23) |
| Serious TEAEs | 0.40 (0.31, 0.51) | 0.49 (0.38, 0.63) | −0.09 (−0.25, 0.06) |
| TEAEs leading to dose interruption | 2.05 (1.76, 2.37) | 2.14 (1.83, 2.48) | −0.09 (−0.54, 0.36) |
| TEAEs leading to dose reduction | 0.68 (0.55, 0.82) | 1.15 (0.96, 1.37) | −0.48 (−0.73, −0.23) |
| TEAEs leading to treatment discontinuation | 0.05 (0.02, 0.09) | 0.22 (0.15, 0.30) | −0.17 (−0.26, −0.09) |
| TEAEs leading to death | 0.03 (0.01, 0.07) | 0.01 (0.00, 0.04) | 0.03 (-0.01, 0.07) |
| Neutropenia | 2.48 (2.13, 2.87) | 2.01 (1.71, 2.35) | 0.47 (−0.02, 0.96) |
| Diarrhea | 1.42 (1.20, 1.67) | 0.41 (0.31, 0.54) | 1.01 (0.76, 1.28) |
| Incidence of TEAEs n (%) | 273 (99) | 269 (97) | – |
| Grade ≥3 | 181 (66) | 171 (62) | – |
| Any-grade leading to dose reduction | 101 (37) | 124 (45) | – |
| Any-grade leading to dose interruption | 181 (66) | 171 (62) | – |
| Any-grade leading to dose discontinuation | 10 (4) | 33 (12) | – |
| TEAEs of interest for SG Median duration (range), days | – | ||
| Neutropenia,a any grade Time to onset of first occurrenceb | 22 (6, 274) n = 187 | 22 (6, 406) n = 158 | – |
| Neutropenia,a any grade Durationc | 9 (2, 49) n=183 | 14 (1, 179) n=155 | – |
| Neutropenia,a grade ≥ 3 Time to onset of first occurrenceb | 22 (7, 720) n = 124 | 29 (7, 295) n = 113 | – |
| Neutropenia,a grade ≥ 3 Durationd | 8 (1, 36) n = 122 | 8 (1, 25) n = 112 | – |
| Diarrhea, any grade Time to onset of first occurrenceb | 13 (1, 427) n = 148 | 26 (1, 296) n = 55 | – |
| Diarrhea, any grade Durationc | 6 (1, 273) n = 130 | 6 (1, 370) n = 48 | – |
| Diarrhea, grade ≥3 Time to onset of first occurrenceb | 67 (6, 356) n = 25 | 210 (110, 310) n = 2 | – |
| Diarrhea, grade ≥3 Durationd | 6 (1, 18) n = 24 | 1 (1, 1) n = 2 | – |
| aNeutropenia includes preferred terms of neutrophil count decreased, neutropenia and febrile neutropenia. bTime to onset of first event is calculated as time from the first dose date of any study drug to the onset date of first event. cDuration is the median duration among multiple episodes of any grade (the end date of event of interest – the onset date of event of interest + 1 day for each episode). | |||
Presentation numberPS1-13-03
Achieving Disparity Reduction in Precision Oncology: A Population-Based Simulation Study
Jack Zeineh, Icahn School of Medicine, New York, NY
J. Zeineh1, R. DeAngel2, E. Grullon2, T. J. Lawton3, K. J. Bloom4; 1Icahn School of Medicine, New York, NY, 2ScopeData, Corona del Mar, CA, 3University of California, Irvine, Irvine, CA, 4Nucleai, Chicago, IL
African-American women with breast cancer receive biomarker testing at lower rates than White women, despite similar overall frequencies of actionable genetic alterations—a gap that contributes to about a 40% higher mortality rate. AI-powered precision oncology platforms, particularly when paired with culturally tailored interventions, hold promise for reducing this inequity, though their race-specific impact has yet to be fully quantified. We modeled AI-enabled clinical decision support combined with equity-focused strategies in a population-based simulation integrating TCGA molecular data with SEER demographics (N=10,000). Baseline testing rates reflected recent literature (2020-2024): BRCA (AA 21.4% vs W 47.9%), PIK3CA (28.6% vs 50.5%), ESR1 (16.6% vs 29.0%). Intervention effects were derived from systematic reviews of patient navigation, cultural competency training, community health workers, and tailored educational materials. Realistic ceilings were applied (85% maximum testing, 70% uptake, 90% sustainability). Economic modeling included program costs ($580/patient). Culturally tailored AI interventions improved testing among African American women. Baseline testing rates showed significant post-intervention improvement: BRCA 21.4% to 32.7% (+53%), PIK3CA 28.6% to 58.6% (+105%), ESR1 16.6% to 42.5% (+156%). Disparity gaps narrowed by 14-50%, with African American women achieving 2-4x greater improvements than White women. The model projected prevention of 4,763 non-concordant treatments and 714 deaths, with African American women representing 21% of lives saved despite being 10.5% of the population. All interventions were highly cost-effective (p<0.0001). Among Black patients, modeled cost per QALY gained was $642- 156x more favorable than the $100,000/QALY benchmark, with results robust to +50% parameter variation. This equity-focused, AI-enabled framework demonstrates the potential to close up to half of existing biomarker testing gaps in breast cancer. By doing so, this approach may prevent thousands of non-concordant treatments and hundreds of deaths while achieving extraordinary cost-effectiveness far beyond accepted thresholds. Importantly, these findings reflect analyses only recently completed and provide new, timely evidence of how equity-focused AI interventions can address one of the most urgent challenges in breast cancer care, racial disparity in biomarker testing and patient outcomes.
Presentation numberPS1-13-04
Fairness-constrained logistic regression achieves superior performance and racial equity in breast cancer survival prediction
Jack Zeineh, Icahn School of Medicine, New York, NY
J. Zeineh1, R. DeAngel2, E. Grullon2, T. J. Lawton3, K. J. Bloom4; 1Icahn School of Medicine, New York, NY, 2ScopeData, Corona Del Mar, CA, 3University of California, Irvine, Irvine, CA, 4Nucleai, Chicago, IL
Machine learning models for clinical prediction often exhibit racial bias, resulting in inferior predictive accuracy and potentially poorer outcomes for African American (AA) breast cancer patients compared to non-AA patients. Such bias can result in delayed diagnosis, inappropriate treatment recommendations, and reduced access to clinical trials, thereby compounding existing survival disparities. Standard optimization approaches prioritize overall performance, risking the amplification of these inequities. We hypothesized that fairness-constrained hyperparameter optimization could enhance overall predictive accuracy while improving equity in AA-specific breast cancer survival prediction. We conducted a controlled study comparing standard logistic regression with a fairness-constrained version for 5-year breast cancer survival prediction using TCGA data (N=1,000, 18.6% AA patients, 70.8% survival rate). The standard model optimized overall predictive accuracy, while the fairness-constrained model also aimed to reduce racial disparities in predictions. Both approaches used identical 10-fold stratified cross-validation, enabling direct comparison. Primary endpoints were overall AUC and AA-specific AUC. The secondary endpoint was AUC difference between non-AA and AA patients as a disparity metric. Statistical analysis used paired t-tests with bootstrap confidence intervals (n=1,000 resamples) and effect size calculation via Cohen’s d. Fairness-constrained logistic regression significantly improved clinical performance and racial equity compared to standard optimization. Overall AUC increased from 0.634 (95% CI: 0.582-0.664) in the control group to 0.641 (95% CI: 0.592-0.669) with fairness constraints, yielding a +0.007 AUC improvement (95% CI: +0.002 to +0.013, p=0.018, Cohen’s d=0.917). AA-specific performance demonstrated even greater enhancement, with AUC increasing from 0.610 (95% CI: 0.518-0.687) to 0.632 (95% CI: 0.528-0.713), representing a +0.021 AUC improvement (95% CI: +0.003 to +0.040, p=0.027, Cohen’s d=0.832). Most importantly, racial disparity was reduced, with AUC difference between non-AA and AA patients improving from +0.029 (indicating disparity favoring non-AA patients) to -0.012 (slight advantage for AA patients), representing a 41-point equity improvement. Non-AA performance was maintained at 0.639 AUC (95% CI: 0.596-0.678) with no significant change (p=0.276), demonstrating that equity gains can be achieved without compromising care for the majority population. Incorporating fairness-constrained hyperparameter optimization into predictive models can simultaneously improve overall accuracy and reduce racial disparities in breast cancer survival prediction. These findings, based on analyses only recently completed, provide new and timely evidence that equitable machine learning is both feasible and clinically advantageous. By demonstrating that fairness constraints can improve African American-specific outcomes without compromising performance in the majority population, this work represents a potential paradigm shift toward more equitable, precision-guided oncology care.
Presentation numberPS1-13-20
Functional precision oncology for metastatic breast cancer (FORESEE): a feasibility trial. Final Results.
Christos Vaklavas, Huntsman Cancer Institute, Salt Lake City, UT
C. Vaklavas, L. Zhao, C.-H. Yang, S. D. Scherer, X. Huang, S. S. Buys, M. Wei, P. Moos, G. Marth, B. E. Welm, A. L. Welm; Huntsman Cancer Institute, Salt Lake City, UT
Background: Precision oncology entails identification of disease driver mutations and administering drugs that target mutated proteins. However, many breast tumors do not harbor actionable mutations. Even if an actionable mutation is identified, it is unknown if inhibition of the target will lead to clinical response. In metastatic breast cancer, mutation testing alone is insufficiently informative to make optimal treatment selections. FORESEE was a pilot study to assess the feasibility of genomic characterization and functional drug response profiling on patient-derived organoids (PDOs) to prospectively inform therapy selection in metastatic breast cancer (MBC). Methods: Women with newly diagnosed metastatic triple negative (TNBC) or hormone receptor positive/Her2 negative (HR+/Her2-) breast cancer who had exhausted endocrine therapy options were eligible. Patients had to have evaluable disease amenable to fresh biopsy. Tumor tissue was used for single cell sequencing and PDO establishment. PDOs were subjected to screening of drugs approved by the FDA or available to the patient in active clinical trials. Circulating tumor (ct) DNA was also collected. After the biopsy, patients began therapy at the discretion of the treating physician. The results from ct and tumor tissue DNA sequencing were compiled along with results from PDO drug screening in a single report that was discussed with the treating physician. The subsequent line of therapy could align with the recommendation of the report (“informed” therapy) or not (“uninformed therapy”); physicians were not required to follow the recommendations. Response assessments were conducted until documented radiographic or clinical progression. The primary objective was to assess the feasibility of comprehensive genomic characterization and functional drug screening in a clinically relevant timeframe (12 weeks). The efficacy of treatment decisions informed by functional genomic assays constituted an exploratory objective. The trial was registered at clinicaltrials.gov (NCT04450706). Results: 15 patients were enrolled (TNBC, n = 7; HR+/Her2-, n = 8; recurrent, n = 11; de novo metastatic, n = 4). Median age was 55.5 years, median number of prior therapies in the HR+/Her2- patients was 3.5. Tumor biopsies were collected successfully from 13 patients; 10 had adequate tumor content (≥20% tumor content). PDOs were successfully established in 4/10 cases (TNBC, n = 2; HR+/Her2-, n = 2). Drug profiling was successfully conducted in all 4. Functional and genomic results were returned within 12 weeks and informed subsequent therapy selection in all cases. In 3 of 4 cases, genomic testing found no mutations matching FDA approved therapies. In 1 case, actionable mutations in PIK3CA and ESR1 were identified; the treatment choice was arbitrated with PDO drug screening. The relapse-free interval (RFI) with the informed line of therapy did not exceed the RFI with the immediate prior line of therapy. In 2 cases, the informed therapy constituted the last therapy that the patients received. Conclusion: The primary endpoint that PDO-based functional drug screening and genomic characterization will identify clinically actionable outcomes in >46% of patients was not met. Although therapy selection informed by functional drug screening can be clinically meaningful, limitations of this approach included the need for a tumor biopsy with adequate tumor content, low PDO establishment rate, and limited efficacy of approved drugs in advanced MBC. Methodological refinements and generation of PDOs earlier in the natural history of the disease to inform therapy selection may overcome these limitations.
Presentation numberPS1-13-05
Reflect: radiomic evaluation for the localization of the excision cavity using tomography
Cesar Federico Lorenzo, RT International Institute, Montevideo, Uruguay
C. F. Lorenzo1, A. Rosich1, E. Sanchez2, G. Silva2, X. Miranda2, S. Guerreros2, C. Bentancur2, E. Rivero2, P. Fernandez2, S. Roldan2, J. Lell1, M. Giordano2, R. Brid2, G. Limachi2, A. Notejane2, L. Ricagni2; 1RT International Institute, Montevideo, URUGUAY, 2Unidad Academica de Radioterapia, Montevideo, URUGUAY.
Introduction: In breast-conserving radiotherapy planning, accurate delineation of the surgical cavity is critical but challenging due to the poor contrast between the cavity and the surrounding breast tissue on CT. Manual contouring remains the standard, but it is time-consuming and subject to interobserver variability. Automated segmentation using deep learning could improve both efficiency and consistency. Methods: We retrospectively analyzed 63 patients with axial CT scans (1.5 mm slice thickness) after lumpectomy. Data were split into training (60%) and validation (40%) sets. We implemented a 3D U-Net with attention blocks and deep supervision, trained with a combined loss and extensive data augmentation. Voxels corresponding to surgical clips were excluded during training to reduce the impact of metal artifacts. As a baseline, a radiomics-based method using the intensity thresholds and texture features was developed. Performance was evaluated against manual contours (ground truth) using Dice similarity coefficient (DSC) and the 95th percentile Hausdorff distance (HD95). Results: The deep learning model achieved a mean DSC of 0.80 ± 0.07 (range: 0.70-0.90), significantly higher than the radiomics approach (0.68 ± 0.09; p = 0.01). The mean HD95 was 9.8 mm, with outliers up to 15 mm, compared to 13.5 mm for the radiomics baseline (p = 0.04). Qualitatively, the neural network performed better in irregular and poorly defined cavities, showing closer agreement with expert delineations.Conclusions: The achieved Dice scores (~0.8) are consistent with state-of-the-art reports. Although further optimization and larger multicenter validation are required, the method shows clear potential to accelerate radiotherapy planning and reduce interobserver variability.
Presentation numberPS1-13-02
Age-dependent mammary tumorigenesis in mice driven by the PIK3CA H1047R oncogene
Thangarajeswari Mohan, UT Health San Antonio, San Antonio, TX
T. Mohan, J. Mackay, M. McLaughlin, Y. Li, A. Nazarullah, L.-Z. Sun; UT Health San Antonio, San Antonio, TX
The incidence of breast cancer increases with age. Hormone receptor positive (HR+) and HER2− tumor is the most prevalent subtype in older women. Approximately 70% of early-stage breast cancers (EBC) are HR+/HER2−, frequently harboring the PIK3CA H1047R mutation, which drives heterogeneous mammary carcinomas. Understanding how PIK3CA H1047R mutant interacts with the aging microenvironment is important for elucidating age-related susceptibility and may enlighten age-specific therapeutic strategies. In this study, we investigated the oncogenic potential of human PIK3CA H1047R across different age groups of FVB/NJ mice (young: 2 mo, n=10; middle-aged: 14 mo, n=8; aged: 22 mo, n=7) following intraductal injection of lentivirus-mediated PIK3CA H1047R expression. Mice were monitored for up to 6 months, and time to palpable tumor formation was used to generate tumor-free survival curves. qPCR was performed to assess PIK3CA H1047R genomic integration and gene expression. Ductal morphology and histopathology were evaluated using carmine whole-mount and hematoxylin and eosin staining. Morphometric analysis was performed using BD FACS Discover S8 Cell Sorter. High-dimensional flowcytometric analysis in FlowJo (t-SNE, FlowSOM, Cluster Explorer) was used to identify and characterize cell populations most susceptible to oncogenic transformation. Our results show that middle-aged and aged mice had significantly higher tumor incidence and developed tumors more rapidly than young mice, supported by higher PIK3CA H1047R integration and expression. Morphometric analysis revealed that luminal cells (CD49flowCD24high) expressed more PIK3CA H1047R than basal cells (CD49fhighCD24low). Importantly, epithelial cells from middle-aged and aged mice showed enrichment of a novel CD24⁺CD49f⁺ cell population with higher PIK3CA expression. The origin of these cells remains unclear and warrants further investigation. Together, these findings indicate that aging is not merely a risk factor but actively modifies PIK3CA-driven oncogenic signaling. These age-associated alterations in mammary epithelial cell populations may inform development of targeted therapies for older patients with HR+/HER2− breast cancer. A limitation of this study is the use of a murine intraductal model, which may not fully capture human breast cancer biology. Further studies incorporating stromal and immune components are needed to comprehensively understand how aging influences oncogenic PIK3CA signaling.
Presentation numberPS1-13-13
Clinical validation of an Artificial Intelligence digital pathology-based prognostic test to predict risk of recurrence using biopsy specimens from patients with invasive breast cancer
Michael Donovan, Icahn School of Medicine at Mount Sinai, PreciseDx, Miami, FL
G. Fernandez1, S. Vaisman2, A. Sainath Madduri2, R. Scott2, M. Prastawa2, X. Zhang2, M. Donovan3; 1Icahn School of Medicine at Mount Sinai, PreciseDx, New York, NY, 2PreciseDx, New York, NY, 3Icahn School of Medicine at Mount Sinai, PreciseDx, Miami, FL
Genomic testing is the standard for guiding adjuvant treatment for patients with early-stage HR+/HER2-IBC. Prognostic testing strategies using the biopsy specimen are needed to individualize patient management in the neoadjuvant and possibly adjuvant setting. The current objective was to clinically validate the PreciseBreast Biopsy test (PDxBRBx) which includes the patient’s age and morphologic features, derived from a standard H&E stained IBC biopsy slide, to predict recurrence risk earlier in the treatment planning and management process. Methods 1788 patients (pts) with IBC from the Mount Sinai Health System were identified (2004-2016) with median follow-up of 6 years (yrs). 60%-40% balanced training (surgical) and validation (biopsy) cohorts were generated. The PreciseBreast Biopsy test was performed using H&E stained slides that were imaged (40X) with a Philips Ultra-Fast scanner. Images were deconstructed to morphologic analytes using an AI-enabled platform to quantify tumor cell and tissue architectural features. Age is the only clinical factor available at the time of diagnostic biopsy and is algorithmically combined with 8 morphologic features (AI-grade model) that stratifies pts into low or high risk of recurrence. Risk stratification for invasive breast cancer free survival (IBCFS) was assessed by concordance index (C-index), area under the curve (AUC), Kaplan-Meier analysis, hazards ratios (HR), sensitivity (Se), specificity (Sp), negative predictive value (NPV), and positive predictive value (PPV). Matched excisional specimens from the biopsy test cohort were evaluated with the PreciseBreast excision test for risk score correlation using Cohen’s Kappa and Odds Ratio. Results In the training surgical cohort (n=1012): median age 59 yrs, 80% grade 2/3, 93% stage 1/2, 77% pN0, 23% pN1-3, ER+(87%)/PR+(81%)/HER2-(88%), 82 (8%) triple negative, 91 (9%) HER2+, with a 14% event rate and median 6-yrs of follow-up. PDxBRBx (Age + AI-grade) yielded a C-index of 0.72 (95% CI, 0.69-0.78) vs age C-index 0.61 (95% CI, 0.55 – 0.66) vs AI-grade 0.69 (95% CI, 0.64 – 0.73). Applying a risk score cut-off of 73 (scale 0-100) stratified patients as low- or high-risk, respectively, with a HR of 4.49 (95% CI 2.96 – 6.8, p<0.001) for predicting IBCFS, with Se 0.70, Sp 0.67, NPV 0.93, and PPV 0.25. In the biopsy validation cohort (n=776): median age 60 yrs, 43% Grade 2, ER+ (87%)/PR+(81%)/HER2-(88%), 56 (7%) triple negative, 97 (12.5%) HER2+ with a 14% event rate. PDxBRBx yielded a C-index of 0.73 (95% CI, 0.69-0.78) vs age 0.64 (95% CI, 0.57-0.70) vs AI-grade 0.69 (95% CI, 0.64-0.74). Patients stratified by a risk score of 73 had a HR of 4.49 (95% CI, 2.96-6.8, p<0.0001) for predicting IBCFS within 6 years, with Se 0.70, Sp 0.67, NPV 0.93, and PPV 0.25. The HR for AI-grade was 2.45 (95% CI 1-68-3.59, p<0.0001). All individual morphologic biopsy features including mitotic figure quantitation, nuclear pleomorphism, tumor-stromal ratio, lymphocytic content, and tumor architecture were statistically significant predictors of event risk (all p<0.05, most p<0.001). Comparison of biopsy (n=776) and matched excisional PreciseBreast risk scores showed substantial agreement, with Cohen’s κ = 0.57 (95% CI 0.51-0.62, p<0.0001) and an odds ratio of 31.4 (95% CI 18.8-55.1, p<0.0001). Conclusion We clinically validated a breast biopsy digital pathology-based AI prognostic test, PreciseBreast Biopsy, which successfully predicted IBCFS within 6 years. The test is designed to assist in the accurate characterization of clinical pathologic risk and patient management at the time of diagnosis. Additional studies in the neoadjuvant and possibly adjuvant settings will further refine the impact of these results on treatment selection.
Presentation numberPS1-13-29
Carcinoembryonic antigen (CEA), Cancer Antigen 15.3 (CA 15.3), and 18-FDG-PET in Early Breast Cancer Follow-up: Findings from the KRONOS Trial
Claudio Zamagni, IRCCS Azienda Ospedaliero-universitaria di Bologna, Bologna, Italy
C. Zamagni1, R. Wirtz2, V. Torri3, D. Sartori4, M. Carapelle1, L. Vivona1, P. Stieber5, J. Hubner6, M. Bergamo7, L. Tondulli8, C. Pizzirani1, S. Coccato4, N. Cacciari1, A. Baldoni4, A. Bernardi1, T. Dalsass6, S. Quercia1, S. Prader7, E. Haspinger8, D. Rubino1, A. Mandrioli1, M. Cubelli1, F. Abbati1, M. Massucci1, R. Pagani1, S. Fanti9, M. Gion10; 1IRCCS Azienda Ospedaliero-universitaria di Bologna, Bologna, ITALY, 2Stratifyer Molecular Pathology and PONS-S Stiftung, Cologne and Munich, GERMANY, 3Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Milan, ITALY, 4UOC Oncologia AULSS3 Serenissima, Mirano, ITALY, 5PONS-S Stiftung, Munich, GERMANY, 6Dept. Gynecology and Obstetrics Ospedale di Merano, Merano-Meran, ITALY, 7Dept. Gynecology and Obstetrics Hospital of Bressanone (SABES-ASDAA), Bressanone-Brixen, ITALY, 8Oncologia Medica Ospedale di Bolzano, Bolzano-Bozen, ITALY, 9IRCCS Azienda Ospedaliero-universitaria di Bologna and Bologna University, Bologna, ITALY, 10AULSS 3 Veneto, Venezia, ITALY.
Background: Current breast cancer (BC) follow-up guidelines for asymptomatic patients recommend only annual mammography and periodic physical exams, based on two trials published in 1994 (JAMA vol. 271). No subsequent randomized controlled trials (RCTs) have evaluated alternative strategies. Advances in BC biology, treatments, and diagnostic tools like 18-FDG-PET have transformed the landscape, prompting a need to reassess surveillance protocols. The role of serum tumor markers (CEA, CA 15.3) and 18-FDG-PET have never been validated in prospective RCTs. Study Design: The KRONOS trial for the first time over 30 years later the above cited RCTs, compares standard follow-up (annual mammography and physical exam; control arm) with an intensified approach (standard plus quarterly serum CEA and CA 15.3, with 18-FDG-PET performed if markers rise critically; experimental arm). The primary endpoint assesses whether intensified surveillance detects recurrences earlier, measured by the difference in restricted mean survival time (RMST). Assuming a 20% five-year recurrence, a 3-month RMST reduction (diagnostic lead time ~10 months) was targeted. If early diagnosis of metastases is demonstrated, the impact on survival will be evaluated as the final endpoint. Secondary endpoints include marker predictive value, PET accuracy, and quality of life. Patients and Methods: Eligible patients had stage I-III BC after adequate surgery, only very low-risk cases and rare histologic types were excluded. The study included two cohorts: newly starting follow-up (cohort 1) and those completing five years without relapse (cohort 2). Participants were randomized 1:1, stratified by nodal status, HER2, and ER status. Follow-up was planned for at least 10 years after surgery. The mean diagnostic anticipation achievable in the experimental arm was assessed by calculating the difference in RMST between the 2 arms and testing such a difference non-parametrically by means of bootstrap resampling. Results: Between October 2014 and November 2021,1507 patients were randomized: 742 to control arm, 765 to experimental arm. Median age was 56 years; 52% stage I, 34.9% stage II, 13.1% stage III. Tumor biology: 64.8% ER-positive/HER2-negative, 25.8% HER2-positive, 9.4% triple-negative. Median follow-up was 7 years; 130 recurrences (8.6%) occurred—101 distant metastases (6.7%) and 29 locoregional relapses (1.9%). The 10-year relapse-free survival was 88.5% in the experimental arm vs. 89.2% in the control arm. Additionally, 89 patients (5.9%) developed new primary cancers, and 20 (1.3%) died without recurrence. The RMST over 10 years was 7.7 years (experimental) vs. 8.3 years (control), indicating an anticipatory benefit of 7.4 months (95% CI -3.1-13.4). Overall survival was similar, with 97 deaths (6.4%) and no significant difference between groups. Subgroups analysis by cohort, stage and biological subtypes will be also presented at the symposium. Conclusions: The 7-year recurrence rate (~8.6%) was lower than expected. Intensified follow-up did not improve overall survival in this unselected BC population. These findings support current standard surveillance, with no evidence of benefit from routine use of serum markers or 18-FDG-PET in asymptomatic patients.
Presentation numberPS1-13-06
Personalised Therapy in Triple Negative breast cancer (TNBC), evaluating predictive performance of Bayesian AI Digital Twins.
Uzma S Asghar, Guys and St Thomas NHS Foundation, London, United Kingdom
N. Somaiah1, R. Kow1, S. Anbalagan1, I. Roxanis1, R. Chauhan1, S. Kannambath1, L. Gothard1, S. Nimalasena2, J. Noble2, A. Johns3, E. Misirlioglu3, B. Plummer4, A. Biankin5, I. Babina4, M. Griffiths4, U. S. Asghar6; 1The Institute of Cancer Research, London, UNITED KINGDOM, 2The Royal Marsden NHS Foundation, London, UNITED KINGDOM, 3Concr, Brisbane, AUSTRALIA, 4Concr, London, UNITED KINGDOM, 5University of Glasgow, School of Cancer Sciences, Glasgow, UNITED KINGDOM, 6Guys and St Thomas NHS Foundation, London, UNITED KINGDOM.
Background: Despite multiple therapeutic strategies, approximately 40% of women with stage 2-3 Triple Negative breast (TNBC) cancer die. Biomarkers are sparse and treatment related toxicities can be significant, especially when drugs fail. Objective: Evaluate FarrSight®-Twin, a predictive tool for treatment response in TNBC based on integrating multi-modal clinico-pathological and genomic data, and assess its ability to prospectively identify patients unlikely to achieve a pathological complete response (pCR) and benefit less from neoadjuvant chemotherapy. Methods: VISION, an observational retrospective clinical study (n=200 RMH, UK) analysed the performance of the predictive algorithm FarrSight®-Twin. Study population included stage 2-3 TNBC, treated with neoadjuvant chemotherapy+/- immunotherapy (IO). Participants were recruited into Arm A (pCR) or Arm B if they had residual disease (non-pCR). Digital or virtual twins of individuals were created using their clinical data, cancer stage and tumour molecular data (WES (n=66), RNA seq n=(43), WES + RNAseq (n=42)) from an FFPE diagnostic biopsy. Treatment response and overall survival were predicted for each individual. Performance and accuracy were assessed by comparing predictions against real outcomes. Results: In this pre-planned analysis, 34 women were recruited into Arm A and 60 into Arm B with a median age of 51 (24-77yrs). 87% stage 2 (n=82) and 13% stage 3 (n=12). Neoadjuvant treatments administered were: anthracycline-taxane (33%), anthracycline-taxane-platinum (34%) or anthracycline-taxane-platinum-IO (21%) or Other. pCR was used to assess treatment response. pCR rates were highest for anthracycline-taxane-platinum-IO (50%). Similar pCR rates were reported for anthracycline-taxane (35%) and anthracycline-taxane-platinum regimens (31%). “Other” regimens consisting of de-escalated regimens following on treat toxicities and these women had pCR rates of 27%. Overall, 95% of women were alive at the 1 year time point (1 death; n=4 not reached), which dropped to 73% (7 deaths; n=19 not reached) alive for the study population. In the first interim analyses we present FarrSight®-Twin accuracy for predicting pCR using the leave one out approach. Following inputs were used: age, tumour type, clinical tumour size (cT), clinical nodal stage (cN) and molecular data from a diagnostic FFPE breast biopsy.
Conclusion:FarrSight®-Twin integrates molecular data from routine diagnostic FFPE samples with limited clinical information, making it both scalable and suitable for incorporation into standard care pathways. In stage II-III TNBC, it accurately predicts an individual patient’s probability of non-response or suboptimal response (i.e., non-pCR) to neoadjuvant chemotherapy, an outcome observed in 50-70% of this population.
Presentation numberPS1-13-21
Multi-layered Inhibition of Metastasis by Targeting TACC3 in Breast Cancer with Centrosome Amplification
Ozgur Sahin, Medical University of South Carolina, Charleston, SC
O. Saatci1, M. Gedik1, K. Calisir1, B. Cerci1, B. Ulukan Altun1, O. Sener Sahin1, R. Hull1, L. Ball1, E. Hill1, C. Lim2, S. Vempati2, B. Caliskan3, E. Banoglu3, I. Chatzistamou4, O. Sahin1; 1Medical University of South Carolina, Charleston, SC, 2A2A Pharmaceuticals, New York, NY, 3Gazi University, Ankara, Turkey, 4University of South Carolina, Columbia, SC
Metastasis is the primary cause of cancer-associated mortality, accounting for about 90% of cancer deaths. Cell migration is the pivotal step in metastasis that enables cancer cells disseminate out of a primary tumor to distant organs. Cell polarization is a fundamental cellular process, allowing cancer cells gain directionality that is critical for migration. Centrosomes direct cell polarity via promoting microtubule assembly and elongation. However, little is known about the molecular mechanisms and drivers of cancer cell polarity which would ultimately provide therapeutic targets to inhibit the deadly metastasis. Here, we found that the mutifunctional adaptor protein TACC3 promotes cell polarity, migration, and invasion in breast cancer cells with amplified centrosomes (CA). We showed that higher TACC3 levels are strongly associated with worse metastasis-free survival in breast cancer patients with CA. By integrating transcriptomics, proximity labeling (APEX2-TACC3) coupled to mass spectrometry, siRNA screening, and functional assays, we demonstrated that TACC3 promotes cancer cell migration and invasion by multilayered regulation of microtubule assembly, actin cytoskeleton, and vesicle trafficking. In addition, we provide evidence that targeting TACC3 via a first-in-class clinically tested small-molecule inhibitor or CRISPR-Cas9 mediated knockout significantly reduces cell migration and invasion. Furthermore, TACC3 inhibition reduces the number of circulating tumor cells and spontaneous metastasis to lungs in the MMTV-PyMT transgenic mouse model and lung-tropic breast cancer xenografts without any toxicity. Overall, our findings demonstrate the potential for TACC3 as a therapeutic target to inhibit metastasis in breast cancer with CA. This work offers new insights into the molecular underpinnings of TACC3’s role in metastatic breast cancer, particularly in CA-driven tumors and underscores its therapeutic relevance.
Presentation numberPS1-13-25
Preliminary data from a global multicohort Phase2 randomized trial of pumitamig (PD-L1 × VEGF-A bsAb) + chemotherapy for 1L/2L+ locally advanced/metastatic TNBC
Peter Schmid, St Bartholomew’s Hospital – Barts Health NHS Trust, London, United Kingdom
P. Schmid1, A. Williams2, S. Aksoy3, M. L. Telli4, S. Loi5, E. Papadimitraki6, G. Vidal7, Ö. Ateş8, J. Vijayakumar9, L. Pan10, S. Koseoglu10, C. Dalle Fratte11, C. Pütter11, D. Akahara10, C.-N. Gann11, P. Rietschel10, C. Brus10, Ö. Türeci11, U. Şahin11; 1St Bartholomew’s Hospital – Barts Health NHS Trust, London, United Kingdom, 2Sarah Cannon Research Institute, London, United Kingdom, 3Hacettepe Universitesi Tip Fakultesi, Ankara, Turkey, 4Stanford University School of Medicine, Palo Alto, CA, 5Peter MacCallum Cancer Centre, Melbourne, Australia, 6University College London Hospitals NHS Foundation Trust, London, United Kingdom, 7The West Clinic, P.C. d b a West Cancer Center, Germantown, TN, 8Sbu Dr. A.Y. Ankara Onkoloji SUAM, Ankara, Turkey, 9Healthpartners Cancer Care, St Louis Park, MN, 10BioNTech US Inc., Cambridge, MA, 11BioNTech SE, Mainz, Germany
Background: Pumitamig (BNT327/BMS986545) is an investigational bispecific antibody targeting PD-L1 and VEGF-A, designed to restore effector T-cell function through PD-L1 binding, while co-localizing VEGF-A neutralization to the tumor microenvironment. Pumitamig + nab-paclitaxel showed encouraging activity in a Phase 1b/2 trial of 1L locally advanced or metastatic triple-negative breast cancer (LA/mTNBC) in China (PS3-08, SABCS 2024). Herein we present the first pumitamig data from a global 1L/2L+ LA/mTNBC population. Methods: In this global Phase 2, randomized, open-label, multicohort trial (NCT06449222), Cohort 1 enrolled patients (pts) with 1L/2L+ LA/mTNBC (50% cap of 2L+ TNBC) who received pumitamig (15 or 20 mg/kg IV Q2W) + nab-paclitaxel until disease progression/unacceptable toxicity. In Cohort 2, pts received the flat dose equivalent of 20 mg/kg pumitamig IV: Arm 1: 1400 mg Q2W + paclitaxel; Arm 2: 2000 mg Q3W + gemcitabine + carboplatin; Arm 3: 2000 mg Q3W + eribulin. Primary endpoints were efficacy (ORR per investigator [RECIST 1.1], best percentage change in tumor size and early tumor shrinkage) and safety (NCI CTCAE v5.0). FoundationOne®Liquid CDx analysis estimated circulating tumor DNA (ctDNA) levels. Results: At cutoff (13 Aug 2025), 69 pts with 1L/2L+ TNBC were enrolled. Median duration of treatment (Tx) was 27.6 wks (range 2.0-41.0) with 27/40 pts still on Tx (Cohort 1) and 8.0 wks (range 3.0-20.4) with all pts on Tx (Cohort 2). In Cohort 1 (enrollment complete, median age: 57.1 yrs; 1L: 21 pts, 2L+: 19 pts), 19 pts received pumitamig 15 mg/kg and 21 pts received 20 mg/kg. In 39 efficacy-evaluable pts, unconfirmed best overall response was PR in 27 pts and SD in 9 pts for an uORR of 69.2% (1L: 71.4%; 2L+: 66.7%) and DCR of 92.3%. Confirmed ORR (cORR) was 56.4%. uORR was 63.2% with pumitamig 15 mg/kg (cORR 47.4%) and 75.0% with pumitamig 20 mg/kg (cORR 65.0%). Of 34 pts with centrally tested PD-L1 levels (22C3 pharmDx assay), 17 had CPS ≥10 (uORR: 70.6%) and 17 CPS <10 (uORR: 64.7%). The mean best percentage change in tumor size was -40.5% with 66.7% of pts achieving early tumor shrinkage. In Cohort 2 (enrollment ongoing, median age: 53.5 yrs, 1L: 9 pts, 2L+: 20 pts), 29 pts received pumitamig + chemotherapy (n=8, Arm 1; n=10, Arm 2; n=11, Arm 3). In 17 efficacy-evaluable pts, uORR/DCR was 64.7%/88.2% (80.0%/80.0% in Arm 1 [n=5], 71.4%/100% in Arm 2 [n=7], 40.0%/80.0% in Arm 3 [n=5]). Updated efficacy, including early DOR and PFS, will be presented. In Cohort 1, baseline (BL) ctDNA detection rate was 94.7% (36/38 pts with cfDNA data), with median maximum allele fraction (mMAF) of 12.5%. At C3D1, mMAF change from BL in 34 evaluable pts was -100%. ctDNA clearance rate was 53% (9/17) for 15 mg/kg and 71% (12/17) for 20 mg/kg pumitamig. Similar results were observed in Cohort 2. Adverse events (AEs) related to either pumitamig or chemotherapy were reported in 39/40 (97.5%) and 23/29 [79.3%] pts, and were Grade ≥3 in 18 (45.0%) and 9 (31.0%) pts in Cohort 1 and Cohort 2, respectively. Pumitamig-related Grade ≥3 AEs were reported in 11 (27.5%) pts in Cohort 1 (5/19 [26.3%] pts receiving 15 mg/kg and 6/21 [28.6%] pts receiving 20 mg/kg) and in 4 (13.8%) pts in Cohort 2. One pt discontinued due to pumitamig-related AEs. No Tx-related deaths occurred. Conclusions: Pumitamig + chemotherapy showed encouraging efficacy independent of CPS levels and manageable safety in 1L/2L+ LA/mTNBC. The efficacy is particularly clinically meaningful in pts with CPS <10, addressing a critical unmet need. No new safety signals were seen compared to previously characterized safety profile of pumitamig. These global 1L/2L+ data align with the 1L TNBC Chinese trial and support further development of pumitamig in LA/mTNBC, being evaluated in the global ROSETTA Breast-01 Phase 3 trial (NCT07173751).
Presentation numberPS1-13-22
Impact of Prior Therapy, Genotype Matching, and Biomarkers in the Bria-ABC Phase 3 Trial
Giuseppe Del Priore, Morehouse School of Medicine, Atlanta, GA
R. Shatsky1, K. McCann2, A. Kahn3, L. Negret4, C. Vaughn5, B. Bayer6, T. Aghajanian6, W. Williams6, G. Del Priore7, C. Nangia8; 1UC San Diego Health Moores Cancer Center, San Diego, CA, 2UCLA Santa Monica Parkside Cancer Care, Santa Monica, CA, 3Yale Cancer Center, New Haven, CT, 4University of Miami, Miller School of Medicine, Miami, FL, 5Hematology and Oncology Associates of Fredericksburg, Fredericksburg, VA, 6BriaCell Therapeutics Corp, Philadelphia, PA, 7Morehouse School of Medicine, Atlanta, GA, 8Hoag Hospital, Newport Beach, CA.
Background Bria-IMT™ is an allogenic whole cell vaccine engineered to express tumor associated antigens and GM-CSF, promoting both adaptive and innate immune responses. The ongoing Bria-ABC trial (NCT06072612) is a multicenter, phase 3 study comparing Bria-IMT based regimens to treatment of physician’s choice (TPC) in pts with late stage metastatic breast cancer who have no conventional options. We explored the feasibility of Bria-IMT in this exceptionally heavily pretreated pt cohort.Methods Pivotal randomized registration trial of Bria-IMT +CPI, Bria-IMT monotherapy, or TPC. Pts randomization 1:1:1 completed; future pts randomized 1:1 into either Bria-IMT + CPI or TPC. The Bria-IMT regimen includes Day -2 cyclophosphamide (300 mg/m²), Day 0 intradermal SV-BR-1-GM (20×106 irradiated cells), and Day 2 pegylated α interferon (0.1 mcg/site). CPI is administered each cycle, q3w. Imaging assessments q6wk (×2) then q8wk. Pts with ECOG < 2, prior checkpoint inhibitor (CPI), antibody drug conjugate (ADC), CDK4/6 inhibitor (CDK4/6i) exposure, and CNS metastases are eligible; no limit on prior lines. This arm blind analysis was conducted to evaluate feasibility, and provide benchmark PFS stratified by prior treatment regimens, HLA alleles, and biomarkers using KM curves, with significance assessed by log rank or Gehan Breslow Wilcoxon tests.Results At data cut off, 186 pts have been screened, 113 randomized, and 107 treated: median age – 59 (range 32-91); BMI -25.1; 75% Caucasian; 20% other; 5% not yet reported; median 6 prior lines (2-15); ECOG 2: 7%, intracranial mets: 11%. 43% of pts HR+, 35% TNBC, 12% HER2+, 7% HER2low and 3% no subtype yet recorded. Among treated pts (n = 107), median PFS (mPFS): 2.9 mo (95% CI, 2.4 – 3.7). mPFS in pts w/ vs. w/o prior ADC; 3.1 vs 6.0 mo(HR: 1.8, 95%CI 0.9-3.6; p=0.09). mPFS in pts w/ vs w/o prior CPI ; 2.3 vs 3.8 mo(HR: 1.8; 95% CI 0.9 – 3.3, p =0.07). Pts w/ vs w/o prior CDK4/6i , mPFS was 3.8 vs 2.1 (HR: 0.5; 95% CI 0.3 – 0.9, p = .03). In treated pts w/ available genotyping (n = 89), those with HLA Class I A2 expression (44%) had a non statistically significant but numerically greater difference in mPFS vs pts expressing other HLA alleles (HR: 1.0; 95% CI 0.6 – 1.7, p = NS). Arm specific HLA based results unreported. Pts w/o detectable CTCs at baseline or follow up, and with a reduction in mean cancer associated macrophage like cell (CAML) size at follow up (n = 22), had a median PFS of 3.8 mo vs 2.3 mo in pts with CTCs present at baseline or follow-up (n = 22) (HR 2.3; 95% CI, 1.1-4.6; p = 0.04). Pts with a disease history of, or emergent, intracranial metastases showed a mPFS of 2.4 vs 3.1 mo in the overall cohort (HR: 1.7; 95% CI 0.7-3.8, p = 0.23). Pts with a favorable neutrophil to lymphocyte ratio (NLR) (≥ 0.7 or ≤ 2.3) after 1st treatment had a mPFS of 4.5 mo vs 2.5 in pts with an unfavorable NLR ( 2.3) (HR: 0.5; 95% 0.3 – 0.9, p = 0.009). At baseline, favorable NLR mPFS 4.9mo vs 2.8 unfavorable (HR: 0.47; 95% CI 0.3 – 0.8, p = 0.004). No treatment discontinuations or dose reductions related to SV-BR-1-GM reported. Majority of adverse events (76%) were grade 1-2, with few (37%) cases > grade 3, including anemia (6.5%), increased GGT (3.7%), and neutropenia (3.7%). Patients demonstrated stable functional status, as assessed by physical functioning domain questions of the EORTC QLQ-C30.Discussion These preliminary findings from the Bria-ABC Phase 3 trial suggest that early sub-group response trends confirm biomarker utility and feasibility as seen in the phase 2 study supporting continued exploration of Bria-IMT™ even in very advanced MBC. Patients in this late stage, phase III trial maintained functional status w/o evidence of decline. This early prospective RCT in late stage metastatic breast cancer following recent drug approvals offers an emerging benchmark with results that may inform future trial design and guide care for pts who have exhausted standard options.
Presentation numberPS1-13-01
Effects of a physical exercise protocol via telehealth on the quality of life and stress of women undergoing chemotherapy: a randomized clinical trial
Giuliano Tavares Tosello, Unoeste, Presidente Prudente, Brazil
D. C. Silva1, G. T. Tosello2, D. G. D. Christofaro1, W. R. Tebar1; 1Unesp, Presidente Prudente, BRAZIL, 2Unoeste, Presidente Prudente, BRAZIL.
Introduction: Although effective, chemotherapy is associated with physical and psychological adverse effects, including stress, which can impair quality of life (QoL). Physical exercise has been shown to effectively reduce stress and improve QoL in cancer patients. Telehealth has emerged as an alternative to expand access to exercise programs, overcoming logistical and financial barriers. Therefore, implementing telehealth-based exercise protocols for women undergoing chemotherapy is relevant to optimize comprehensive care in this population. Objetives: To evaluate the effect of a telehealth-based exercise protocol on quality of life and stress in women undergoing chemotherapy. Methods: This randomized clinical trial was approved by the Research Ethics Committee (47329721.6.0000.5402) and registered with the ReBEC (RBR-9mqyz6j). Forty-four women (48.36 ± 12.39 years) diagnosed with solid tumors and undergoing chemotherapy were randomized into an intervention group (IG, n=22) and a minimal intervention group (MIG, n=22). The IG performed a supervised telehealth exercise program, including flexibility, muscular endurance, cardiorespiratory fitness, and balance, three times per week for 20 minutes per session via video call. The MIG received guidance and follow-up to address questions regarding physical activity. QoL was assessed using the EORTC QLQ-C30 questionnaire (Global Health Status, Functional, and Symptom scales), where higher scores indicate better functioning and higher symptom burden. Physical and emotional stress was evaluated using Lipp’s Stress Symptoms Inventory for Adults (alert ≥7 symptoms, resistance ≥4 symptoms, exhaustion ≥8 symptoms). For statistical analysis, ANOVA adjusted for age, sex, and socioeconomic status was applied to the EORTC QLQ-C30. Descriptive frequency analysis was used for the Lipp questionnaire, and the Smallest Worthwhile Change (SWC) index was applied to both questionnaires to identify clinically meaningful changes, complementing statistical tests. Results: The telehealth-based exercise protocol did not show statistically significant differences in QoL between groups. However, SWC analysis revealed slight improvements or maintenance in global health domains in the IG (improved: 8, maintained: 6, worsened: 8), as well as in the functional scale. Maintaining QoL values is considered a positive outcome, suggesting that interventions may help mitigate chemotherapy’s adverse effects. Evidence indicates that during chemotherapy, patients’ QoL tends to deteriorate regardless of treatment. Regarding stress, both groups showed increased symptom presence. SWC analysis revealed small clinical changes in the exhaustion phase and more pronounced improvements in the resistance phase in the IG (improved: 9, maintained: 4, worsened: 9) compared to the MIG (improved: 7, maintained: 0, worsened: 15). Conclusions: These findings suggest that the exercise protocol may have a more pronounced effect in the early stages of stress. The clinical relevance of this study lies in demonstrating that the telehealth-based exercise protocol may be effective in maintaining or improving QoL and reducing stress in this population.
Presentation numberPS1-13-14
Trypanosoma cruzi Extracts Modulate Diverse Death Signaling Pathways in Triple Negative Breast Cancer
Destiny D Ball, Meharry Medical College, Nashville, TN
D. D. Ball, K. J. Rayford, A. Cooley, S. Briseno-Gonzalez, P. Nde, A. Sakwe; Meharry Medical College, Nashville, TN
The protozoan parasite Trypanosoma cruzi (T. cruzi) causes American trypanosomiasis or Chagas disease. While chronic T. cruzi infection is associated with the development of gastrointestinal and esophageal cancers, protein components of T. cruzi parasite extracts, as well as cell surface antigens, do exhibit anti-tumor effects. This novel study focuses on triple-negative breast cancer (TNBC), that lacks estrogen and progesterone receptors as well as the human epidermal growth factor receptor-2. Compared to other invasive breast cancer types, TNBC patients are often resistant to conventional methods of treatment. Thus, there is a need for nonconventional therapeutics to antagonize tumor progression. In this study, we investigated how T. cruzi parasite extracts affect the viability of TNBC cell models. We isolated membrane and cytosolic fractions of T. cruzi trypomastigotes and treated basal-like MDA-468 and mesenchymal BT-549 triple-negative breast cancer cell lines with the whole parasite, cytosolic and membrane fractions. We then performed cell proliferation, autophagy, apoptosis and DNA damage assays to evaluate T. cruzi-induced cell death. These parasite extracts significantly induced PARP1 cleavage in MDA-468 cells and upregulated the expression of autophagic marker, LC3B-II in BT-549 cells. To identify potential molecular mechanisms, we isolated RNA for RNA sequencing to evaluate alterations in the transcriptome. Our data show that both T. cruzi membrane and cytosolic extracts inhibit the viability of mesenchymal-like and basal-like TNBC cells. These parasite extracts significantly affected several pathways including cellular stress, and proinflammatory signaling pathways. Collectively, our findings demonstrate that T. cruzi membrane and cytosolic extracts suppress TNBC cell viability through the activation of multiple cell death pathways. These results highlight the potential of T. cruzi-derived components as nonconventional therapeutic agents for the treatment of triple-negative breast cancer. This project was supported, in part, by NIH/NIGMS SC1GM139814, NIH/NIAID T32AI007281, NIH U54MD007586 and an Education Gift from Dr. Bernard Crowe.
Presentation numberPS1-13-09
Fasting During Ramadan in Breast Cancer: Practices and Outcomes
Ahmad Al-Bitar, Damascus University, Damascus, Syrian Arab Republic
A. Al-Bitar1, A. Nasra1, L. Alsaoub1, G. Tannous1, N. Rajjoub2, A. Kouli1, O. Johar1, F. Kalam3, M. Hafez4; 1Damascus University, Damascus, Syrian Arab Republic, 2Homs University, Homs, Syrian Arab Republic, 3Ohio State University Comprehensive Cancer Center, Columbus, OH, 4St. Luke’s University Health Network, Bethlehem, PA
AbstractBackground:Fasting has emerged as a potential modifier of cancer outcomes through effects on metabolism, treatment tolerance, and survivorship. For Muslim patients, Ramadan represents a month of religiously prescribed fasting, but clinical recommendations are limited for this population. clarify how patients approach Ramadan fasting and factors influencing safety, and given the lack of contemporary data, this study of 238 patients is one of the largest surveys to date to examine these practices in a Middle Eastern breast cancer cohort. We evaluated fasting practices, predictors, and patient-reported outcomes among breast cancer patients during Ramadan.Methodology:A cross-sectional survey was conducted at Al-Bairouni University Hospital, Syria’s national cancer center. All patients from the breast cancer unit diagnosed with breast cancer were included (N=238; 99.6% female; mean age 50.0 years). Measures included demographics, disease status, treatments, fasting behaviors, and factors influencing decisions. Predictors of fasting (none/partial/full) were evaluated with multivariable proportional-odds logistic regression.Results:Overall, patients’ fasting patterns varied: 71.0% fasted for the entire month, 14.3% fasted only on certain days (defined as partial fasting), and 14.7% did not fast at all. Religious commitment was the primary motivator for fasting (85.8%). Despite their strong religious motivation, patients expressed a need for better medical guidance. Although 63.4% of patients desired clear medical guidance, only 43.8% of them consulted their oncologist about fasting. Among patients who fasted for the full month, self-reported adverse effects were generally mild, and no serious adverse events were reported. Fatigue was the most common mild symptom (32.5%), while 55.0% reported no health problems. Furthermore, 96.4% of the participants reported no negative impact on their treatment adherence, with some even reporting improved adherence. In adjusted models, older age was linked to lower odds of achieving full fasting (aOR 0.94 per year, 95% CI 0.91−0.97, p<.001). Lower fasting odds were also predicted by metastatic disease (aOR 0.28; 95% CI 0.08−0.97, p=.046) and recurrent disease (aOR 0.17; 95% CI 0.04−0.91, p=.039). Conversely, patients who reported having equal trust in both physicians and religious leaders had threefold higher odds of fasting (aOR 3.0; 95% CI 1.5−5.8, p=.001).Conclusion:The findings suggest that most breast cancer patients can fast safely if they receive appropriate guidance; however, there is a clear need for evidence-based guidelines. Oncology teams should proactively address fasting in care plans through culturally tailored counseling to ensure effective patient care. Furthermore, collaboration with patients’ religious leaders may enhance informed decision-making, a valuable and novel approach to improving patient support.
Presentation numberPS1-13-30
Concordance of Mammographic Studies Between Artificial Intelligence and Expert Breast Radiologists
RONALD SERGIO LIMON, OncoBolivia, santa cruz, Bolivia, Plurinational State of
R. S. LIMON; OncoBolivia, santa cruz, BOLIVIA, PLURINATIONAL STATE OF.
Background. Various artificial intelligence (AI) programs have been specifically designed and trained for breast cancer screening and prediction, and their use is steadily increasing. However, tools such as ChatGPT—an AI not originally developed for imaging analysis—are being increasingly employed for this purpose, particularly by non-medical personnel. This situation may generate mistrust and uncertainty among patients when comparing AI-generated reports with those of medical specialists. Therefore, the proper use of these tools requires awareness of their intended scope and limitations.Methods. Random mammograms performed in August 2025 on asymptomatic women undergoing preventive screening were included. All studies were interpreted and reported by expert breast radiologists. For AI evaluation, images were uploaded using an iPhone 16 Pro Max into ChatGPT, which generated a structured mammography report. Concordance was assessed across three parameters: breast density, BI-RADS classification, and recommendations. Results A total of 50 mammograms met the inclusion criteria. Concordance for breast density was 88%, with a predominance of pattern C in 65% of cases. BI-RADS classification showed 93% concordance, most commonly BI-RADS 2. Recommendations demonstrated 90% concordance, particularly regarding complementary breast ultrasound and annual follow-up when clinically indicated. Conclusions. Our findings suggest that ChatGPT, despite not being specifically designed for imaging interpretation, demonstrated a high level of concordance with expert breast radiologists. These results should be confirmed in larger case series to validate the potential role of non-imaging AI tools in breast cancer screening support.
Presentation numberPS1-13-23
Survival Results of Phase II Bria-IMT Allogenic Whole Cell-Based Cancer Vaccine
Giuseppe Del Priore, Morehouse School of Medicine, Atlanta, GA
C. Nangia1, S. Chumsri2, K. Rowland3, L. Negret4, J. Knecth5, B. Bayer6, T. Aghajanian6, W. Williams6, G. Del Priore7, C. Calfa4; 1Hoag Hospital, Newport Beach, CA, 2Mayo Clinic, Jacksonville, FL, 3Carle Cancer Center, Urbana, IL, 4University of Miami Sylvester Comprehensive Cancer Center, Miami, FL, 5Tranquil Clinical Research, Friendswood, TX, 6BriaCell Therapeutics Corp, Philadelphia, PA, 7Morehouse School of Medicine, Atlanta, GA.
BackgroundManagement of metastatic breast cancer (MBC) continues to present obstacles, as many pts develop resistance to or cannot tolerate standard of care treatments. Bria-IMT combines two modalities to both overcome immune exhaustion and minimize the toxic safety profiles commonly observed in contemporary late stage therapies: an allogeneic whole-cell vaccine (SV-BR-1-GM) and checkpoint inhibition (CPI). The SV-BR-1-GM cell line originates from breast cancer cells and are engineered to secrete GM-CSF and irradiated before administration. These cells upregulate surface presentation of tumor associated antigens on both HLA class I and II, enhancing tumor recognition by T cells. Coadministration w/ an anti-PD-1 CPI, the vaccine response is reinforced, counteracting the immuno-suppressive tumor microenvironment.MethodsThis phase I/II study evaluated the Bria-IMT regimen: low-dose cyclophosphamide (Day –2) followed by SV-BR-1-GM and CPI (pembrolizumab or retifanlimab), then low-dose local peg- interferon α. Cycles q3w. In the phase II study, pts were randomized 1:1 to start CPI immediately at cycle 1 (C1) or delayed start at cycle 2 (C2). There were 2 formulations of SV-BR-1-GM w/ IFNγ stimulation (IFNγ) or unstimulated (PH3). Cancer-associated macrophage-like cells (CAMLs), circulating tumor cells (CTCs), and PD-L1 expression scores on these cells were monitored as biomarkers. Candida skin test assessed anergy status before cycle 1, and a delayed type hypersensitivity (DTH) skin test to SV-BR-1-GM was assessed at each cycle w/ a small SV-BR-1-GM dose before the full dose. ResultsResults of the randomized phase II study cohort are reported (n = 32). Retifanlimab was co-administered w/ SV-BR-1-GM at C1 or C2. Median age 61 (41-80) years; median number of prior regimens was 6 (2-13). 20(63%) pts were ER+/PR+/HER2-; 3(9%) HER2+, and 9 (28%) triple-negative (TNBC). The Bria-IMT regimen was well tolerated, w/ no drop-outs attributable to Bria-IMT. The majority of adverse events (93.8%) grade 1-2, w/ (40.6%) grades 3 and 4. Common AEs included injection site reactions (53%), nausea (43.8%), and fatigue (34%). The median progression free survival (PFS) was 3.5 (range: 1.8-24.5) and overall survival (OS) 9.5 (2.4-31.6) months. One patient remains on therapy, approaching 25 months of PFS. PFS and OS among pts who received immediate vs. delayed start of CPI (PFS 3.7 vs. 3.3 mo, HR: 0.59; 95% CI 0.3-1.2, p=0.16 | OS 13.3 vs. 7.4 months, HR: 0.78; 95% CI 0.34-1.8, p = 0.56). Pts who received the SV-BR-1-GM Ph3 formulation (n=21) had an increase in both PFS (4.1 vs 2.6 months, p = 0.005) and OS (16.6 vs. 9.1 months, HR: 0.6; 95% CI 0.2 – 1.4, p = 0.24); best objective response rate was 12% and the clinical benefit rate was 53%. In ER+/PR+/HER2-, overall OS was 11.7 (3.5-31.6) mo, 17.3 mo (3.5-27.9) in Ph3; HER2+ 7.2 (4.7-24.5) mo, all received Ph3; TNBC 9.9 (2.4-23.0) mo, 16.6 mo (2.4-23.0) in Ph3. In pts w/ available data, pts w/ post- dose CTC count 5 (16.6 vs. 5.5 months, p = 0.0003). Pts w/ positive DTH response had significantly improved PFS (3.6 vs. 2.4 months, p = 0.01) and OS (11.9 vs. 4.7 months, p < 0.0001). ConclusionsIn heavily pretreated MBC pts, the Bria-IMT regimen demonstrated promising results and a favorable safety profile across all subtypes of breast cancer. A clinically meaningful, though not statistically significant, difference in OS was observed between pts receiving CPI at C1 versus C2. Superior outcomes were observed in pts receiving SV-BR-1-GM w/o IFNγ incubation, guiding selection of Ph3 formulation for ongoing and subsequent clinical trials. Pts w/ post-dose CTC count < 5, and positive DTH response had significantly improved outcomes. On basis of these results, a randomized phase III trial comparing the Bria-IMT regimen to physician’s choice is ongoing (NCT06072612). Trial information: NCT03328026.
Presentation numberPS1-11-24
Effectiveness of Initial Denosumab Versus Sequential Bisphosphonate-to-Denosumab Therapy in Preventing Skeletal-Related Events in Breast Cancer Patients With Bone Metastases: A Retrospective Single-Center Study
Jun Cao, Fudan University Shanghai Cancer Center, Shanghai, China
J. Cao; Fudan University Shanghai Cancer Center, Shanghai, China
Effectiveness of Initial Denosumab Versus Sequential Bisphosphonate-to-Denosumab Therapy in Preventing Skeletal-Related Events in Breast Cancer Patients with Bone Metastases: A Retrospective Single-Center StudyJun Cao; Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China Background: Skeletal-related events (SREs)—including pathological fractures, spinal cord compression, radiation to bone, and bone surgery—significantly impair quality of life in breast cancer patients with bone metastases. Denosumab, a monoclonal antibody targeting RANKL, differs mechanistically from bisphosphonates and is not renally cleared, potentially offering clinical advantages. However, in clinical practice, an increasing number of patients transition from bisphosphonates to denosumab. The comparative efficacy and safety of sequential therapy versus initial denosumab therapy in SRE prevention remain unclear. Methods: This retrospective single-center study analyzed 165 breast cancer patients with radiologically confirmed bone metastases treated between January 2019 and April 2024. Patients were grouped by Bone-targeting agent (BTA) strategy: initial denosumab treatment (n=67) or sequential bisphosphonate-to-denosumab therapy (n=98). Patients treated before 2019 were excluded, as denosumab was approved in China in 2019. The primary endpoint was the 1-year SRE rate, with SRE defined as the time to first on-treatment SRE. Baseline clinical characteristics including tumor subtype, stage, visceral metastases, bone lesion burden, and systemic therapy history were analyzed. Kaplan-Meier methods estimated SRE-free survival, and Cox regression identified factors associated with SRE risk. Results: The median age at bone metastasis diagnosis was 54.7 years. Median intervals from initial breast cancer diagnosis to bone metastasis varied by stage: 30.9 months (stage I, n=5), 50.2 months (stage II, n=14), 44.8 months (stage III, n=11), and 29.8 months (stage IV, n=125). Follow-up durations were 22.5 months in the sequential therapy group versus 11.3 months in the initial denosumab group. The number of bisphosphonate and denosumab administrations in the sequential group was 10 and 8, respectively, while that in the initial denosumab group was 9. A higher proportion of patients in the initial denosumab group received ≥2 lines of treatment (P < 0.001). The median time from bone metastasis confirmation to BTA initiation was 0.9 months in both groups. After BTA initiation, the 12-month cumulative SRE incidence was lower with initial denosumab (5.9%; 95% CI, 0–12.3%) than with sequential therapy (15.7%; 95% CI, 8.1–22.7%). Pathological fractures (46.4%) and bone radiotherapy (50.0%) accounted for most SREs. The median time to first SRE was not reached in either group.Univariate Cox regression showed that second-line systemic therapy was associated with an increased SRE risk (HR=2.651, P=0.021). The 1-year incidence of adverse events, including hypercalcemia and medication-related osteonecrosis of the jaw (MRONJ), was lower in the initial denosumab group. Conclusions: This single-center retrospective study demonstrates that initiating denosumab treatment is associated with a lower incidence and delayed SRE onset compared to sequential bisphosphonate-to-denosumab therapy in breast cancer patients with bone metastases, with fewer adverse events. Second-line systemic therapy was associated with an increased SRE risk. These findings support early initiation of denosumab as a preferred BTA strategy.
Presentation numberPS1-03-21
Patient-reported measures of taxane-induced peripheral neuropathy (TIPN) and risk of dose reductions or worsening quality of life in Black women with breast cancer: analysis from ECOG-ACRIN EAZ171
Tarah J. Ballinger, Indiana University, Indianapolis, IN
T. J. Ballinger1, F. Zhao2, G. Jiang1, F. Shen1, D. Cella3, D. Peipert4, K. D. Miller1, A. Wolff5, A. DeMichele6, B. P. Schneider1, L. Wagner7; 1Medicine, Indiana University, Indianapolis, IN, 2School of Public Health, Dana Farber Cancer Institute, Boston, MA, 3Medicine, Northwestern, Chicago, IL, 4Applied Health Sciences, University of Birmingham, Birmingham, UNITED KINGDOM, 5Medicine, Johns Hopkins, Baltimore, MD, 6Medicine, University of Pennsylvania, Philadelphia, PA, 7Health Policy and Management, University of North Carolina, Chapel Hill, NC.
Introduction: Black patients with breast cancer experience significantly higher rates of taxane-induced peripheral neuropathy (TIPN), potentially impacting quality of life (HRQoL), physical functioning, and dose delivery of curative chemotherapy. Here, we utilize the prospective trial ECOG-ACRIN EAZ171 to determine whether patient-reported TIPN outcome measures (PROMs), and which PROM, predict dose reductions of taxane therapy and longer-term detriment to HRQoL and physical functioning in Black patients who are most impacted by TIPN. Methods: EAZ171 enrolled 249 Black patients planned to receive (neo)adjuvant taxane therapy. This PRO analysis includes patients receiving at least one dose of therapy with available PRO data (n= 239). PRO assessments included FACT-GOG/NTx 4-item and 11- item neurotoxicity subscales and PRO-CTCTAE numbness/tingling and interference at baseline and each cycle. FACT-G and PROMIS Physical Function short form 10a were administered at baseline, during treatment, and 1 year post treatment initiation. Occurrence of patient-reported TIPN was defined as a decrease in FACT-GOG/NTx 4-item subscale of 2 or more. Other measures of TIPN explored included NTx item 1, NTx item 2, total FACT-GOG/NTx 11-item subscale, PRO-CTCTAE numbness/tingling, and PRO-CTCAE neuropathy interference. Reduction in HRQoL or physical function at 12 months were defined as a decrease in FACT-G total score of >/= 7 or decrease in PROMIS PF T score of >/= 5, respectively. Multivariate logistic mixed effect models with random intercept were used to determine the association of patient-reported TIPN with 1) subsequent dose reduction of taxane therapy due to neuropathy, and 2) subsequent reduction in HRQoL or physical function at one year, Covariates included baseline neuropathy, taxane type, age, ECOG PS, BMI, disease stage, nodal status, and HgbA1c level. Results: Receipt of paclitaxel and ECOG PS of 1 (vs 0) were associated with dose reduction. All measures of neuropathy were correlated with dose reduction, with the FACT-GOG/NTx 4-item subscale being the most highly correlated in multivariate analysis (OR 10.8, 95% CI 4.5-25.90). Dose reductions were also correlated with increasing neuropathy by total FACT-GOG/Ntx score (OR 5.02, 95% CI 2.55-9.88), NTx item 1 (OR 6.10, 95% CI 2.96-12.57), NTx item 2 (OR 3.88, 95% CI 2.00-7.50), PRO-CTCTAE severity (OR 8.16, 95% CI 3.65-18.25), and PRO-CTCTAE interference (OR 5.64, 95% CI 2.89-10.98). PRO data available at 12 months was limited (n=140; 59%). Occurrence of patient-reported neuropathy by FACT-GOG/Ntx-4 during treatment was not associated with a reduction in HRQoL or physical function at 12 months; however, persistent neuropathy at 12 months was associated with reduced HRQoL (OR 5.05, 95% CI 1.33-19.17). Conclusions: The majority of Black women reported moderate or severe TIPN during treatment, which significantly predicted dose reduction of curative therapy. The FACT-GOG/NTx 4-item subscale correlated most highly with dose reduction, supporting its use in future trials. Patient-reported TIPN during therapy did not correlate with longer- term reductions in HRQoL or physical function at 12 months, perhaps due to lack of specificity of these items or low sample size and differential attrition. These findings support the potential impact of and need for a focus on careful evaluation of toxicity. We now plan to utilize the measures identified here to select patients in need of early supportive care to reduce TIPN’s impact on dose delivery in Black patients, potentially improving survival and racial equity in breast cancer.
Presentation numberPS1-10-06
Impact of prior fulvestrant use and endocrine therapy duration on real-world outcomes of elacestrant treatment in ER+/HER2- metastatic breast cancer
Suresh Reddy, Cancer Care Specialists, Reno, NV
S. Reddy1, R. Choksi2, V. Gorantla2, F. Kudrik3, D. Patt4, S. Reganti1, S. Rosenfeld5, G. Cioffi6, F. Yang6, D. Parris6, A. Rui6, M. Gart6, C. Wall6, B. Wang6, P. Varughese6, J. Donegan6, L. Morere6, R. Geller6, J. Scott6, R. Mahtani7; 1Cancer Care Specialists, Reno, NV, 2University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3South Carolina Oncology Associates, Columbia, SC, 4Texas Oncology, Austin, TX, 5Highlands Oncology Group (HOG), Fayetteville, AR, 6IntegraConnect, West Palm Beach, FL, 7Miami Cancer Institute, Baptist Health South Florida, Miami, FL
Background: Endocrine therapy (ET) combined with a CDK4/6 inhibitor (CDK4/6i) is the standard first-line treatment for patients (pts) with estrogen receptor-positive (ER+), HER2-negative (HER2-) metastatic breast cancer (mBC). However, many pts eventually develop endocrine resistance, often driven by acquired ESR1 mutations (ESR1m). Elacestrant is the first oral selective estrogen receptor degrader approved for pts with ESR1-mutated ER+/HER2- mBC. Approval was based on the phase III EMERALD trial, which demonstrated significantly improved progression-free survival (PFS) compared to standard-of-care ET in pts with ESR1m. Notably, ~29% of pts received prior fulvestrant, and subgroup analyses showed a PFS benefit with elacestrant even in this subgroup. Exploratory analyses suggest that pts with longer disease control on prior ET + CDK4/6i may derive greater benefit from elacestrant. Despite these findings, real-world evidence on how prior ET exposure, including fulvestrant use and ET duration, impacts elacestrant outcomes is limited. This study assessed the association of prior fulvestrant use and prior ET duration with time to next treatment (TTNT) and real-world overall survival (rwOS) among pts who received elacestrant monotherapy. Methods: The IntegraConnect PrecisionQ de-identified electronic health record database, (>3 million cancer pts, >500 care sites) was utilized to identify pts treated with elacestrant monotherapy between 1/27/2023 and 1/31/2025. All pts received ≥1 ET before elacestrant initiation. ESR1m was not assessed but assumed present. Pts were stratified by prior use of fulvestrant monotherapy, prior use of fulvestrant + CDK4/6i, and duration of prior ET (<12 vs. ≥12 mo, ET could be other than fulvestrant). Kaplan-Meier survival curves were used to assess TTNT and rwOS across subgroups. Multivariable Cox proportional hazards regression was used to evaluate the association of prior fulvestrant use and ET duration with TTNT and rwOS, adjusting for clinical and demographic covariates including age at index date, race, ethnicity, Eastern Cooperative Oncology Group performance status, and metastatic line of therapy. Results: A total of 589 pts met study inclusion criteria: 77 pts (13%) received fulvestrant monotherapy previously, 294 (50%) received fulvestrant + CDK4/6i previously, and 329 (56%) had ≥12 mo duration of prior ET. No significant differences in TTNT or rwOS were observed by prior fulvestrant monotherapy (prior fulvestrant vs. no prior fulvestrant median TTNT, 6 vs. 5.3 mo; median rwOS, 18.3 vs. 17.3 mo) or CDK4/6i combination therapy (prior CDK4/6i + fulvestrant vs. no prior CDK4/6i + fulvestrant, median TTNT 5.2 vs. 5.7 mo; median rwOS, 18.3 vs. 17.3 mo). Pts with ≥12 mo of prior ET had significantly longer TTNT (6 vs. 4.6 mo, log-rank p<0.001) and improved rwOS (23.0 vs. 13.8 mo; log-rank p<0.001). In multivariable analyses, pts with ≥12 mo prior ET were 31% less likely to initiate a subsequent treatment (TTNT hazard ratio [HR], 0.69; 95% confidence interval [CI] 0.56-0.85). The impact on rwOS was attenuated after adjustment (HR, 0.77; 95% CI, 0.58-1.03, p=0.079). Conclusions: In this real-world cohort of pts treated with elacestrant, prior fulvestrant use did not significantly impact clinical outcomes. However, longer duration of prior ET (≥12 mo) was associated with delayed TTNT, even after adjusting for key clinical factors. These findings are consistent with a post-hoc analysis from the EMERALD trial. Our real-world results further support the hypothesis that longer endocrine sensitivity may enrich for elacestrant-responsive disease and inform treatment sequencing decisions in clinical practice.