Poster Session 2
Session Details
Presentation numberPS2-01-01
Preoperative Neutrophil-to-Lymphocyte and Platelet-to-Lymphocyte Ratios as Predictors of Capsular Contracture after Nipple-Sparing Mastectomy with Implant-Based Breast Reconstruction
Jongwon Kang, Seoul national university bundang hospital, Seongnam, Korea, Republic of
J. Kang, S. Jung, Y. Yoon, J. Oh, H. Koh, H. Shin, E. Kim, K. Yoon; Surgery, Seoul national university bundang hospital, Seongnam, KOREA, REPUBLIC OF.
Preoperative Neutrophil-to-Lymphocyte and Platelet-to-Lymphocyte Ratios as Predictors of Capsular Contracture after Nipple-Sparing Mastectomy with Implant-Based Breast Reconstruction Jongwon Kang¹, Sungjin Jung¹, Yena Yoon¹, Jeong-Hun Oh¹, Hyoung Won Koh¹, Kyung-Hwak Yoon¹, Hee-Chul Shin¹, Eun-Kyu Kim*¹ 1Department of Surgery, Seoul National University College of Medicine, Breast Care Center, Seoul National University Bundang Hospital, Korea BackgroundCapsular contracture is a major complication following nipple-sparing mastectomy (NSM) with direct-to-implant (DTI) breast reconstruction. While inflammation is a known contributor to capsular contracture, the role of systemic inflammatory markers such as the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) remains unclear. This study aimed to evaluate their predictive value for capsular contracture.MethodsWe retrospectively reviewed 376 patients who underwent NSM with immediate DTI reconstruction. Patients were grouped by Baker’s classification into the non-contracture (grades 1–2) and the contracture (grades 3–4) groups. Clinical and pathological factors including preoperative NLR and PLR were analyzed. The optimal cut-off values for NLR and PLR were determined using receiver operating characteristic (ROC) curve analysis.ResultsAmong the 376 patients, 43 (11.4%) developed grade 3 or 4 capsular contracture during follow-up. The contracture group showed significantly higher rates of radiotherapy, chemotherapy, and elevated NLR/PLR. Multivariate analysis identified radiotherapy (OR 12.142, p<0.001) and elevated NLR and PLR (OR 5.287, p=0.001) as independent risk factors of capsular contracture. In subgroup analysis, NLR and PLR were not significant predictors among patients who did not receive radiotherapy, but remained significant among those who received radiotherapy (OR 7.334, p=0.002).ConclusionElevated NLR and PLR are associated with an increased risk of capsular contracture in patients undergoing NSM with DTI reconstruction, particularly in patients receiving radiotherapy. These findings suggest that systemic inflammation and radiation-induced fibrosis may contribute synergistically to capsular contracture development. As these markers are routinely available, they may serve as useful tools for preoperative risk stratification and individualized patient counseling in reconstructive breast surgery. Key wordsBreast cancer, Nipple sparing mastectomy, Direct-to-implant reconstruction, Capsular contracture, Systemic inflammation, Neutrophil-to-lymphocyte ratio, Platelet-to-lymphocyte ratio, Radiation therapy
Presentation numberPS2-01-02
Magnetic seed for preoperative localization of non‑palpable breast cancer: impact on care pathway organization and re-excision rate
Delphine Hequet, Institut Bourdonnais, Paris, France
D. Hequet1, A. Hababou2, G. Aubry1, R. Salmon3, S. Harguem2, M. Bou Antoun2, J. Seror1; 1Surgery, Institut Bourdonnais, Paris, FRANCE, 2Radiology, Groupe Union Imagerie, Paris, FRANCE, 3Surgery, Clinique Saint Jean de Dieu, Paris, FRANCE.
Introduction: Preoperative localization of non-palpable breast lesions, whether performed the day before or on the day of surgery, can be anxiety-inducing for patients. The scheduling in a busy operating theater for short procedures is highly sensitive to even minor delays. Localization appointments may cause such delays, as well as raise potential timing confusions between surgery time, arrival time at the care center, and localization time. Moreover, wire-guided localization requires specific radiology sessions and coordination with radiologists specialized in breast imaging. For these reasons, we progressively implemented preoperative localization using a magnetic clip. Placement is done a few days before surgery, which also allows a comprehensive re-review of the radiologic images. The objective is to describe the organizational impact and the re-excision rate of the systematic use of magnetic seeds in a single institution.Methods: Retrospective single-center study. We describe the characteristics of cases with magnetic seed localization and compare these features, as well as ambulatory stay durations, to a historical control group that underwent wire-guided localization. For this comparison, only consecutive patients who had unifocal, unilateral breast cancer and were treated on an outpatient basis were included.Results: Between March 2024 and June 2025, 261 patients were referred to the radiology department for magnetic seed placement before surgery. During imaging review, additional lesions were detected in 21 patients (8%), leading to total mastectomy in 5 cases. Excluding 8 patients (3%) who were scheduled for inpatient admission, 232 underwent magnetic seed placement, including 35 (15%) for benign lesions. We compared 197 patients who had magnetic seed localization for cancer between March 2024 and June 2025 to 60 patients in the historical control group who underwent wire localization for cancer between November and December 2023. The median age in both groups was 62 years (p = 0.98). The control group had a higher proportion of in situ ductal carcinoma (n = 18, 30% vs. n = 27, 15%, p = 0.02), while the magnetic seed group underwent more axillary procedures (n = 170, 86% vs. n = 42, 70%, p = 0.009). Tumor size was slightly larger in the magnetic seed group (10 mm vs. 8 mm, p = 0.04). Fewer re-excisions for positive margins occurred in the magnetic seed group (n = 3, 1.5% vs. n = 8, 13%, p < 0.01). This result was confirmed in multivariate analysis : use of magnetic seed reduced independently the rate of re-excision with an OR of 0.37 (IC95%[0.002-0.001], p=0,0018). Operating time was equivalent in both groups (32 min vs. 33 min, p = 0.87), but ambulatory stay was reduced by more than 2 hours in the magnetic seed group (5h 14 vs. 7h 19, p < 0.01).Conclusion: The use of magnetic seeds enables time savings on the day of surgery, streamlining the operating theater schedule and reducing patient anxiety associated with pre-surgical delays. Re-excision rates is dramatically decreased when using magnetic seeds.
Presentation numberPS2-01-03
Is Breast Conserving Surgery a Viable Option for Non-Inflammatory T3/T4 Breast Cancer?
Leena Almaghrabi, KFSHD, Dammam, Saudi Arabia
L. Almaghrabi1, J. Alazhri2, F. Aldulaijan3, N. Almana2, S. Alajmi2, M. Alduhaileb2, A. Abbas3; 1General Surgery, KFSHD, Dammam, SAUDI ARABIA, 2Breast Surgery, KFSHD, Dammam, SAUDI ARABIA, 3Breast and Endocrine surgery, KFSHD, Dammam, SAUDI ARABIA.
Background: Breast conserving surgery for locally advanced breast cancer remains a controversial approach,Historically mastectomy has been the preferred surgical intervention for patients with T3/T4 breast cancer.However,the utilization of neoadjuvant systemic therapy (NAST), development of oncoplastic breast conserving techniques rendered BCS an increasingly appealing option for patients and surgeons.Methodology: Study aims to evaluate the surgical and oncological safety of BCS for non-inflammatory, non-multicentric T3/T4 non-metastatic breast cancer through a retrospective analysis of prospectively maintained data. 75 female patients diagnosed between 2017 and 2024. Analysis encompassed clinico-pathological characteristics, treatment modalities and surgical outcomes, including surgical margins, rates of margin re-excision and conversion to mastectomy and pathologic complete response (PCR) or down-staging to a lower tumor stage after NAST. Oncological outcomes included disease recurrence and overall survival.Result: Out of 75 patients, 43 (57%) had T3 lesion,32 (43%) had a T4a-c lesion. Mean age of patients was 52 years. Most of patients had an invasive ductal histology (95%) of high grade (71%), node positive (60%). Luminal type comprised 41%,37% triple negative and 21% were HER2neu positive. BCS associated with positive surgical margins in 2 patients (2.6%) one underwent margin re-excision (1.3%), other patient (1.3%) converted to mastectomy for margin clearance. Most of the patients received NAST; 63 (84%) chemotherapy, 6 (8%) hormonal therapy. Majority (93%) received adjuvant radiotherapy. PCR observed in 31(45%) patients,35 (50%) achieved down-staging to smaller tumor size, including patients with triple negative or HER2 positive. 3 patients (5%) with luminal type failed to achieve pathological response to NAST and had identical clinical and pathological tumor stages. On multivariate analysis, patients with TN (70%) and HER2 positive (50%) were likely to achieve PCR after NAST, compared to patients with luminal type (8%) (P<0.001). with average follow up duration of 34 months, our cohort demonstrated an overall survival rate of 95%. While the majority of patients remained disease-free, Tumor recurrence was observed in 10 (13%) patients. No isolated local recurrences. Most recurrences were systemic (11%), compared to one systemic and local, and another Locoregional recurrence (1% each). Systemic recurrences were predominantly associated with HER2 positive (50%) grade 3 (60%) and characterized by the lack of PCR (78%) after NAST. However, this did not correlate to statistical significance. Discussion: Data demonstrates that BCS for T3/T4 non inflammatory breast cancer after NAST is associated with low rates of positive surgical margins and conversion to mastectomy, while maintaining acceptable outcomes after oncoplastic surgery, including level 1 and 2, therapeutic reduction mammoplasty, and chest wall perforator flap reconstruction. Isolated local recurrence was rare, and most recurrences were systemic, suggesting that treatment failure was primarily systemic rather than regional. Interestingly, the two patients who experienced local recurrence had negative surgical margins following BCS; however, they did not receive adjuvant radiation, and both had a triple positive, the association between local recurrence and HER2-positive subtype, high tumor grade, and lack of PCR highlights the impact of tumor biology on prognosis conclusion, BCS for non-inflammatory T3/T4 breast cancer appears to be both surgically and oncologically safe after NAST. Therefore, it should be considered and thoroughly discussed with the patient as a viable treatment alternative to mastectomy.
Presentation numberPS2-01-04
Association of Long-term Oncologic Prognosis With Internal Mammary Sentinel Lymph Node Biopsy In Breast Cancer Patients
Pengfei Qiu, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
Z. Shi, R. Jia, Y. Wang, P. Qiu; Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, CHINA.
Background: Internal mammary sentinel lymph node biopsy (IMSLNB) is a minimally invasive diagnostic technique for assessing regional lymph nodes, which provides precise lymph node staging and informs adjuvant treatment decisions. However, its prognostic impact remains uncertain, resulting in ongoing debate regarding its clinical application. This study aims to investigate the long-term prognostic outcomes of IMSLNB in patients with early-stage breast cancer.Methods: This study performed a retrospective cohort analysis involving 10,923 breast cancer patients who visited our hospital between January 1, 2013, and December 31, 2022. Following propensity score matching, the patients were categorized into two groups: the IMSLNB group and the no-IMSLNB group. The prognostic outcomes of these two groups were compared. The primary endpoint of this study was disease-free survival (DFS), while secondary endpoints included overall survival (OS), regional recurrence-free survival (RRFS), local recurrence-free survival (LRFS), and distant metastasis-free survival (DMFS).Results: A total of 1608 patients were included in the final analysis, with 536 in the IMSLNB group and 1072 in the no-IMSLNB group. In the IMSLNB group, 93 patients were identified with IMSLN metastasis, yielding a metastasis rate of 17.4%. Among these, 82 had concurrent axillary lymph node (ALN) metastasis, while 11 exhibited isolated IMSLN metastasis. The median follow-up duration was 60.9 months. The findings demonstrated that the 5-year DFS (95.9% vs. 93.9%; HR, 0.562; 95% CI, 0.364-0.869; P=0.010) was significantly higher in the IMSLNB group compared to the no-IMSLNB group. However, there was no statistically significant difference in the 5-year overall survival (OS) rates between the two groups (98.9% vs. 98.5%; HR, 0.487; 95% CI, 0.208-1.141; P=0.098).The 5-year RRFS (99.4% vs 98.4%, HR, 0.360; 95% CI, 0.145-0.890; P=0.027) and LRFS (99.6% vs 98.5%, HR, 0.380; 95% CI, 0.118-0.800; P=0.016) in the IMSLNB group were significantly superior to those in the no-IMSLNB group; however, there was no statistically significant difference observed in the 5-year DMFS (96.8% vs 97.0%, HR, 0.843; 95% CI, 0.472-1.508; P=0.565) between the two groups.Exploratory subgroup analysis of disease-free survival (DFS) revealed that patients within specific subgroups, including diagnostic age (>50 years), mastectomy, absence of lymphovascular invasion (LVI), pathological type as invasive ductal carcinoma, and negative axillary lymph node (ALN) status, demonstrated significant benefit from IMSLNB (P<0.05).Conclusion: IMSLNB enables more precise regional lymph node staging for early-stage breast cancer, facilitates the optimization of adjuvant radiotherapy strategies, thereby enhancing RRFS, LRFS, and DFS outcomes. It can be recommended as a minimally invasive technique for regional lymph node staging.
Presentation numberPS2-01-05
Improving radical resection rates in patients with breast cancer by intraoperative imaging using bevacizumab-IRDye800CW – the MARGIN-2 study
Joni J. Nijveldt, University Medical Center Groningen, Groningen, Netherlands
B. Keizers1, J. J. Nijveldt2, T. S. Nijboer3, H. H. Boersma4, F. J. Voskuil3, A. H. de Haas5, S. Kruijff2, P. J. van der Zaag6, W. Kelder7; 1Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, NETHERLANDS, 2Surgery, University Medical Center Groningen, Groningen, NETHERLANDS, 3Oral and Maxillofacial Surgery, University Medical Center Groningen, Groningen, NETHERLANDS, 4Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, NETHERLANDS, 5Pathology and Medical Biology, Academic Breast Center Groningen, location Martini Hospital, Groningen, NETHERLANDS, 6Molecular Biophysics, University of Groningen, Zernike Institute, Groningen, NETHERLANDS, 7Surgery, Academic Breast Center Groningen, location Martini Hospital, Groningen, NETHERLANDS.
Introduction Tumor-positive margins are reported in 10-11% of patients undergoing breast-conserving surgery (BCS) for breast cancer and require additional therapy (boost radiotherapy or re-excision) to reduce local recurrence risk. Fluorescence imaging (FI) shows promise in reducing tumor-positive margins by intraoperative tumor tissue visualization using tumor-specific tracers. This clinical study aims to identify tumor-positive margins intraoperatively using FI with bevacizumab-IRDye800CW, and assess the feasibility of implementing FI in high-volume breast cancer centers. Methods Patients with early-stage breast cancer scheduled for BCS were included. Bevacizumab-IRDye800CW was administered intravenously 2-4 days prior to surgery. After the lumpectomy, fluorescence images of both the surgical cavity and resection specimen were obtained. While surgeons proceeded with the sentinel node procedure, the fluorescence images were analyzed and a tumor-to-background (TBR) ratio of higher fluorescence intensity spots were calculated. A TBR >1.5 was considered a risk of a tumor-positive margin and re-excision of the corresponding area was performed in consultation with the surgeon. All fluorescence images were correlated to histopathology results. Results In total, 53 patients completed all study procedures. Five patients (9.4%) initially had a tumor-positive margin. FI identified all those patients. Re-excision was performed in the same session, resulting in tumor-negative margins in all these cases. In 35 other patients re-excisions were performed because of a high TBR. In hindsight, they had negative margins, but the re-excision was generally performed at the side with the closest margin based on histopathology assessment. Incorporation of FI prolonged the average BCS time by a maximum of five minutes. Conclusion Targeted-fluorescence guided re-excisions prevented tumor-positive margins in 9.4% of patients undergoing BCS for breast cancer. In our study, no tumor-positive margins were missed and tumors were confirmed to be completely resected. With only a five-minute increase in surgery time, FI seems feasible to implement in a high-volume breast cancer center. Further optimization of tracer specificity and signal depth quantification is needed to improve accuracy and clinical applicability.
Presentation numberPS2-01-07
A Multicenter Retrospective Observational Study on the Safety and Timing of Radiotherapy After Oncoplastic Breast-Conserving Surgery
Rena Yamakado, Mie medical University, Tsu, Japan
R. Yamakado1, A. Noro1, R. Ito1, N. Imai1, M. Shibusawa1, M. Kimoto1, M. Yoshikawa1, K. Nakamura1, E. Hatakawa1, S. Watanabe1, M. Yoshida1, T. Mitsui1, E. Matsumoto2, C. Mizumoto2, N. Hanamura3, T. Nishimine3, Y. Kashikura3, M. Yamashita4, R. KoJima4, Y. Nomoto1, T. Ogawa2, K. Kawaguchi1; 1breast center, Mie medical University, Tsu, JAPAN, 2breast center, Japanese Red Cross Ise Hospital, Ise, JAPAN, 3breast center, Saiseikai Matsusaka General Hospital, Matsusaka, JAPAN, 4breast center, Mie prefectural general Medical center, Yokkaichi, JAPAN.
Background:Whole-breast irradiation after breast-conserving surgery (BCS) is a standard treatment for breast cancer. Boost radiation is recommended in cases with positive surgical margins, and recently, its application has expanded to younger patients with negative margins to reduce local recurrence. At our center and affiliated institutions in Mie Prefecture, oncoplastic breast-conserving surgery (OPBCS) has been actively implemented over the long term, aiming to achieve both oncological safety and favorable cosmetic outcomes. OPBCS includes volume displacement techniques using intra-breast tissue and volume replacement techniques using autologous tissue, which may obscure the tumor bed and margins due to extensive tissue rearrangement, raising concerns regarding radiotherapy targeting accuracy.Methods:This multicenter retrospective observational study included 4215 patients who underwent primary breast cancer surgery from January 2017 to December 2023. The study aimed to evaluate the safety and treatment outcomes of postoperative radiotherapy in patients who received OPBCS. The primary endpoint was ipsilateral breast tumor recurrence (IBTR), while secondary endpoints were overall survival (OS), event-free survival (EFS), the interval from surgery to initiation of radiotherapy, and the incidence of radiotherapy-related complications.Results:The median observation period was 55 months in the OPBCS group and 54 months in the non-OPBCS group. Patient characteristics revealed a higher rate of re-excision in the OPBCS group. IBTR showed no significant difference between the groups (p = 1.0; OR, 0.97; 95% CI, 0.16-4.59). OS was significantly better in the OPBCS group (p = 0.0284; OR, 0.22; 95% CI, 0.025-0.95), while EFS was comparable between the groups. The median interval from surgery to radiotherapy initiation was longer in the OPBCS group (48 days) than in the non-OPBCS group (40 days) (p < 0.001); however, no significant difference was observed for delays beyond 16 weeks. Multivariate analysis identified OPBCS as an independent factor associated with delayed initiation of radiotherapy. The OPBCS group also exhibited a significantly higher incidence of postoperative complications (p < 0.001; OR, 2.24; 95% CI, 1.52-3.31), which was considered a major contributor to the delay. Notably, the incidence of radiotherapy-related complications, excluding grade 1 dermatitis, did not differ between groups (p = 1.0). Discussion:Postoperative complications likely contributed to delayed radiotherapy in the OPBCS group. Surgeons may also intentionally delay radiotherapy initiation to preserve vascular supply in rearranged tissue. However, with a median delay of only 48 days (~7 weeks), and no difference in IBTR rates, OPBCS does not appear to negatively impact oncologic outcomes and should not be considered a barrier to its broader adoption.
Presentation numberPS2-01-08
A Decade Later: Our Updated Systematic Review and Meta-Analysis of Overall Survival, Disease-Free Survival, Local Recurrence, and Nipple-Areolar Recurrence Following Nipple-Sparing Mastectomy
Rachel N Rohrich, Medstar Georgetown University Hospital, Washington, DC
R. N. Rohrich1, H. Soltani1, I. A. Snee1, K. L. Fan1, L. De La Cruz2; 1Plastic and Reconstructive Surgery, Medstar Georgetown University Hospital, Washington, DC, 2Division of Breast Surgery, Medstar Georgetown University Hospital, Washington, DC.
Background: Since our 2015 manuscript, nipple-sparing mastectomy (NSM) continues to be an increasingly adopted technique in breast cancer management. However, concerns regarding long-term oncologic safety still persist. This systematic review and meta-analysis provides a ten year update on oncologic safety associated with NSM, specifically overall survival (OS), disease-free survival (DFS), local recurrence (LR), and nipple-areolar complex recurrence (NACR). Methods: A systematic review of the literature was performed according to PRISMA guidelines to identify studies reporting outcomes after NSM. Studies with internal comparison arms evaluating therapeutic NSM versus skin-sparing mastectomy (SSM) and/or modified radical mastectomy (MRM) were included in a meta-analysis of OS, DFS, and LR. Studies lacking comparison arms were only included in the systematic review, wherein pooled analyses for NSM patients were conducted for OS, DFS, LR, and NACR using a random-effects model. Weighted averages and 95% confidence intervals (CI) were calculated, and studies were stratified by duration of follow-up: 10 years. Results: A total of 60 studies representing a total of 7,752 patients undergoing NSM were included. The meta-analysis included 11 studies with comparison arms (Figure 1). Nine studies reported OS with a significant pooled risk difference of 2.1% favoring NSM over MRM/SSM (95% CI: 0.0-4.3%, p=0.047). Seven studies reported DFS, with a nonsignificant pooled risk difference of 4.8% favoring NSM (95% CI: -0.8-10.3%, p=0.093). Eleven studies reported LR, with a nonsignificant pooled risk difference of -0.7% (95% CI: -2.1-0.8%, p=0.35). The systematic review included all 60 studies. Weighted average follow-up ranged from 25.6 to 133.4 months across groups. Among patients with 10 years follow-up (n=391), OS was 82.0% (80.9-93.2), DFS 60.7% (58.8-62.7), LR 15.1% (13.9-16.2), and NACR 1.1% (1.0-1.1). Conclusions: This study continues to support that NSM is associated with excellent overall and disease-free survival with low local recurrence rates comparable to that of SSM/MRM as well as low NAC recurrence rates among appropriately selected patients. This updated data provide additional support for the long-term safety of NSM.
Presentation numberPS2-01-09
The Structural Mastectomy: Application of circum-mammary ligament preservation in nipple-sparing mastectomy
Rachel N Rohrich, Medstar Georgetown University Hospital, Washington, DC
L. De La Cruz1, R. N. Rohrich2, A. Junn2, M. E. Currin2, D. H. Song2, K. L. Fan2; 1Division of Breast Surgery, Medstar Georgetown University Hospital, Washington, DC, 2Plastic and Reconstructive Surgery, Medstar Georgetown University Hospital, Washington, DC.
Background: The anatomic boundaries of conventional nipple-sparing mastectomy (NSM) is poorly defined. As such, deeper support structures, such as the circummammary ligament, the anterior lamellar fascia, and adjacent perforators, lymphatics and nerves are disrupted, compromising perfusion, wound healing, and reconstructive outcomes. The structural mastectomy preserves these structures to enhance perfusion, reduce complications, and facilitate direct-to-implant (DTI) reconstruction without acellular dermal matrices in the prepectoral plane. This study evaluates reconstructive outcomes associated with structural versus conventional NSM with immediate DTI reconstruction. Methods: A retrospective review was conducted of all patients who underwent NSM with DTI reconstruction at a single institution between November 2020 and July 2024. Patients were stratified by mastectomy technique into those who received a structural NSM (s-NSM) and those who underwent conventional non-structural NSM. s-NSM was characterized by the preservation of the circummammary ligament, superficial fascial system, and associated neurovascular bundles through dissection along well-visualized planes through a 6 cm incision. Primary outcome was short-term (≤30 days) and long-term (>30 days) reconstructive complications. Results: A total of 122 patients (225 breasts) underwent conventional NSM (135 breasts, 60.0%) or s-NSM (90 breasts, 40.0%) with immediate DTI reconstruction. Baseline demographics, comorbidity profiles, and oncologic indications were similar between groups. Compared with conventional NSM, s-NSM was associated with shorter operative time (p=0.002), lower ADM use (55.6% vs. 94.1%, p<0.001), and greater rates of NAC reinnervation (70.0% vs. 26.7%, p<0.001). Oncologic outcomes were equivalent between groups. Short-term complications occurred less frequently following s-NSM (5.6% vs. 20.0%, p=0.003), including a significantly lower rate of partial nipple-areola complex necrosis (1.1% vs. 8.2%, p=0.030). On multivariate analysis, s-NSM remained independently protective against short-term reconstructive complications (OR 0.09, 95% CI 0.02-0.50, p=0.006), with good model discrimination confirmed by an area under the curve value (AUC) of 0.80. Long-term complication rates were also significantly less common following s-NSM (2.2% vs. 11.1%, p=0.018), though this did not remain statistically significant on multivariate analysis. The s-NSM was also associated with a significantly lower incidence of capsular contracture (2.2% vs. 11.1%, p=0.018). While fewer patients in the s-NSM cohort underwent elective aesthetic revision (11.1% vs. 20.0%, p=0.078), this did not reach statistical significance. Rates of positive margins (NSM: 3.7% vs. s-NSM: 3.6%, p=1.000), local recurrence (3.0% vs. 0.0%, p=0.529), distant metastasis (6.1% vs. 0.0%, p=0.117), and cancer-related death (5.3% vs. 0.0%, p=0.107) did not differ significantly. No differences in the instance of residual disease, tumor size, nodal metastasis, extranodal extension, or pathologic staging was observed between groups upon evaluation of final tumor pathology. Conclusion: For the first time, we demonstrate that structural preservation in breast surgery allows for efficacious single-stage reconstruction, limits ADM use, and improves reconstructive complication profiles early 1-year follow-up. While oncologic outcomes were not the primary focus of this study, overall survival and local recurrence rates were similar to those reported in prior series. The structural NSM may represent a paradigm shift in oncoplastic breast surgery in appropriately selected patients contributing to more favorable reconstructive outcomes.
Presentation numberPS2-01-10
De-escalation of sentinel lymph node biopsy beyond the Choosing Wisely guidelines
Tiffany Cheung, Cleveland Clinic, Cleveland, OH
T. Cheung1, S. Patil2, M. Nelis3, S. A. Valente1, J. E. Lang4; 1Breast Surgery, Cleveland Clinic, Cleveland, OH, 2Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, 3Obstetrics and Gynecology, Cleveland Clinic, Cleveland, OH, 4Breast Surgery and Cancer Biology, Cleveland Clinic, Cleveland, OH.
Background: The Choosing Wisely (CW) campaign issued by the Society of Surgical Oncology recommends omitting sentinel lymph node biopsy (SLNB) in women over age 70 with early-stage hormone receptor (HR) positive, HER2 negative breast cancer. The publication of the SOUND and INSEMA trials recently expanded indications for considering the omission of SLNB in younger women. We hypothesized that the rate of omission of SLNB has increased over time. Our primary objective was to define predictors of omitting SLNB and secondarily, to determine the incidence of patients beyond the CW population having omission of SLNB. Methods: We utilized the National Cancer Database (NCDB) to query between 2018-2021 for patients with cT1 or cT2 invasive breast cancer treated with breast surgery without use of neoadjuvant therapy. We recorded surgical procedure types, biomarkers, grade, the number of clinical and pathologic positive nodes, Charlson-Deyo Score, race/ethnicity, age, urban/rural setting, histology, and academic/community setting. Univariate analysis with the Pearson’s Chi-squared test and multivariable analysis with logistic regression were performed to identify factors associated with omission of SLNB. Univariate analysis was also performed to identify variables associated with rates of breast conserving surgery (BCS) versus mastectomy. Results: Overall, 433,737 patients met study inclusion criteria with 313,201 (72%) treated with BCS while 120,536 (28%) were treated with mastectomy. In our cohort, 133,169 (31%) were over age 70 and 368,228 (86%) were HR+/HER2-. Only 4219 patients (1%) had omission of SLNB, of which 3599 (88%) were HR+/HER2-, 247 (6.0%) were HER2+, and 238 (5.8%) were triple negative. Omission of SLNB did not increase over time with rates ranging from 0.06%-1.6% of the overall study population by year. On multivariable analysis, predictors of omitting SLNB were age >70 (OR 1.51, CI 0.86-0.97), non-zero Charlson-Deyo Score (p<0.001), HR+/HER2- (OR 1.09, CI 1.06-1.13), year of diagnosis 2021 (OR 1.56, HR 1.52-1.6), non-lobular histology (p<0.001), academic setting (p<0.001), and metropolitan setting (p<0.001). White race was associated with omitting SLNB but was only marginally significant on univariate analysis (p=0.047). Type of surgery as BCS was associated with omission of SLNB (p<0.001) on univariate analysis only. Most patients who had omission of SLNB had cT1 disease (n=3409 or 83% of patients having no nodal surgery while fewer patients with cT2 status had no nodal procedure (n=722, 17%), p<0.001). Of patients having no SLNB, 98% were cN0, 2% were cN1, 0.2% were cN2 and <0.1% were cN3. Conclusions: Rates of the omission of SLNB did not increase over time from 2018-2021 despite the publication of the Choosing Wisely recommendations in 2016 to consider omitting SLNB in patients over age 70 with HR positive, HER2 negative early-stage breast cancer. Only 1% of breast cancer patients had omission of SLNB despite 31% of the cohort being over age 70. Age >70, non-zero Charlson-Deyo Score, HR+/HER2- status, year of diagnosis 2021, non-lobular histology, academic and metropolitan setting were associated with omission of SLNB. These data provide estimates that may be used in the future to compare the impact of the SOUND and INSEMA trial on rates of omission of SLNB based on prospective randomized controlled trials that expand criteria for omission of SLNB beyond the Choosing Wisely guidelines.
Presentation numberPS2-01-11
Cumulative Medical Costs of Prepectoral versus Subpectoral Implant-Based Breast Reconstruction: A Retrospective Analysis of a Single Academic Center
David B Lipps, University of Michigan, Ann Arbor, MI
D. B. Lipps1, B. D. Baglien2, M. N. Saunders3, J. Vidergar4, G. L. Maica3, P. L. Myers2, M. Banerjee4, A. O. Momoh2; 1School of Kineisology, University of Michigan, Ann Arbor, MI, 2Plastic Surgery, University of Michigan, Ann Arbor, MI, 3Medical School, University of Michigan, Ann Arbor, MI, 4School of Public Health, University of Michigan, Ann Arbor, MI.
Background: Post-mastectomy breast reconstruction is a crucial component in improving psychosocial outcomes and quality of life for breast cancer survivors. Prepectoral implant-based breast reconstruction has gained traction over the past decade due to its potential to enhance aesthetic results and reduce muscle disinsertion-related complications. Wider adoption is limited by concerns over potentially higher healthcare costs compared to the previously traditionally favored subpectoral approach. Prior research on the cost implications of these methods has produced mixed results, often lacking comprehensive analyses of cumulative perioperative medical expenses. Methods: This retrospective cohort study analyzed data from women who underwent unilateral or bilateral mastectomy followed by immediate implant-based reconstruction at a single academic center between July 2017-June 2022. The study period marked a transition in institutional practice from predominantly subpectoral to prepectoral procedures. We examined billing charges from the index surgery and the downstream costs of patient care up to six months post-operation. Regression analyses assessed total index surgery costs, downstream costs, and cumulative costs, adjusting for variables such as age, BMI, and racial demographics. Results: The study analyzed data from 185 patients—86 underwent prepectoral and 99 underwent subpectoral reconstruction. Demographic analysis revealed no significant differences in baseline characteristics such as age, BMI, or intraoperative time between groups. Subpectoral reconstructions had significantly lower index surgery costs, with a 30.5% decrease in costs compared to prepectoral reconstruction (p < 0.001). While downstream costs up to six months post-surgery did not significantly differ, a trend towards reduced expenses was noted following subpectoral procedures (p = 0.094). Cumulative costs, driven by initial surgery expenses, were 29.2% lower in subpectoral cases (Table, p < 0.001). The increased use of acellular dermal matrix in prepectoral procedures was identified as a major factor contributing to higher costs. Conclusions: Our findings demonstrate that prepectoral implant-based reconstruction is associated with significantly higher surgical and cumulative costs compared to subpectoral reconstruction. No substantial differences were observed in postoperative care costs, suggesting that the increased expense of prepectoral approaches is primarily driven by the higher cost of surgical adjuncts during the index surgery. These results provide critical cost-related insights as prepectoral techniques, including the need to use lower-cost surgical adjuncts. Addressing these cost variables could facilitate broader implementation of prepectoral procedures, leveraging their potential patient-centered benefits.
| Predictor | Estimate | Std.Error | t-value | p-value |
| (Intercept) | 10.945 | 0.172 | 63.531 | <0.001 |
| Surgery:Subpectoral | -0.292 | 0.059 | -4.947 | <0.001 |
| Race:Other | -0.101 | 0.088 | -1.156 | 0.25 |
| BMI | -0.013 | 0.005 | -2.85 | 0.005 |
| Age | -0.003 | 0.002 | -1.234 | 0.22 |
Presentation numberPS2-01-12
Evaluation of different methods of targeting axillary lymph node for axillary surgery in patients with clinically node-positive breast cancer: A retrospective multicenter study
Cho Eun Lee, Samsung Medical Center, Seoul, Korea, Republic of
C. Lee1, J. Ryu1, S. Ahn2, J. Lee3, H. Shin4, J. Lee5, Y. Kwak6, H. Lee7, S. Nam1, S. Kim1, J. Lee1, J. Yu1, B. Chae1, S. Lee1, W. Park1, K. Kim1, S. Lee1; 1Department of Surgery, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 2Department of Surgery, Gangnam Severance Hospital, Seoul, KOREA, REPUBLIC OF, 3Department of Surgery, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF, 4Department of Surgery, Myongji Hospital, Seoul, KOREA, REPUBLIC OF, 5Department of Surgery, Soonchunhyang University Hospital, Seoul, KOREA, REPUBLIC OF, 6Department of Surgery, Chung-Ang University Hospital, Seoul, KOREA, REPUBLIC OF, 7Department of Surgery, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF.
Background: In patients with clinically node-positive breast cancer undergoing chemotherapy, sentinel lymph node biopsy has been associated with a false-negative rate up to 14% according to previous studies. However, when target axillary dissection techniques, such as the placement of clips or markers in biopsy-proven metastatic lymph nodes, are employed, false-negative rates have been reported to decrease dramatically to approximately 1.4%, as shown in a pivotal study published in 2016. Despite this evidence, there has been no large-scale evaluation of axillary lymph node targeting methods in Korean clinical setting. This study aims to establish foundational data for targeted axillary strategies in Korea by comparing different localization methods used in patients with initially node-positive breast cancer. Methods: This study was conducted as a retrospective multicenter study involving patients with biopsy-proven node-positive breast cancer, who underwent surgery between January 2015 and June 2024. Patients were enrolled from seven institutions based on the following inclusion criteria; primary unilateral breast cancer, clinical tumor stage cT1-T4, biopsy-proven metastatic axillary lymph node with marker placement, and age over 18 years. Detailed analyses were conducted regarding the extent of axillary surgery, the type of marker used (clip or tattoo), and the use of localization techniques such as wire localization. Results: A total of 331 patients were included and the mean age was 49.3 years with 31 months of median follow-up. According to marking methods, targeted lymph nodes were successfully identified in 127 out of 145 patients (87.6%) who received clip markers, and in 178 out of 184 patients (96.7%) who underwent tattoo marking. There were no significant differences in concordance between sentinel lymph node and targeted lymph node across the clip and tattoo group (66.2% vs. 79.3%, p=0.298). Discordance between the sentinel lymph node and targeted lymph node was observed in 12.1% of total patients, 13.8% of patients in the clip marker group and 10.9% in the tattoo marker group. Among the clip group, patients who underwent wire localization showed a non-significant trend toward lower concordance between sentinel lymph node and targeted lymph node compared to those without wire localization (66.7% vs. 83.3%, p=0.069). Demographics and treatment characteristics, including tumor stage, surgery type, and systemic therapy, did not differ significantly between different markers. Conclusions: This is the first multicenter analysis in Korea focusing on targeting methods for metastatic axillary lymph nodes. No significant difference was observed in sentinel lymph node-targeted lymph node concordance and targeted lymph node detection between the clip and tattoo marker groups, suggesting that both methods have comparable feasibility in planning targeted axillary surgery. Interestingly, tattoo-based marking was used at a frequency comparable to clip marking, which contrasts with global trends. This may be attributed to the limited use of clip markers in Korea due to reimbursement restrictions. Also, these findings are clinically meaningful when considered in the context of the well-established false-negative rate of approximately 14% reported in node-positive breast cancer patients undergoing sentinel lymph node biopsy, suggesting that appropriate lymph node targeting strategies may help reduce discordance and improve surgical accuracy.
Presentation numberPS2-01-13
Pixels to Prognosis: Predicting Response to NAST Using Multiparametric Imaging Combining Tomosynthesis, Shear Wave Elastography and Doppler
Gaurav Agarwal, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Lucknow, India
G. Agarwal1, S. Dariya1, S. Mayilvaganan1, G. Chand1, A. Mishra1, N. Mohindra2, V. Agrawal3, P. Mishra4; 1Department of Endocrine & Breast Surgery, SGPGIMS, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Lucknow, INDIA, 2Department of Radiology, SGPGIMS, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Lucknow, INDIA, 3Department of Pathology, SGPGIMS, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Lucknow, INDIA, 4Department of Biostatistics, SGPGIMS, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Lucknow, INDIA.
BACKGROUND: Predicting response to NAST may help tailor treatment, maximize pCR rates and de-escalate surgery (BCS, SLNB). We tested if multiparametric imaging- using mammography breast tomosynthesis (DBT), ultrasound shear wave elastography (SWE) & color Doppler after 2 NAST cycles (post2NAST) of their changes from baseline correlate with NAST response and predict pCR. METHODS: In this prospective study, 100 non-metastatic breast ca pts (39 TNBC, 31 HER2 enriched, 28 Luminal B like) receiving NAST underwent imaging at baseline, post2NAST, after completing NAST. Changes in tumor dimensions, volume, density, stiffness, vascularity were correlated with pCR & RCB after surgery. RESULTS: 40% achieved pCR. Post2NAST DBT density changes predicted pCR (p<0.0001); 92% patients with pCR had decreased density. Density post2NAST was strongly associated with response- 87% low-density tumors had low RCB (0-1), c.w. 88% persistent high-density tumors had poor response (RCB 2-3)- 87% sensitivity, 80% specificity. Post2NAST, ≥17.6% reduction in max diam predicted pCR with 82.5% sensitivity, 70% specificity (AUC = 0.817), while ≥39.4% vol. reduction predicted pCR with 87.5% sensitivity, 70% specificity (AUC = 0.824). Mean & max SWE values from tumor core, peritumoral zone & large ROI were studied. Only mean, max peritumoral SWE baseline→ post2NAST reduction was significantly assoc. with pCR (p=0.038, 0.020 respectively). Absolute SWE values post2NAST or post NAST across all regions didn’t distinguish responders. Baseline→ post2NAST reduction in Adler vascular score (semi-quantitative Doppler grading that classify vascularity/angiogenesis) was significantly assoc. with pCR, p=0.002. Patients achieving ‘No flow’ had highest pCR (71.4%). A ≥2-point drop demonstrated high 87% specificity for pCR. Mean 13-point vascular imaging score declined significantly (baseline 9.6→ 6.9 post2NAST). On ROC analysis, score ≤6 post2NAST predicted pCR with 77% sensitivity, 60% specificity (AUC = 0.695, p=0.001). Subtype analysis: HER2+ tumors had greatest DBT density reduction after NAST (87.1%, p=0.038), closely mirroring their highest pCR rates. TNBC &HER2+ subtypes had greater stiffness reductions on SWE post2NAST, esp. in peritumoral mean/max & large ROI values (AUC 0.70). While Adler score regression (to “no flow”) was commonest in TNBC, subtype difference didn’t reach significance. Luminal B tumors consistently showed least favorable imaging response. CONCLUSIONS: Early imaging response post2NAST, especially DBT density/volume reduction & peritumoral SWE stiffness reduction strongly predict pCR, outperforming absolute values. Multiparametric imaging combining DBT, SWE & Doppler vascular scores (Adler & 13-point) offers a non-invasive, early surrogate for treatment response, enabling timely therapy tailoring, and predicting pCR.
| Imaging Modality | Parameter | Area under curve (AUC) | Cut-off Value/ % | Sensitivity (%) | Specificity (%) | Significance (P value) | ||||||||||||||
| Digital Breast Tomosynthesis (DBT) | Ellipsoidal volume reduction | 0.824 | ≥39.4% | 87.5% | 70% | <0.001 | ||||||||||||||
| DBT | Max diameter reduction | 0.817 | ≥17.6% | 82.5% | 70% | <0.001 | ||||||||||||||
| Shear Wave Elastography (SWE) | Tumor SWE mean | 0.641 | ≤85.9 kPa | 72.5% | 49.2% | 0.017 | ||||||||||||||
| SWE | Tumor SWE max. | 0.631 | ≤85.0 kPa | 72.5% | 50.8% | 0.028 | ||||||||||||||
| SWE | Peritumoral SWE mean reduction | 0.680 | ≥31 kPa | ~85% | ~65-70% | 0.038 | ||||||||||||||
| SWE | Peritumoral SWE max. reduction | 0.700 | ≥32 kPa | ~85% | ~65-70% | 0.020 | ||||||||||||||
| Color Doppler | Adler score | – | No flow (score 0) | 25% | 86.7% | 0.002 | ||||||||||||||
| Color Doppler | 13-point vascular score | 0.695 | ≤6 | 77.5% | 60% | 0.001 |
Presentation numberPS2-01-14
Omission of sentinel lymph node biopsy among women age 50-70: applying updated ASCO guidelines to a real-world population
Meghan R Flanagan, University of Washington, Seattle, WA
E. C. Namoglu1, A. Meisner2, M. R. Flanagan3, E. F. Gillespie1; 1Department of Radiation Oncology, University of Washington, Seattle, WA, 2Public Health Sciences Division, Fred Hutch Cancer Center, Seattle, WA, 3Department of Surgery, University of Washington, Seattle, WA.
Background: In April 2025 the American Society for Clinical Oncology (ASCO) published practice-changing guidelines recommending the omission of sentinel lymph node biopsy (SLNB) in patients with early-stage breast cancer over the age of 50. We aimed to evaluate factors associated with a positive SLNB and the impact on adjuvant therapy among patients meeting updated ASCO Guideline recommendations for SLNB omission. Methods: A single-institution retrospective cohort study of patients 50-70 years old diagnosed with early-stage invasive breast cancer (cT1N0, grade 1-2, ER+, HER2-) between 2022-2024 who underwent SLNB. All patients were cN0 and pre-operative axillary ultrasound (axUS) was not required. The primary outcome was SLNB positivity (SLNB+), defined as at least pN1mi, and secondary outcome was adjuvant treatment. Categorical covariates were compared in univariate analysis using Fischer’s exact test. Multivariable logistic regression was used to evaluate factors associated with SLNB+, adjusting for relevant clinical characteristics. Results: Among 282 patients meeting ASCO guideline criteria, 275 (97.5%) had SLNB results available for analysis. Among the cohort with SLNB, median age was 63 years (IQR 58-66) and the majority (81.0%) were ductal histology (Table 1). Adjuvant therapy included hormonal therapy (87.0%), whole breast radiation (73.0%), partial breast radiation (19.0%) and chemotherapy (5.8%). SLNB was positive in 17 (6.2%) patients (9 pN1mi, 8 pN1a, 0 pN2). Among those with pN1a, two patients had 2 positive nodes (one was pT2 and one had Oncotype Recurrence Score [RS] of 29) and no patients had 3 nodes. Factors associated with SLNB+ included tumor grade, pathologic tumor stage and Oncotype RS. In multivariable analysis, younger age and Oncotype RS were associated with SLNB+ but not tumor grade or tumor stage (Table 1). Among the 17 patients with SLNB+, 5 (29.4%) received chemotherapy of which 4 had a high Oncotype RS and 1 was pT2 on final pathology. Per 2024 Updated ASTRO Guideline, APBI would have been suitable for 8, and cautionary or not recommended for 6 patients with SLNB+ based on risk factors alone (3 pT2, 2 lobular histology, 1 with lymphovascular invasion [LVI]), and potentially 3 other patients with Oncotype RS >25. Among 8 patients with pN1a, 2 had a high-risk feature (both Ki67>20%) indicating eligibility for adjuvant CDK4/6i per 2024 ASCO Guideline. Conclusion: In this cohort of 50- to 70-year-old patients with cT1N0 tumors meeting ASCO Guidelines for SLNB omission in the real-world setting of inconsistent pre-operative axUS, 6.2% were SLNB+. No decisions about chemotherapy would have been affected by the omission of SLNB. The positive SLNB could have impacted a total of 10 (3.6%) patients who may have received APBI instead of WBRT or not been eligible for CDK4/6i.
| Overall (N = 275) n(%) | SLNB-(N = 258) n(%) | SLNB+(N = 17)n(%) | p-value (Fischer’s exact test) | Adjusted Odds Ratio of SLNB+ (95%CI) | |||||||
| Age at Diagnosis | 0.249 | Continuous | |||||||||
| 50-54 | 27 (9.8) | 24 (89.0) | 3 (11.0) | 0.898 (0.802, 0.998) | |||||||
| 55-59 | 66 (24.0) | 60 (91.0) | 6 (9.1) | ||||||||
| 60-64 | 69 (25.0) | 65 (94.0) | 4 (5.8) | ||||||||
| 65-70 | 114 (41.0) | 109 (96.0) | 4 (3.5) | ||||||||
| Histology | 0.886 | ||||||||||
| Ductal | 223 (81.0) | 208 (93.0) | 15 (6.7) | ||||||||
| Lobular | 38 (14.0) | 35 (95.0) | 2 (5.4) | ||||||||
| Mixed/Other | 15 (5.4) | 15 (100.0) | 0 (0.0) | ||||||||
| Pathologic T Stage | 0.011 | ||||||||||
| PT1a | 42 (15.0) | 41 (98.0) | 1 (2.4) | Reference | |||||||
| PT1b | 99 (36.0) | 96 (98.0) | 2 (2.0) | 0.303 (0.021, 7.513) | |||||||
| PT1c | 121 (44.0) | 110 (91.0) | 11 (9.1) | 0.602 (0.07, 13.124) | |||||||
| PT2 | 14 (5.1) | 11 (79.0) | 3 (21.0) | 2.699 (0.217, 67.323) | |||||||
| Pathologic Grade | 0.023 | ||||||||||
| 1 | 141 (51.0) | 137 (97.0) | 4 (2.8) | Reference | |||||||
| 2 | 135 (49.0) | 121 (90.0) | 13 (9.7) | 3.06 (0.934, 12.188) | |||||||
| Oncotype Recurrence Score (RS) | <0.001 | ||||||||||
| High | 18 (6.5) | 14 (78.0) | 4 (22.0) | Reference | |||||||
| Intermediate | 90 (33.0) | 80 (90.0) | 9 (10.0) | 0.289 (0.071, 1.285) | |||||||
| Low | 28 (10.0) | 25 (89.0) | 3 (11.0) | 0.416 (0.066, 2.363) | |||||||
| Unknown/Not Done | 140 (51.0) | 139 (99.0) | 1 (70.0) | 0.021 (0.001, 0.211) | |||||||
| Lymphovascular Invasion (LVI) | 0.511 | ||||||||||
| Not Present | 265 (96.0) | 248 (94.0) | 16 (6.1) |
Presentation numberPS2-01-15
Long-term outcomes of sentinel lymph node biopsy alone for breast cancer with residual axillary nodal disease (ypN+) following neoadjuvant chemotherapy: a systematic review and meta-analysis.
Mariam Rana, University of Saskatchewan, Regina, SK, Canada
M. Rana1, A. C. Weiss2, A. D. Laws3, C. Mita4, T. King5; 1Surgery, University of Saskatchewan, Regina, SK, CANADA, 2Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 3Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB, CANADA, 4Countway Library, Harvard University, Harvard Medical School, Boston, MA, 5Department of Surgery, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Boston, MA.
Background: The optimal management of the axilla following neoadjuvant chemotherapy (NAC) for initially node-positive breast cancer (cN+) remains contentious, although there has been a trend towards de-escalating morbid axillary surgery under certain conditions. Clinically node-positive breast cancer that converts to node-negative (ypN0) following NAC is increasingly being managed by sentinel lymph node biopsy (SLNB) alone rather than by axillary lymph node dissection (ALND). Recent consensus by experts in St. Galen [Ditsch et al, 2025] suggest SLNB alone may also be acceptable for low volume residual axillary nodal disease (ypN1) following NAC, although data to support the oncologic safety of this approach are limited. The aim of this meta-analysis was to evaluate the long-term oncologic outcomes of SLNB alone for residual axillary nodal disease following NAC. Methods: A systematic review and meta-analysis was conducted according to PRISMA guidelines. Medline (Ovid), Embase, and Cochrane Central Registry were systematically searched for studies comparing women undergoing SLNB or ALND following NAC for initially clinically node-positive breast cancer (cN+). Included studies reported one of the following outcomes: axillary recurrence (AR), locoregional recurrence (LRR), distant recurrence (DR), disease-free survival (DFS) or overall survival (OS). A random effects meta-analysis was used to calculate weighted pooled effect estimates (risk ratios, RR) for all outcomes, comparing SLNB alone with ALND for patients with residual nodal disease (cN+/ypN+). Variability across studies due to heterogeneity was estimated using I2 statistics. Results: Fourteen observational studies were eligible for meta-analysis, providing data for 9,518 patients. The median age of the women included in the studies ranged from 47 to 53 years. Seven studies included only cN1 cases (pre-NAC), with 8 studies reporting outcomes for only ypN1 cases. The median number of sentinel lymph nodes retrieved in each study ranged from 2 to 6. The median follow-up time ranged from 28 to 108 months. The rates of axillary recurrence ranged from 0.2% to 7.8% across all included studies. Distant recurrence rates ranged from 15.7% to 38.6%. No significant differences were observed in AR between patients undergoing SLNB alone versus ALND following NAC: pooled RR 1.12 (95% CI: 0.72-1.74, I2=0.0%). Similarly, no significant differences were observed in LRR (RR 0.97, 95% CI: 0.68-1.37, I2=0.0%) nor overall mortality (RR 0.96, 95% CI: 0.65-1.42, I2=58.3%) between the two groups. However, the pooled risk of distant recurrence was lower for SLNB alone compared to the ALND group (RR 0.75, 95% CI: 0.60-0.95, I2=0.0%). Conclusions: This meta-analysis suggests that long-term outcomes of SLNB alone for breast cancer with residual axillary nodal disease following NAC are not inferior to those of ALND. AR rates were low across all included studies; the majority of recurrences were distant, questioning the need for aggressive axillary surgery for patients with low volume residual nodal disease. We await prospective data from several randomized clinical trials, including Alliance A011202 and MAC 19, to further guide axillary management in this group of patients. References:Ditsch et al (2025) St. Gallen/Vienna 2025 Summary of Key Messages on Therapy in Early Breast Cancer from the 2025 St. Gallen International Breast Cancer Conference. https://doi.org/10.1159/000546080
Presentation numberPS2-01-16
Feasibility of Systematic Breast-Conserving Surgery Under Local Anesthesia and Sedation: A Large Single-Center Study
Delphine Hequet, Institut Bourdonnais, Paris, France
D. Hequet1, M. Cittanova2, R. Salmon3, M. Wilhelm1, G. Aubry1, C. Gout Duracher2, J. Seror1; 1Surgery, Institut Bourdonnais, Paris, FRANCE, 2Anesthesia, Clinique Saint Jean de Dieu, Paris, FRANCE, 3Surgery, Clinique Saint Jean de Dieu, Paris, FRANCE.
Background : Local anesthesia with sedation (LAS) is widely used in other surgical disciplines but remains underutilized in breast-conserving surgery (BCS), where general anesthesia (GA) is still the standard [1, 2]. Recent reports have demonstrated the potential of LAS in selected breast surgery cases [3-5]. We aimed to evaluate the feasibility of systematically performing BCS under LAS in a high-volume breast cancer center.Methods : We conducted a retrospective study including all patients who underwent BCS ± axillary surgery from September 2021 to December 2023 in our institution. Patients operated under GA during the initial period (Sept 2021-March 2022) were compared to those under LAS (June 2022-Dec 2023), excluding the transition phase. LAS combined intravenous sedation (midazolam, dexmedetomidine, ketamine, remifentanil) with local infiltration (naropeine, lidocaine with epinephrine, saline). Feasibility endpoints included conversion to GA, intraoperative tolerance, and early surgical complications.Results : Of the 948 BCS procedures performed during the study period, 708 (75%) were carried out under LAS. Among the eligible patients during the LAS period, 12 refused local anesthesia and were operated under GA. Only 2 conversions to GA occurred during LAS procedures (0.3%), confirming excellent feasibility. We planned more preoperative wire in the LAS group (80% vs. 59%) because of the aqueous environment making small lesions more difficult to feel. Re-operation rates (for positive margins, hematoma, or infection) did not differ between LAS and GA groups (11% in LAS group vs. 13% in GA group, p=0.34). Axillary procedures were performed in both groups (52% in LAS group and 58% in GA group, n=0.06). Oncoplastic techniques were successfully performed under LAS without compromising safety or outcomes. Conclusions : Our results confirm that systematic BCS under LAS is feasible, safe, and applicable to a wide range of breast surgical procedures, including oncoplastic and axillary surgeries. To our knowledge, this represents the largest reported single-center series of breast-conserving surgery performed under LAS, supporting its broader implementation in routine clinical practice and perioperative optimization strategies.References
- D’Alonzo M, Martincich L, Biglia N, et al. Feasibility of breast surgery under local anesthesia in elderly patients. J Surg Oncol. 2020;121(3):446-453.
- Fischer JP, Nelson JA, Sieber B, et al. Propensity-matched analysis of local versus general anesthesia for outpatient breast surgery. Ann Surg Oncol. 2014;21(10):3274-3280.
- Boughey JC, Goravanchi F, Pockaj BA, et al. Anesthesia considerations in breast surgery. Breast J. 2020;26(4):727-732.
- Peled AW, Foster RD, Esserman LJ. Increasing role of local anesthesia with sedation in breast surgery. Ann Surg Oncol. 2015;22(10):3336-3342.
- Fernandez-Rodriguez M, Castano-Oreja MT, et al. Local anesthesia and sedation in breast-conserving surgery: A valid alternative to general anesthesia. Clin Transl Oncol. 2022;24(3):473-480.
Presentation numberPS2-01-17
Long term outcome of patients having Breast Conserving Surgery and Immediate Lipofilling
James Lucocq, Western General Hospital, Edinburgh, United Kingdom
J. Lucocq, R. Swan, A. Klenjnotowska, J. Dixon; Edinburgh Breast Unit, Western General Hospital, Edinburgh, UNITED KINGDOM.
Background: The impact of immediate lipofilling in the setting of breast conserving surgery on oncological outcomes are unknown. There is concern from in-vitro studies that adipose-derived stem cells may promote cancer recurrence through their proliferative properties. The aim of the present study was to investigate the long-term safety of breast conservation surgery (BCS) combined with immediate lipofilling. Methods: The patient cohort included 117 consecutive patients with invasive breast cancer or ductal carcinoma in situ (DCIS) who had BCS plus immediate lipofilling from 2011 to 2019 and 248 controls. Control patients had BCS without lipofilling in the same week as patients having BCS with lipofilling. Propensity score matching (PSM) was performed to determine the impact of immediate lipofilling on outcome where treatment (lipofilling) and control (no lipofilling) groups were matched for pathological and treatment variables. Multivariate survival analysis was performed to investigate the impact of lipofilling in subgroups according to T-stage, N-stage and tumour grade. The median follow up was 127 months. Results: There were 3 (2.5%) in-breast recurrences in lipofilled patients and 9 (3.6%) in the control group.When comparing lipofilled and non-lipofilled groups, there was no significant difference in overall survival (10-year, 94.8% vs. 93.3%; p=0.392), local recurrence (3.6% vs. 3.8%; p=0.601), regional recurrence (1.0% vs. 1.3%; p=0.761) and distant metastasis (5.8% vs. 3.3%; p=0.361). After propensity matching, lipofilling was not associated with mortality (log-rank, p=0.14), local recurrence (p=0.64), regional recurrence (p=0.91), or distant recurrence p=0.73). Across all pathological subgroups investigated, immediate lipofilling had no impact on overall survival or local recurrence (p>0.05). Conclusion: There is no evidence that immediate lipofilling performed at the same times as breast conserving surgery has any impact on mortality or on local, regional, systemic recurrence. This study shows the oncological safety of BCS plus immediate lipofilling.
Presentation numberPS2-01-18
Targeted Axillary Dissection after Neoadjuvant Systemic Chemotherapy in patients with Early Breast Cancer: Estimating the Real-World Impact of Omitted Axillary Lymph Node Dissection using the Canto Cohort
Angelica Conversano, GUSTAVE ROUSSY, Villejuif, France
A. Conversano1, F. Loi2, J. Blanc3, E. El Rassy2, A. Di Meglio2, J. M Ribeiro2, A. Viansone2, A. Puchar1, J. Zeitoun1, A. Turpin1, A. Martin4, M. Fournier5, M. Gutowski6, C. Cheymol7, B. Sauterey8, P. Cottu9, A. Bertaut3, B. Pistilli2; 1Department of Breast and Plastic Surgery, GUSTAVE ROUSSY, Villejuif, FRANCE, 2Department of Medical Oncology, GUSTAVE ROUSSY, Villejuif, FRANCE, 3Methodology and Biostatistics Unit, Centre Georges François Leclerc, Dijon, FRANCE, 4Data and Partnerships Department, UNICANCER, Paris, FRANCE, 5Oncology Department, Institut Bergonié, Bordeaux, FRANCE, 6Department of Surgical Oncology, Institut Régional du Cancer, Montpellier, FRANCE, 7Oncology Department, Centre Oscar Lambret, Lille, FRANCE, 8Oncology Department, Institut de Cancérologie de L’Ouest, Angers, FRANCE, 9Oncology Department, Institut Curie, Paris, FRANCE.
Background Targeted Axillary Dissection (TAD), which combines sentinel lymph node biopsy with excision of a pre-marked target lymph node, has been shown in clinical trials to improve staging accuracy and reduce false-negative rates in patients with early breast cancer (EBC) and clinically positive lymph nodes (cN+) undergoing neoadjuvant systemic therapy (NAST). By identifying NAST responders with limited residual nodal burden, TAD would enable omission of ALND in selected cases, thus reducing surgical morbidity. However, its potential applicability and impact in unselected real-world populations remain unclear. Methods Using data from the CANTO cohort (NCT01993498), a large French prospective study including patients with stage I-III EBC, we consecutively identified patients with cT1-T4 cN+ EBC treated with NAST between May 2012 and August 2022 who underwent ALND according to national guidelines implemented during the same period. The primary outcome was the proportion of patients who would have avoided ALND if TAD had been available, defined as the percentage of patients with cN+ EBC treated with NAST who achieved axillary pCR, according to changes in surgical management of the axilla consistent with ongoing international trials (NCT05462457, NCT04373655, NCT06092892, NCT04744506) and future guideline proposals. Additional outcomes included 3-year and 5-year rate of axillary recurrence, any invasive recurrence and the impact of ALND on quality of life (QoL) using arm symptom score on QLQ BR23. Results In total, 370 patients with cN+ EBC treated with NAST in the CANTO cohort were included, of whom 56 (15.1%) stage T1, 220 (59.5%) T2, 94 (25.4%) T3. Most of them were N1 (335 pts, 90.5%), 32 (8.7%) N2 and 3 (0.8%) N3. Pathological axillary response was available for 241 patients and 82/241 (34.0%) achieved complete pathological axillary response. Rates of complete response by BC subtype were as follows: 23/65 (35.4%) with HR-/HER2-, 37/76 (48.7%) with HER2+ and 22/100 (22.0%) with HR+/HER2- EBC. Pathological breast response was available for 255 patients and 100/255 (39.2%) had complete breast pCR. A dissociated pCR (axilla-breast) was observed in 62 patients of whom, 37 with residual cancer in axilla and 25 in the breast. Adjuvant treatments were: 356 (96.2%) locoregional radiotherapy, 27 (7.3%) chemotherapy, 224 (60.7%) hormonotherapy, 1 (0.3%) immunotherapy, 28 (7.6%) HER2-directed therapy (TDM-1, pertuzumab, trastuzumab). At a median follow-up of 93 months, 3 year-DFS was 78.3% (95%CI,73.8%;82.2%), with axillary recurrence 7.3% (n= 19/101 (18.8%) with HR-/HER2-, n= 2/122 (1.6%) with HER2+ and n=6/146 (4.1%) with HR+/HER2-; p<0.001), local recurrence 2.4%, distant relapse 16.5%. 5 year-DFS was 72.2% (95%CI, 67.2%; 76.5%), with axillary recurrence 8.4% (n=19/101 (18.8%) with HR-/HER-, n=3/122 (2.5%) with HER2+ and n=9/146 (6.2%) with HR+/HER2-; p<0.001), local recurrence 3.2%, distant relapse 20.8%. Overall, 108/323 (33.4%) patients experienced lymphoedema/arm pain after ALND. Conclusions This analysis showed that TAD could have been considered instead of ALND in around one third of patients treated with NAST, particularly those with HR-/HER2- or HER2+ EBC, having a higher likelihood of complete pathological response in the axilla. These findings underline the potential of TAD to replace ALND, especially when guided by clinical response and tumor subtype. Bibliography 1. Vaz-Luis I. ESMO Open. 2019;4(5):e000562. 2. Kuemmel S. Ann Surg. 2022 Nov 1;276(5):e553-e562. 3. Cabioglu. Ann Surg Oncol. 2025 Feb;32(2):952-966. 4. Zatecky L. World Journal of Surg Oncol, (2023) 21:252. 5. Nijveldt JJ. Ann Surg Oncol. 2024 Jul;31(7):4477-4486. 6. Kuemmel S. JAMA Surg. 2023 Aug 1;158(8):807-815
Presentation numberPS2-01-20
Unexpected invasive breast cancer after mastectomy for in situ carcinoma and implication on axillary management – data from a real-world collective
Elna Kuehnle, Hannover Medical School, Hannover, Germany
E. Kuehnle1, L. Steinkasserer1, C. Mueller1, H. Kuehnle2, K. Luebbe3, K. Noeding4, S. Noeding5, M. Arfsten6, P. Hillemanns1, T. Park-Simon1; 1Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, GERMANY, 2Department of Human Centered Design and Engineering, University of Washington, Seattle, WA, 3Breast Center, DIAKOVERE Henriettenstift, Hannover, GERMANY, 4Department of Gynecology and Obstetrics, HELIOS Klinikum Hildesheim, Hildesheim, GERMANY, 5Department of Gynecology and Obstetrics, Gynäkoonkologische Praxis am Pelikan Platz, Hannover, GERMANY, 6Breast Center Schaumburg, Kreiskrankenhaus Stadthagen, Stadthagen, GERMANY.
Purpose: Mastectomy for large in situ carcinoma is a commonly used therapy. Currently most national and international guidelines recommend sentinel lymph node biopsy because unexpected invasive breast cancer after mastectomy is an indication for sentinel lymph node staging. This cannot be performed after mastectomy and complete axillary dissection would be necessary in these cases. SLN as well as axillary dissection both can cause lifelong complications, especially arm movement impairment. Therefore, the necessity for this additional procedure needs to be addressed. We sought to gain insight into the frequency of unexpected upstaging to invasive breast cancer in case of mastectomy for in situ carcinoma and invasive nodal involvement in a real-world collective. Methods: Data collection was conducted from 2012-2024 in six certified breast cancer centers using a personal interview and data from the patients’ medical records. All patients had histologically confirmed pure DCIS or LCIS prior to mastectomy. All preoperatively diagnosed invasive breast cancer cases were excluded from final analysis. Descriptive statistics and multivariate analyzes were utilized to identify risk factors for upstaging from in situ carcinoma to invasive breast cancer. Results: The study collective included 4307 patients with primary breast cancer or in situ carcinoma. 237 preoperatively diagnosed in situ carcinomas were analyzed. 42/237 (18%) cases were upstaged to invasive breast cancer and positive lymph nodes were found in 8/237 (3%) cases. 71 cases of primary and secondary mastectomy were performed with an upstaging to invasive breast cancer occurring in 19 cases (31%). SLN biopsy in combination with mastectomy was performed in 60 patients and 3 patients with positive lymph nodes were detected. Looking into risk factors for unexpected upstaging on multivariate analyzes only a Ki67 higher then 20% was significantly correlated with an upstaging to invasive carcinoma (p = 0.033). Conclusions: The frequency of unexpected detection of invasive breast cancer in mastectomy for in situ carcinoma, is relatively low, despite comprehensive clinical work up including core needle biopsy. The likelihood of positive nodal involvement is also rather low but remains apparent and has clinical implications for adjuvant therapies. Still, postoperative and long-term complications of SLN biopsy remain an issue. Therefore, omission of SLN biopsy during mastectomy for in situ carcinoma should be discussed individually. Risk factors for upstaging need to be identified and long-term follow up is needed. Novel approaches to a two stage SLN biopsy offer surgical strategies to avoid unnecessary axillary interventions.
Presentation numberPS2-01-21
Risk Factors for Fat Necrosis Following Abdominally Based Autologous Breast Reconstruction
Michael Choi, University of California, San Francisco, San Francisco, CA
M. Choi, A. J. Lopes, C. M. Vargas, P. Tewari, D. S. Rouhani, R. Behnam-Hanona, J. Brown, M. Lem, A. Gozali, N. Parmeshwar, M. L. Piper; Plastic and Reconstructive Surgery, University of California, San Francisco, San Francisco, CA.
Background Fat necrosis is one of the most common complications following autologous breast reconstruction. The clinical presentation of fat necrosis can mimic that of a cancerous recurrence, inducing patient anxiety and necessitating further imaging and biopsies. Furthermore, severe cases can compromise cosmetic outcomes, often requiring revisional surgery. Despite its prevalence and clinical significance, identification of specific risk factors for the development and severity of fat necrosis is poorly understood. This study, therefore, aims to identify and examine these risk factors to improve patient outcomes. Methods We performed a retrospective review of patients who underwent autologous breast reconstruction with an abdominally based free flap at a single academic institution between September 2005 through January 2024. Data collected included demographics, surgical details, and post-operative complications (fat necrosis, infection, delayed wound healing, seroma, hematoma, necrosis, revision surgery). Chi-square and independent t-test analysis were performed to identify differences between groups, and multivariate logistic regressions were performed to assess predictors of fat necrosis. RESULTS A total of 434 patients (706 breasts) were included in this study. The median length of follow up after reconstruction was 4.2 years. 157 breasts (22%) developed fat necrosis, of which 68 breasts (43%) which underwent surgical excision. Average age (p=0.03) and BMI (p=0.01) were both significantly higher in the cohort that developed fat necrosis. Neither smoking status nor the presence of pre- or post-mastectomy radiation impacted rates of fat necrosis. Increased venous coupler size (p=0.01) and use of a bipedicled flap (p=0.000) were also significantly associated with the cohort that developed fat necrosis. Flap type (DIEP, msTRAM, SIEA) did not impact rates of fat necrosis. No significant differences in demographics or surgical details were found between patients that developed fat necrosis that did or did not warrant surgical management. On multivariate logistic regression, age (p=0.009, OR: 1.02), BMI (p=0.003, OR: 1.06), venous coupler size (p=0.04, OR: 1.58), and bipedicled flap reconstruction (p=0.00, OR: 6.05), were identified as independent predictors of developing fat necrosis. CONCLUSION This study identifies advanced age, higher BMI, the use of larger venous couplers, and bipedicled flap as independent risk factors for the development of fat necrosis following autologous breast reconstruction. These findings can help guide preoperative patient counseling and intraoperative decision-making.
| Predictor of Any Fat Necrosis | Odds Ratio | 95% CI | p |
| Age |
1.04 |
1.01-1.06 |
0.001 |
| BMI |
1.07 |
1.03-1.11 |
0.002 |
|
Venous Coupler Size (mm) |
1.62 |
1.03-2.54 |
0.04 |
|
Bipedicled Flap Reconstruction |
6.74 |
2.80-16.23 |
0.000 |
Presentation numberPS2-01-22
Changes in reoperation rate after publication of guidelines defined positive margins for breast conserving surgery: Collaboration study of Japanese Breast Cancer Society
Takehiko Sakai, Cancer Institute Hospital of JFCR, Tokyo, Japan
T. Sakai1, M. Ishitobi2, Y. Sagara3, A. Yoshida4, Y. Takahashi5, T. Tsukioki5, K. Takada6, Y. Ono7, Y. Kimura1, T. Osako8; 1Breast Surgical Oncology, Breast Center, Cancer Institute Hospital of JFCR, Tokyo, JAPAN, 2Department of Breast Surgery, Osaka Habikino Medical Center, Habikino, JAPAN, 3Breast Surgical Oncology, Sagara Hospital, Hakuaikai Medical Corporation, Kagoshima, JAPAN, 4Breast Surgical Oncology, St. Luke’s International Hospital, Tokyo, JAPAN, 5Breast Surgical Oncology, Okayama University Hospital, Okayama, JAPAN, 6Department of Breast Surgery, Osaka Metropolitan University, Osaka, JAPAN, 7Department of Radiation Oncoloty, Kyoto University, Kyoto, JAPAN, 8Division of Pathology, Cancer Institute of Japanese Foundation for Cancer Research, Tokyo, JAPAN.
Background: In 2014, the SSO-ASTRO consensus guidelines redefined a positive margin requiring re-excision after breast-conserving surgery (BCS) as “tumor on ink.” The adoption of this standard has been reported internationally to significantly reduce the rate of surgical re-excision for positive margins. In Japan, this definition was explicitly incorporated into the 2018 Japanese Breast Cancer Society clinical practice guidelines. To clarify changes in re-excision rates before and after guideline publication in Japan, we conducted a multicenter collaborative study. Method 1: A nationwide questionnaire survey was distributed to representatives of 521 Japanese Breast Cancer Society-accredited institutions to assess current practices related to BCS, including imaging, surgical and pathological assessment, rates of positive margins, and re-excision. Method 2: Multicenter cohort study was performed using databases from seven participating institutions, examining trends in positive margin and re-excision rates among patients undergoing BCS from 2008 to 2022. Result 1: Questionnaire Survey The questionnaire survey collected responses from 262 out of 521 Japanese Breast Cancer Society-accredited institutions, representing a response rate of 55.7%. Most institutions (60%) performed between 100 and 300 breast surgeries annually, and the majority (64%) had one or two breast surgeons. A full-time pathologist was present in 89% of the institutions, and preoperative MRI was utilized in 97% of cases for disease extent assessment. Notably, the proportion of institutions reporting a positive margin rate of 0-10% increased from 47% to 63% around 2018, reflecting a shift in clinical practice following the introduction of new guidelines. However, only 18% of institutions noted a decrease in re-excision rates after a positive margin post-2018, while 77% reported no change. Despite this, the proportion of institutions with a re-excision rate of 0-10% has increased slightly from 45% to 49% since 2018, indicating that more facilities have decreased their reoperation rates. Result 2: Multicenter Cohort Survey The multicenter cohort survey analyzed data from 18,688 cases of breast-conserving surgery performed between 2008 and 2022 across seven participating institutions. Over this period, the positive margin rate showed a consistent decline, decreasing from 10.7% in 2008-2014 to 8.8% in 2015-2018, and further to 6.9% in 2019-2022. Similarly, the re-excision rate dropped from 7.6% to 6.6% and then to 4.9% across the same intervals. Multivariate analysis identified several significant predictors of re-excision: younger patient age, diagnosis of invasive lobular carcinoma, larger tumor size, and presence of lymph node metastasis. Additionally, surgeries performed before 2014 (OR 1.34, 95% CI 1.34-1.88, P<0.001) and before 2018 (OR 1.33, 95% CI 1.10-1.60, P=0.003) were associated with a higher likelihood of re-excision compared to those performed after 2019, underscoring the impact of guideline implementation on surgical practice. Conclusions: Our findings suggest considerable diversity in the approach to BCS margin assessment among Japanese breast surgeons. Following the publication of both international and domestic guidelines regarding positive margins, a significant reduction in re-excision rates after BCS was observed in Japan. This trend was gradual, corresponding to the 2014 and 2018 consensus statements, indicating that formal guideline publication has contributed to reducing re-excision rates after breast-conserving surgery.
Presentation numberPS2-01-23
The accuracy of sentinel lymph node biopsy in locally advanced breast cancer
Alexander Petrovsky, N.N. Blokhin Russian Cancer Research Center, Moscow, Russian Federation
A. Petrovsky, M. Kurbanova, M. Frolova, V. Amosova, E. Artamonova, I. Stilidi; Breast cancer, N.N. Blokhin Russian Cancer Research Center, Moscow, RUSSIAN FEDERATION.
Introduction: Sentinel lymph node biopsy (SLNB) is the standard procedure for early breast cancer (BC), however its role in patients with locally advanced tumors (cT4N0) after neoadjuvant hormonal or chemotherapy remains a subject of discussion. The data on this type of patients is limited and the rate of false-negative results exceeds 10%/ Purpose of the study: to assess the diagnostic accuracy SLNB in patients with cT4N0M0 (ycN0) breast cancer after neoadjuvant therapy. Materials and methods: Our single-center prospective cohort study included 50 consecutive patients with cT4N0M0 breast cancer who received NALT followed by mastectomy with SLNB followed by level I-II axillary lymph node dissection (ALD). A radioguided method (99mTc-albumine nano colloid) was used to identify sentinel lymph nodes. The detection rate of sentinel lymph nodes, the level of false-negative results (FNR), sensitivity, and negative predictive value were evaluated. The univariate regression analysis was conducted to determine the factors that affect the frequency of false-negative results. Results: The detection rate was 96% (48/50). Metastatic lymph nodes were detected in 15 of 48 patients 31.3% during SLNB. After ALD metastasis were detected in 19 of 48 patients (39,5%). False negative rate (FNR) was 8,3% it the cohort. Sensitivity was 82,6%, Specificity 100%, and negative predictive value was 91,5%. Total accuracy was 92,3%. If 1 or 2 sentinel lymph nodes were removed FNR was 30,8% (4/13), if 3 or more lymph nodes were detected, FNR was 0% (0/35) (p=0,012). In univariate regression analysis only in the number of removed SLN increases the frequency of FNR. Patient’s age, BMI, molecular subtype, tumor grade, type of neoadjuvant therapy (chemo/hormonal) didn’t affect the accuracy of SLMB. Conclusion: The radioguided SLMB demonstrates acceptable diagnostic accuracy in patients with cT4N0 breast cancer after neoadjuvant therapy, which allows it to be an alternative to ALD, if at least three SLN are removed.
Presentation numberPS2-01-24
Efficacy and safety of intercostal nerve anastomosis in immediate biplane breast reconstruction after nipple-areola-sparing mastectomy: a randomized, controlled, open-label clinical study
Zhang Juan, Tangshan People’s Hospital, Tangshan, China
Z. Juan1, H. Cai1, Y. Liang2; 1Breast surgery, Tangshan People’s Hospital, Tangshan, CHINA, 2Ultrasound, Tangshan People’s Hospital, Tangshan, CHINA.
Efficacy and safety of intercostal nerve anastomosis in immediate biplane breast reconstruction after nipple-areola-sparing mastectomy: a randomized, controlled, open-label clinical study AbstractPurpose: to investigate the efficacy and safety of intercostal nerve anastomosis among breast cancer patients who undergo immediate biplane breast reconstruction after nipple-areola-sparing mastectomy.Methods: From 2023 to 2025, 20 to 60 year-old female breast patients (stage I-IIIA), who required and were willing to undergo immediate biplane breast reconstruction after nipple areola-sparing mastectomy, were screened and assigned to take the operation with (treatment group) or without (control group) intercostal nerve anastomosis (selected and dissociated the nerves with appropriate length and thickness among the 2nd to 4th intercostal nerves, then anastomosed to the posterior areola tissue). A radial incision at the surface projection of the tumor location was used. Using Semmes-Weinstein monofilaments to evaluate the patients’ breast local before the operation as well as at 15 days, 3 months, and 6 months postoperatively. Additionally, the patients’ quality of life was subjectively assessed using the BREAST-Q scale at 6 months after surgery. Adverse events, operation duration, drainage volume, and the duration of drainage tube carrying time were also monitored and recorded.Results: Compared to the control group, intercostal nerve anastomosis significantly improved the local sensation at 15 days, 3 months, and 6 months postoperatively (P<0.001). Along with the time an improvement was seen in the patients’ local sensation, however, we also noticed a significant decrease in local sensation in both groups (P < 0.001). Whereas the treatment group showed noticeable recovery in sensation at 3 and 6 months postoperatively, while the control group showed a significant reduction. Also, the results of the BREAST-Q scale showed that intercostal nerve anastomosis significantly improved the patients’ quality of life at 6 months postoperatively (P<0.001). Two groups held no significant differences in general characteristics (age, BMI, and subtypes). Intercostal nerve anastomosis did not affect the volume or duration of postoperative drainage tube usage, while it did increase the duration of operation by around 25 min (P < 0.001).Conclusion: This study demonstrates intercostal nerve anastomosis improved the local sensation and quality of life of patients who underwent immediate biplane breast reconstruction after nipple-areola sparing mastectomy.
Presentation numberPS2-01-25
Does surgery in a specialized clinic improve locoregional recurrence rates in Inflammatory Breast Cancer?
Vanessa N. Sarli, UT MD Anderson Cancer Center, Houston, TX
V. N. Sarli1, S. Meas1, M. Kai2, M. Desai1, N. Erry1, S. X. Sun3, H. M. Johnson3, W. A. Woodward4, A. Lucci3; 1Breast Surgery Research, UT MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, 3Breast Surgical Oncology, UT MD Anderson Cancer Center, Houston, TX, 4Breast Radiation Oncology, UT MD Anderson Cancer Center, Houston, TX.
Background: Our institution has a specialized Inflammatory Breast Cancer (IBC) clinic (SIBCC) where almost all patients undergo trimodality therapy. Surgical management for IBC at SIBCC routinely consists of modified radical mastectomy to negative margins along with an axillary lymph node dissection after completion of neoadjuvant systemic therapy. We sought to evaluate the differences in surgical outcomes between patients who underwent surgery at SIBCC versus patients who underwent surgery at outside institutions (OSI). Methods: A retrospective analysis was performed on patients with IBC enrolled in a prospective registry from 2003-2025 at SIBCC. All patients underwent surgery for primary tumor removal either at SIBCC or an outside institution. Chi-squared test and Kaplan-Meier method were used for statistical analysis. Results: Of the 920 patients included, 620 had their surgical procedures completed at SIBCC, while 300 had surgery at OSI. Patients underwent the following surgical procedures: modified radical mastectomy (SIBCC: 620, 100%; OSI: 249, 83%), total mastectomy (SBICC: 0; OSI: 35, 11.7%), skin sparing mastectomy (SIBCC: 0; OSI: 5, 1.7%), and segmental mastectomy (SIBCC: 0; OSI: 5, 1.7%). Axillary lymph node dissections were performed in 96% of cases at SIBCC and 83% of cases at OSI. Pathologic nodal positivity was higher in the OSI group (40%) compared to the SIBCC group (31% p=0.021). Patients treated at outside institutions were significantly more likely to have positive surgical margins (63.2%) compared to those treated at SIBCC (2.5%, P<0.001). Median number of lymph nodes removed was higher at SIBCC (20 vs 11) (p<0.001). Of the 620 patients who completed surgery at SIBCC, 40% had a recurrence or progression, while 72% of surgical cases completed outside (216/300) had a recurrence or progression. Median overall survival was significantly longer among patients treated at SIBCC compared to those treated at OSI (153 vs 56 months, 95% CI: 141.2-164.8 vs 48.5-63.5). Similarly, patients treated at SIBCC had significantly improved relapse-free survival compared to those treated OSI (median RFS: 108 vs 16 months; p < 0.001). Locoregional recurrence-free survival was significantly longer in patients treated at SIBCC compared to those treated at OSI (mean 185.8 months vs. 92.0 months, p<0.001), highlighting the critical role of specialized surgical care in improving local disease control in IBC. Conclusion: Patients with inflammatory breast cancer had better outcomes when their surgery was performed at a specialized IBC clinic. This study demonstrated that a specialized surgical approach is associated with significantly lower rates of positive margins and improved locoregional recurrence-free survival.
Presentation numberPS2-01-27
A retrospective cohort study on axilla management in ipsilateral breast tumor recurrence after partial mastectomy with sentinel node biopsy
Takahiro Nakayama, Osaka International Cancer Institute, Osaka, Japan
T. Nakayama1, R. Nakamura2, T. Onishi3, H. Yasojima4, J. Ando5, H. Jinno6, T. Ogawa7, T. Aihara8, H. Matsumoto9, N. Masuda10, M. Kawada11, S. Kuba12, Y. Shinden13, N. Wada14, M. Kitada15, M. Saito-Oba16, J. Sakamoto17, S. Imoto18; 1Breast and Endocrine Surgery, Osaka International Cancer Institute, Osaka, JAPAN, 2Breast Surgery, Chiba Cancer Center, Chiba, JAPAN, 3Breast Surgery, National Cancer Center Hospital East, Kashiwa, JAPAN, 4Breast Surgery, National Hospital Organization Osaka National Hospital, Osaka, JAPAN, 5Breast Surgery, Tochigi Cancer Center Hospital, Utsunomiya, JAPAN, 6Breast Surgery, Teikyo University Hospital, Tokyo, JAPAN, 7Breast Center, Mie University Hospital, Tsu, JAPAN, 8Breast Surgery, Aihara Hospital, Minoh, JAPAN, 9Breast Surgery, Osaka International Cancer Institute, Kitaadachi-gun, JAPAN, 10Breast Surgery, Kyoto University Hospital, Kyoto, JAPAN, 11Breast and Thoracic Surgery, Tonan Hospital, Sapporo, JAPAN, 12Breast and Endocrine Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JAPAN, 13Breast and Thyroid Surgery, Kagoshima University Hospital, Kagoshima, JAPAN, 14Surgery, Tokyo Dental College Ichikawa General Hospital, Ichikawa, JAPAN, 15Breast Surgery, Asahikawa Medical University Hospital, Asahikawa, JAPAN, 16Clinical Research & Education Promotion Division, National Center of Neurology and Psychiatry, Tokyo, JAPAN, 17Clinical Research Department, Tokai Central Hospital, Kakamigahara, JAPAN, 18Breast Surgery, Kyorin University Hospital, Hachioji, JAPAN.
Background: Sentinel node biopsy (SNB) has been the standard of care for patients with clinically node-negative breast cancer (BC) since the 2000s. Recent ASCO guidelines now permit SNB to be omitted under specific conditions in clinical T1N0 BC. While axillary lymph node dissection (ALND) remains a regional control option for early BC, optimal axillary management, including repeat SNB, for ipsilateral breast tumor recurrence (IBTR) remains undefined. To address this, the Japanese Society for Sentinel Node Navigation Surgery (SNNS), established in 1999 to advance sentinel node research in solid tumors, conducted a retrospective cohort study (UMIN No. 000049737) on axillary management in IBTR cases. Patients and Methods: We included patients with clinical Tis-4N0 BC who underwent partial mastectomy and SNB, and whose IBTR was diagnosed between January 2010 and August 2022. Exclusion criteria included clinically node-positive BC, bilateral BC, and stage IV BC. Adjuvant therapy decisions were at the physician’s discretion. We analyzed clinicopathological data, focusing on lymphatic mapping and repeat SNB, from primary BC and IBTR medical records. This study received approval from the Kyorin University School of Medicine institutional review board. Results: Our study enrolled 314 eligible cases from 20 Japanese institutions. At primary BC diagnosis, 66 cases were stage 0, 178 were stage IA/B, 66 were stage IIA/B, 1 was stage IIIA, and 3 had unknown stages. SNB was successful in 312 cases (99.3%), with only 12 cases (3.8%) showing positive sentinel lymph nodes (SLN). Breast and regional nodal irradiation were performed in 237 and 8 cases, respectively. IBTR was diagnosed between 3 and 259 months (median: 70 months) after primary BC surgery. Total mastectomy was planned for 243 cases, and partial mastectomy for 64. Sixteen cases also underwent breast reconstruction. Repeat SNB was performed in 216 cases (68.7%), with successful SLN identification in 181 (83.7%). Of the 168 cases that underwent radioisotope-guided lymphatic mapping, hot spots were visualized in 112 (66.6%) via lymphoscintigraphy. SLN localization showed 175 in the ipsilateral axilla, 6 in the contralateral axilla, and 4 in the ipsilateral internal mammary lesion. Positive SLN rates were 9.1% (11 cases) for ipsilateral axilla, 1.6% (1 case) for contralateral axilla, and 0% (0 cases) for ipsilateral internal mammary lesion. Following axillary management for IBTR, repeat SNB was performed in 194 cases (61.7%), lymph node sampling in 2 (0.6%), ALND in 37 (11.7%), and no axillary surgery in 81 (25.7%). Post-IBTR axillary management, 20 cases experienced regional or contralateral lymph node recurrence, and 23 cases died. Conclusion: Repeat SNB is feasible and provides valuable information for de-escalation surgery in IBTR cases. However, axillary management should be carefully considered, taking into account both loco-regional and systemic dissemination at the time of IBTR diagnosis.
Presentation numberPS2-01-28
Factors influencing surgical decision-making in breast cancer using patient-reported outcomes- A multicenter collaborative study-
Hirohito Seki, Kyorin University School of Medicine, Tokyo, Japan
H. Seki1, T. Komiya2, M. Kato3, Y. Sowa4, J. Takano5, Y. Nishida1, N. Tamura6, M. Saiga7; 1Breast Surgery, Kyorin University School of Medicine, Tokyo, JAPAN, 2Plastic and Reconstructive Surgery, Tokyo Medical University, Tokyo, JAPAN, 3Plastic Surgery, Aichi Medical University Hospital, Aichi, JAPAN, 4Plastic Surgery, Jichi Medical University, Tochigi, JAPAN, 5Plastic Surgery, Saitama Medical Center, Saitama, JAPAN, 6Breast and Endocrine Surgical Oncology, Toranomon Hospital, Tokyo, JAPAN, 7Plastic Surgery, Okayama University Hospital, Okayama, JAPAN.
Background: Surgical decision-making in breast cancer involves not only clinical and oncological considerations but also psychosocial factors such as personal values, social support, and anticipated quality of life. Although shared decision-making (SDM) is increasingly emphasized, factors influencing patients’ surgical choices remain unclear. This study aimed to identify determinants associated with the selection of total mastectomy (MT), breast-conserving surgery (BCS), or immediate breast reconstruction (IBR) using patient-reported outcomes. Patients and Methods: A multicenter study was conducted across six institutions in Japan. We included 577 female patients who underwent breast cancer surgery between 2008 and 2018. Using patient-reported outcomes, we collected data on sociodemographic background and 15 decision-influencing factors, including opinions of healthcare providers, family, and peers; radiation therapy; fear of recurrence; hospitalization; and perceived impacts on childcare, work, and hobbies. Patients were categorized into MT, BCS, or IBR groups. Chi-square tests were used for group comparisons, and multinomial logistic regression was performed using the IBR group as the reference. Results: Among 577 patients, 194 underwent MT, 185 BCS, and 198 IBR. The mean age was 53.6 years (±11.5), with the IBR group significantly younger (48.2) than MT (58.1) and BCS (54.8) (p<0.001). The mean interval from surgery to questionnaire response was 56.0 months. IBR patients were more likely to have avoided chemotherapy (p<0.001), be non-smokers (p<0.001), have smaller tumors (p<0.001), no nodal metastasis (p=0.003), and be full-time employed (p<0.001). The incidence of postoperative complications (grade ≥3) was highest in the IBR group (p<0.001). The most reported influence was the surgeon’s opinion (92.2%), followed by fear of recurrence (43.8%), partner’s opinion (35.5%), family (22.4%), and breast cancer survivors (21.0%). Patients influenced by nurses were more likely to select IBR (MT: 3.1%, BCS: 4.5%, IBR: 10.8%; p=0.004). Other significant differences included partner’s opinion (p=0.012), hospitalization (p=0.022), radiation therapy (p<0.001), recurrence fear (p=0.002), and concerns about hobbies (p=0.004) and work (p=0.025). Univariable analysis showed that IBR was more likely in patients who were younger (p<0.001), had smaller tumors (p<0.001), no nodal metastasis (p=0.036), no chemotherapy (p=0.007), were non-smokers (p=0.001), employed full-time (p<0.001), and had higher income (p=0.004). IBR was also associated with influence from nurses (p=0.005), partners (p=0.005), hospitalization (p=0.009), hobbies (p=0.017), and work concerns (p=0.011). Recurrence fear was associated with higher IBR selection over BCS (p=0.017). In multivariable analysis, independent predictors of selecting IBR over both MT and BCS included younger age (p<0.001), non-smoking status (p=0.012), being influenced by nurses’ opinions (p=0.022), and absence of fear of cancer recurrence (p=0.042). Conclusions: This study is the first large-scale, multicenter collaborative study to identify factors influencing surgical decision-making in Japanese breast cancer patients. Predictors influencing the choice of IBR compared with MT and BCS were age, smoking status, nurses’ opinions, and anxiety about recurrence. The results of this study suggest the importance of multidisciplinary collaboration and surgical decision-making support that considers the individual background of patients.
Presentation numberPS2-01-29
Post-operative complications of breast cancer surgery among women with and without obesity in the U.S. Military Health System
Brooke A. Engelman, Landstuhl Regional Medical Center, Landstuhl, Germany
B. A. Engelman1, Y. L. Eaglehouse2, S. Darmon2, M. Nealeigh2, R. W. Krell2, C. D. Shriver2, K. Zhu2; 1General Surgery, Landstuhl Regional Medical Center, Landstuhl, GERMANY, 2Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD.
Background: Obesity is known to increase risk of postoperative complications for patients undergoing general and cardiovascular surgery. The impact of obesity on risk of complications among women with breast cancer is less understood. Access to medical care may be associated with management of obesity and its related conditions, as well as in the diagnosis and treatment of cancer. Thus, it is unknown what the association between obesity and risk of short-term complications and readmission after breast cancer surgery is when controlled for access to medical care. To address this gap in knowledge, we aimed to study the association of obesity with risk of general and breast-specific complications among women with breast cancer in the U.S. Military Health System which provides access to care for all eligible Department of Defense beneficiaries with little financial barriers. Methods: Women aged 18 and older with non-metastatic invasive breast cancer between 2001 and 2014 and who underwent breast conservation or total mastectomy without immediate reconstruction were included. Obesity status was determined by the presence of International Classification of Diseases (ICD)-9th edition diagnosis codes corresponding to body mass index (BMI) greater or equal to 30 in the medical encounter data prior to breast cancer diagnosis. General and breast surgery-specific complications, breast reoperation, and hospital readmissions were assessed in the 30-day postoperative period. Modified Poisson regression was used to compare the risk of the outcomes between women with and without obesity expressed as adjusted risk ratios (ARRs) with 95% confidence intervals (CIs). Results: The study included n=1,094 women with obesity (mean age 57.5 +/-10.9 years) and n=6,741 women without obesity (mean age 55.9 +/-13.0 years) receiving surgery without immediate reconstruction for stage I-III breast cancer. Overall, the frequency of general complications was 2.5% among women with obesity and 1.9% among women without obesity (Chi-square p=0.22). The risk of general complications (e.g. myocardial infarction, pneumonia) was not statistically different by obesity status (ARR=0.95, 95% CI=0.61, 1.49 for obese vs. non-obese) in multivariable analyses with adjustment for potential confounders including age, race-ethnicity, tumor stage, and total comorbidity burden. The frequency of breast-specific complications was 8.9% among women with obesity and 6.3% among women without obesity (Chi-square p=0.03). In Poisson models, women with obesity had a 29% higher risk of breast complications (ARR=1.29, 95% CI=1.03, 1.61) relative to women without obesity. For specific complications, the risk among women with obesity was higher relative to those without obesity for surgical site infection (ARR=1.56, 95% CI=1.10, 2.21) and lymphedema (ARR=1.71, 95% CI=1.02, 2.88) and not statistically different for hematoma or seroma after adjusting for patient, tumor, and surgery characteristics. There were no statistical differences in risk of reoperation (ARR=0.93, 95% CI= 0.80, 1.10) or readmission (ARR=1.04, 95% CI=0.79, 1.36). Conclusion: In the universal Military Health System, women with obesity had an increased risk of site-specific postoperative complications following breast cancer surgery, especially surgical site infection and lymphedema. Our results highlight the importance of weight management in breast surgical oncology care and indicate that efforts are needed to identify effective interventions which may reduce both obesity and risk of postoperative complications among women with breast cancer.
Presentation numberPS2-01-30
Patients with small, clinically and imaging node-negative, HR-positive/HER2-negative early breast cancer undergoing mastectomy do not show increased node-positivity and missed adjuvant systemic treatment options
Martin Heidinger, University Hospital Basel, Basel, Switzerland
M. Heidinger1, G. Montagna2, F. Halbeisen3, N. Maggi1, M. Frevert1, R. Kiblawi1, J. Loesch1, F. D. Schwab1, C. Kurzeder1, T. A. Zwimpfer4, W. P. Weber1; 1Breast clinic, University Hospital Basel, Basel, SWITZERLAND, 2Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 3Surgical Outcome Research Center, University Hospital Basel, Basel, SWITZERLAND, 4Gynaecology & Obstetrics, University Hospital Basel, Basel, SWITZERLAND.
Introduction The SOUND and INSEMA trials included patients who underwent breast-conserving surgery (BCS). Mastectomy offers equivalent local control; however, tumor stage is often more advanced, potentially impacting adjuvant treatment decisions. Adjuvant cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) provide oncological benefits to patients with hormone receptor (HR)-positive and human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC). Their indication is based on clinical parameters yet irrespective of the type of breast surgery performed. Consequently, it remains unclear whether in SOUND/INSEMA eligible patients with HR-positive/HER2-negative BC undergoing mastectomy versus BCS proportions of node-positivity differ, and whether omission of sentinel lymph node biopsy (SLNB) would impact treatment candidacy for CDK4/6i differently. Methods This single-center, retrospective cohort study included patients with clinically T1 and T2, cN0/iN0, HR-positive/HER2-negative BC who underwent SLNB between 01/2014 and 12/2024. Clinicopathological features, nodal involvement, and CDK4/6i eligibility were compared between patients who underwent mastectomy and BCS. CDK4/6i eligibility was evaluated using the monarchE (≥4 positive lymph nodes, or 1-3 positive lymph nodes and either grade 3 disease, tumor size ≥5cm, or Ki67 ≥20%) and NATALEE inclusion criteria. Ribociclib eligibility based on node-criteria was defined as either pT1pN1 (excluding pN1mi) or pT2pN1, provided that no additional criteria would render the pT2 status per se eligible for treatment. Multivariable logistic regression analysis was used to identify independent predictors of node-positivity and CDK4/6i eligibility. Results A total of 454 patients were included, of whom 31.9% (145/454) underwent a mastectomy. Patients who underwent a mastectomy had clinically larger tumors (cT2 37.2% vs. 27.8%, p=0.05) without differences in pathological tumor stage (p=0.185). Tumors of patients who underwent a mastectomy were more frequently multifocal (23.5% vs. 9.7%, p<0.001), of lobular histotype (21.4% vs. 11.3%, p=0.008), and exhibited a higher tumor grade (grade II and III 73.8% vs. 63.8%, p=0.019). The median number of positive lymph nodes was comparable between groups (0, IQR 0-0 vs. 0, IQR 0-0, p=0.283) without differences in pathological nodal stage (pN1 16.6% vs. 14.6%, p=0.168). Predictors of node-positivity were found to be lymphovascular invasion (odds ratio [OR] 6.8, 95%CI 3.7-12.9, p<0.001) and premenopausal status (OR 2.0, 95%CI 1.0-3.8, p=0.034). Mastectomy was not found to be a predictor of node-positivity (OR 1.1, 95%CI 0.6-1.9, p=0.719).Eligibility for any CDK4/6i was numerically higher in patients who underwent a mastectomy (29.7% vs. 21.4%, p=0.06). However, eligibility based on nodal criteria did not show any differences between the groups (abemaciclib 7.6% vs. 6.2%, p=0.550; ribociclib 9.7% vs. 7.8%, p=0.586). Predictors for CDK4/6i eligibility were found to be ≥pT2 (OR 12.9, 95%CI 6.5-26.5, p<0.001), Ki67 ≥20% (OR 5.3, 95%CI 2.9-10.0, p<0.001), and lymphovascular invasion (OR 5.1, 95%CI 2.6-10.1, p<0.001). Mastectomy was not found to be a predictor of CDK4/6i eligibility (OR 1.3, 95%CI 0.7-2.3, p=0.481). Conclusion In patients with cT1-T2, cN0/iN0, HR-positive/HER2-negative BC, type of surgery did neither impact node positivity nor CDK4/6i eligibility. These findings support the consideration of SLNB omission in appropriately selected patients undergoing mastectomy, particularly postmenopausal women with low-risk tumors. Further investigation of the extension of SLNB omission eligibility to patients undergoing mastectomy is warranted.
Presentation numberPS2-03-01
Nipple Delay Improves Nipple-Areolar Complex Viability in High-Risk NSM Patients
Koshi Matsui, University of Toyama, Toyama, Japan
K. Matsui, S. Nagasawa, E. Kanaya, M. Araki, S. Sekine, T. Fujii; Department of Surgery and Science, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, JAPAN.
BackgroundNipple-sparing mastectomy (NSM) is increasingly utilized in breast cancer surgery due to its superior cosmetic outcomes and patient satisfaction. However, ischemic complications, particularly necrosis of the nipple-areolar complex (NAC), remain a significant concern, with reported incidence ranging from 6% to 30%. Several patient- and surgery-related factors such as large breast volume, periareolar incisions, prior breast surgery, radiation exposure, and smoking have been identified as contributors to NAC necrosis. Nipple delay (ND) is a surgical technique performed approximately 1-2 weeks prior to NSM, designed to improve NAC perfusion and reduce ischemic complications in patients considered high risk. While the efficacy of ND has been demonstrated in Western populations, there are no published studies evaluating this technique in an Asian population. To our knowledge, this is the first report from Japan to assess the impact of ND in a cohort of Japanese patients undergoing NSM with immediate autologous reconstruction. Methods We conducted a retrospective review of patients who underwent either ND followed by NSM (ND-NSM) or NSM alone between January 2020 and March 2025 at a single high-volume breast cancer center in Japan. All patients underwent one-stage autologous breast reconstruction using abdominal flaps. ND was performed approximately 14 days prior to NSM under local or general anesthesia, using a semicircular periareolar incision and skin undermining to precondition the NAC. Baseline characteristics, including body mass index (BMI), history of breast surgery, radiation therapy, and smoking status, were collected. Surgical variables such as incision type, mastectomy specimen weight, and postoperative complications were analyzed. NAC necrosis was graded by severity, and statistical comparisons were performed using Fisher’s exact test or Mann-Whitney U test, as appropriate. Results A total of 100 patients were included, with 31 in the ND-NSM group and 69 in the NSM-only group. There were no significant differences between the two groups in terms of BMI, prior breast surgery, radiation exposure, or smoking history. The ND-NSM group had a significantly higher frequency of periareolar incisions (73.5% vs. 33.3%, p < 0.01) and larger mastectomy weights (median 350g vs. 275g, p = 0.02), indicating higher surgical risk. The overall incidence of NAC necrosis did not differ significantly between groups (22.6% in ND-NSM vs. 23.2% in NSM-only). However, when stratified by necrosis grade, the ND-NSM group demonstrated a significantly lower rate of grade ≥2 necrosis (p = 0.0276). Importantly, no cases of grade ≥2 necrosis were observed in the ND group. Other complications such as skin necrosis, infection, thrombosis, and flap-related events were similar between groups. Conclusion Despite higher-risk features, the ND-NSM group showed a markedly reduced severity of NAC necrosis. Our findings suggest that the nipple delay procedure may allow patients with traditionally high-risk characteristics to safely undergo NSM with complication rates comparable to lower-risk patients. This is the first study from Japan to evaluate the clinical utility of ND, and our results indicate that this approach is both feasible and beneficial in the Japanese population. ND should be considered a valuable adjunct to NSM in patients at elevated risk for ischemic complications, particularly when immediate autologous reconstruction is planned.
Presentation numberPS2-03-02
Breast surgery for metastatic breast cancer a update of Cochrane systematic review
Giuliano Tosello, Unoeste, Presidente Prudente, Brazil
G. Tosello1, R. Riera2, M. Torloni2, T. Neeman3, M. Cruz4, I. Freitas5, D. Christofaro5, T. Paulo6, C. Oliveira7, B. Mota8; 1Gynecology and Obstetrics, Unoeste, Presidente Prudente, BRAZIL, 2Adjunct Professor of Evidence-Based Medicine at Escola Paulista de Medicina UNIFESP, Unifesp, Sao Paulo, BRAZIL, 3Statistics Department, Australian National University, Canberra, AUSTRALIA, 4Medical Oncology, Hospital Sírio-Libanês, Sao Paulo, BRAZIL, 5Department of Physical Education, Unesp, Presidente Prudente, BRAZIL, 6Department of Physical Education, Universidade Federal do Rio Grande do Norte, Natal, BRAZIL, 7Department of Medicine, Unoeste, Presidente Prudente, BRAZIL, 8Gynecology and Obstetrics, ICESP FMUSP, Sao Paulo, BRAZIL.
TitleBreast surgery for metastatic breast cancer a update of Cochrane systematic review.ObjectivesTo assess the effects of breast surgery on women with de novo metastatic breast cancer.MethodologyThe inclusion criteria were randomized controlled trials of women with de novo metastatic breast cancer that compared breast surgery plus systemic therapy versus systemic therapy alone in these databases: the Cochrane Breast Cancer Specialised Register, CENTRAL, MEDLINE (by PubMed), and Embase (by OvidSP) on 19 April 2023. The primary outcomes were overall survival (OS) and quality of life. Secondary outcomes were progression-free survival (local and distant control) and toxicity from local therapyTwo review authors independently conducted trial selection, data extraction, and ‘Risk of bias’ assessment (using Cochrane’s ‘Risk of bias’ tool), which was checked by a third review author. We used the GRADE tool to assess the quality of the body of evidence, the risk ratio (RR) to measure the effect of treatment for dichotomous outcomes, and the hazard ratio (HR) for time-to-event outcomes. We also calculated the 95% confidence intervals (CI) for these measures. We used the random-effects model, as we expected clinical or methodological heterogeneity, or both, among the included studies. We reported continuous outcomes, including quality of life, as mean differences (MD) with 95% CIs. We planned to use standardised mean differences (SMD) if outcomes were reported on different scales. ResultsThis is an update of the Cochrane systematic review published in 2018, which was approved by the ethics committee and had its research protocol published in 2014.We included five randomized clinical trials including 1368 women with de novo metastatic breast cancer in the review. Breast surgery not reduce mortality in women with de novo metastatic breast cancer (HR 0.89; 95% CI 0.75 to 1.05, P = 0.09; 5 studies, 1368 participants; I2 = 50%; moderate certainty of evidence). In subgroup analyses of overall survival for women with luminal tumours, the addition of breast surgery to systemic treatment appears to increase the OS, reducing the risk of death by 18% (HR 0.82; 95% CI 0.69 to 0.96; 4 studies; 841 women; P = 0.72; I2 = 0%; moderate certainty of evidence). Breast surgery reduces the risk of local disease progression (HR 0.43; 95% CI 0.32 to 0.58; 4 studies; 1093 women; I2 = 31%; high certainty of evidence) and doesn’t improve metastatic disease control (HR of 1.19, 95% CI 0.86 to 1.18; 3 studies; P = 0.29; I2 = 69%; moderate certainty of evidence). The quality of life of women undergoing locoregional treatment is similar when compared to women undergoing systemic treatment alone in 24 months of follow-up (MD 2.74; 95% CI -2.22–7.70, I2 =0%, p = 0.28, 2 studies, low certainty of evidence).
Overall survival
Overall survival (Luminal subgroup analysis)
Local progression-free survival
ConclusionsEvidence from five randomised controlled trials suggests that adding breast surgery to the treatment of de novo metastatic breast cancer improves local disease control. The impact on survival may vary depending on the immunohistochemical profile of the tumour. Breast surgery might not affect quality of life, toxicity and distant progression-free survival.
Presentation numberPS2-03-03
Retrospective analysis of axillary recurrence in patients with breast cancer receiving radiotherapy to the axillary after positive sentinel lymph node biopsy
Chandana Gavisiddappa, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, United Kingdom
C. Gavisiddappa, R. Parmeshwar, P. McManus, S. Somasundaram; Breast Surgery, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, UNITED KINGDOM.
Retrospective analysis of axillary recurrence in patients with breast cancer receiving radiotherapy to the axilla after positive sentinel lymph node biopsy Background: Breast cancer surgery currently focuses on de-escalating treatment without compromising oncological safety. Axillary lymph node dissection (ALND) has been the standard of care in all sentinel lymph node biopsy (SLNB) positive patients. Although axillary lymph node dissection provides excellent regional control, there are additional side effects like lymphoedema, shoulder stiffness, numbness in the arm. Studies show axillary radiotherapy is an alternate and equally effective treatment for SLNB-positive patients with reduced morbidity. However, this is not a standard of practice in all breast centers in UK. Methodology: A retrospective, single center study between June 2015 and June 2022 following the results published from AMAROS (After Mapping of the Axilla: Radiotherapy Or Surgery) trial in 2014. All patients with breast cancer and clinically and radiologically negative axilla underwent SLNB along with either breast conserving surgery or mastectomy as a routine procedure. Patients with positive SLNB were later offered further axillary management (ALND vs Axillary radiotherapy). Patients who opted for axillary radiotherapy were then analyzed in a retrospective study. Patients’ demographics, disease distribution, nodal positivity, adjuvant treatment were studied. The primary end point was axillary recurrence at the end of 5 years (in line with the AMAROS trial i.e. not more than 4% in axillary radiotherapy group). The secondary end point was arm morbidity and overall survival. Results: A total of 186 patients had positive sentinel lymph node biopsy during the study period and out of them 112 patients, opted for axillary radiotherapy as their mainstay of axillary management. 40(35.7%) patients had T1 disease, 57(50.89%) patients had T2, and 15(13.39%) patients had T3 disease. 13.39% had Grade1 disease, 53.57% had Grade2, and 33.92% had Grade 3 disease. The median number of nodes being 1. The mean age was 63 years, and the mean tumor size of 14.33mm. The mean follow-up was 5.58 years. Axillary recurrence was seen in 3 patients (2.67%), and 1 patient (0.89%) developed lymphedema. 9 patients (8.03%) had distant metastasis. Death due to breast cancer was slightly lower in our study as compared to AMAROS trial (7.84% vs 10.3%) and similar overall death rates (19.04% vs 16.4%) Conclusion: Our study shows that the 5-year axillary recurrence rate is comparable with the AMAROS trial (2.76% vs 1.19%) with similar follow up period (5.58years vs 6.1years). Our study has limitations of small numbers but aligns well with the AMAROS trial. Hence, axillary radiotherapy is non-inferior to axillary lymph node dissection in patients with positive SLNB with significantly less morbidity.
Presentation numberPS2-03-04
Re-defining Breast Cancer care in an LMIC- a single center study on long term oncological and cosmetic outcomes of Oncoplastic Breast Surgery
Safna Naozer Virji, Aga Khan University Hospital, Karachi, Karachi, Pakistan
S. N. Virji, L. Vohra, S. Khan, I. Khan; Breast Surgery, Department of Surgery, Aga Khan University Hospital, Karachi, Karachi, PAKISTAN.
Background: Surgical management has greatly evolved from a radical mastectomy to the most recent advancement of a more conservative oncoplastic breast surgery (OBS). It has not only proven to be safe, but also has shown superior cosmetic outcomes compared to standard breast conservation surgery as it is able to address larger and even multi-focal tumors. Nevertheless, this is a relatively novel concept in our developing country with a limited number of professionals trained in the field. Furthermore, there is a financial aspect and stigma to take into consideration. Patients have to pay out of pocket for treatment which deters them from risking the possibility of a second surgery – incase of incomplete resection margins. It also adds the burden on the surgeon to deliver optimal cosmetic outcomes with no intervention to the contralateral breast. The purpose of this study is to determine the long-term oncological and cosmetic outcomes of patients with early-stage breast cancer who underwent OBS at our institution. Methodology: A single-center retrospective study was conducted on adult female patients with biopsy proven Stage I to III breast cancer who underwent OBS from January 2017 to June, 2022. Patients who underwent bilateral breast surgery were excluded. A total of 194 women were eligible for the study and data was extracted from patient records to determine the long term oncologic outcomes— breast cancer recurrence, disease-free survival (DFS), and overall survival (OS). Cosmetic outcomes were evaluated via patient-reported satisfaction using the Harvard breast cosmesis scale, through telephonic interview after obtaining informed verbal consent. The study was approved by the institutional Ethical Review Committee (ERC 2025-11482-34955). Results: The mean age of women at diagnosis was 48.3 ± 13.2 years with 81% having invasive ductal carcinoma. Sixty-seven percent of the women had hormone receptor positive breast cancer, while 20% had triple negative disease. Among all the patients who were included in the study, half of the patients received neoadjuvant chemotherapy before OBS, followed by adjuvant radiation therapy. Of note, only 6 (3.1%) out of 194 women had a positive margin for which they underwent a second procedure. Level 1 OBS was performed among 77% of the patients. Among those who underwent Level 2 OBS a variety of local perforator flaps were performed. We had a 68% response rate among the patients who were approached via telephone to determine the self-reported cosmetic outcomes, with 91% of the participants reporting ‘Good’ (51%) or ‘Excellent’ (40%) cosmetic outcomes when compared to the contralateral breast. Eight (4.1%) out of 16 patients had an ipsilateral recurrence, while the remaining developed distant metastases. The Kaplan Meier analysis showed that the mean OS of the study participants was 96 months (95% CI=92.2-99.7) since diagnosis, with death as the outcome event. The mean DFS was 93 months (95% CI=89.5 – 97.6) since diagnosis until recurrence as the outcome event. There were significant differences in survival times for patients by recurrence (log rank test, p=0.000) and hormone receptor status (log rank test, p=0.015). There was no significant difference in survival times for patients by type of OBS (log rank test, p=0.860). Conclusion: Oncoplastic breast surgery is a safe and effective option for early-stage breast cancer, with low margin positivity and favorable long-term outcomes. Our study demonstrates excellent survival and high patient-reported cosmetic satisfaction, especially with Level 1 OBS. These findings support its broader use—even in resource-limited settings—as a balanced approach to oncologic safety and aesthetic outcomes.
Presentation numberPS2-03-05
Evaluating the Role of Preoperative Magnetic Resonance Imaging and Intraoperative 3D Tomosynthesis in Achieving Negative Resection Margins in DCIS
STYLIANI PARPOUDI, Anticancer Hospital Theageneio, Thessaloniki, Greece
S. PARPOUDI1, R. Iosifidou1, C. Kontos1, M. Paida2, C. Geranou3, E. Skourtanioti3, V. Xatziravdeli4; 1Surgical Breast Oncology Unit, Anticancer Hospital Theageneio, Thessaloniki, GREECE, 2Gynaecology Department, Ippokrateio General Hospital of Thessaloniki, Thessaloniki, GREECE, 3Radiology Department, Anticancer Hospital Theageneio, Thessaloniki, GREECE, 4Sport Medicine Department, KAT Attica General Hospital, Athens, GREECE.
Title:Evaluating the Role of Preoperative Magnetic Resonance Imaging and Intraoperative 3D Tomosynthesis in Achieving Negative Resection Margins in DCIS Introduction:The increased implementation of breast cancer screening programs has led to a rise in the detection of ductal carcinoma in situ (DCIS). This, however, has introduced new challenges in the surgical management of DCIS, particularly in achieving negative resection margins. The impact of preoperative breast magnetic resonance imaging (MRI) on reducing reoperation rates remains debatable. Furthermore, the use of intraoperative 3D tomosynthesis (3D mammography) to assess surgical specimens presents a potential method for ensuring margin adequacy during surgery. Materials and Methods:From 2017 to 2024, a total of 280 patients diagnosed with DCIS were treated at the Breast Surgical Oncology Department of Anticancer Hospital Theagenio. Among them, 87 were women under 50 years of age and 193 were over 50. The study examined DCIS grade (1, 2, 3), mammographic breast density (ACR categories A-D), and family history. Patients were categorized based on whether they underwent preoperative breast MRI and whether 2D or 3D tomosynthesis was used on the surgical specimen. Reoperation rates were then compared across these groups. Results:Out of the 280 patients, 136 did not undergo preoperative breast MRI, while 144 did. Reoperations were required in 16 patients without MRI and 9 with MRI. Although the overall analysis showed no statistically significant association between MRI use and reoperation rate, age and breast density were significantly correlated with reoperation rates (p=0.018 and p=0.001, respectively). Subgroup analysis revealed that breast density and MRI use significantly influenced reoperation outcomes (p=0.001 and p=0.005, respectively). Additionally, the use of intraoperative 3D tomosynthesis was significantly associated with achieving negative margins (p=0.002). Conclusions:Preoperative breast MRI did not significantly reduce reoperation rates across the entire patient cohort. However, it showed beneficial effects in younger women with dense breast tissue. Intraoperative use of 3D tomosynthesis also contributed to improved surgical outcomes. The combination of these two modalities may represent an effective strategy for reducing reoperation rates in the treatment of DCIS. Keywords: DCIS, breast MRI, intraoperative 3D tomosynthesis, surgical margins, reoperation.
Presentation numberPS2-03-06
Prevalence and Impact of Treatment-Related Pain on Quality of Life in Brazilian Breast Cancer Patients
Rosemar Macedo Sousa Rahal, UNIVERSIDADE FEDERAL DE GOIAS, Goiania, Brazil
R. Macedo Sousa Rahal1, H. Garcia Gomes de Andrade1, L. Pádua Oliveira1, D. Calheiros Campelo Maia2, F. Cardoso Marques3, C. Dias Caminha Gentile4, M. Macambira Noronha5, P. Costa Passos4, V. Oliveira Costa Filho4; 1DEPARTMENT OF GYNECOLOGY, UNIVERSIDADE FEDERAL DE GOIAS, Goiania, BRAZIL, 2DEPARTMENT OF CLINICAL ONCOLOGU, HOSPITAL UNIVERSITÁRIO WALTER CANTIDIO, Fortalez, BRAZIL, 3UNDERGRADUATE, UNIVERSIDADE FEDERAL DO CEARÁ, Goiania, BRAZIL, 4UNDERGRADUATE, UNIVERSIDADE FEDERAL DO CEARA, FORTALEZA, BRAZIL, 5UNDERGRADUATE, UNIVERSIDADE FEDERAL DO CEARÁ, Fortaleza, BRAZIL.
Introduction: Breast cancer (BC) remains the most common malignancy among women and is the fourth leading cause of mortality worldwide. Despite advances in screening, surveillance, and treatment that have improved survival rates, most patients continue to experience treatment-related adverse effects that significantly impact their quality of life(QoL). Objective: This study aims to describe the prevalence and risk factors associated with chronic pain, as well as its impact on QoL, in patients with BC treated at a single institution in Brazil. Methodology: This cross-sectional study included patients with BC who received treatment in the mastology department of Hospital das Clínicas da Universidade Federal de Goiás, Brazil in 2022. Data collected included clinical characteristics (age, menopausal status, comorbidities) and treatment characteristics (type of surgery, radiotherapy, chemotherapy, and treatment toxicity). Standardized questionnaires assessing chronic pain, such as the McGill Pain Questionnaire and the Visual Analogue Scale (VAS), were administered. Statistical analyses were performed using R Studio v 3.6, employing descriptive statistics, Pearson correlation, and regression analyses to evaluate the study data. Results: 122 patients were included in the analysis, with a median age of 55 years. 84% were postmenopausal. Breast reconstruction was performed in 53% of the patients, the majority of whom (92%) underwent immediate reconstruction. Regarding systemic therapy, 51% of patients received neoadjuvant chemotherapy and 29% received adjuvant chemotherapy. Additionally, 81% of the patients received radiotherapy. Lymphedema was present in 20% of the cohort and seroma was reported in 15% of all patients. The total McGill score had a median of 30 (IQR: 22-38). On the Visual Analogue Scale, the median pain intensity was 5.5 (IQR: 3-8). We conducted Wilcoxon rank-sum tests to assess whether pain intensity, measured by the VAS scale, varied according to clinical and treatment-related variables. Patients who underwent mastectomy reported significantly higher pain scores than those who received breast-conserving surgery (p = 0.0088). Higher VAS scores were also observed among patients with lymphedema (p = 0.0069) and those who experienced wound dehiscence (p = 0.026). Interestingly, patients who received radiotherapy reported significantly lower pain intensity compared to those who did not undergo this treatment (p = 0.016). We also examined the association between clinical characteristics and pain perception using the total score from the McGill Pain Questionnaire. Among the variables analyzed, wound dehiscence was significantly associated with higher McGill scores (p = 0.011). Conclusion: Chronic pain is highly prevalent and significantly impairs health-related quality of life in breast cancer patients treated in our cohort in Brazil, especially those with high-risk surgical profiles and comorbidities.
Presentation numberPS2-03-07
Oncoplastic and Reconstructive Breast Surgery: Experience in a Public Healthcare Center of the Brazilian Unified Health System (SUS)
Carolina Souza Vasconcelos, Hospital Cancer of Pernambuco, Recife, Brazil
C. S. Vasconcelos, I. V. Araújo, M. Salgado, D. S. Viana, L. Torres, C. R. Jesus, C. C. Anunciação; Breast Cancer, Hospital Cancer of Pernambuco, Recife, BRAZIL.
AbstractTitle: Oncoplastic and Reconstructive Breast Surgery: Experience in a Public Tertiary Hospital of the Brazilian Unified Health System (SUS)Introduction: Breast cancer is the most common malignancy among women worldwide (excluding non-melanoma skin cancer) and the leading cause of cancer-related death in this population. Advances in surgical techniques have progressively shifted treatment paradigms toward breast-conserving approaches. Oncoplastic surgery merges oncologic safety with plastic surgery principles, enabling broader indications for breast conservation and enhancing aesthetic and psychological outcomes for patients.Objective: To evaluate the clinical and epidemiological profile of patients undergoing oncoplastic breast surgery with immediate or delayed reconstruction in a Brazilian public tertiary hospital, as well as to analyze postoperative complications and compare results with current literature.Methods: A cross-sectional, descriptive, and analytical study was conducted with 100 female patients who underwent oncoplastic breast surgery at the Pernambuco Cancer Hospital (HCP) between March 2023 and July 2024. Data were collected from medical records and included demographics, tumor characteristics, surgical procedures, adjuvant treatments, and postoperative complications. Statistical analysis was performed using SPSS v27 with a 5% margin of error.Results: The patients’ mean age was 50.2 years. Of the 100 cases, 59% underwent oncoplastic quadrantectomy, while 39% had mastectomy with immediate reconstruction. Only 2 patients underwent delayed reconstruction. The most common techniques included therapeutic mammoplasty (17%) and the Burrow triangle (12%). Symmetrization was performed in 29% of cases and fat grafting in 5%. The majority of tumors were stage T1 or T2 (72%) and axilla-negative (73%). Most patients were luminal subtype (61%), and 13% had in situ lesions. Systemic treatment included chemotherapy (60%), radiotherapy (64%), hormone therapy (57%), and HER2-targeted therapy (14%).Postoperative complications occurred in 14% of patients, with wound dehiscence being the most frequent (7%). Five patients required reoperation, including two prosthesis removals and three margin enlargements. No statistically significant association was found between complications and clinical variables, although patients undergoing mastectomy showed a higher absolute rate of complications compared to those who had conservative surgery (22% vs. 8.5%).Conclusion: Oncoplastic breast surgery proved to be a safe and effective strategy for both breast conservation and post-mastectomy reconstruction in a resource-limited public healthcare setting. The complication rate was low and aligned with international standards, despite the absence of plastic surgeons in most cases. The findings support the inclusion of oncoplastic training in mastology residency programs as a way to increase access to reconstruction and improve quality of life for patients treated within the public health system. This approach, beyond restoring anatomy, promotes emotional and psychological well-being, reinforcing its role as a key component in the holistic management of breast cancer. Keywords: Breast cancer; Oncoplastic surgery; Breast reconstruction; Public health; Surgical outcomes
Presentation numberPS2-03-08
Survival outcomes after lumpectomy versus therapeutic mammoplasty in breast cancer patients: a Brazilian cohort study
Anne Dominique Nascimento Lima, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
A. Dominique Nascimento Lima1, F. Pimentel Cavalcante2, A. Mattar3, F. Pereira Zerwes4, M. Antonini5, M. Leite Kraft6, A. Oliveira de Alencar2, A. de Queiroz Germano2, D. Pitanga Torres7, E. Goulart Carneiro8, C. Freitas de Lima8, R. Zocchio Torresan6, F. Palermo Brenelli6, M. Lichtenfels9, A. Frasson9, J. Bines10, E. Camargo Millen8; 1Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 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 (PUCRS), Porto Alegre, BRAZIL, 5Breast Surgery, Hospital do Servidor Público Estadual, Sao Paulo, BRAZIL, 6Breast Surgery, Universidade Estadual de Campinas, Campinas, BRAZIL, 7Breast Surgery, Instituto Américas – Ensino, Pesquisa e inovação, Rio de Janeiro, BRAZIL, 8Breast Surgery, Instituto Americas – Ensino, pesquisa e inovação, Rio de Janeiro, BRAZIL, 9Breast Surgery, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 10Clinical Oncology, IDOR – Instituto D’or de Ensino e Pesquisa, Rio de Janeiro, BRAZIL.
Background: Breast cancer treatment increasingly incorporates breast-conserving approaches that integrate oncologic control with superior cosmetic outcomes. Among these, oncoplastic breast-conserving surgery (OBS), such as therapeutic mammoplasty, has gained popularity, particularly in patients with larger tumors or moderate-to-large breasts. However, robust comparative data on its oncologic safety versus standard breast-conserving surgery (BCS) remain limited, especially in low- and middle-income countries like Brazil. This multicenter cohort study aimed to compare recurrence-free survival (RFS), locoregional recurrence-free survival (LRRFS), breast cancer-specific survival (BCSS), progression-free survival (PFS), and overall survival (OS) between patients undergoing therapeutic mammoplasty and BCS.Methods: A retrospective cohort study was conducted across six Brazilian institutions (five private and one public hospital). Eligible patients were women aged 18 years or older with clinical stage 0-III breast cancer who underwent either BCS or therapeutic mammoplasty between 2016 and 2022. Data collected included demographics, tumor and treatment characteristics, and type of surgical approach. Kaplan-Meier analysis was used to estimate survival outcomes, and Cox proportional hazards models were employed to identify independent predictors of recurrence and survival. Logistic regression analysis evaluated factors associated with surgical technique selection. The study was approved by the institutional ethics committee (IDOR reference 6,907,736). Results: A total of 685 patients were included, of whom 550 (80.3%) underwent BCS and 135 (19.7%) underwent therapeutic mammoplasty. Patients who underwent therapeutic mammoplasty were significantly younger (mean age 53.3 vs. 59.5 years) and had more advanced tumor characteristics, including higher clinical T-stage (T2: 36.3% vs. 24.5%; T3: 8.1% vs. 2.5%) and greater frequency of stage IIB or higher (24.8% vs. 10.8%). Multivariate analysis showed that higher tumor stage (OR=2.71; p=0.001) and treatment in public hospitals (OR=0.41; p=0.001) were independent predictors for receiving therapeutic mammoplasty. Despite these baseline differences, survival outcomes were similar between groups. No statistically significant differences were observed for RFS (HR=0.36; p=0.091), LRRFS (HR=0.35; p=0.085), BCSS (HR=0.36; p=0.091), PFS (HR=0.52; p=0.169), or OS (HR=0.53; p=0.314). Five-year RFS rates were excellent in both groups (97.3% for therapeutic mammoplasty vs. 96.0% for BCS), as were LRRFS rates (96.6% vs. 92.2%). Positive surgical margin rates were low overall (1.5% for therapeutic mammoplasty and 4.2% for BCS), likely due to careful surgical planning, intraoperative frozen section, and larger volume resections in oncoplastic procedures. Conclusion: In this large Brazilian multicenter cohort, therapeutic mammoplasty demonstrated oncologic safety equivalent to standard breast-conserving surgery, even in patients with more advanced tumors. These results support the integration of oncoplastic techniques in breast cancer surgical care, particularly when individualized according to tumor characteristics, patient anatomy, and healthcare setting. While short- to medium-term outcomes are reassuring, prospective studies with longer follow-up are warranted to further validate the oncologic equivalence of therapeutic mammoplasty.
Presentation numberPS2-03-09
Is it safe to offer breast-conserving surgery after neoadjuvant chemotherapy? A Brazilian multicenter cohort study
Anne Dominique Nascimento Lima, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
A. Dominique Nascimento Lima1, A. Mattar2, F. Pimentel Cavalcante3, F. Pereira Zerwes4, M. Antonini5, M. Leite Kraft6, A. Oliveira de Alencar3, A. de Queiroz Germano3, D. Pitanga Torres7, E. Goulart Carneiro7, C. Freitas de Lima7, R. Zocchio Torresan6, F. Palermo Brenelli6, M. Lichtenfels8, A. Frasson8, J. Bines9, E. Camargo Millen10; 1Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 2Breast Surgery, Hospital da Mulher SP, São Paulo, BRAZIL, 3Breast Surgery, Hospital Geral de Fortaleza, Fortaleza, BRAZIL, 4Breast Surgery, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 5Breast Surgery, Hospital do Servidor Público Estadual, São Paulo, BRAZIL, 6Breast Surgery, Universidade Estadual de Campinas, Campinas, BRAZIL, 7Breast Surgery, Instituto Americas – Ensino, pesquisa e inovação, Rio de Janeiro, BRAZIL, 8Breast Surgery, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 9Clinical Oncology, IDOR – Instituto D’or de Pesquisa e Ensino, Rio de Janeiro, BRAZIL, 10Breast Surgery, Instituto Américas – Ensino, pesquisa e inovação, Rio de Janeiro, BRAZIL.
Background: Neoadjuvant chemotherapy (NAC) has become a cornerstone in breast cancer treatment, especially for locally advanced tumors and aggressive subtypes such as triple-negative and HER2-positive disease. NAC can reduce tumor size, increase breast-conserving surgery (BCS) eligibility, improve cosmetic outcomes, and allow early systemic control. However, concerns persist regarding whether BCS following NAC achieves oncologic outcomes comparable to those of BCS followed by adjuvant chemotherapy (AC). This study aimed to compare long-term survival outcomes in patients undergoing BCS after NAC versus AC in a real-world Brazilian cohort. Methods: We conducted a retrospective, multicenter cohort study across six Brazilian institutions (five private, one public). Women aged 18 years or older with clinical stage 0-III breast cancer who underwent BCS (either standard lumpectomy or therapeutic mammoplasty) between 2016 and 2022 were included. Patients were divided into two groups according to whether they received NAC or AC. Data on demographics, tumor characteristics, and treatments were collected from institutional registries. The outcomes assessed were locoregional recurrence-free survival (LRRFS), distant recurrence-free survival (DRFS), breast cancer-specific survival (BCSS), progression-free survival (PFS), and overall survival (OS). Survival outcomes were estimated using Kaplan-Meier methods, and Cox proportional hazards models were used to assess hazard ratios (HRs) and 95% confidence intervals (CIs). This study was approved by the IDOR Ethics Committee (reference 6,907,736). Results: A total of 268 women underwent breast-conserving surgery; 138 (51.5%) received NAC and 130 (48.5%) received AC. The mean age of the cohort was 53.1 years (SD 12.1), with patients in the NAC group being slightly younger (mean 51.1 vs. 55.2 years). Over half of the patients (56.8%) were over 50 years old, and 51.1% identified as white. Invasive ductal carcinoma was the predominant histological type (84.7%), and the majority of patients (73.8%) presented with early-stage disease (≤ stage IIA). Most underwent lumpectomy (72.4%), while 27.6% had therapeutic mammoplasty. In the NAC group, 57.2% had tumors classified as stage >IIB. After a median follow-up of 60 months, no statistically significant differences were observed in survival outcomes between the NAC and AC groups. The hazard ratio for LRRFS was 1.29 (95% CI: 0.28-5.88; p=0.816), for BCSS was 0.52 (95% CI: 0.10-2.61; p=0.473), for DRFS was 1.05 (95% CI: 0.31-3.52; p=0.955), for PFS was 1.19 (95% CI: 0.36-3.96; p=0.869), and for OS was 0.73 (95% CI: 0.18-2.91; p=0.710). Conclusion: In this multicenter Brazilian cohort, breast-conserving surgery following neoadjuvant chemotherapy was associated with survival outcomes comparable to those of surgery followed by adjuvant chemotherapy. These findings support the oncologic safety of BCS after NAC and reinforce its role as a safe and effective option in the individualized surgical management of breast cancer.
Presentation numberPS2-03-10
Safety of nipple/skin-sparing mastectomy in breast cancer patients who had breast implants
Seongjin Jeong, Seoul National University Bundang Hospital, Seongnam-si, Korea, Republic of
S. Jeong; General surgery, Seoul National University Bundang Hospital, Seongnam-si, KOREA, REPUBLIC OF.
Background With advances in breast cancer surgery, the proportion of breast-conserving surgery (BCS), nipple-sparing mastectomy (NSM), skin-sparing mastectomy (SSM), and breast reconstruction has increased compared to the past, when total mastectomy was the predominant approach. The introduction of NSM and SSM has enabled patients with more extensive disease to expect improvements in psychological and emotional recovery, cosmetic satisfaction, and quality of life. However, NSM and SSM are known to have a higher risk of wound complications, such as skin necrosis and infection, compared to BCS or total mastectomy. In particular, previous studies have reported that the risk of ischemic complications increases as the thickness of the skin flap decreases. Recently, due to increasing aesthetic demands and advances in medical technology, implant-based breast augmentation has become more common. Clinically, it is often observed that patients who have undergone breast augmentation with implants tend to have thinner skin flaps due to compression of the breast tissue and subcutaneous fat. Therefore, this study aims to compare the incidence of postoperative wound complications after implant-based breast reconstruction following NSM or SSM between patients with and without a history of implant-based breast augmentation. Methods In total, 817 breast cancer patients treated with nipple-sparing mastectomy (NSM) or skin-sparing mastectomy (SSM) followed by implant-based breast reconstruction between March 2019 and March 2025 at Seoul National University Bundang Hospital in South Korea were included in this study. Patients were divided into two groups: those with a history of breast implant augmentation prior to their breast cancer diagnosis (N=53) and those without such a history (N=764). To adjust for confounding variables, 1:4 propensity score matching was performed. Postoperative wound complications were classified as acute (within 4 weeks after surgery) or late (after 4 weeks), and the incidence of seroma, infection, necrosis, bleeding/hematoma, wound dehiscence, and capsular contracture was compared between the two groups using the chi-square test. Results Using 1:4 propensity score matching, 51 breast cancer patients with pre-existing implants were matched to 175 patients without implants. Patients with pre-existing implants demonstrated numerically higher rates of acute infection (13.7% vs. 5.7%, P=0.108), acute wound dehiscence (5.9% vs. 3.4%, P=0.703), late infection (5.9% vs. 4.6%, P=0.990), and late wound dehiscence (2.0% vs. 1.1%, P=1.000) compared to those without implants. Conversely, patients without implants exhibited numerically higher rates of acute seroma (5.7% vs. 0, P=0.174), acute bleeding (2.9% vs. 2.0%, P=1.000), acute necrosis (1.7% vs. 0, P=0.806), late seroma (2.9% vs. 0, P=0.497), and capsular contracture (11.4% vs. 7.8%, P=0.636). However, none of these differences were statistically significant (all P > 0.05). Conclusion While complication profiles varied between patients with and without pre-existing implants undergoing nipple- or skin-sparing mastectomy with implant-based reconstruction, these differences did not reach statistical significance. With the rising prevalence of breast augmentation, the number of breast cancer patients with pre-existing implants is expected to increase. Careful preoperative assessment and individualized surgical planning are therefore essential. Further large-scale, multi-center studies are needed to validate these findings.
Presentation numberPS2-03-11
Current Practices and Perspectives in the Management of Multicentric and Multifocal Breast Cancer: A Survey-Based Analysis
Revathy Krishnamurthy, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
R. Krishnamurthy1, S. Joshi2, R. Pathak3, A. Das Sheth2, T. Wadasadawala1, B. Kothari2, D. Hoysal4, A. Deshpande4, R. Sarin3; 1Radiation Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, INDIA, 2Surgical Oncology, Tata Memorial Hospital, Mumbai, INDIA, 3Radiation Oncology, Tata Memorial Hospital, Mumbai, INDIA, 4Surgical Oncology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, INDIA.
BackgroundBreast conservation for multifocal (MF) and multicentric (MC) breast cancer remains contentious, particularly with evolving surgical and radiotherapy techniques. We conducted a cross-sectional survey to assess current practices, definitions, and attitudes among surgical oncologists (SOs) and radiation oncologists (ROs) regarding MF/MC breast cancer.MethodsAn online 28-item questionnaire was disseminated to consultant SOs and ROs. Responses from 82 participants across 76 institutions representing 16 Indian states and 3 union territories were analyzed descriptively. Items assessed included diagnostic definitions, indications for breast-conserving surgery (BCS), imaging, boost strategies, and multidisciplinary involvement.ResultsROs and SOs were equally represented (n=41 each), with 93% specializing in breast cancer. Most respondents were from private hospitals (74.4%), followed by government institutions (18.3%) and trust/NGO setups (7.3%).A high-volume cohort, 37% treated >50 breast cancer cases per month. MF disease was encountered ‘frequently’ or ‘very frequently’ by 40% of respondents, and MC by 34%. Most (76%) felt MF/MC incidence is rising due to advanced imaging.BCS was routinely offered for MF by 52% and conditionally by 40%; for MC, 13.6% offered it routinely, and 51% conditionally. Definitions of MC varied: 45% used quadrant-based criteria; 46% used the ≥5 cm separation rule; and 16% defined it as lesions requiring different incisions.Oncoplastic approaches were used to manage tissue deficits in 85% of MF and 76% of MC cases. Five surgeons had never offered BCS for MC, and extreme oncoplastic procedures were performed or encountered in 61% of practices. MRI was considered mandatory by 30%, while 26.5% used contrast-enhanced mammography. Biopsy of both lesions was deemed essential by 35%, with some emphasizing the importance of evaluating biological heterogeneity.Radiation strategies varied: for MF, 10% gave whole-breast irradiation (WBI) without boost, 60% gave a full boost, and 8.5% selectively boosted part of the bed. For MC, 57% boosted both tumor beds. Hypofractionated WBI (40 Gy in 15 fractions) was preferred (74%), while 11% used ultra hypofractionation selectively.IMRT/VMAT was the most commonly used boost technique (54.9%), followed by 3DCRT (38%), electrons (35.4%), and mini-tangents (20.7%). Boost decisions were typically based on tumor size, grade, nodal status, age, and receptor status, with 17.1% using all five factors.Key challenges included uncertainty in target delineation (73.2%), concerns about poor cosmesis (54.9%), and technical complexity (62.2%). Acute toxicity was also reported as a concern (42.7%). Multidisciplinary team (MDT) involvement before surgery was reported by 42.7% and postoperatively or selectively by 44%. Only 2.4% lacked access to an MDT. In 64.6% of practices, the surgical team was involved in boost planning only when the RO found it challenging; routine joint delineation was rare (7.3%). A majority (93.9%) expressed willingness to participate in a prospective study to develop consensus guidelines for boost contouring in MF/MC cases.ConclusionThis survey highlights substantial heterogeneity in surgical and radiation practices for MF/MC breast cancer across India. While breast conservation is cautiously expanding, standardized definitions and contouring guidelines are lacking. Multidisciplinary planning and tailored radiation strategies are essential. These findings underscore the urgent need for consensus protocols and prospective studies to unify practice.
Presentation numberPS2-03-12
Impact of non-sequential lymphatic spread on breast cancer prognosis
Pinxuan Zhu, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
P. Zhu, L. Zhu, Y. Feng, Y. Ye, S. Xu, L. Zhou, Y. Lin, Y. Wang, Y. Du, J. Lu, W. Yin; Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, CHINA.
Background:Skip metastasis, defined as lymphatic spread that bypasses anatomically contiguous lymph nodes, is frequently observed in breast cancer. Previous studies have reported that skip metastasis occurs in approximately 2.6% of invasive breast cancer cases1.Lymphatic dissemination typically follows a sequential pattern, with axillary level I nodes being the most common initial site, followed by levels II, III, and occasionally other nodal basins2. However, the precise patterns and underlying mechanisms of lymphatic spread remain unclear and warrant further investigation.Methods:This retrospective study included adult patients with unilateral, invasive breast cancer who underwent axillary lymph node dissection at Renji Hospital, School of Medicine, Shanghai Jiao Tong University, between October 2013, and December 2023. Patients with de novo stage IV disease were excluded.We developed a novel classification system that stratified patients based on the involvement of axillary, interpectoral (Rotter’s), and infraclavicular lymph nodes. Patients were categorized into three groups, lymph node negative (LN-), lymph node sequentially positive (LNs+) and lymph node non-sequentially positive (LNns+). LN- represents no lymph node metastasis. LNs+ means lymph node metastases following a typical anatomic sequence, from proximal to distal. While LNns+ was defined as involvement of distal nodes without proximal node involvement. Kaplan-Meier analysis and Cox proportional hazards models were used to compare disease-free survival (DFS), the primary endpoint, and recurrence-free survival (RFS) and overall survival (OS) as secondary endpoints. Results:A total of 2,838 patients diagnosed with invasive breast cancer and treated with axillary lymph node dissection were included. Based on the classification, 1,833 patients were assigned to the LN- group, 858 to the LNs+ group, and 147 to the LNns+ group, among whom 9.5% patients presented with isolated Rotter’s lymph node metastasis, 6.8% patients had isolated infraclavicular lymph node involvement and 81.0% patients with axillary nodal metastasis, infraclavicular node metastasis occurred without Rotter’s node involvement. Additionally, 4 patients exhibited combined Rotter’s and infraclavicular lymph node metastases without axillary lymph node involvement.Compared with the other two groups, the LNns+ group showed significantly different characteristics in terms of age, pathological tumor stage, hormone receptor status, human epidermal growth factor receptor 2 (HER2) status, molecular subtype, and the number of infraclavicular lymph nodes dissected (P < 0.05).In the univariate analysis, the LNns+ group consistently exhibited the poorest prognosis across DFS, RFS, and OS (P<0.001). A similar trend was observed in the multivariate Cox regression analysis (P<0.001).Conclusions: Non-sequential lymphatic metastasis may indicate more aggressive biological behavior than either the absence or sequential spread of nodal involvement. These patients may benefit from more potent treatmentsand closer surveillance during follow-up. References:1. Chung HL, Sun J, Leung JWT. Breast Cancer Skip Metastases: Frequency, Associated Tumor Characteristics, and Role of Staging Nodal Ultrasound in Detection. AJR American Journal of Roentgenology 2021; 217(4): 835-44.2. Bitencourt A, Rossi Saccarelli C, Morris EA, et al. Regional Lymph Node Involvement Among Patients With De Novo Metastatic Breast Cancer. JAMA Network Open 2020; 3(10): e2018790.
Presentation numberPS2-03-13
Breast surgery and systemic treatment continuation for patients with de novo metastatic breast cancer and locoregional oligoprogression: a cohort study.
Nadia Bianco, European Institute of Oncology, IRCCS, Milan, Italy
N. Bianco, C. Valenza, M. Milano, D. Trapani, S. Di Bella, A. Sciandivasci, I. Minchella, P. Veronesi, V. Galimberti, M. Intra, C. Sangalli, E. Munzone, G. Curigliano, M. Colleoni; Division of Medical Senology, European Institute of Oncology, IRCCS, Milan, ITALY.
Background: In patients with de novo metastatic breast cancer (mBC), two randomized clinical trials demonstrated that upfront breast surgery does not improve clinical outcomes. However, the role of breast surgery in patients with a locoregional oligoprogression (OPD), in the breast or locoregional lymph nodes, who previously had a response to systemic treatment is unknown. This approach may allow to maintain the same systemic treatment and prolong the time to treatment failure. Methods: We conducted a prospective-retrospective, single-center, cohort study including consecutive patients with de novo mBC, who experienced an OPD in the breast and/or locoregional lymph nodes (with other sites maintaining the best response), underwent breast surgery, and continued the same systemic treatment after surgery, according to the indication of the multidisciplinary tumor board at the European Institute of Oncology (Milan, Italy) from January 2007 to March 2025. The primary endpoint was post-surgery progression-free survival (pS-PFS), defined as the time from the first radiographic evidence of locoregional OPD to subsequent progression or death. Results: 59 patients were included: 28 (47%) had Hormone Receptor (HR)+/HER2- disease, 30 (51%) HER2+, and 1 (2%) triple-negative. When OPD occurred, the median line of treatment was 1 (IQR: 1-2), and the median pre-OPD PFS was 15.2 months (95% CI, 12.0-18.4). In the HR+/HER2- subgroup 22/28 (79%) patients were receiving an endocrine therapy-based therapy, in the HER2+ subgroup 19/30 (63%) were on trastuzumab/pertuzumab maintenance. The median time between OPD and surgery was 1.5 months (95% CI, 1.3-1.7). 12 (20%) patients underwent lumpectomy, 45 (75%) mastectomy, 5 (9%) sentinel lymph node biopsy, and 27 (46%) lymph node dissection. After a median follow-up of 54.1 months (95% CI 44.2-64.0), 38 pS-PFS events occurred in all patients, 21 in the HR+/HER2- subgroup, and 15 in the HER2+ subgroup. The median pS-PFSs were 15.0 months (95% CI, 3.7-26.3) in all patients, 9.0 months (95% CI, 4.9-13.1) in the HR+/HER2- subgroup, and 24.2 months (95% CI, 8.2-40.3) in the HER2+ subgroup.Among patients with a pre-OPD PFS ≥12 months, the median pS-PFSs was 21.4 months (95% CI, 10.2-32.6), as compared with 10.5 months (95% CI, 4.4-16.6) among those with a pre-OPD PFS <12 months (log-rank test: p=.027).Conclusions: This study suggests that selected patients with de novo mBC and locoregional OPD can benefit from breast surgery while maintaining the same systemic treatment, especially in case of HER2+ disease or a longer a pre-OPD PFS.
Presentation numberPS2-03-14
Autologous versus Implant-Based Breast Reconstruction: Complication and corrective surgery rates with and without Radiotherapy
Romi Cencelj-Arnez, Institute of Oncology Ljubljana, Ljubljana, Slovenia
R. Cencelj-Arnez1, T. Arnez2, A. Perhavec1, D. Ribnikar3, I. Ratosa4; 1Division of surgery, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 2Department of Plastic surgery, University Medical Centre Ljubljana, Ljubljana, SLOVENIA, 3Division of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 4Division of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA.
Background: Although radiotherapy (RT) significantly improves oncological outcomes of patients with high-risk breast cancer, it also increases complication rates and the need for corrective surgical procedures in patients undergoing breast reconstruction. The authors hypothesize that RT impacts complication rates and the need for corrective surgical procedures, especially in implant-based breast reconstruction (IBR) compared to autologous breast reconstruction (ABR). Methods: A retrospective review of consecutive patients treated at the Institute of Oncology Ljubljana and University Medical Centre Ljubljana with mastectomy and ABR or IBR in the year 2019, with or without RT, was conducted. Complication and corrective surgery rates were compared between ABR and IBR. The associations between categorical variables were investigated using Pearson’s chi-square test and multivariate analysis. A two-tailed test was used to identify significant differences with a p-value of ≤0.05. Results: In 2019, we identified 219 patients who underwent breast reconstruction; among them, 153 (69.5%) had unilateral reconstructions and 66 (30.5%) had bilateral reconstructions, resulting in a total of 285 breast reconstructions, with 106 (37.2%) being ABR and 179 (62.8%) being IBR. Most patients (87.7%) had immediate breast reconstruction. Risk-reducing surgery was done in 30.2% of reconstructions. Eighty-nine (31.2%) breast reconstructions underwent RT, of whom 46 had IBR and 43 had ABR. Twenty-eight (9.8%) reconstructed breasts received preoperative RT. We detected 22.1% of early complications and 6.3% of late complications. Patients who had preoperative RT and IBR experienced early complications more often, with 18.8% of them affected compared to only 4.8% of those who did not have preoperative RT in IBR, p=0.014. For the analysis of corrective surgery rates, we excluded breast reconstructions that had delayed-immediate ABR (3.2%) and included 276 breast reconstructions for analysis. Patients who had ABR or IBR had similar rates of corrective procedures when compared, regardless of RT (25.8% ABR no RT vs 25.6% IBR no RT; 42.5% ABR with RT vs 36.6% IBR with RT). Overall, patients who received RT had more corrective surgeries (39.5% with RT vs 25.6% no RT, p=0.502), but the difference was not statistically significant. In subgroup analysis of unilateral reconstruction, we excluded patients that later received risk-reducing contralateral mastectomy with reconstruction, and detected a difference in the symmetrization rate according to the type of reconstruction (16.1% in ABR vs 50.8% in IBR; p<0.0001). Although the difference in the symmetrization rate was detected between patients who received RT (36.8%) and those who did not (25.8%) (p=0.196), statistical significance was not observed. Conclusions: In patients with preoperative RT, IBR was associated with a higher probability of early complications. Breast reconstruction type selection does not influence the need for further corrective surgical procedures in patients receiving RT according to this analysis. Symmetrization surgery was more common after IBR. RT increases the need for symmetrization surgery in both reconstruction types.
Presentation numberPS2-03-15
Breast Sensitization Techniques in Mastectomies with An Implant-Based Reconstruction: A Systematic Review
Jasmine El-Taraboulsi, Imperial College Healthcare NHS Trust, London, United Kingdom
J. El-Taraboulsi, D. Leff, P. Thiruchelvam; Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, UNITED KINGDOM.
Introduction: Implant-based reconstructions are the most prevalent oncoplastic breast reconstruction globally. Nevertheless, the vast majority of patients undergoing nipple-sparing mastectomies (NSM) or skin-sparing mastectomies (SSM) experience total or significant sensory loss of the breast and nipple-areolar complex (NAC), as well as post-mastectomy breast pain, both key predictors of poor post-operative psychosocial and sexual wellbeing. In recent years, novel surgical techniques have been introduced to preserve sensation or neurotize the breast and NAC in NSMs and SSMs. However, the ease of implementation, reproducibility, and efficacy of such methods remain under investigation. This systematic review aims to evaluate the current literature describing surgical techniques for sensation preservation or neurotization in mastectomies with implant-based reconstructions. This includes a review of sensory outcomes from the available case-control studies, and an analysis of emerging surgical approaches, aiming to guide clinical implementation, and identify future areas of research. Method: A systematic review of open-access, English-language literature published between January 2000 and June 2025 on implant-based reconstructions offering a sensation preservation or neurotization technique was conducted across PubMed, the Cochrane Library, Google Scholar and an external search, utilizing a keyword-based advanced search. Of the 139 screened records, six prospective studies were assessed and included in the review. Results: Four surgical techniques are described across the included records, namely, intercostal nerve preservation, nerve allografting, nerve autografting, and muscle reinnervation. Despite small sample sizes, variation in follow-up duration, lack of surgical controls, and discrepancies in subjective and objective measurements of sensation across the studies, all proposed techniques improve sensory outcomes or pain reduction. Across all four proposed techniques, the data suggests that treatment groups demonstrated superior sensory outcomes. However, larger-scale studies, with prolonged follow-up periods, consistent measures of sensation and patient quality-of-life, and robust complication reporting are required to establish evidence-based guidelines for sensation preserving mastectomies with implant-based reconstruction. Conclusion: Despite limited case-control data, there is growing evidence to support that utilizing one of the following surgical techniques: intercostal nerve preservation, nerve allografting, nerve autografting and muscle reinnervation, improves breast and NAC sensation following a mastectomy with an implant-based reconstruction. This reduces the physical and psychosocial burden associated with diminished or eliminated breast sensation postoperatively. In addition, the reported complications related to these techniques are minimal, suggesting these methods are safe for clinical use.
Presentation numberPS2-03-16
Endoscopic Nipple Sparing Mastectomy: Early Outcomes and Learning Curve from a Unicentric Series of 21 Cases
Julien Seror, Institut Bourdonnais, Paris, France
J. Seror1, D. Hequet1, G. Aubry1, M. Wilhelm1, M. Cittanova2, C. Gout-Duracher2, B. Sarfati3; 1Surgery, Institut Bourdonnais, Paris, FRANCE, 2Anesthesia, Clinique Saint Jean de Dieu, Paris, FRANCE, 3Surgery, Institut Gustave Roussy/Groupe hospitaliser Ambroise Paré Hartmann, Villejuif/Neuilly, FRANCE.
Background: Endoscopic nipple-sparing mastectomy (eNSM) is a minimally invasive technique offering improved cosmetic outcomes while preserving oncologic safety. As its international use expands, mastering the procedure presents technical and ergonomic challenges, and few studies have described the early learning curve or setup innovations [1,2].Objective: To evaluate the learning curve, perioperative outcomes, and technical refinements of eNSM in a unicentric early experience.Methods: We conducted a retrospective, descriptive study of 21 consecutive eNSM procedures between November 2024 and June 2025. The procedure was offered to patients undergoing mastectomy with immediate implant-based reconstruction. Patients with prior ipsilateral breast irradiation were excluded. We collected data on patient characteristics, indications, operative time, hospital stay, adjuvant treatments, and complications. A specific technical feature of our approach is the use of a high horizontal axillary incision, parallel to the natural axillary crease, providing optimal access and cosmetic concealment. For bilateral procedures, a single video tower positioned at the foot of the patient allowed both sides to be treated successively without reinstallation. In one case, we performed a simultaneous bilateral eNSM using two surgical teams and dual video towers, achieving a total operative time of 137 minutes for both sides.Results: A total of 21 eNSM procedures were performed: 9 unilateral and 6 bilateral. The median age was 46 years (range: 26-64). Indications included 8 multifocal tumors and 8 prophylactic mastectomies. Among the 13 invasive cancers: 9 were HR+/HER2-, 1 HR+/HER2+, and 2 triple-negative. The median tumor size was 13 mm (range: 6-30 mm), and associated DCIS was present in 4 cases (median size: 30 mm). Two patients had nodal involvement. Neoadjuvant chemotherapy was administered in 2 patients, and 1 received adjuvant chemotherapy. Two patients underwent adjuvant radiotherapy. Operative time for unilateral eNSM was a median of 105 minutes (range: 105-180), and for bilateral procedures 160 minutes (range: 137-210). A trend toward shorter operating times was observed over the series, especially with team coordination and ergonomics optimization. The fastest bilateral case (137 minutes) was achieved using a double-surgical team, double-video tower approach. One intraoperative complication occurred (bleeding requiring conversion to open surgery), during the initial phase. No postoperative complications were reported. All patients underwent immediate implant-based reconstruction (median volume: 310 cc, range: 260-525). Median hospital stay was 2 nights (range: 2-4).Conclusion: Our experience confirms the feasibility and safety of eNSM in selected patients, with a short learning curve. Technical optimizations—such as high axillary incisions and ergonomically placed video towers—facilitated bilateral procedures without repositioning. Simultaneous dual-team surgery further reduced operative time. These innovations may help standardize and streamline eNSM in high-volume centers [3].References:1. Toesca A, Peradze N, Galimberti V, et al. Robotic nipple-sparing mastectomy for the treatment of breast cancer: Feasibility and safety study. Breast. 2017;31:51-56.2. Lai HW, Chen ST, Lin SL, et al. Robotic versus conventional nipple-sparing mastectomy and immediate breast reconstruction: a case-control comparison study from Taiwan. Ann Surg Oncol. 2020;27(4):1191-1202.3. Leff DR, Vashisht R, Yongue G, et al. Endoscopic nipple-sparing mastectomy with implant-based reconstruction: a systematic review. Eur J Surg Oncol. 2021;47(1):25-32.
Presentation numberPS2-03-17
Dose reductions in metastatic breast cancer patients: Frequency, strategies, and patient experience
Amy Beumer, Patient Centered Dosing Initiative, Denver, CO
A. Beumer1, M. Carlson1, J. Maues2, J. Cowden1, S. Walker1, A. Veach1, J. Collins1, K. Shanahan1, M. E. Burkard3, K. Kalinsky4, A. Bardia5, H. Rugo6, M. Lustberg7, E. L. Papautsky8, A. Arcuri9, J. Brown9, M. Roberson9; 1None, Patient Centered Dosing Initiative, Denver, CO, 2NA, Patient Centered Dosing Initiative, Denver, CO, 3Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, 4Winship Cancer Institute, Emory University, Atlanta, GA, 5Johnsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 6Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, 7School of Medicine, Yale, New Haven, CT, 8College of Applied Health Sciences, University of Illinois, Chicago, Chicago, IL, 9Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC.
Introduction: Many people with metastatic breast cancer (MBC) experience life-limiting side effects from MBC treatments, with most patients remaining on treatment indefinitely. While supportive medications are often used to mitigate toxicities, they may introduce additional side effects and financial cost. Individualized dosing, particularly dose reductions and frequency adjustments, is a strategy to improve tolerability with the goal of maintaining treatment efficacy. The objective of this study was to elicit MBC patient experiences with treatment toxicity, including dosing strategies and side effect management. Methods: As a follow up to a 2020 survey to characterize real-world experiences with dosing strategies (Loeser et al. 2024), the Patient-Centered Dosing Initiative (PCDI) conducted a new cross-sectional survey to elicit patient experiences and preferences related to toxicity management. The survey, distributed electronically to a convenience sample of people living with MBC in the United States, combined closed and open-ended questions. Recruitment occurred through support organization e-mail lists and newsletters, as well as closed social media groups. The survey included items associated with side effect management, knowledge and experiences of dose modifications, and supportive medication use among other topics. The survey opened April 14th 2025 and data were pulled on June 22nd for analysis, although data collection is still ongoing. Descriptive statistics, including frequency counts and percentages, were used to summarize participant responses. Associations between knowledge of dose modifications and care primarily by a breast specialist were evaluated using chi-squared tests. Results: As of the abstract submission, 340 responses were available for analysis. Surveyed patients reported all treatment drugs taken in the metastatic setting, including cytotoxic chemotherapy (35%), CDK 4/6 inhibitors (62%), HER2-targeted antibodies (24%), antibody-drug conjugates (23%), and PI3K pathway inhibitors (11%). Of the 26% who started an MBC drug at a lower dose, 69% reported that the oncologist initiated the conversation and 16% said they initiated it themselves. There was no statistically significant difference in who initiated the conversation based on whether the participant was primarily treated by a breast specialist or not (p=0.43). Overall, 57% of the respondents had their dose reduced due to side effects, whether they started at full or reduced dose, and 72% felt better on the lower dose. In addition, 27% of patients reported that their oncologist reduced the frequency of drug administration; 67% of these patients felt better with the reduced frequency. 88% of surveyed patients were aware that an MBC drug with significant side effects may have an approved dose reduction schedule. Further, 59% believed that the highest dose of a drug is not always more effective than a lower dose. Conclusions: Dose modifications are common among people with MBC, often driven by treatment toxicity and side effect management. Experiences with dose reductions did not significantly differ based on receiving care from a breast cancer specialist, suggesting broader awareness and acceptance of dose adjustments in patient and clinical communities. Notably, 83% of respondents reported starting on a lower dose, having the dose reduced, or both, indicating a need for further study into the efficacy of lower doses and for clinical trial designs in MBC that include multiple doses tested in later phases. Given that patients rarely initiated these conversations, further research is needed to understand barriers to discussing dose adjustments and to support more patient-clinician dialogue.
Presentation numberPS2-03-18
Postoperative cavity remodeling technique in breast-conserving surgery: a case series
Yuxia Chen, Maoming People’s Hospital, Maoming, China
Y. Chen1, Y. Wang2, X. Jiang3, R. Huang1, H. Liu3; 1Department of Breast Surgery, Maoming People’s Hospital, Maoming, CHINA, 2Department of Breast Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, CHINA, 3Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, CHINA.
Background: Restoring breast contour in Oncoplastic Breast-Conserving Surgery (OBCS) with substantial glandular excision is challenging, as traditional implants or flaps are often invasive, costly, and carry risk of complications. We assessed the feasibility of postoperative cavity remodeling technique (PCRT) that promotes autologous tissue regeneration through mechanical stabilization of the lumpectomy cavity. Methods: This retrospective case series analyzed breast cancer patients with glandular defects ≥50% who underwent PCRT. The procedure involved intraoperative suture fixation of the residual cavity, and seroma-guided endogenous remodeling, supplemented by a mini dermal-fat fascial flap for incision reinforcement. Volume and resolution of postoperative serous effusion, incision-related complication, postoperative pain using visual analog scale (VAS), aesthetic evaluation (Harris score), and patient satisfaction (BREAST-Q) were assessed. Results: Among 60 patients included, 42 (70.0%) had moderate glandular defects (50%-69%) and 18 (30.0%) had severe defects (≥70%). Postoperative fluid accumulation peaked 1 week after drain removal, with 28.3% of patients requiring aspiration. Incision-related complications occurred in 10% of patients (wound dehiscence: 5.0%; suture-site reactions: 5.0%) at 3 months after drain removal, all cases were managed with supportive care. Moderate to severe pain (VAS score of 4-6 points) was reported in 73.3% of patients 1 week postoperatively, declining to 6.7% at 3 months. Aesthetic assessment was rated as good/excellent in 81.6% of patients, and 86.7% reported high satisfaction BREAST-Q score >80 while 78.3% reported very satisfied (Likert scale) at 12 months. Conclusions: PCRT might be a minimally invasive, implant-free reconstructive approach with favorable aesthetic and patient-reported outcomes in patients with significant breast tissue loss.
Presentation numberPS2-03-19
Foreseeing Fluid: Risk Factors for Seroma Formation Following Skin- and Nipple-Sparing Mastectomy with Immediate Reconstruction.
Kerstin Pfister, University Hospital Ulm, Ulm, Germany
K. Pfister, H. Schäffler, E. Leinert, K. Veselinovic, J. Haager, S. Lukac, S. Huesmann, B. Rack, W. Janni, V. Fink; Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, GERMANY.
Background: Mastectomies with immediate implant-based reconstruction are increasingly common, particularly among younger breast cancer (BC) patients and individuals undergoing risk-reducing surgery. Skin-sparing and nipple-sparing mastectomies are now well-established techniques; however, they are associated with specific postoperative complications. Notably, prolonged drain duration and seroma formation can hinder recovery, increasing the risk of infection, potentially requiring further intervention. Identifying risk factors for seroma formation is crucial to improving postoperative outcomes and guiding surgical planning. Methods: All nipple-sparing and skin-sparing mastectomies performed at a high-volume German breast center between January 2017 and June 2024 were retrospectively analyzed. A total of 1,298 mastectomies in 756 patients were included. Patient demographics, clinical characteristics, and perioperative data were evaluated. Seroma formation was defined as either a drain remaining in place beyond postoperative day 7 or the need for seroma aspiration following drain removal. Results: Among the 1,298 mastectomies, 389 (30.0%) met the criteria for seroma formation. Oncologic indications (e.g., DCIS or invasive BC) were significantly associated with higher rates of seroma formation compared to risk-reducing procedures. Older age, higher BMI, and the presence of medically treated hypertension were also significantly associated with increased seroma risk. No significant associations were found with neoadjuvant chemotherapy, diabetes, smoking status, or anticoagulant use. The type of mastectomy (skin-sparing vs. nipple-sparing), the use of mesh for stabilization, and the plane of reconstruction (prepectoral vs. retropectoral) did not affect the incidence of seroma. However, higher implant weight and greater drain output within the first three postoperative days were strongly correlated with seroma formation (p < 0.001). Patients in the seroma group experienced significantly more postoperative infections (8.0% vs. 2.6%, p < 0.001), although rates of revision surgery and delayed wound healing were not significantly different. Conclusion: This large retrospective cohort study identifies key clinical and surgical factors associated with seroma formation following mastectomy with immediate reconstruction. These findings underscore the importance of individualized risk assessment and may inform patient counseling and surgical decision-making to reduce postoperative morbidity.
Presentation numberPS2-03-20
Feasibility of the omission of axillary surgery in node-negative early breast cancer: a systematic review and meta-analysis
James Lucocq, Western General Hospital, Edinburgh, United Kingdom
J. Lucocq1, H. Baig1, K. Elder1, G. Urquhart2, R. Sharma3, L. Romics4, B. Elsberger5; 1Edinburgh Breast Unit, Western General Hospital, Edinburgh, UNITED KINGDOM, 2Oncology Department, Aberdeen Royal Infirmary, Edinburgh, UNITED KINGDOM, 3Oncology Department, Aberdeen Royal Infirmary, Aberdeen, UNITED KINGDOM, 4Department of Academic Surgery, University of Glasgow and National Health Service Greater Glasgow and Clyde, Glasgow, UNITED KINGDOM, 5Breast Unit, Aberdeen Royal Infirmary, Aberdeen, UNITED KINGDOM.
Introduction: Recent and historical trials have suggested that the omission of axillary surgery is oncologically safe in node-negative early breast cancer. This meta-analysis investigates the feasibility of the omission of axillary surgery (SLNB or ALND) in terms of oncological outcomes and adjuvant treatment decisions. Method: A systematic search of Medline, Embase and Cochrane Central was conducted. Random-effect meta-analysis was conducted to compare recurrence and survival outcomes between omission of axillary surgery and SLNB or ALND. Differences in real-world adjuvant treatment decisions and patient reported outcomes were investigated. Results: Ten studies (omission, n=3716; SLNB/ALND, n=4604/785) investigated oncological outcomes (pooled rates, T1 – 88%; Grade 1-2, 80%; ER-positive 91.2%; HER2-positive 4.4%; ductal carcinoma 74%). In the omitted group, the pooled rates of local, axillary and distant recurrence (follow-up, 8 years 9 months) were 3.6% (95%CI,2.0-6.5%), 2.5% (95%CI,1.3-4.8%; 5year, 1.0%) and 3.6% (95%CI,2.0-6.5%; 5year, 2.7%), respectively. In comparison to SLNB/ALND, there were no differences in local recurrence (OR 0.91; 95%CI,0.56-1.50), distant metastasis (OR 0.91; 95%CI,0.56-1.50), BCM (OR 1.00; 95%,CI 0.63-1.60), OS (HR 0.88; 95%CI,0.65-1.19) or DFS (HR 0.91; 95%CI,0.76-1.11). Axillary recurrence was higher in the omission group compared to SLNB/ALND (OR 4.42, 95%CI,1.5-12.8) but not in SLNB alone (OR 2.85 95%CI 0.1-133.8). Omission of axillary surgery was associated with lower rates of adjuvant chemotherapy, radiotherapy and hormonal treatment but quality of evidence was poor. Conclusion: Omission of axillary surgery is safe in node-negative early breast cancer. Prospective data is required to investigate the impact of omission on adjuvant treatment decision.
Presentation numberPS2-03-21
Trends in axillary surgical strategies for breast cancer patients with clinical complete response after neoadjuvant systemic therapy: a population-based retrospective cohort study
Yizi Zheng, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
Y. Zheng, Y. Chen, E. Xia, O. Wang; Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, CHINA.
Objectives: Over the last decade, axillary management strategies for patients with breast cancer have experienced several paradigm shifts toward surgical de-escalation, especially in exceptional responders who have achieved a complete response after neoadjuvant systemic therapy (NST). Current trends in axillary surgery for breast cancer patients who achieved clinical complete response (cCR) to neoadjuvant systemic therapy (NST) are unknown.Methods: In this retrospective cohort study, female patients diagnosed with breast cancer who achieved cCR after NST were selected from the Surveillance, Epidemiology, and End Results database (2010-2021). The proportions of patients undergoing each axillary treatment approach were summarized by year of diagnosis to assess the trend of axillary management strategy. The Cox proportional hazard models with the inverse probability of weighting (IPW) were used to evaluate the effects of axillary surgery on overall survival (OS) and disease-specific survival (DSS). A multivariate logistic regression model was used to explore factors associated with axillary pathological (p)CR.Results: In total, 15,111 patients were included. The nodal positivity rate after NST in cCR patients with initially clinical node-negative breast cancer was only 0.56%, while the nodal negativity rate after NST in cCR patients with initially clinical node-positive breast cancer was 49.57%. Between 2010 and 2021, axillary lymph node dissection (ALND) rates decreased from 55.27% to 29.07% while sentinel lymph node biopsy (SLNB) rates increased from 41.04% to 64.84%. From 2010 to 2021, the percentage of patients with initially node-negative disease receiving ALND after NST decreased from 32.92% to 13.03%, whereas those receiving SLNB increased from 66.24% to 80.82%.Patients who received ALND had worse OS than those who received SLNB (hazard ratio [HR] = 1.662, 95% confidence interval [CI], 1.339-2.063, P < .001). After applying the inverse probability weighting (IPW), patients in the ALND group still had a worse OS than those in the SLNB group (HR = 1.491, 95%CI, 1.164-1.911, P < .001). Meanwhile, patients who received ALND were associated with worse DSS than those who received SLNB (HR = 1.909, 95%CI, 1.485-2.453, P < .001). Despite the application of IPW, the DSS of patients in the ALND group remained inferior to that of the SLNB group (HR = 1.654, 95%CI, 1.243-2.202, P < .001).The logistic regression analysis revealed that initial cN0 was a strong predictor of axillary pCR. Patients who were initially diagnosed with N1 (OR, 0.024; 95% CI, 0.022-0.027), N2 (OR, 0.036; 95% CI, 0.029-0.043), and N3 (OR, 0.028; 95% CI, 0.023-0.034) stages had a decreased odds of achieving axillary pCR.Conclusions: In summary, the results of this study indicated that ALND was administered to up to 13% of patients with originally node-negative breast cancer who achieved cCR following NST. Compared to SLNB, ALND may reduce the survival of patients who obtained nodal pCR, indicating potential overtreatment. In this subgroup of patients, we recommend carrying out SLNB first to determine the node status before the decision to undergo ALND.
Presentation numberPS2-03-22
Retromamamry breast-conserving surgery and volume displacement with diced acellular dermal matrix for operable breast cancer
Jean Schneider, Texas Tech University Health Sciences Center, Lubbock, TX
J. Schneider1, Y. An2, H. Choi3, Y. Suh4; 1Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 2Diagnostic radiology, The Catholic University of Korea St. Vincent’s Hospital, Suwon, KOREA, REPUBLIC OF, 3Pathology, The Catholic University of Korea St. Vincent’s Hospital, Suwon, KOREA, REPUBLIC OF, 4Surgery, The Catholic University of Korea St. Vincent’s Hospital, Suwon, KOREA, REPUBLIC OF.
Objective: In a previous clinical study, we demonstrated that it is feasible to perform volume displacement using diced acellular dermal matrix (dADM) immediately after breast-conserving surgery (BCS) in patients with operable breast cancer. However, we realized that unexpected non-infectious erythema could present as a complication, which led us to develop a new approach to address this issue. Summary Background Data: Previous study proved dADM was effective to fill the defect in the breast after BCS, however, some patients showed intramammary fat necrosis during adjuvant chemotherapy or postoperatively following non-infectious erythema on the skin over the cavity filled with dADM. We take into consideration of embryological characteristics of breast to minimize this complication by changing approach route through retromammary plane, while maintaining its proven advantage. Methods: After institutional review board approval, we prospectively recruited 530 breast cancer patients requiring BCS from August 2019 to June 2022. Incision was made along the periphery of breast mound including axilla or inframammary fold, where the minimum distance was expected from the incision to the index tumor. After breast cancer removal, cavity was filled with dADM. After surgery, all patients received adjuvant local irradiation treatment, systemic chemotherapy, targeted agent or endocrine treatment according to the recommendations of international treatment guidelines according to the type of molecular subtype. All enrolled patients were followed up at 2 months after completion of both chemotherapy and external radiation treatment and up to 3 years from the date of surgery, including systemic evaluation to assess the occurrence of any complications, including non-infectious erythema, and oncologic outcome related to locoregional recurrence or systemic metastasis. Results: Of 530 patients enrolled, only 5 patients showed intramammary fat necrosis. Locoregional recurrence and systemic metastasis rates were also not statistically different from previous results not included in this study.Conclusions: Retromammary BCS and volume displacement with dADM for operable breast cancer is effective for reducing fat necrosis postoperatively, even after repetitive episodes of neutropenia during systemic chemotherapy, and is easy to preserve preoperative breast mound volume and shape. Although further clinical studies are needed in a larger number of patients, we believe that this surgical method could be another oncoplastic BCS that can overcome the limitations of conventional BCS.
Presentation numberPS2-03-23
What really drives seroma formation? – Insights from a retrospective study of 244 patients undergoing mastectomy.
Laura Weydandt, University hospital Leipzig, Leipzig, Germany
L. Weydandt, K. Hein, S. Briest, I. Nel, B. Aktas; Department of Gynecology, University hospital Leipzig, Leipzig, GERMANY.
Purpose: Seroma is the most common postoperative complication following mastectomy, often prolonging hospital stays and increasing the need for additional treatment. Despite its high prevalence, the etiology and predictive factors remain unclear. Therefore, identifying reliable predictors and potential causes remains a critical focus in research. This study aimed to identify potential risk factors associated with seroma formation following mastectomy. Methods: We conducted a retrospective analysis of 244 patients who underwent mastectomy at the University Hospital Leipzig between 2019 and 2023. Data was extracted from the patient’s medical record and from pathology reports. The analysis focused on selected factors including epidemiological patient’s characteristics, preoperative treatments (e.g. neoadjuvant chemotherapy), tumor status (e.g. TNM classification), perioperative (e.g. surgery duration) and postoperative variables (e.g. drainage duration). Seroma formation was assessed based on the flow rate from wound drains placed in the breast and axilla and follow-up visits. Statistical methods included Spearman correlation, t-tests, ANOVA, and multivariate regression to identify risk factors associated with seroma development. Results: The median patient age was 51 years (range 18-88). A total number of 301 mastectomies procedures were performed on 244 patients. Of these mastectomies, 215 (71.4%) were conducted due to breast cancer, 24 (8.0%) due to ductal carcinoma in situ and 62 (20.6%) as a prophylactic intervention. In 145 (48.2%) cases a mastectomy with immediate implant-reconstruction was performed, in 156 (51.8%) a mastectomy without reconstruction was performed. In 123 (40.9%) cases the mastectomy was combined with a sentinel node biopsy procedure, in 89 cases (29.6%) with an axillary dissection and in 89 cases (29.6%) no axillary treatment was performed. The median amount of total seroma was 585 ml (range 35-3625 ml). Epidemiological factors found to be significant were patients’ age (r=0.19; p<0.001), BMI (r=0.24; p<0.0001), hypertension (T=-3.52; p<0.001), and diabetes (T=-2.18; p<0.05). Among preoperative treatments found to be significant was previous breast surgery (T=2.25; p<0.05). Furthermore, significant factors regarding tumor status were, tumor size (r=0.33; p<0.001) and histology (F=3.71; p<0.001), removed specimen weight (r=0.31; p<0.0001), number of metastatic lymph nodes (r=0.42; p<0.0001), lymphovascular invasion (r=0.27; p<0.0001) and nodal status (r=0.41; p<0.0001). Perioperative variables found to be significant were the number of removed lymph nodes regardless of metastastic state (r=0.55; p<0.0001), duration of surgery (r=0.13; p<0.05) and inserted implant size (r=0.27; p<0.001). Significant postoperative parameters were C-reactive protein (CRP) -value on the first postoperative day (r=0.24; p<0.0001), amount of seroma in the first 24 hours (r=0.47; p<0.0001) and the duration of drainage (r=0.68; p<0.0001). Conclusion: These results suggest that both patient-related and treatment-related factors significantly influence postoperative development of seroma following mastectomy. Higher seroma volumes were associated with older age, higher BMI, comorbidities (such as hypertension and diabetes) and lymph node removal. Recognizing these risk factors may help guide postoperative management to reduce complications.
Presentation numberPS2-03-24
De-escalation of Axillary Surgery in Patients with Residual Nodal Disease After Neoadjuvant Chemotherapy: A Systematic Review and Meta-Analysis
Natalia Polidorio, Rede Americas Oncology, São Paulo, Brazil
N. Polidorio1, R. Frederice1, G. Azevedo Gabriele Carlos1, T. Dassie1, R. Sousa-Barroso2; 1Breast Surgery, Rede Americas Oncology, São Paulo, BRAZIL, 2Breast Medical Oncology, Rede Americas Oncology, Brasília, BRAZIL.
Background: Axillary lymph node dissection (ALND) remains the standard of care for patients with breast cancer with residual nodal disease (ypN+) after neoadjuvant chemotherapy (NAC). While the results of randomized controlled trials on the safety of omitting ALND are still awaited, retrospective studies suggest that de-escalation is already incorporated into clinical practice. This systematic review and meta-analysis aimed to evaluate oncological outcomes of sentinel lymph node biopsy (SLNB) alone versus ALND in patients with residual nodal disease after NAC. Methods: A systematic search of PubMed, Embase, and the Cochrane Library was performed. Eligible studies included randomized controlled trials and observational cohort studies of patients with clinically node-positive breast cancer who received NAC and were found to have ypN+ disease at surgery. Studies reporting outcomes of axillary recurrence, distant recurrence-free survival (DRFS), and/or overall survival (OS) were included. Hazard ratios (HRs) and 95% confidence intervals (CIs) comparing SLNB to ALND were extracted or estimated from Kaplan-Meier curves using validated methods. A random-effects meta-analysis was conducted. Results: Nine retrospective studies encompassing a total of 13,160 patients were included in the analysis. Of these, 3,831 patients underwent SLNB alone, while 9,329 underwent ALND. Axillary recurrence outcomes were reported in six studies. The pooled axillary recurrence rate was 5.91% (95% CI: 3.47%-9.92%) in the SLNB group and 5.24% (95% CI: 3.66%-7.44%) in the ALND group. SLNB was associated with a statistically significant reduction in axillary recurrence compared to ALND (HR = 0.94, 95% CI: 0.90-0.99), although the difference may not be clinically meaningful. Five studies reported DRFS with no significant difference between SLNB and ALND (HR = 0.99, 95% CI: 0.83-1.18), though substantial heterogeneity was present (I² = 69.7%), suggesting variability that may affect outcomes. Eight studies reported OS outcomes and no significant difference in OS between the two groups (HR = 1.08, 95% CI: 0.94-1.24, p = 0.26). Notably, all these findings may reflect selection bias, as patients selected for SLNB alone may have been more likely to have favorable baseline characteristics. Conclusion: This meta-analysis of retrospective studies suggests that omission of ALND in ypN+ patients does not compromise axillary control and may be associated with comparable regional and distant control with similar survival. However, due to the inherent limitations of retrospective data—including potential selection bias and unmeasured confounding—these findings should be interpreted with caution. Prospective randomized trials are necessary to confirm the safety of omitting ALND and to identify which patients may be appropriate candidates for axillary surgery de-escalation.
Presentation numberPS2-03-25
Serratus anterior fascia flap versus muscle flap for implant coverage in immediate subpectoral breast reconstruction
Lilian de Sá Paz, Aristides Maltez Hospital; Oncoclínicas&Co –Medica Scientia Innovation Research (MEDSIR), Salvador, Brazil
L. d. Paz1, J. V. Biazús2; 1Department of Breast Surgery, Aristides Maltez Hospital; Oncoclínicas&Co –Medica Scientia Innovation Research (MEDSIR), Salvador, BRAZIL, 2Postgraduate Program in Health Sciences: Obstetrics and Gynecology, Federal University of Rio Grande do Sul, Porto Alegre, BRAZIL.
Background Breast reconstruction (BR) is an essential component of breast cancer treatment, contributes to quality of life. Implant-based techniques are the most commonly used for immediate reconstruction following mastectomy. Subpectoral implant placement offers benefits such as reduced implant palpability and decreased risk of rippling. Complete implant coverage also provides greater protection and prevents lateral migration of the implant. Traditionally, the inferolateral coverage in total submuscular reconstruction is achieved using the serratus anterior muscle flap (SMF). However, this approach is associated with potential donor site morbidity. Alternatives include synthetic meshes, acellular dermal matrices (ADMs), dermal flaps, and the serratus anterior fascia flap (SFF). The high cost limits the widespread use of ADMs. The SFF has emerged as a promising autologous alternative. Despite its potential, limited data exist on the use of SFF in immediate BR. This study aims to compare clinical, surgical and oncologic outcomes, including time to first adjuvant therapy between patients undergoing subpectoral BR with either the SFF or the SMF for inferolateral implant coverage. Methods A retrospective review was conducted of 209 medical records of women aged 18 years or older diagnosed with stage I-III invasive breast carcinoma. All patients underwent mastectomy followed by immediate subpectoral implant-based BR and received subsequent adjuvant chemotherapy or radiotherapy at Aristides Maltez Hospital, Salvador-BA, Brazil from January 2018 to December 2022. Patients were grouped based on the type of inferolateral implant coverage used: SFF (n=53; 25.4%) or SMF (n=126; 60.3%). Thirty patients with unrecorded flap details were excluded from analysis. Clinical, oncologic, and demographic data were extracted from medical records. The time from surgery to the initiation of adjuvant therapy (defined as either first chemotherapy cycle or first radiotherapy session) was calculated. Results The median age was 44 years (IQR 38-50), with a predominantly black women population (90.5%). The SFF group received significantly larger implants [375 mL (IQR 305-475) vs. 337.5 mL (IQR 300-390); p=0.01] and had a higher rate of direct-to-implant reconstruction (98.1% vs. 61.2%; p<0.01). No significant differences were observed between groups regarding age, menopausal status, clinical stage. The median time from surgery to initiation of adjuvant therapy was 11 weeks in both groups. Conclusions The SFF is a viable and cost-effective alternative to SMF or ADMs in immediate BR. It supports the use of larger implants, does not delay the initiation of adjuvant therapy. These findings highlight the broader applicability of SFF in reconstructive planning, particularly in resource-constrained settings.
| Characteristics |
All patients n = 179 Nº (%) |
Serratus fascia n = 53 Nº (%) |
Serratus muscle n=126 Nº (%) |
p-value |
| Age at surgery years median | 44.0 | 44.0 | 44.5 | 0.91 |
|
Menopausal Status n=158 Premenopause, Postmenopause |
122 (72.2), 36 (22.8) | 40 (88.9), 5 (11.1) | 82 (72.6), 31 (27.4) | 0.05 |
| BMI kg/m2 n=128 median | 25.38 | 25.40 | 25.36 | 0.88 |
| Clinical stage n=149 I, II, III | 23 (15.4), 89 (59.7), 37 (24.8) | 9 (20.0), 24 (53.3), 12 (26.7) | 14 (13.5), 65 (62.5), 25 (24.0) | 0.05 |
| Mastectomy type Total, SSM, NSM | 53 (29.6), 99 (55.3), 27 (15.1) | 8 (15.1), 35 (66.0), 10 (18.9) | 45 (35.7), 64 (50.8), 17 (13.5) | 0.02 |
| Axillary surgery SLNB, ALND | 78 (43.6), 101 (56.4) | 29 (54.7), 24 (45.3) | 49 (38.9), 77 (61.1) | 0.07 |
| Implant volume mL n=126 median | 350 | 375 | 330 | 0.01 |
| Implant type n=173 Direct-to-implant, Tissue expander | 125 (72.3), 48 (27.7) | 51 (98.1), 1 (1.9) | 74 (61.2), 47 (38.8) | < 0.01 |
Presentation numberPS2-03-26
Lymphatic-sparing sentinel lymph node biopsy to prevent breast cancer-related lymphedema
Eleni Kohilakis, Montefiore Medical Center, Bronx, NY
E. Kohilakis1, S. Chow2, N. Habboosh2, M. Bajwa2, M. McEvoy2, S. Feldman2; 1Internal Medicine, Montefiore Medical Center, Bronx, NY, 2Surgery, Montefiore Medical Center, Bronx, NY.
Background: Breast cancer-related lymphedema (BCRL) remains a persistent and impactful morbidity associated with axillary surgery. Even with sentinel lymph node biopsy (SLNB), the reported incidence of BCRL is 5-7%. As surgical oncology shifts toward minimizing treatment-related morbidity—particularly in patients with clinically node-negative, early-stage breast cancer—techniques that reduce the risk of BCRL are increasingly valuable. Large trials such as SOUND and INSEMA support de-escalation of axillary surgery, demonstrating that SLNB may be safely omitted in select low-risk patients. These findings reinforce efforts to refine surgical techniques for those who still require axillary staging. Technique: We propose a novel, broadly applicable surgical approach: lymphatic-sparing sentinel lymph node biopsy (LSSNBX). This technique preserves and reapproximates the afferent and efferent lymphatic channels after sentinel node excision to facilitate lymphangiogenesis and reduce postoperative lymphedema risk. Unlike axillary reverse mapping (ARM)-dependent approaches, LSSNBX avoids additional mapping procedures, making it more feasible in varied practice settings. The procedure begins with dual-agent mapping (Technetium-99m, indocyanine green [ICG], or blue dye). After identification of the sentinel lymph node, an axillary crease incision is used to access the node. During excision, the afferent and efferent lymphatic bundles are carefully preserved and prevented from retracting. These lymphatic channels are then reapproximated using an absorbable suture, promoting potential reconnection and continuity of lymphatic flow. Importantly, this method simplifies prior ARM-based strategies by performing lymphatic preservation routinely, regardless of ARM findings. Results: A retrospective review at our institution included 103 patients: 52 underwent LSSNBX and 51 underwent standard SLNB without lymphatic preservation. Patient demographics indicated the LSSNBX group was older and had a lower mean BMI. At three-month follow-up using bioimpedance spectroscopy (SOZO®), no cases of BCRL were identified in either group. Notably, the LSSNBX technique added minimal operative time and required no specialized equipment, supporting its feasibility and cost-effectiveness. Conclusion: LSSNBX is a safe, simple, and scalable modification of standard SLNB that aligns with the current movement toward minimally invasive, morbidity-reducing surgical oncology. By preserving and reapproximating lymphatic channels intraoperatively, this technique may reduce the incidence of BCRL and improve long-term patient quality of life. As axillary surgery continues to evolve, the LSSNBX technique represents a promising step in optimizing outcomes. Further studies with larger cohorts and extended follow-up are warranted to validate these preliminary findings.
Presentation numberPS2-03-27
Comparison of Robot-assisted and Conventional Partial Mastectomy in Breast Cancer Patients: A Pilot Study
Si Yeon Lee, Yonsei University College of Medicine, Seoul, Korea, Republic of
S. Lee1, J. Ahn1, S. Lim2, H. Alanazi1, H. Park1; 1Surgery, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Surgery, Inha University Colleage of Medicine, Incheon, KOREA, REPUBLIC OF.
Background : Robotic-assisted breast surgery has emerged as a minimally invasive option to achieve oncologic safety and favorable cosmetic outcomes. While robotic-assisted nipple-sparing mastectomy has been studied, data on robotic-assisted partial mastectomy (RAPM) remain limited. This pilot study aimed to compare surgical and patient-reported outcomes between RAPM and conventional partial mastectomy (CPM) in patients with early-stage breast cancer. Methods : This was a non-randomized, prospective comparative pilot study conducted at Severance Hospital between March and July 2024. Ten patients were enrolled, with five undergoing RAPM and five undergoing CPM. Clinicopathologic characteristics, operative outcomes, complication rates, costs, and patient-reported outcomes were analyzed. Fisher’s exact test and the Mann-Whitney test were used to compare categorical and continuous variables, respectively. Results : There were no significant differences in age (median 54.0 vs 48.0 years, p = 0.095), BMI (24.9 vs 21.7, p = 0.095), menopausal status, hormone receptor status, HER2 expression, Ki-67 index, histologic grade, or TNM stage between groups (all p>0.05). The incision size was significantly smaller in the RAPM group (2.74cm vs. 5.20cm, p=0.008). Operative time, blood loss, drain output, and postoperative complications were similar. One patient in the RAPM group experienced a hematoma and wound dehiscence. Surgical cost was approximately 5,000 USD higher in the RAPM group, but the difference was not statistically significant (p=0.140). Patient satisfaction and health-related quality of life (HRQoL) were assessed using the BREAST-Q and EQ5D_5L. Postoperative satisfaction improved in both groups. BREAST-Q scores were slightly higher in the CPM group (100.2 vs. 95.2, p = 0.690), while EQ5D_5L index scores were slightly higher in the RAPM group (0.8498 vs. 0.8276, p = 1.000). No statistically significant differences were observed. Conclusion : RAPM was shown to be a feasible and safe alternative to CPM for early-stage breast cancer patients. While both techniques yielded comparable clinical and patient-reported outcomes, RAPM demonstrated a significant cosmetic advantage due to a smaller incision size. These findings support further evaluation of RAPM in larger, long-term studies.
Presentation numberPS2-03-28
An Investigation to Identify the Factors for Negative Sentinel Lymph Node in Early Breast Cancer Cases
Reiki Nishimura, Fukuda Hospital, Kumamoto City, Japan
R. Nishimura1, Y. Ueda2, K. Kiyohara2, M. Funagayama2, N. Ikeda2, A. Kato2, H. Tokiniwa2, R. Higashi3, M. Nakahara3, H. Ifuku3, T. Hayashi4, Y. Sagara5, Y. Sagara5, S. Ohno5; 1Department of Breast Surgery, Fukuda Hospital, Kumamoto City, JAPAN, 2Department of Breast Surgery, Sagara Hospital Miyazaki, Miyazaki City, JAPAN, 3Department of Radiology, Sagara Hospital Miyazaki, Miyazaki City, JAPAN, 4Department of Pathology, Sagara Hospital Miyazaki, Miyazaki City, JAPAN, 5Department of Breast and Thyroid Surgical Oncology, Sagara Hospital, Kagoshima City, JAPAN.
Background: Sentinel Lymph Node Biopsy (SLNB) in breast cancer is a standard procedure used to determine whether breast cancer has spread to the axillary lymph nodes. It has replaced full axillary lymph node dissection (ALND) in early-stage clinically node-negative (cN0) breast cancer patients. However, SLNB is associated with some rare complications such as pain, seroma and lymphedema. Omission of SLNB in breast cancer is an evolving area in clinical practice and may be a safe and reasonable option. To identify cases in which SLNB was negative, we conducted a retrospective study using early breast cancer data from our hospital registry. Methods: A total of 2,589 breast cancer patients underwent SLNB between June 2016 and April 2025. Of these, 1,956 cases of cN0 invasive breast cancer who did not undergo preoperative chemotherapy were included in this retrospective study. Pathological diagnosis using frozen section or one-step nucleic acid amplification (OSNA) method was used for the diagnosis of SLNs. The number of SLNs examined ranged from 1 to 7, with a mean of 1.8 nodes. The disease-free survival (DFS) was calculated using the Kaplan-Meier method and analyzed using the log-rank procedure. The multivariate analysis of factors for negative SLN was performed using logistic regression analysis. Results:1. Metastasis to the SLNs was observed in 379 cases (19.4%) including micro-metastasis. The median number of metastatic SLN was 1.0. SLN metastasis was observed in 139 (14.5%) cases with breast conserving surgery (Bp) and 240 (24.1%) cases with total mastectomy (Bt). Of cases with positive SLN, 378 cases (87.9%) received ALND. 2. Cases with invasive ductal carcinoma (IDC) had a 19.9% positive SLN rate and cases with invasive lobular carcinoma (ILC) and invasive micropapillary carcinoma (IMPC) had higher positive rates than IDC. 3. SLN metastasis was not associated with ER, PgR and HER2 status. However, significant differences were observed in age, tumor size, Ki-67 index value and histological grade. The incidence of SLN metastasis was significantly lower in patients aged 50 years or older (postmenopausal), a tumor size of 2 cm or less, a Ki-67 index value 50 years of age, a tumor size of ≤ 2 cm and a HG of 1 was favorable regardless of the presence or absence of SLN metastasis (99.5% for negative SLN and 100% for positive SLN). Conclusion: Metastasis to SLN was found in 19.4% of cN0 breast cancer cases. Factors related to negative SLN were age (50 years or older), tumor size (2cm or less), and HG (1), and the positive SLN rate was significantly higher in cases with the Luminal B subtype. The prognosis for cases with the above factors was good, and recurrences were rarely observed. In the future, whether SLNB is required will depend on the above-mentioned patient characteristics.
Presentation numberPS2-03-29
Evaluating Prognosis According to Axillary Surgery in Breast Cancer Patients with Isolated Internal Mammary Lymph Node Metastasis (cN2b): A Retrospective Cohort Study
Yoshiharu Ishizaka, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
Y. Ishizaka, T. Sakai, M. Kasahara, M. Kai, A. Kanazawa, E. Taniguchi, Y. Ito, Y. Kimura, U. Nakadaira, Y. Inoue, T. Maeda, N. Yamashita, A. Kataoka, N. Uehiro, T. Ueno; Breast Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN.
Background Metastasis to the internal mammary lymph nodes (IMLN) plays a critical role in the regional staging and prognosis of breast cancer. Although IMLN involvement often occurs alongside axillary lymph node (AXLN) metastasis, isolated IMLN metastasis without axillary involvement (cN2b) is rare, and the optimal surgical approach for these patients remains uncertain. While axillary lymph node dissection (ALND) is generally recommended for node-positive disease, its necessity in cases of cN2b is debatable. This study aims to clarify the prognostic significance of AXLN status and to assess whether ALND can be safely omitted in breast cancer patients presenting with isolated IMLN metastasis. Methods We retrospectively analyzed patients with IMLN metastases confirmed preoperatively by cytology, who underwent surgery at our institution between 2007 and 2021. Inclusion required cytological confirmation of IMLN metastasis. Exclusion criteria included clinical T4 tumors, supraclavicular or distant metastases, and bilateral breast cancer. Patients were stratified into two groups: cN2b (isolated IMLN metastasis, n=21) and cN3b (IMLN with AXLN metastasis, n=104). Clinicopathological characteristics and 5-year disease-free survival (DFS) were compared between groups. In the cN2b cohort, oncologic outcomes were further evaluated according to the type of axillary surgery performed: sentinel lymph node biopsy (SLNB) alone versus ALND. Results Of 15,231 surgically treated breast cancer patients, 230 had IMLN metastasis. After exclusions, 125 (0.8%) were included: 21 (0.1%) with cN2b and 104 (0.7%) with cN3b. The median follow-up was 59 months. The cN2b group demonstrated a significantly higher frequency of medial tumor location (52% vs. 30%, p=0.046) and tended to present with smaller tumor size compared to the cN3b group (p=0.039). Histologically, invasive ductal carcinoma was the predominant subtype in both groups. Hormone receptor-positive tumors accounted for the majority of cases in both cN2b and cN3b groups (62% and 60%, respectively, p=0.85). HER2-positive tumors were seen in 19% of cN2b and 26% of cN3b cases (p=0.50).The 5-year DFS was significantly better in cN2b patients compared to cN3b (94.7% vs. 64.0%, p=0.0049), indicating favorable prognosis in the absence of axillary involvement. Among cN2b patients, 10 underwent SLNB alone and 10 underwent ALND. The remaining patient did not undergo any axillary surgery due to personal preference. DFS rates did not significantly differ between these subgroups (88.9% for SLNB vs. 100% for ALND, p=0.32). In the cN2b group, 20 patients (95.2%), and in the cN3b group, 101 patients (97.1%) received postoperative radiotherapy, with irradiation fields including the regional lymph node areas in all cases. One SLNB-only patient developed distant bone metastasis, but no regional axillary recurrence occurred in any group. Two ALND patients had previously undetected axillary metastases though it is unclear whether these findings had any effect on DFS. Conclusions This study suggests that breast cancer patients with isolated IMLN metastasis and no axillary involvement (cN2b) have significantly better outcomes than those with combined IMLN and AXLN metastasis (cN3b). Furthermore, omission of ALND in cN2b cases may be feasible and did not appear to compromise disease control, with no locoregional failures observed in the SLNB-only group. These findings support the potential safety and efficacy of a less invasive axillary management strategy—SLNB alone—for appropriately selected cN2b patients. Considering the rarity of this clinical subset, larger prospective studies are warranted to further validate these findings and refine treatment guidelines for isolated IMLN metastasis.
Presentation numberPS2-03-30
Age at breast cancer diagnosis and short-term postoperative outcomes for women treated in a universal-coverage health system
Gabrielle Falco, San Antonio Military Medical Center, Fort Sam Houston, TX
G. Falco1, Y. L. Eaglehouse2, S. Darmon2, M. Nealeigh2, R. W. Krell2, C. D. Shriver2, K. Zhu2; 1Surgery, San Antonio Military Medical Center, Fort Sam Houston, TX, 2Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD.
Background: Breast cancer is the most commonly diagnosed cancer among women—either active duty, retirees, or dependents—in the U.S. Military Health System (MHS). Surgical treatment is an essential component of treatment for patients without metastatic disease. Differences in insurance coverage and access to care by patient age in the general U.S. population may affect breast cancer diagnosis and surgery. The impact of age on surgical outcomes in breast cancer under conditions of universal healthcare access remains unclear. This study compared postoperative outcomes following breast cancer surgery according to patient age at diagnosis in the U.S. MHS, which provides universal access to care to all its beneficiaries with no or minimal out-of-pocket cost. Methods: We identified women aged 18 and older with stage I-III breast cancer between 2001 and 2014 undergoing partial or total mastectomy without reconstruction in the MilCanEpi database. Multivariable Poisson regression estimated the adjusted risk ratios (ARRs) with 95% confidence intervals (CIs) in association with age at diagnosis (18-39, 40-49, 50-64, and >65) for 30-day general and breast complications (surgical site infection, seroma, hematoma, or lymphedema), reoperation, and hospital readmission while controlling for potential confounders. Results: The study included 7,835 women who were 18-39 (9.2%), 40-49 (23.6%), 50-64 (42.7%), and >65 (24.5%). Women 18-39 were more likely to present with stage III tumors (20.2%) and high-grade tumors (51.5%) than other age groups. While older women (>65) had higher unadjusted rates of any complication (RR=1.34; 95% CI=1.12-1.60) or general complication (RR=1.77; 95% CI=1.25-2.50) relative to women diagnosed at age 50-64, these differences were not statistically significant after multivariable adjustment for demographic, tumor, and treatment variables and time to surgery. The overall risk of any breast complication did not differ significantly by age. However, women aged 40-49 had a statistically increased adjusted risk of seroma (ARR=1.50; 95% CI=1.03-2.18) compared to women age 50-64. No significant age-related differences were observed for reoperation or hospital readmission after adjustment. Conclusion: In the universal-access Military Health System, the risk of 30-day postoperative complications and hospital readmissions varied by age among women undergoing surgery without reconstruction for non-metastatic breast cancer. However, these differences were attenuated after adjustment for clinical and demographic factors, suggesting that age may play a limited role in short-term surgical outcomes for breast cancer. Surgical decision-making should continue to prioritize tumor characteristics, comorbidity burden, and other clinical factors rather than age alone when assessing perioperative risk. Disclaimer: The contents of this abstract are the sole responsibility of the authors and do not necessarily reflect the views, assertions, opinions, or policies of the Uniformed Services University of the Health Sciences, the Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., the Department of Defense, or the Departments of the Army, Navy, or Air Force. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. government.
Presentation numberPS2-05-01
Pro-tigr-matrix-prepectoral-trial
Marc Antonius Maria Thill, Agaplesion Markus Krankenhaus, Frankfurt, Germany
M. A. Thill1, E. Klein2, T. Lantzsch3, D. Rezek4, L. Bauer5, K. Kelling1; 1Klinik für Gynäkologie und Gynäkologische Onkologie, Agaplesion Markus Krankenhaus, Frankfurt, GERMANY, 2Klinik für Gynäkologie und Geburtshilfe, Technische Universität München, München, GERMANY, 3Klinik für Frauenheilkunde und Geburtshilfe, Krankenhaus St. Elisabeth und ST. Barbara Halle (Saale) GmbH, Halle (Saale), GERMANY, 4Senologie und Ästhetische Chirurgie, Evangelisches Krankenhaus Wesel GmbH, Wesel, GERMANY, 5Klinik für Gynäkologie und Gynäkologische Onkologie, GNR-Klinik Weinheim, Weinheim, GERMANY.
Background: Implant reconstruction after breast removal is a common method of restoring the appearance of the breast. For many years, the subpectoral implant position was considered the gold standard. However, due to the advancement of technology, the current standard is the prepectoral implant position. To support this technique, resorbable or non-absorbable meshes or acellular dermal matrices can be used in the reconstruction to stabilize the implant, fix it and preform a capsule. Meta-analyses have shown that neither the pre- nor the subpectoral position is an advantage for the patient in terms of complication rate. With regard to the use of meshes or matrices, there is still no head-to-head comparison between a mesh, an ADM or nothing. The aim of this study is to determine the patient-reported and cosmetic outcome, as well as the complications associated with the use of the fully absorbable TIGR mesh in the prepectoral implant reconstruction of the breast.Trial design: The PRO-TIGR-Matrix-PREPECTORAL study is a national, prospective, multicentre post market surveillance study on „Patient Reported Outcome“ of implant based primary or secondary reconstructive breast surgery after mastectomie using the complete resorbable synthetic Matrix (TIGR-Matrix, Novusscientific Sweden). The observation period is 3 years with a focus on patient reported satisfaction (as measured by BreastQTM), cosmetic outcome and complication rate. The study period will be 54 months, per patient: 36 months plus optional further follow-up surveys as part of the usual follow-up until the end of the study. The recruitment phase will be 18 months, the follow-up phase 36 months. 135 patients will be recruited.Primary endpoints: The primary endpoint of this clinical investigation is to assess quality of life after 12 months determined by the questionnaire BreastQTM. The study is considered successful, if and only if non-inferiority can be established for all four domains. Secondary endpoints: The secondary endpoints are patient reported outcome/quality of life measured by BreastQ after 6, 24, and 36 months, the feasibility, efficacy, safety of the heterologous complete resorbable TIGR-Matrix in prepectoral implant-based breast reconstructive surgery. This includes the number and rate of occurrence of adverse events and a tabular list of adverse events that have occurred; cosmetic result reported by doctor and patient.Primary Measurements: Comparison of the pre- and postoperative (12 months) mean score of the BreastQ category “breast satisfaction” (well-being-breast); Patient-Reported-Outcome (PRO) measured with the specific questionnaire for breast reconstruction BreastQ 12 months after surgery.Secondary measurements: Comparison of the pre- and postoperative (6, 12, 24, 26 months) mean score of the BreastQ category “breast satisfaction” (well-being-breast); Means scores of all BreastQ categories pre- and postoperatively after 6, 12, 24, 36 months. Complication rate: defined as major complications, minor complications; specific complications, such as seroma-volume, need for aspiration in symptomatic patients and number of aspirations in postoperative follow up, reconstructive failure (defined as unplanned implant loss), cosmetic outcome (domains of cosmesis of BreastQ) and rate of unplanned conversion operations (to mastectomy without reconstruction; to mastectomy with reconstruction; to autologous reconstruction).Current status:20 patients are recruited.First Interim analysis planned: 40 evaluable patients with 6 months FU data availableNo uncommon complications.Sponsor: AWOgynFunding: Novus Scientific
Presentation numberPS2-05-03
Lateral intercostal artery perforator (LICAP) flap: a new surgical approach for granulomatous lobular mastitis
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen, B. Zhu, J. Xue, L. Zheng; Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA.
Background: Granulomatous lobular mastitis (GLM) is characterized by nonspecific chronic inflammation concentrated in breast lobules. Surgical resection is one of the most common treatment options for GLM. Here we report our 1-year experience with a new treatment approach for GLM that involves the use of a lateral intercostal artery perforator (LICAP) flap. Methods: During March 2024 to May 2024, we enrolled 20 GLM patients who underwent breast surgery with the use of LICAP flap. All patients were women; most of the patients were 18-50 years old; and all the clinical manifestation of GLM was breast mass. Then, we evaluated operative data (time, blood loss, and intraoperative complications), primary healing time, and recurrence, and we also analyzed the safety of day-case surgery and patient-reported outcomes (cosmetic outcome, and improvement in dressing change and bathing) at 12 months. We recorded the occurrence of common postoperative complications of the breast and documented the follow-up conditions with photographs. Results: Median age of the patients was 42 years (range: 27 to 50 years). Duration of GLM was about 1 month, and median size of the lesions was 4.75 cm (range: 3.6 to 8 cm). No significant intraoperative complications occurred. All wounds healed by primary intention. The patients’ satisfaction with the breast shape was as follows: excellent (85%), good (15%), acceptable (0%), and poor (0%). No recurrence was recorded. Day-case surgery was proven to be safe and feasible, and could reduce hospital stay. Conclusion: For GLM patients refractory to conservative therapy or former unsatisfactory surgical management whose lesion is larger than 3 cm, LICAP is a suitable approach to fill the post-operation defect and achieve a relatively satisfactory cosmetic outcome.
Presentation numberPS2-05-04
Single-dye sentinel lymph node mapping for targeted axillary dissection in breast cancer patients after neoadjuvant chemotherapy: a prospective study
Wuzhen Chen, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
W. Chen, Y. Jini, P. Liwei, N. Chao, H. Jian; Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, CHINA.
Background: Accurate axillary staging after neoadjuvant chemotherapy (NAC) in initially node-positive breast cancer remains controversial, particularly in settings where radioisotopes are unavailable. We prospectively evaluated the performance of single-blue-dye sentinel lymph node biopsy (SLNB) combined with clip-guided targeted axillary dissection (TAD).Methods: Consecutive patients with biopsy-proven cN1-2 disease received standard NAC. The biopsy-positive node was clipped prior to treatment. During surgery, 2-4 mL of methylene blue was injected peritumorally; all blue-stained nodes were resected as sentinel lymph nodes (SLNs), followed by selective excision of the clipped (marked) node. Completion axillary lymph node dissection was performed only in cases where residual nodal disease was identified. The primary endpoints were the SLN identification rate (IR), concordance of pathologic status between the marked lymph node (MLN) and SLNs, and the false-negative rate (FNR; negative SLNs with metastasis in non-SLNs).Results: Twenty-nine women (median age 48 years; range, 31-68 years) were enrolled between July 2022 and February 2024. SLNs were successfully identified in 28/29 patients, yielding an IR of 96.6% (95% CI 82.8-99.4). The MLN was retrieved as an SLN in 28/29 cases (96.6 %), and it constituted the first SLN removed in 19/28 (67.9 %). Residual nodal disease was present in 11/29 patients (37.9%). One of these 11 patients had negative SLNs but positive non-SLNs, leading to an overall FNR of 9.1% (1/11; 95% CI 0-41.3). Nodal pathological complete response (pCR) rates varied by molecular subtype: 90% in triple-negative tumors, 66.7 % in HER2-positive tumors, and 44.4 % in HR+/HER2-negative tumors. No dye-related adverse events were observed.Conclusions: The combination of single-blue-dye SLNB and clip-guided TAD demonstrated a high identification rate, excellent MLN-SLN concordance, and FNR below the 10% benchmark. This pragmatic and minimally invasive approach provides a reliable alternative to routine axillary lymph-node dissection in post-NAC patients when dual tracers or radioisotopes are unavailable.
Presentation numberPS2-05-05
The Anatomic Basis of Thoracodorsal Artery Perforator-based Flap: Cadaveric and Angiographic Study
Ramita Mukherjee, All India Institute of Medical Sciences, New Delhi, Delhi, India
R. Mukherjee1, S. K. Pattashanee1, B. K. Singh1, A. Krishna1, S. Soren1, D. Kandasamy2, S. V1; 1Department of Surgical Disciplines, All India Institute of Medical Sciences, New Delhi, Delhi, INDIA, 2Department of Radiology, All India Institute of Medical Sciences, New Delhi, Delhi, INDIA.
INTRODUCTION: Breast cancer surgery has shifted towards breast conservation surgery. Perforator-based flaps play a pivotal role. The thoracodorsal artery perforator-based (TDAP) flap is one such. In TDAP, varied amount of skin and subcutaneous fat can be used. It can reach any quadrant of the breast. It can even be converted to a musculocutaneous flap, if a larger volume is required. In this study, the anatomical and vascular basis of TDAP were analysed using computed tomography angiographic (CTA) and cadaveric dissection (CD). METHODS: A prospective observation study was carried out over 2 years in AIIMS, New Delhi. TDAP was dissected in 10 cadavers (20 sides). As part of metastatic workup, CTA was done in 23 locally advanced breast cancer (LABC) patients along with bone scan. Fresh, Theil cadavers, cadavers ≥ 18 years of age, and female BC patients >18 years were included. Injury/ scarring of chest wall, previous axillary surgery and cadavers/ patients <18 years were excluded. Primary objective was to determine course, precise localization and diameter of TDAPs. Secondary objective was to analyse and compare pattern of distribution of TDAPs: Dominant (DP) and Non-Dominant (NDP), in CD and CTA. Perforators were marked in sequence as TDAP 1, 2, 3 and so on. Analysis parameters of TDAP, in CD and CTA were distance from apex of axilla, distance from anterior border of latissimus dorsi (LD), diameter of perforator at its entry into muscle and perforator’s length. DPs and NDPs were analysed. Data was recorded in MS Excel and analysed in SPSS v23. Descriptive statistics were elaborated as means/standard deviations, medians/IQRs for continuous variables. Frequencies and percentages for categorical variables, group comparisons of continuously distributed data using independent sample ‘t’ test, Chi-squared test for group comparisons of categorical data, Paired t-test for paired analysis for continuous variables and Wilcoxon Signed Rank test for non-parametric data were used. RESULTS: A total of 52 TDAPs (20 in males and 32 in females) in cadavers and 57 TDAPs in 23 LABC patients were identified. Spatial distribution-wise (CD), 20 (38.5%) were TDAP1, 20 (38.5%) were TDAP2 and 12 (23.1%) TDAP3. From CTA, 45 (78.9%) were TDAP1 and 8 (14%) were TDAP2. Only 2 perforators were found as TDAP 3 and 4 in CTA. At least 1 DP was identified in CD and CTA. TDAP detection frequency and distance from the anterior border of LD were more in CD. In CD and CTA, the mean length of TDAP and distance from the apex of the axilla were comparable. The diameter of the perforator was significantly more in CTA. Table 1 shows the comparison of CTA and CD findings (significant ones).
| Parameters [***Significant at p < 0.05, 1: Chi-square test, 2: Wilcoxon Mann Whitney U test] | CT-angiography (n = 57 perforators in 23 patients) | Cadaveric dissection (n = 52 perforators in 10 cadaver) | p value | ||||
| Dominant TDAP *** | 45 (78.9%) | 20 (38.5%) | <0.0011 | ||||
| Non-Dominant TDAP *** | 12 (21.1%) | 32 (61.5%) | <0.0011 | ||||
| Diameter Of Perforator (mm)*** | 1.48 ± 0.32 | 1.10 ± 0.25 | <0.0012 | ||||
| Distance From Anterior Border Of LD (cm)*** | 0.84 ± 0.62 | 2.36 ± 0.87 | <0.0012 |
CONCLUSION: TDAPs have a consistent distribution pattern, adequate length and vessel calibre, when analysed in CD and CTA. The anatomical and vascular basis of TDAP (DPs and NDPs) are important for better perforator-based reconstructions.
Presentation numberPS2-05-06
Safety and Oncologic Outcomes of Immediate Breast Reconstruction Following Neoadjuvant Chemotherapy
Mai Tomiguchi, Kumamoto University Hospital, KUMAMOTO, Japan
M. Tomiguchi, K. Hidaka, L. Goto-Yamaguchi, Y. Yamamoto; Breast and Endocrine Surgery, Kumamoto University Hospital, KUMAMOTO, JAPAN.
Background; Although the Japanese Breast Cancer Clinical Practice Guideline (2022) allow immediate breast reconstruction (IBR) after neoadjuvant chemotherapy (NAC) with caution, concerns remain regarding delayed would healing, increased surgical complications, and unknown oncologic safety. We aimed to evaluate the safety and oncologic outcomes of IBR following NAC by comparing patients with and without NAC at our institution. Methods: We retrospectively reviewed 228 breast reconstructions performed at the time of mastectomy from July 2013 to September 2024. Of these, 29 breasts (13.0%) underwent IBR after NAC (NAC group), and 194 breasts (87.0%) were reconstructed without NAC (control group). We analyzed clinicopathological characteristics, operative time, blood loss, length of hospital stay, postoperative complications, recurrence, and survival. Median follow-up was 73 months (range, 3-134). Results: Patient age and surgical procedures (including nipple-sparing mastectomy rates: 41.4% vs 44.3%) were comparable. Autologous reconstruction was more frequent in the control group (27.3% vs 10.3%, p=0.06). No significant differences were observed in: operative time: 254 vs 271.5 min (p=0.71), blood loss: 65 vs 63 mL (p=0.71), hospital stay: 13 vs 14 days (p=0.26), overall complication rate: 37.9% (NAC) vs 30.4% (control) (p=0.42), major complications requiring reoperation: 13.3% vs 13.0% (p=1.00). Local recurrence occurred in 1 (3.4%) vs 7 (3.6%) cases (p=1.00). Distant recurrence was significantly more common in the NAC group (17.2% vs 0.52%, p=0.0005), as was breast cancer-specific mortality (10.3% vs 0%, p=0.002). These outcomes likely reflect the higher baseline recurrence risk of the NAC group. Conclusions: Immediate breast reconstruction following NAC was not associated with increased surgical risk. Although oncologic outcomes were worse in the NAC group, this is likely attributable to patient selection with inherently higher recurrence risk. With proper patient counseling and risk stratification, IBR after NAC can be considered a safe and feasible option. Further prospective studies incorporating patient-reported outcomes are warranted.
Presentation numberPS2-05-07
Evaluation of fusion imaging guidance in radiofrequency ablation for early-stage breast cancer
CHIE YAMANAKA, Kindai University, Osaka, Japan
C. YAMANAKA, S. MATSUZAKI, Y. HAYAKAWA, H. MANABE, M. KUBOTA, T. ITO, Y. KOMOIKE; Department of Surgery, Kindai University, Osaka, JAPAN.
Background: Radiofrequency ablation (RFA) for early-stage breast cancer with tumors ≤15 mm in diameter was approved for coverage under Japan’s national health insurance in December 2023, following favorable results from several clinical trials. Clinical RFA procedures commenced at our institution in May 2024. Small early-stage breast cancers—especially non-mass lesions—are often difficult to visualize or delineate using conventional B-mode ultrasound (US) alone. Additionally, gas formation during RFA may obscure the lesion, complicating precise localization for additional ablation. This study aimed to evaluate the clinical utility of fusion imaging combining contrast-enhanced breast magnetic resonance imaging (MRI) with US and/or contrast-enhanced US (CEUS) to improve lesion targeting and procedural accuracy during RFA. Patients and Methods: Fourteen patients with early-stage breast cancer underwent RFA guided by fusion imaging. A volume navigation system (LOGIQ E10x, GE Healthcare) was used to fuse dynamic contrast-enhanced MRI with US, aligning anatomical landmarks such as the nipple to ensure accurate lesion localization. CEUS using Sonazoid® was performed to evaluate intratumoral microvascularity. A Cool-tip needle (Medtronic, CO, USA) was inserted with a planned 1-cm safety margin around the tumor. Intratumoral impedance was measured upon insertion, and 10-20 mL of 5% glucose was injected into the retromammary space to prevent heat diffusion. Ablation was initiated at 5 W and continued until automatic roll-off occurred, due to increasing impedance. Therapeutic efficacy was assessed ≥4 weeks post-procedure using follow-up imaging and ultrasound-guided vacuum-assisted biopsy (US-VAB) of the ablation zone. Results: Patient ages ranged from 34 to 82 years (mean: 57 years), with tumor sizes from 7 to 15 mm (mean: 12.4 mm). Histopathologic findings included ductal carcinoma in situ (DCIS; n=8) and invasive ductal carcinoma (IDC; n=6). Molecular subtypes included Luminal A-like (n=4) and Luminal B-like (n=2). Tumors were located in the upper outer quadrant (n=8), upper inner quadrant (n=2), lower outer quadrant (n=2), and deep medial or axillary tail regions (n=1 each). Fusion imaging significantly improved visualization and delineation of lesions that were challenging to identify with B-mode US alone. It also enabled accurate re-localization of the original lesion during additional ablation sessions, resulting in successful complete ablation in all cases. Initial impedance ranged from 116 to 245 Ω (mean: 169 Ω), maximum ablation temperature from 83°C to 100°C (mean: 93°C), and ablation time from 14 to 32 minutes (mean: 20 minutes). One case of postoperative nipple retraction was observed; no other major complications occurred. No viable cancer cells were detected in any US-VAB specimens. Conclusion: Fusion imaging combining contrast-enhanced MRI with US/CEUS enhances the precision, safety, and efficacy of RFA for early-stage breast cancer, particularly for lesions that are poorly visualized on conventional US. Further accumulation of clinical cases is needed to standardize fusion imaging protocols and optimize outcomes.
Presentation numberPS2-05-08
Refusal of sentinel lymph node biopsy in patients with luminal A subtype early breast cancer
Petr Vladimirivich Krivorotko, FSBI “N.N. Petrov National Medical Research Center of Oncology” of the Russian Ministry of Health, Saint Petersburg, Russian Federation
A. O. Gorina1, P. V. Krivorotko1, A. S. Emelyanov1, D. A. Enaldieva1, E. K. Zhiltsova1, L. P. Gigolaeva1, T. T. Tabagua1, A. V. Komyakhov1, V. V. Mortada2, P. S. Pesotsky1, D. G. Ulrikh1, N. S. Amirov1, V. F. Semiglazov1; 1The department of breast tumours, FSBI “N.N. Petrov National Medical Research Center of Oncology” of the Russian Ministry of Health, Saint Petersburg, RUSSIAN FEDERATION, 2The department of oncology and reconstructive plastic surgery, FSBI “N.N. Petrov National Medical Research Center of Oncology” of the Russian Ministry of Health, Saint Petersburg, RUSSIAN FEDERATION.
BackgroundThe indications for and extent of axillary surgery in breast cancer have evolved significantly over the past two decades and remain an area of active research and debate. Sentinel lymph node biopsy (SLNB) is currently the standard approach for axillary staging in patients with early-stage breast cancer. However, emerging data suggest that axillary surgery may be safely omitted in select patients aged ≥60 years with early-stage, luminal A, clinically node-negative breast cancer. This prospective study aimed to assess the applicability of this strategy in patients with hormone receptor-positive (HR+), HER2-negative (HER2−) tumors. MethodThis prospective study included 100 patients diagnosed with cT1-2N0M0, HR+HER2- breast cancer (BC) who underwent combined treatment from 2023 to 2025 at the FSBI «N.N.Petrov National Medical Research Center of Oncology» of the Russian Ministry of Health. All patients had clinically negative axillary lymph nodes confirmed via ultrasound, mammography and mammoscintigraphy/breast molecular imaging with 99mTc-MIBI. Surgical axillary staging was omitted in this patient population. Preoperative sentinel lymph node localization for adjuvant radiation treatment planning was performed using single-photon emission computed tomography (SPECT) lymphoscintigraphy. ResultsAmong 100 patients with cT1-2N0M0 HR+HER2- breast cancer median age was 67 [59; 86] years. Median tumor size was 15 [4; 30] mm and tumors were grade 1 or 2. Estrogen receptor (ER) expression ranged from 90-100% in 95 cases and 70-80% in 5 cases. Progesterone receptor (PR) expression was 0-10% in 17 cases, 20-40% in 4, and 60-100% in 79. The Ki-67 proliferation index was 1-10% in 29 cases, 11-20% in 46, and 21-30% in 25. Histological subtypes included 94 cases of invasive carcinoma of no special type (NST), 3 – lobular carcinomas, and 3 – mucinous carcinomas. All patients in the study received adjuvant radiotherapy, administered in hypo- or ultra-hypofractionation regimens at the discretion of the radiation oncologist for breast and sentinel lymph nodes. Specifically, 15 patients received a 5-fraction regimen, 50 patients received 15 fractions, 35 patients received 16 fractions, and 2 patients underwent high-dose brachytherapy additionally. Adjuvant hormone therapy was recommended for all patients: 79 received Aromatase Inhibitors and 21 received Tamoxifen. No patient was recommended to receive adjuvant chemotherapy. ConclusionWe believe that these findings support careful implementation of omission of surgical staging of the axilla in postmenopausal patients with cT1-2N0, HR+HER2− breast cancer and a negative axillary lymph nodes. At our institution, this approach is currently implemented in patients over 59 years of age with ECOG performance status 20%, and Ki-67 index <30%.
Presentation numberPS2-05-09
Preoperative Assessment for the Omission of Sentinel Node Biopsy in Luminal/HER2-Negative Breast Cancer in Low-Resource Settings
Julia Dias do Prado, Hospital Universitário Antônio Pedro (HUAP-UFF), Niterói, Brazil
J. D. Prado1, S. A. Fialho2, A. V. Silva1, J. Moraes3, A. A. Vasconcelos4, G. R. Donald5, I. C. Rocha5, C. F. Monteiro1, V. V. Pignataro1, R. J. Vieira6, C. E. Barbosa2; 1Breast Surgery, Hospital Universitário Antônio Pedro (HUAP-UFF), Niterói, BRAZIL, 2Maternal and child care – Gynecology discipline, Universidade Federal Fluminense, Niterói, BRAZIL, 3Statistics Department, Universidade Federal Fluminense, Niterói, BRAZIL, 4Diagnostic imaging unit, Hospital Universitário Antônio Pedro (HUAP-UFF), Niterói, BRAZIL, 5Oncology and Hematology, Hospital Universitário Antônio Pedro (HUAP-UFF), Niterói, BRAZIL, 6Gynecology Department, Fundacao Oswaldo Cruz, Rio de Janeiro, BRAZIL.
Introduction: Breast cancer is the second most common malignancy in women worldwide after non-melanoma skin cancer and remains the leading cause of cancer-related mortality in this population. In Brazil, approximately 74,000 new cases are projected annually through 2025. Sentinel lymph node biopsy is the standard axillary management for early-stage breast cancer. However, emerging evidence suggests that patients with negative preoperative axillary involvement may not benefit significantly from axillary surgery.Objective: The aim of this study was to determine if the preoperative assessment can identify patients with absent or low axillary tumor burden who could safely omit sentinel lymph node biopsy during surgical treatment of luminal/HER2-negative breast cancer.Methods: We conducted a prospective observational study at a public university hospital operating within the Brazilian National Health System (SUS). Eligible candidates were women with early-stage (T1-T2 N0) luminal/HER2-negative breast cancer who exhibited no clinical or ultrasonographic evidence of axillary disease on preoperative assessment and subsequently underwent sentinel lymph node biopsy. Histopathological findings from surgery specimens were then correlated with the clinicopathological parameters during preoperative assessment.Results: Among the 40 participants, 12 (30%) exhibited axillary involvement, and none demonstrated extensive axillary disease (≥ 4 positive lymph nodes). The absence of tumor lymphovascular invasion was significantly associated with a negative axillary status (OR = 9.286; p = 0.018). Age ≥ 60 years was also associated with a negative axilla, although at a 10% significance level (OR = 3.5; p = 0.082).Conclusion: Negative preoperative axillary assessment is an effective strategy for selecting patients without extensive axillary disease in luminal/HER2-negative breast cancer. The absence of lymphovascular invasion and age ≥ 60 years increase the likelihood of a negative axilla. Clinicopathological parameters in early-stage luminal/HER2-negative breast cancer appear to provide sufficient data for selecting candidates for omission of sentinel lymph node biopsy, especially when the presence of low axillary tumor burden does not influence systemic therapy choice.Keywords: Sentinel lymph node biopsy; Breast cancer; Invasive ductal carcinoma; Sentinel lymph node; Lymphatic metastasis. Table: Logistic regression results for estimating the probability of a negative axillary status.
| Univariate logistic model | |||
| Clinicopathologic Characteristics | OR | IC95% | P value (Wald) |
| Menstrual Status | |||
| Premenopausal | 0,385 | (0,048; 3,113) | 0,371 |
| Postmenopausal | 1 | – | – |
| Tumor size (preoperative) | |||
| < 2 cm (T1) | 0,952 | (0,242; 3,748) | 0,944 |
| 2 to 5 cm (T2) | 1 | – | – |
| Age | |||
| ≥ 60 years | 3,5 | (0,854; 14,342) | 0,082 |
| < 60 years | 1 | – | – |
| Luminal subtype (postoperative) | |||
| Luminal A | 0,422 | (0,076; 2,341) | 0,324 |
| Luminal B | 1 | – | – |
| Histological grade | |||
| I | 1,667 | (0,365; 7,607) | 0,51 |
| II or III | 1 | – | – |
| Time between biopsy and surgery (days) | |||
| ≤ 121 days (median) | 0,375 | (0,091; 1,543) | 0,174 |
| > 121 days (median) | 1 | – | – |
| BMI | |||
| BMI ≥ 30 kg/m2 | 0,663 | (0,164; 2,676) | 0,564 |
| BMI < 30 kg/m2 | 1 | – | – |
| Diagnosis | |||
| screening | 0,556 | (0,141; 2,187) | 0,401 |
| Physical examination | 1 | – | – |
| lymphovascular invasion | |||
| No | 9,286 | (1,475; 58,467) | 0,018 |
| Yes | 1 | – | – |
| Multifocality | |||
| No | 0,418 | (0,043; 4,024) | 0,45 |
| Yes | 1 | – | – |
| Sentinel lymph nodes removed | |||
| < 3 | 0,647 | (0,166; 2,527) | 0,531 |
| ≥ 3 | 1 | – | – |
| *n=40 patients; CI, confidence interval; BMI, body mass index; OR, odds ratio. |
Presentation numberPS2-05-10
Comparative microbiome analysis of biofilm in contracted breast capsules: a cross-sectional study
Yang Jung Dug, School of Medicine, Kyungpook National University, Daegu, Korea, Republic of
Y. Jung Dug1, T. Park1, H. Cho1, Y. Chang1, M. Kim2, J. Shin2, J. Lee1, J. Lee3, H. Park3, T. Jung4; 1Department of Plastic and Reconstructive Surgery, School of Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 2Department of Applied Biosciences, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 3Department of Surgery, School of Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 4Department of Physical Medicine and Rehabilitation, School of Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF.
Despite the implication of bacterial biofilms in the etiology of capsular contracture, there is limited research on the microbial structure of the biofilm in patients with capsular contracture. Moreover, the potential role of the skin microbiome in capsular contracture remains undefined. Therefore, we conducted this study to characterize the microbiome of the breast capsular contracture by comparing it with the microbiome of biofilms without capsular contracture. We also investigated the associations between the microbial structures of the skin surrounding the breast and those of the breast capsule. We collected 25 capsules, including 14 samples from biofilms without capsular contracture and 11 samples from biofilms with capsular contracture. Beta diversity analysis demonstrated that the microbial structures in biofilms were distinctly different from those in the skin. The microbial composition of the biofilms was more influenced by individual variation than by the progression of the capsular contracture. Biofilms with capsular contracture showed a higher relative abundance of Staphylococcus, correlating positively with total bacterial load. Capsular contracture is associated with an escalation in total bacterial load and an increase in the abundance of opportunistic pathogens, such as Staphylococcus. This comparative analysis of biofilms in contracted breast capsules emphasizes the management of pathogens, considering bacterial load, in the occurrence of capsular contracture.
Presentation numberPS2-05-11
Early oncologic outcomes of robot-assisted versus conventional nipple-sparing mastectomy: a propensity score matching analysis
Seung Ah Lee, Samsung Medical Center, Gangnam-gu, Seoul, Korea, Republic of
S. Lee; Division of Breast Surgery, Samsung Medical Center, Gangnam-gu, Seoul, KOREA, REPUBLIC OF.
Background Robot-assisted nipple-sparing mastectomy (R-NSM) was developed to minimize visible scarring and improve the quality of life in breast cancer patients. However, despite its increasing adoption, data on its oncologic outcomes remain limited. This study aimed to compare the oncologic outcomes of R-NSM and conventional nipple-sparing mastectomy (C-NSM). Methods We retrospectively reviewed patients who underwent NSM with immediate breast reconstruction (IBR) for breast cancer between 2019 and 2022. Risk-reducing cases were excluded. R-NSM and C-NSM groups were compared after 1:5 propensity score matching using nearest-neighbor methods (caliper = 0.2). Results After 1:5 propensity score matching, most clinicopathologic variables were well balanced between groups. The median follow-up duration was 37.5 months for the R-NSM group and 42.6 months for the C-NSM group. No locoregional recurrence, distant metastasis, or death occurred in the R-NSM group. All oncologic events occurred in the C-NSM group, including 3 locoregional recurrences (1.5%), 3 distant metastases (1.5%), and 7 total recurrences (3.5%). Kaplan-Meier analysis showed no statistically significant differences in LRFS (p = 0.162), DMFS (p = 0.255), or DFS (p = 0.050), though all adverse events occurred in the C-NSM group. Event-based comparison revealed a significantly better DFS for R-NSM (p = 0.002). In multivariable Cox analysis, older age (HR = 0.14, p = 0.040) and positive nodal stage (N1: HR = 4.7; N3: HR = 31.9, p = 0.042) were associated with increased recurrence risk. R-NSM showed a lower recurrence hazard in univariable analysis (HR = 0.12, p = 0.177). Conclusion R-NSM showed oncologic outcomes comparable to those of conventional NSM, with no recurrence or mortality observed despite longer follow-up. These results support R-NSM as a safe and effective surgical option for appropriately selected breast cancer patients.
Presentation numberPS2-05-16
Prognostic value of TILs variation in breast cancer undergoing neoadjuvant chemotherapy
Alessandra Amatuzzi Fornazari, Hospital de Clínicas, Curitiba, Brazil
A. A. Fornazari1, I. Rabinovich1, A. Sebastião1, S. Boscoli2, I. C. Soares1, J. M. Jakobson3, E. M. Kac3, F. Cohene3, C. de Souza3, C. Urban4, C. Spautz4, K. F. Anselmi4, F. F. Kuroda4, M. T. Doria4, L. P. Nissen4, E. T. Schunemann4, R. S. Lima4; 1Universidade Federal do Paraná, Hospital de Clínicas, Curitiba, BRAZIL, 2Pathology, Hospital Universitário Evangélico Mackenzie, Curitiba, BRAZIL, 3Medicine, Universidade Federal do Paraná, Curitiba, BRAZIL, 4CDM, Centro de doenças da mama de Curitiba, Curitiba, BRAZIL.
Background/Objective: Pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) is associated with improved event-free and overall survival, particularly in triple-negative and HER2-positive breast cancer. Tumor-infiltrating lymphocytes (TILs) are promising prognostic and predictive biomarkers, correlating with higher pCR rates in these subtypes. However, the clinical relevance of TILs variation before and after NAC remains unclear. This study aimed to investigate associations between TILs variation, pCR, Residual Cancer Burden (RCB), and pre-NAC TILs with clinical outcomes. Methods: Retrospective cohort of 302 patients treated with NAC followed by surgery at two tertiary centers in Curitiba, Brazil, from 2016 to 2020. Data were extracted from electronic medical records. Pre-NAC core biopsies and post-NAC surgical specimens were reviewed by a dedicated breast pathologist. TILs were assessed per International TILs Working Group guidelines, and analyzed as categorical variables (low ≤10%, intermediate 11-49%, high ≥50%). Post-NAC TILs were quantified in stroma adjacent to the residual tumor bed. Pathologic response was defined using the RCB classification. Statistical analysis included the Wilcoxon signed-rank and Stuart-Maxwell tests. ROC curves identified outcome-related cut-offs. Results: Of 302 patients, 30% achieved pCR (Table 1). TILs were evaluated in 183 paired samples. TILs levels significantly declined post-NAC (p25% were associated with reduced distant recurrence risk. No significant association was found with locoregional recurrence or mortality (Table 2). Conclusion: TILs declined after NAC, particularly in pCR cases. Pre-NAC TILs >25% were linked to lower risk of distant recurrence, supporting their prognostic value in the neoadjuvant setting.
|
Variable |
n (%) |
|
Mean age ‡ SD (yr.) |
50,4 ‡ 12,7 (26-89) |
|
Pre menopausal |
150 (51,5%) |
|
Post menopausal |
141 (48,5%) |
|
Histological type |
|
|
Ductal |
275 (92%) |
|
Mixed ductal |
13 (4,3%) |
|
Lobular |
4 (1,3%) |
|
Others |
7 (2,3%) |
|
Molecular subtype |
|
|
Luminal B |
113 (39%) |
|
Triple negative |
65 (22,3%) |
|
Luminal B- Her |
49 (16,8%) |
|
Her-2 pure |
39 (13,4%) |
|
Luminal A |
26 (9%) |
|
T1 |
23 (8,3%) |
|
T2 |
164 (59,2%) |
|
T3 |
53 (19,1%) |
|
T4 |
37 (13,4%) |
|
N0 |
100 (36,1%) |
|
N1 |
145 (52,3%) |
|
N2 |
26 (9,4%) |
|
N3 |
6 (2,2%) |
|
Pathologic complete response (pCR) – RCB 0 |
90 (30%) |
|
Residual disease |
210 (70%) |
|
Outcome |
Median Follow-up (months) |
Median Time to Event (months) |
Pre-NAC TILs >25% |
Pre-NAC TILs ≤25% |
SHR (95% CI) |
p-value |
|
Locoregional recurrence |
53.9 |
25.0 |
6 (4.7%) |
3 (2.3%) |
1.99 (0.49–8.10) |
0.336 |
|
Distant recurrence |
54.8 |
25.4 |
8 (6.2%) |
18 (14.1%) |
4.43 (1.83–10.7) |
0.001 |
|
Death |
55.9 |
20.6 |
6 (4.7%) |
9 (7.0%) |
1.51 (0.54–4.25) |
0.433 |
Presentation numberPS2-05-17
Pathologic response in invasive lobular carcinoma versus invasive ductal carcinoma following neoadjuvant therapy: A single institution study
Tristan Jordan, Yale New Haven Hospital, New Haven, CT
T. Jordan, D. L. Rimm, H. Zhan; Pathology, Yale New Haven Hospital, New Haven, CT.
Objective: A subset of invasive lobular carcinoma (ILC) is managed with neoadjuvant therapy (NAT). The clinical and pathologic features of ILC following NAT are not well characterized. The aim of this study is to characterize the pathologic response in patients with ILC treated with NAT, including neoadjuvant endocrine therapy (NET), neoadjuvant chemotherapy (NCT), and Herceptin-targeted NCT (H-NCT). Methods: We conducted a retrospective cohort study of 65 patients with ILC and 290 patients with invasive ductal carcinoma (IDC) who received neoadjuvant therapy followed by resection between 2019 and 2024. Clinicopathologic parameters were recorded, including age, tumor grade, histologic type, staging, ER, PR, and HER2 status. Residual cancer burden (RCB) was evaluated on the post NAT resection specimens through an online calculator (www.mdanderson.org/breastcancer_RCB). Cases were categorized into three subtypes: ER+/HER2-, HER2+, and triple negative. Statistical analyses were performed to compare ILCs and IDCs with the same immunohistochemical (IHC) profiles. Results: Patients with ER+/HER2- and HER2+ ILCs were older at diagnosis compared to those with ER+/HER2- and HER2+ IDCs (p<0.01). Among patients with ILCs, 41 (63%) had ER+/HER2- tumor and received NET or NCT, 6 (9%) had triple negative tumor and received NCT only, and 18 (28%) had HER2+ tumor and received H-NCT. The pathologic complete response (pCR) rate was 44% in HER2+ ILCs and 60% in HER2+ IDCs. No pCR was achieved in triple negative and ER+/HER2- ILCs and IDCs. The majority of ER+/HER2- ILCs (87%) and all triple negative ILCs (100%) were classified as RCB II or RCB III, comparable to IDC cases with the same immunohistochemical subtypes (Table 1). Conclusion: ILCs demonstrated a similar response to neoadjuvant therapy as IDCs when matched by IHC subtype. These findings suggest that the effectiveness of neoadjuvant therapy is primarily determined by IHC-based tumor subtype rather than histologic subtype.
| Immunohistochemical subtype | ER+/HER2- ILC (n=41) | ER+/HER2- IDC (n=66) | p value | Immunohistochemical subtype | HER2+ ILC (n=18) | HER2+ IDC (n=168) | p value | Immunohistochemical subtype | TNBC ILC (n=6) | TNBC IDC (n=56) | p value |
| Age (median) | 64.8 | 49.3 | <0.001 | Age (median) | 63.3 | 54.1 | 0.002 | Age (median) | 64.5 | 58.3 | 0.33 |
| Histologic grade | 0.32 | Histologic grade | 1 | Histologic grade | 1 | ||||||
| Grade 1 | 6 (15%) | 5 (8%) | Grade 1 | 0 (0%) | 1 (1%) | Grade 1 | 0 (0%) | 0 (0%) | |||
| Grade 2 | 30 (73%) | 42 (64%) | Grade 2 | 14 (78%) | 64 (38%) | Grade 2 | 2 (33%) | 10 (18%) | |||
| Grade 3 | 5 (12%) | 19 (29%) | Grade 3 | 4 (22%) | 103 (61%) | Grade 3 | 4 (67%) | 46 (82%) | |||
| Classic | 33 (80%) | Classic | 8 (44%) | Classic | 1 (17%) | ||||||
| Pleomorphic | 8 (20%) | Pleomorphic | 10 (56%) | Pleomorphic | 5 (83%) | ||||||
| RCB categories | 1 | RCB categories | 0.314 | RCB categories | 1 | ||||||
| RCB 0 | 0 (0%) | 1 (2%) | RCB 0 | 8 (44%) | 100 (60%) | RCB 0 | 0 (0%) | 0 (0%) | |||
| RCB 1 | 5 (12%) | 6 (9%) | RCB 1 | 1 (6%) | 21 (13%) | RCB 1 | 0 (0%) | 18 (32%) | |||
| RCB 2 | 17 (41%) | 38 (58%) | RCB 2 | 7 (39%) | 38 (23%) | RCB 2 | 3 (50%) | 29 (52%) | |||
| RCB 3 | 19 (46%) | 21 (32%) | RCB 3 | 2 (11%) | 9 (5%) | RCB 3 | 3 (50%) | 9 (16%) |
Presentation numberPS2-05-18
Distinct Biological Profiles of Mucinous Carcinoma with a Solid Papillary Carcinoma Component Compared to Other Histological Types
Yuki Hara, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
Y. Hara1, R. Yamaguchi2, M. Matsumoto1, M. Oshi3, S. Urakawa1, A. Tanaka1, M. Akashi4, S. Kuba4, R. Otsubo1, S. Eguchi4, K. Matsumoto1; 1Department of Surgery, Division of Surgical Oncology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JAPAN, 2Department of Diagnostic Pathology, Nagasaki University Hospital, Nagasaki, JAPAN, 3Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 4Department of Surgery, Division of Digestive Surgery and Transplantology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, JAPAN.
[Introduction] Mucinous carcinoma (MC) of the breast is histologically defined by the World Health Organization as either pure (comprising >90% mucinous component) or mixed (comprising 10-90% mucinous component) subtypes. Typically, the tumor nests within MC display conventional cluster morphology or invasive micropapillary carcinoma (IMPC) architecture. Intriguingly, some MCs contain hypercellular solid nests with neuroendocrine features that closely resemble solid papillary carcinoma (SPC), a distinct entity characterized by mucin production and fibrovascular cores. Given the overlapping morphological and secretory features between SPC and some forms of MCs, we hypothesized that a subset of MCs may arise from or share a lineage with SPC. To explore this, we stratified MCs based on the presence or absence of SPC-like histological features and conducted a comprehensive molecular characterization using data from The Cancer Genome Atlas Breast Invasive Carcinoma Collection (TCGA-BRCA). This study aims to elucidate the biological distinctions between SPC-related and non-SPC MC subtypes, potentially refining our understanding of MC heterogeneity and its clinical implications. [Material & Method] We systematically re-evaluated all available hematoxylin and eosin (H&E) digital slides from the TCGA-BRCA cohort (n=1084). Tumors exhibiting >10% mucinous component were selected and subclassified into two groups based on histopathological features: those with a SPC component (MC-SPC) and those without a SPC component, including IMPC patterns (MC-OTHER). Clinicopathological parameters were collected and compared between the groups using the chi-square or Fisher’s exact test, as appropriate. To explore transcriptional differences, differentially expressed genes (DEGs) were identified using a false discovery rate-adjusted p-value 1. DEGs were visualized using principal component analysis (PCA), volcano plots, and heatmaps. Functional enrichment analyses were performed using the clusterProfiler R package, referencing the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. R (version 4.4.3) was used for all statistical analyses. [Result] We identified 29 mucinous carcinoma cases from the TCGA-BRCA cohort with > 10% mucinous component, including 18 with MC-SPC and 11 with MC-OTHER. The median age was 67.5 years (range, 41-85) in the MC-SPC group and 55 years (range, 38-79) in the MC-OTHER group. There was no significant difference in age between the two groups (p = 0.16). The HER2-positive subtype was significantly more frequent in MC-OTHER compared to MC-SPC (p = 0.014). Transcriptomic analysis revealed distinct molecular profiles between the two groups, as demonstrated by volcano plots and a heatmap of the top 30 differentially expressed genes, although PCA did not show clear separation. Functional enrichment analysis showed that MC-OTHER tumors exhibited upregulation of pathways related to extracellular matrix (ECM) organization and cell adhesion, suggesting a more mesenchymal-like phenotype. In contrast, MC-SPC tumors were enriched for pathways associated with synaptic signaling and neurotransmitter transport, reflecting potential neuroendocrine-associated biology. [Conclusion] This study identifies two transcriptionally distinct subtypes of mucinous carcinoma, MC-SPC and MC-OTHER, based on histopathological features. These subtypes exhibit divergent biological characteristics, including mesenchymal-like features in MC-OTHER and neuroendocrine-associated pathways in MC-SPC. This subclassification may have clinical relevance and warrants further investigation in larger cohorts.
Presentation numberPS2-05-19
Immuno- and Molecular-Profiling of Breast Cancer Metastasis in the CNS
Niklas Abele, University Erlangen, Erlangen, Germany
N. Abele1, M. Berner1, R. Stöhr1, H. Arndt1, C. Mawrin2, R. Erber3; 1Institute of Pathology, University Erlangen, Erlangen, GERMANY, 2Institute of Neuropathology, University Magdeburg, Magdeburg, GERMANY, 3Institute of Pathology, University Regensburg, Regensburg, GERMANY.
Introduction: Breast cancer brain metastasis (BCBM) represents a significant clinical challenge with poor prognosis. Certain breast cancer subtypes, notably HER2-positive and triple-negative breast cancer (TNBC), are associated with a higher risk of brain metastasis. Understanding these unique properties, distinct from primary tumors, is crucial for therapeutic strategies. In this study, we aimed to characterize key biomarkers in BCBM to enhance understanding of their distinct molecular and immunological profiles. Methods: We retrospectively evaluated 36 intracranial BCBM from 36 patients for hormone receptor (ER/PR), HER2-status (including HER2-low and -ultra-low), proliferation (Ki-67), PD-L1 expression, tumor-infiltrating lymphocytes (TILs) as well as PIK3CA mutations. Biomarker frequencies were compared against published data from primary breast cancer cohorts (TCGA, METABRIC, SEER, KEYNOTE-355). Results: The BCBM cohort exhibited a significantly skewed subtype distribution compared to primary breast cancer. While primary breast cancer is predominantly HR+/HER2- (Luminal A/B HER2-) (approx. 74.4%), this subtype comprised only 16.7% (6/36) of our BCBM cases. Conversely, TNBC (44.4%, 16/36) and HER2-positive subtypes (38.9%, 14/36) were notably overrepresented in our cohort compared to their prevalence in primary breast cancer (approx. 11.4% and 14.2%, respectively). This significant difference suggests a differential propensity for brain metastasis among subtypes and is in line with previous findings. The overall mean Ki-67 was 42.6% and most cases (88.9%, 32/36) showed high proliferation (Ki-67 ≥ 20%), with 75.0% (27/36) demonstrating very high proliferation (Ki-67 ≥ 30%). Subtype-specific Ki-67 levels generally reflected the expected proliferative activity of corresponding primary breast cancer subtypes. PD-L1 expression (22C3, CPS ≥ 10%) was very low, found in only 2.8% (1/36) of cases. A slightly larger subset (8.3%, 3/36) met the IC positivity criterion (IC ≥ 1%) with SP142. This low prevalence of actionable PD-L1 expression in BCBM contrasts sharply with the higher rates observed in primary TNBC (e.g., 38% for CPS ≥ 10% in KEYNOTE-355). Furthermore, TILs were significantly lower compared to primary breast cancer and across all subtypes; only 5.9% cases had high TILs (>=60%) compared to the 19% for primary found in literature. This suggests a very different tumor microenvironment for metastasis to the CNS, with implications for immunotherapy efficacy. PIK3CA analysis revealed 20% (6/30) of cases with mutations and no significant deviation from the expected frequencies in respect of the molecular subtype. Conclusion: Our findings indicate that BCBMs exhibit a distinct molecular landscape characterized by an enrichment of TNBC and HER2-positive subtypes with high proliferation (Ki-67). PD-L1 expression and TILs appears to be markedly lower than in primary breast cancer, especially in TNBC. These insights highlight the unique biology of BCBM, emphasizing the need for biomarker analysis on metastatic tissue to guide personalized therapeutic strategies.
Presentation numberPS2-05-20
Evaluation of novel diagnostic kits for detecting metastatic lymph nodes in breast cancer patients treated with neoadjuvant chemotherapy using the semi-dry dot-blot method: A single-center prospective study
Ryota Otsubo, Nagasaki University Hospital, Nagasaki, Japan
R. Otsubo1, Y. Hara1, S. Urakawa1, A. Tanaka1, M. Akashi1, S. Kuba1, M. Matsumoto1, R. Yamaguchi2, K. Matsumoto1; 1Surgery, Breast and Endocrine Surgery, Nagasaki University Hospital, Nagasaki, JAPAN, 2Pathology, Nagasaki University Hospital, Nagasaki, JAPAN.
Background:The semi-dry dot-blot (SDB) method is a diagnostic technique used to detect lymph node (LN) metastases by identifying cytokeratin (CK) proteins in the lavage fluid of sectioned LNs. This method is based on the principle that CK, an epithelial marker, is absent in normal lymphatic tissue. We prospectively evaluated novel SDB kits incorporating a newly developed anti-CK19 antibody and an automatic reader for diagnosing LN metastases in breast cancer patients. In a previous analysis of 1734 sentinel LNs (SLNs) from patients not receiving neoadjuvant chemotherapy (NAC), the kit demonstrated a sensitivity of 93.0%, specificity of 97.1%, and overall agreement of 96.7% for detecting macrometastases (>2.0 mm). However, the impact of NAC—especially in triple-negative and HER2-positive subtypes—on the diagnostic performance of the SDB method remains unclear, given that chemotherapy can cause cancer cell degeneration. This study aimed to evaluate whether the SDB kit could accurately detect macrometastases in patients undergoing NAC. Methods:A total of 127 LNs were collected from 74 breast cancer patients who underwent NAC followed by surgery between May 2022 and December 2024 at Nagasaki University Hospital. Both patients with and without axillary LN metastases were eligible. Eligible LNs were defined as all SLNs in patients who underwent SLN biopsy and two axillary LNs in patients who underwent axillary LN dissection. Each LN was sliced at 2-mm intervals and rinsed with phosphate-buffered saline. The resulting lavage fluid was centrifuged, and the cells were lysed to extract protein, which was then analyzed using the SDB kit and automatic absorbance reader. Pathologists, blinded to the SDB results, independently diagnosed LN metastasis using hematoxylin and eosin (H&E) staining. SDB-based diagnoses were compared with permanent histopathological diagnoses. Results:Among the 127 LNs, 56 were obtained via SLN biopsy and 71 via axillary LN dissection. Histological evaluation identified 16 macrometastases and 111 non-macrometastases (including 4 micrometastases and 1 case with isolated tumor cells). Using a CK19 absorbance cutoff of 11.9 milli-absorbance units (mAbs) for detecting macrometastases, the SDB kit correctly identified 15 of the 16 macrometastases and all 111 non-macrometastases. The resulting sensitivity, specificity, and overall agreement were 93.8%, 100%, and 99.2%, respectively. The single false-negative case involved a sparsely distributed residual tumor measuring 2.1 mm. The diagnostic process required approximately 20 minutes and cost less than USD 30 per kit. Conclusions:The novel SDB kits combined with an automatic reader provided rapid, accurate, and cost-effective detection of LN macrometastases without tissue loss, even after NAC. These findings support the potential utility of this method in clinical decision-making. A prospective multi-center study is planned to further validate clinical performance.
Presentation numberPS2-05-21
Altered expression of catenins and cadherins in lobular breast cancer: A focus on beta-catenin and E-cadherin
Taesung Jeon, Korea University Guro Hospital, Seoul, Korea, Republic of
T. Jeon1, Y. Sung2, J. Lee2, J. Oh1, J. Ahn3, A. Kim1; 1Department of Pathology, Korea University Guro Hospital, Seoul, KOREA, REPUBLIC OF, 2Department of Pathology, Korea University Anam Hospital, Seoul, KOREA, REPUBLIC OF, 3Department of Pathology, Ewha Womans University Mokdong Hospital, Seoul, KOREA, REPUBLIC OF.
Background: Dysfunction of the E-cadherin/catenin complex is a hallmark of invasive lobular carcinoma (ILC) of the breast, which is associated with poorer long-term outcomes compared to invasive ductal carcinoma (IDC). However, accurate histologic diagnosis of ILC remains challenging due to complex morphologic and immunohistochemical (IHC) patterns. This study aimed to compare the expression patterns of E-cadherin, beta-catenin, and P120 catenin in breast carcinomas, focusing on aberrant patterns and their clinicopathologic significance. Methods: We retrospectively analyzed 264 breast carcinoma cases with suspected lobular morphology. Tissue microarray (TMA) blocks were constructed from surgical specimens. IHC for E-cadherin, beta-catenin, P120 catenin, N-cadherin, and P-cadherin was performed on TMA sections, and staining patterns were classified as membranous, aberrant, or loss. Results: Morphologically, 43 cases (16.3%) showed IDC features and 221 (83.7%) showed ILC features. After integrating IHC results, 18 cases (6.8%) were classified as IDC and 246 (93.2%) as ILC based on aberrant or loss of marker expression. The distribution of staining patterns was similar among E-cadherin, beta-catenin, and P120 catenin, with a high concordance (95.1%) between E-cadherin and beta-catenin. Beta-catenin demonstrated a distinct Golgi pattern in aberrant cases, enhancing interpretability and sensitivity compared to E-cadherin and P120 catenin. Interpretation of P120 staining was sometimes difficult due to the small size of lobular carcinoma cells and the presence of intracytoplasmic vacuoles. Twelve cases (4.5%) showed discordant results between E-cadherin and beta-catenin, with beta-catenin being more sensitive in detecting aberrant/loss patterns. Aberrant beta-catenin expression was significantly associated with higher N and M stage, higher histologic grade, lymphovascular invasion, lower estrogen receptor (ER) expression, higher HER2 expression, and increased N-cadherin and P-cadherin expression. Conclusions: Beta-catenin IHC is more sensitive and interpretable than E-cadherin or P120 catenin for distinguishing ILC from IDC, due to its unique localization patterns. Aberrant expression of adhesion complex molecules, especially beta-catenin, is associated with adverse clinicopathologic features and may reflect atypical features among breast carcinomas. These findings support incorporating beta-catenin IHC into the diagnostic workup of breast carcinomas with ambiguous morphology.
Presentation numberPS2-05-22
Assessment of the tumor bed area in patients with breast cancer and RCB class I after neoadjuvant therapy: comparison of original and modified method.
Nikolay Amirov, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, Russian Federation
N. Amirov1, N. Mikhaskova2, P. Krivorotko1, A. Kudaybergenova2; 1Breast Tumor Department, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, RUSSIAN FEDERATION, 2Pathology Department, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, RUSSIAN FEDERATION.
Background: One of the important prognostic and predictive markers in the histological assessment of breast cancer (BC) after neoadjuvant therapy (NAT) is the Residual Cancer Burden (RCB) index. In the original formula developed by MD Anderson Cancer Center the linear dimensions of the tumor bed are taken into account, which makes it impossible to assess fragmented material, for example, when performing vacuum-assisted biopsy. Considering this limitation, an attempt was made to modernize the method for evaluating tumor response to NAT by recalculating the RCB index using the area of the tumor bed based on digital images (scans) of histological slides obtained from sectoral resections in patients with early-stage BC. Methods: A single-center retrospective study included 20 patients with ypT1a-cN0 stage breast carcinoma who had undergone neoadjuvant systemic therapy (FAC-T — 3 patients, AC-T — 8, AC — 2, DHP — 1, T — 2, EC+D+TRAST — 1, TCARB — 1, TCHP — 2) and were classified as RCB class 1. Digital scans of histological slides were obtained using the 3D HISTECH PANORAMIC 1000 scanner. Measurements of the tumor bed were carried out using two methods: traditional MD Anderson technique, which defines the tumor bed area with the assessment of the two largest dimensions of the tumor bed, and a modified in-house technique, which implies manual delineation of the entire area of tumor bed on the histological scans using a stylus within the 3D HISTECH SlideViewer 2.5.0 software. The measured areas were then used to recalculate the RCB index according to the original formula, which includes extracting the square root of the area. Results: The mean tumor bed area calculated using the MD Anderson method was 76.67 mm² (range: 14.2-258.7), whereas for the modified method, the mean was 153.65 mm² (range: 35.6-535). The mean RCB index for the standard method was 1.034 (range: 0.804-1.316), and for the modified method — 1.102 (range: 0.829-1.461). The obtained data was normally distributed in both groups. A high correlation was found between the two methods, indicating a high consistency between the methods (Pearson’s Correlation r = 0.96, p<0.001; Spearman’s Correlation ρ = 0.95, p<0.001). The mean difference in RCB index between two methods was 0.068 (95% CI: +0.040 to +0.096). According to the t-test, the difference between the two methods was statistically significant (p < 0.01). The modified method demonstrated systematically higher values of RCB index. Moreover, when applying the modified method, 2 out of 20 patients (10%) were reclassified from RCB class 1 to RCB class 2. Conclusion: The proposed modified method of tumor bed assessment correlates highly with the classical approach in calculating the RCB index. Larger studies are needed to determine the clinical significance of a persistent slight overestimation of the RCB index using the modified method.
Presentation numberPS2-05-23
Immune Heterogeneity in Breast Cancer Natural History: Benign Breast Disease to Breast Cancer
Amy Zhang, UNC Medical School, Chapel Hill, NC
A. Zhang1, A. Cozzo2, L. Olsson2, E. Kirk2, X. Gao2, S. Nyante3, B. Calhoun1, R. Vierkant4, M. Sherman4, M. Troester2; 1Department of Pathology, UNC Medical School, Chapel Hill, NC, 2Department of Epidemiology, UNC Medical School, Chapel Hill, NC, 3Department of Radiology, UNC Medical School, Chapel Hill, NC, 4Department of Biostatistcs, Mayo Clinic, Rochester, MN.
Benign breast disease (BBD) confers elevated risk for breast cancer, yet current tools lack precision in identifying lesions likely to progress. Immune mechanisms may influence progression, but the spatial and molecular features of immune responses in BBD versus invasive cancer remain poorly characterized. We applied Digital Spatial Proteomics (DSP) to evaluate immune-related protein expression across 1,540 regions of interest (ROIs) from breast tumors (n=58, Carolina Breast Cancer Study [CBCS]), a UNC BBD cohort (n=20), and a Mayo BBD cohort (n=177). Using Consensus Clustering in CBCS and UNC BBD, we identified two stable immune phenotypes: “High Immune,” enriched in tumors and high-risk benign lesions, and “Quiet Immune,” predominant in low-risk BBD. ROIs were classified by immune marker expression, and patients were labeled as “High” if ≥50% of their ROIs were High Immune. In CBCS, High Immune profiles were associated with ER-negative status (OR=2.92 [0.99-8.65] participant-level; OR=3.28 [1.34-8.04] ROI-level). In BBD, High Immune profiles trended toward higher odds of high-risk lesions (OR=1.00 [0.15-6.70] participant-level; OR=1.68 [0.57-4.97] ROI-level), though this pattern was not replicated in the Mayo cohort. BBD samples exhibited substantial spatial heterogeneity in immune expression (40% within-patient ROI agreement vs. 74% in tumors), potentially limiting biomarker reliability. Nonetheless, four immune proteins (CD44, CD80, CD14, CD66b) were significantly associated with future cancer development in the Mayo cohort, though these findings require validation due to within-person variability. Our findings highlight the challenges posed by spatial heterogeneity in assessing immune biomarkers in BBD and suggest that immune activation may emerge later in disease progression. These results raise important questions about the timing and consistency of immune responses in the natural history of breast cancer.
Presentation numberPS2-05-24
Histopathological Grade as a Prognostic Marker in de novo Metastatic HER2-Positive Breast Cancer Treated with First-Line Docetaxel, Trastuzumab, and Pertuzumab
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
M. Kubeczko1, M. Mianowska-Malec1, A. Polakiewicz-Gilowska1, K. Swiderska1, A. Lesniak1, B. Lanoszka1, B. Grandys1, K. Chomik1, N. Lisovska1, B. Pycinski2, E. Stobiecka3, J. Simek3, E. Chmielik3, A. Prat4, M. Jarzab1; 1Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 2Faculty of Biomedical Engineering, Silesian University of Technology, Gliwice, POLAND, 3Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 4Cancer Institute and Blood Disorders, Hospital Clínic; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute; Reveal Genomics; SOLTI Cancer Research Group; University of Barcelona, Barcelona, SPAIN.
Background: The TPH regimen (docetaxel, trastuzumab, pertuzumab) has long been the standard first-line treatment for HER2-positive metastatic breast cancer (MBC). However, recent findings from the DESTINY-Breast09 trial have challenged this paradigm, underscoring the need for predictive biomarkers to personalize treatment selection. Estrogen receptor positivity (ER+) may inform the use of CDK4/6 inhibitors, such as palbociclib, during the maintenance phase. While molecular classifiers—such as ERBB2 expression within the HER2Dx signature—have shown promise in identifying patients likely to benefit from prolonged TPH treatment, the prognostic potential of routinely available histopathological parameters remains insufficiently explored. This study aimed to evaluate whether histological grade, Ki67 proliferation index, and ER status predict progression-free survival (PFS) in patients receiving first-line TPH. Methods: We conducted a retrospective analysis of 134 female patients with de novo HER2-positive MBC, treated with first-line TPH between 2017 and 2024, all of whom had available breast tumor biopsy samples for histopathological evaluation. The median age was 60.2 years (range, 27.1-84.3 years). ECOG performance status was 0 in half of the patients, while the other half had a status of 1 or 2. Oligometastatic disease (OMD, ≤5 metastatic lesions) was present in 46.3%, liver metastases in 32.8%, and lung metastases in 24.6%. Histological grade was G1 in 2.2%, G2 in 44.8%, and G3 in 53.0%; G1 and G2 were grouped as G1-2. ER positivity was observed in 55.2% of cases, and the median Ki67 index was 32% (IQR: 20-60%). All evaluations were based on the initial breast tumor biopsy. Prognostic factors were assessed using Cox proportional hazards models; variables with p < 0.2 in univariate analysis were included in the multivariate model. Statistical significance was defined as p < 0.05. Results: The median PFS in the entire cohort was 38.3 months. Histological grade correlated strongly with outcome: patients with G1-2 tumors had a median PFS of 79.1 months versus 25.2 months for those with G3 tumors. The 2-year PFS rate was 71.2% (95% CI: 57.7-81.1%) for G1-2 and 52.1% (95% CI: 39.7-63.2%) for G3. In univariate analysis, G3 grade (HR 1.93; 95% CI: 1.05-3.53; p = 0.034) and OMD (HR 0.49; 95% CI: 0.29-0.81; p = 0.006) were significantly associated with PFS. ER-positive status (HR 0.65; 95% CI: 0.38-1.11; p = 0.115) and Ki-67 index (HR 1.01; 95% CI: 0.996-1.021; p = 0.178) met the threshold for inclusion in multivariate analysis. In the multivariate model, both OMD and G3 grade remained independently associated with PFS. High histological grade was confirmed as a significant negative prognostic factor (HR 2.40; 95% CI: 1.08-5.28; p = 0.030). Conclusions: Histological grade emerged as an independent prognostic factor, whereas ER status and Ki67 did not reach statistical significance. This finding highlights the prognostic relevance of traditional histopathological features in de novo HER2-positive MBC treated with TPH. High-grade tumors were associated with significantly shorter PFS, suggesting the need for intensified or alternative therapeutic strategies in this subgroup. While histopathology offers valuable prognostic insights, molecular profiling holds the potential to deliver a deeper biological understanding and enables more precise treatment stratification in this heterogeneous disease.
Presentation numberPS2-05-25
Slide-free histology for breast cancer core-needle-biopsy diagnostics at point-of care in Ghana
Richard Levenson, UC Davis Health, Sacramento, CA
R. Levenson1, E. Seibel2, B. Wiafe-Addai3, F. Fereidouni4; 1Dept. of Pathology and Laboratory Medicine, UC Davis Health, Sacramento, CA, 2Mechanical Engineering, University of Washington, Seattle, WA, 3Surgery, Peace and Love Hospital, Kumasi, GHANA, 4Pathology and Laboratory Medicine, Emory University, Atlanta, GA.
Fluorescence imitating brightfield imaging (FIBI) and related modes provide near-instantaneous slide-free histology. Benefits include greatly accelerated patient-lesion assessment in major clinical center, but also accessible diagnostics for lower- and middle-income countries. In addition, the technology can be an improvement over standard histology, as it achieves visualization of novel tissue features not readily assessed on standard slides. The approach used is termed FIBI (fluorescence imitating brightfield imaging), which features inexpensive and robust hardware, automated specimen scanning and forthcoming computer-aided diagnosis. This innovative, non-destructive technique employs affordable optics and sensors to image intact tissues within seconds to minutes. A previously published preliminary validation study demonstrated approximately 97% clinical concordance across diverse tumor types. Complementing FIBI, we are developing a sample management platform to stain and image core-needle biopsy specimens with minimal physical manipulation, preserving tissue integrity.While this set of features will be helpful in well-resourced medical centers, they are also well-suited to responding to patient care needs in lower resource settings. One example: lower- and middle-income countries (LMICs) face an urgent need for point-of-care breast cancer diagnosis and treatment planning. With no mammography screening available, many patients present with late-stage tumors that are typically evaluated using core-needle biopsies. However, it is not possible to obtain complete time-of-procedure diagnostic feed-back, as formalin-fixation, embedding, sectioning, and pathologist examination are typically involved, and the facilities required are scarce and unevenly distributed. Even when presenting with large lesions, patients often endure weeks to months of waiting before receiving a diagnosis and commencing appropriate treatment. FIBI can be implemented with automated specimen scanning and, soon, computer-aided diagnosis. A prototype system for needle biopsy staining and automated specimen scanning using FIBI slide-free surface imaging has been developed at UC Davis in conjunction with Emory University, and a version of this designed at the University of Washington has been deployed to the Peace & Love Hospital in Kumasi, Ghana. Images are captured within a few minutes after core-needle biopsy acquisition and have similar appearance to standard 20X H&E-stained slides. However, while the main histological features in FIBI derive from optical absorbance of superficial hematoxylin and eosin stains, there is information also being detected arising from the simultaneous fluorescence of tissue features such as elastin and collagen. This gives rise to images with a color gamut that goes beyond the pinks and blues usually visible in standard H&E-stained slides. In addition, because FIBI images thick specimens, some extended structures, such as blood vessels, are captured with much improved structural fidelity.
Presentation numberPS2-05-26
Analytical performance of an image analysis algorithm developed as an adjunctive aid for determining HER2 gene status from in situ hybridization
Keith A Wharton Jr, Roche Diagnostics Ltd, Boston, MA
N. Thampy1, K. Young-Zvandasara1, C. M. Bacon2, C. Chen3, N. Harris1, M. Howe1, A. Long1, L. Mansfield1, J. Ness1, K. A. Wharton Jr4, S. Dance5, L. J. Inge3; 1Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UNITED KINGDOM, 2Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UNITED KINGDOM, 3Ventana Medical Systems, Roche Diagnostics Ltd, Tuscon, AZ, 4Oncology, Genetics and Pathology, Roche Diagnostics Ltd, Boston, MA, 5Medical Science Liaison Oncology, Roche Diagnostics Ltd, West Sussex, UNITED KINGDOM.
Background: Accurate detection of HER2 gene amplification via in situ hybridization (ISH) is critical for guiding targeted therapy use in patients with breast cancer. Traditionally, this involves manual quantification of the HER2-to-chromosome 17 (HER2/CHR17) signal ratio; however, this process is often time-consuming and may be complicated by factors such as chromosome 17 polysomy or intratumoral heterogeneity. The uPath HER2 Dual ISH image analysis algorithm is a tool for research use only, developed to assist medical laboratory scientists with determining HER2 gene status from images of formalin-fixed, paraffin-embedded (FFPE) neoplastic breast tissue. The algorithm was previously validated with samples stained using the VENTANA BenchMark ULTRA platform, but its performance across alternative staining platforms has not yet been evaluated. Methods: This study was designed to evaluate agreement between the BenchMark ULTRA and XT staining platforms, and between manual and algorithm-assisted methods for scoring across both platforms. In total, 120 advanced breast cancer FFPE samples with varying levels of HER2 amplification were selected for analysis (amplified [HER2/CHR17 ratio > 2.6], n = 50; non-amplified [HER2/CHR17 ratio < 1.4], n = 50; borderline amplified [HER2/CHR17 ratio ≥ 2.0 and ≤ 2.5], n = 10; borderline non-amplified [HER2/CHR17 ratio ≥ 1.5 and ≤ 1.9], n = 10). Tissue sections were prepared and stained using the VENTANA HER2 Dual ISH DNA probe cocktail using both the BenchMark ULTRA and XT platforms. Two trained medical laboratory scientists performed blinded manual quantification of HER2 ISH across all samples. These results were then checked by pathologists (n = 3). Following a washout period, these pathologists repeated the analysis using the algorithm. Results: High inter-platform agreement was observed with the algorithm across 118 evaluable samples, with an overall percent agreement (OPA) of 94.9% (95% confidence interval [CI], 89.3-97.6); positive percent agreement (PPA) and negative percent agreement (NPA) were 95.1% (95% CI, 86.5-98.3) and 94.7% (95% CI, 85.6-98.2), respectively. High agreement was observed between the algorithm and manual reads irrespective of the platform used for staining (Table 1). Perfect agreement was observed between manual reads across the two platforms. Conclusions: These data demonstrate that the uPath HER2 Dual ISH image analysis algorithm offers similar performance to manual scoring across samples stained using either the BenchMark ULTRA or XT platforms, and across samples with varying levels of HER2 amplification. With further validation, the algorithm may provide pathologists with a reliable adjunctive tool to support the diagnosis of patients with breast cancer.
| PPA n/N (%) | PPA 95% CI | NPA n/N (%) | NPA 95% CI | OPA n/N (%) | OPA 95% CI | |
| Manual ULTRA vs algorithm XT | 57/59 (96.6) | 88.5–99.1 | 55/59 (93.2) | 83.8–97.3 | 112/118 (94.9) | 89.3–97.6 |
| Manual ULTRA vs manual XT | 59/59 (100.0) | 93.9–100.0 | 59/59 (100.0) | 93.9–100.0 | 118/118 (100.0) | 96.8–100.0 |
| Manual XT vs algorithm ULTRA | 58/59 (98.3) | 91.0–99.7 | 56/59 (94.9) | 86.1–98.3 | 114/118 (96.6) | 91.6–98.7 |
| Manual XT vs algorithm XT | 57/59 (96.6) | 88.5–99.1 | 55/59 (93.2) | 83.8–97.3 | 112/118 (94.9) | 89.3–97.6 |
Presentation numberPS2-02-01
Improved Antiemetic Outcomes with NEPA vs (Fos)aprepitant in Patients with Breast Cancer Receiving Antibody-Drug Conjugates: A 12-Month Real-World Claims Analysis
Lee S. Schwartzberg, Renown Health, Reno, NV
L. Schwartzberg1, T. Gillespie2, E. Roeland3, T. Tyler4, R. Agarwal5, A. Alonzi6, T. Fralich7, P. Mudumby8, G. Aweh8, M. Fisher8, H. Rugo9; 1William N. Pennington Cancer Institute, Renown Health, Reno, NV, 2Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, 3Division of Hematology and Medical Oncology, Oregon Health and Sciences University, Portland, OR, 4Desert Regional Medical Center, Comprehensive Cancer Center, Palm Springs, CA, 5Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, TN, 6Medical Affairs, Helsinn Healthcare, Lugano, SWITZERLAND, 7Medical Affairs, Helsinn Therapeutics, Inc., Iselin, NJ, 8Real-World Evidence, STATinMED, Dallas, TX, 9Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA.
Background: Antibody-drug conjugates (ADCs) are increasingly used to treat breast cancer (BC) and are often administered over prolonged periods. While nausea and vomiting (NV), including long-delayed nausea, are well-established side effects, limited real-world evidence exists evaluating antiemetic outcomes over extended treatment durations. This analysis evaluated 12-month real-world clinical and economic outcomes in U.S. patients with BC receiving ADCs, using national claims data. Outcomes were assessed for NCCN-guideline recommended antiemetic agents: NEPA (netupitant/palonosetron), distinguished by its prolonged NK1 receptor occupancy and extended half-life, and fos(aprepitant). Methods: A retrospective claims analysis using the STATinMED RWD Insights all-payer medical and pharmacy claims database evaluated patients with BC treated between January 1, 2019, and December 31, 2023. The index date was defined as the earliest claim for either NEPA or (fos)aprepitant. Eligible patients were ≥18 years with a BC diagnosis who received one of three ADCs—trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd), or sacituzumab govitecan (SG)—and maintained continuous medical and pharmacy benefits for 12 months before and after the index date. To minimize confounding, 1:1 propensity score matching was conducted to create balanced cohorts for NEPA vs fos(aprepitant) based on demographics and clinical variables including sex, geographic region, payer type, and comorbidity burden. Clinical outcomes were compared for NEPA vs (fos)aprepitant and included claims for nausea or vomiting or related complications (i.e., dehydration, IV fluid use, and/or electrolyte imbalance) occurring at least once during the 12-month evaluation period. Nausea occurring >72 h after the index date was also assessed. Outpatient hospital services (i.e., hospital-based care not involving admission or emergency treatment) and outpatient and emergency room costs were also determined for the two cohorts. NV-related utilization and costs were identified based on the presence of a NV diagnosis code in any position on the medical claim. Statistical analyses included descriptive statistics and hypothesis testing to evaluate differences between cohorts. Results: In the matched cohort analysis of 209 patients per group (n = 192 T-DM1, n = 154 T-DXd, and n = 72 SG of total population), NEPA had improved clinical and economic outcomes compared with (fos)aprepitant. During the 12-month post-index period, significantly fewer NEPA patients (82.3%) experienced nausea or vomiting or related clinical complications than (fos)aprepitant patients (90.4%; p=0.0154). Nausea rates >72 h were significantly lower for NEPA (27.8%) compared to (fos)aprepitant (40.2%; p=0.0073). Outpatient hospital services related to NV were also significantly lower with NEPA (43.0%) compared to (fos)aprepitant (69.4%; p<0.0001). The mean NV-related outpatient and emergency room cost for NEPA was $14,884, less than half the cost for (fos)aprepitant ($30,665; p<0.0001). While claims data offer valuable insights into real-world treatment patterns and outcomes, they have inherent limitations including potential coding inaccuracies, lack of clinical detail, and incomplete capture of patient-reported outcomes or uninsured care. Conclusion: These findings highlight clinical and economic benefits of NEPA-based antiemetic prophylaxis in patients with BC receiving ADCs. Given that patients often undergo prolonged ADC treatment, selecting an antiemetic regimen with sustained efficacy─such as NEPA in this database analysis─may be particularly important to minimize complications, lower healthcare utilization and cost, preserve quality of life, and support treatment continuity.
Presentation numberPS2-02-02
Safety and efficacy of trastuzumab deruxtecan and concomitant radiation therapy in breast cancer patients: an international retrospective cohort study
Luca Visani, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
L. Visani1, I. Ratosa2, R. Bartsch3, B. Linderholm4, G. Griguolo5, C. Becherini1, M. Valzano1, V. Salvestrini1, A. Starzer3, P. Kus6, M. Lambertini7, V. Guarneri5, L. Livi8, I. Meattini8; 1Radiation Oncology & Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Firenze, ITALY, 2Medical faculty and Division of Radiotherapy, University of Ljubljana and Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 3Clinical Research Unit, Division of Oncology, Department of Medicine I, Medical University of Vienna, Wien, AUSTRIA, 4Medical Oncology Unit, Department of Oncology, Sahlgrenska Academy and University Hospital, Göteborg, SWEDEN, 5Department of Surgery, Oncology and Gastroenterology, Division of Oncology 2, University of Padova and Istituto Oncologico Veneto IRCCS, Padova, ITALY, 6Department of Oncology, University Medical Centre Maribor, Maribor, SLOVENIA, 7Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, ITALY, 8Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, Firenze, ITALY.
Background. Trastuzumab deruxtecan (T-DXd) has emerged as the standard treatment for patients with metastatic HER2-positive (HER2+) breast cancer (BC) following disease progression on first-line therapy containing taxanes and trastuzumab. Results from DESTINY-Breast04 have extended the approval of T-DXd to include metastatic BC patients with HER2-low expressing tumours. Radiation therapy (RT) is an integral component of multimodal treatment in the metastatic setting. This retrospective study aims to evaluate the safety of the concurrent use of T-DXd and RT in a consecutive, multicentre international cohort. Material/Methods. A retrospective analysis was conducted on patients with metastatic HER2+ or HER2-low BC treated at six leading European institutions. Ablative RT was defined based on a biological dose threshold of a minimum of 50Gy EQD2(10) delivered in no more than 12 fractions, as per the European Society for Radiotherapy and Oncology (ESTRO) OligoCare study. The primary objective was to assess the association between RT administration and adverse events (AEs) greater than grade 2 (G2). In all cases, RT was administered within one month prior to cycle 1 or during systemic treatment, without any interruption of T-DXd. Results. Data from 147 consecutive patients were evaluated. Sixty-seven patients received RT immediately before or during T-DXd treatment, amounting to 71 concurrent RT treatments, while 80 patients did not receive RT. The median age was 53 years old (range 28-88), and the median follow-up from T-DXd initiation was 15 months (range 1-46). Most patients received T-DXd as fourth or beyond line of systemic therapy (52.4%, N=77/147). The median total RT dose was 44Gy (range 8-48), delivered over a median of 4 fractions (range 1-20). The median EQD2 dose was 82Gy (range 12-104), and the median BED was 98Gy (range 14-125). The central nervous system was the most frequently treated site (58.2%; N=39/67), followed by bone (31.3%; N=21/67). Thirty-six patients (53.7%) received RT with ablative intent, and 31 patients (46.3%) received palliative RT. The relationship between RT and the development of >G2 overall toxicity was not statistically significant (p=0.30), nor when considering acute toxicity >G2 (p=0.31) and late toxicity >G2 (p=0.59) separately. There was no significant association between EQD2 (<50Gy or ≥50Gy) and BED (G2 and any grade acute and late toxicity. The only factor associated with an increased risk of acute toxicity was age ≥50 years old (p=0.035). Grade 2 interstitial lung disease (ILD) or G2-3 pneumonitis were observed in 7 cases in the RT group (10.5%) and 7 cases in the no-RT group (8.8%). Only one case of radionecrosis was reported among the 39 patients treated with intracranial RT (median follow up of 11 months). Overall, 19 patients permanently discontinued T-DXd due to toxicity, 8 in the RT group (11.9%) and 11 in the no-RT group (13.8%). Conclusion. Our preliminary findings suggest that the combination of T-DXd and concurrent RT does not increase the risk of severe acute and late toxicity.
Presentation numberPS2-02-03
Real-world treatment patterns in 1954 US patients with HER2-negative early breast cancer and germline BRCA mutations (2021-2025)
Sagar Sardesai, The Ohio State University Comprehensive Cancer Center, Columbus, OH
S. Sardesai1, M. Ru2, X. Ma3, C. Keane3, J. Wang3, M. Miranda4, X. Xu2, C. Isaacs5; 1Internal Medicine, Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 2Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 3Flatiron Health, Inc., New York, NY, 4Oncology Outcomes Research, AstraZeneca, Cambridge, UNITED KINGDOM, 5Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
Background: In 2022, the US FDA approved adjuvant olaparib for patients (pts) with germline pathogenic/likely pathogenic mutations in BRCA1 and/or BRCA2 (gBRCAm) and high-risk HER2-negative (HER2−) early breast cancer (eBC), based on significant survival improvements with olaparib vs placebo in OlympiA. In clinical practice, select pts with high-risk HER2− eBC may also receive CDK4/6 inhibitors (CDK4/6i) or immuno-oncology (IO) therapies; however, data are limited on treatment preference and sequencing in these pts. We describe real-world treatment patterns in US pts with gBRCAm HER2− eBC. Methods: Data from pts aged ≥18 years diagnosed with HER2− eBC (stage I-III) between January 1, 2021, and January 31, 2025, and with gBRCAm, were captured in the deidentified, electronic health record-derived Flatiron Health Research Database. Demographics, clinical characteristics, and treatment patterns were described across tumor subtypes (hormone receptor-positive [HR+] or triple-negative eBC [TNBC]). Pts with high-risk eBC were defined using criteria adapted from OlympiA (Tutt et al. NEJM 2021). Results: A total of 1954 pts with gBRCAm HER2− eBC were included (Table). Median age at diagnosis was 51 years among 1241 pts with HR+ eBC and 47 years among 713 pts with TNBC. Of 1010 pts with HR+ eBC who received adjuvant therapy, 217 (21%) received olaparib (including 19 pts who received olaparib before, 2 with, and 5 after CDK4/6i); 61 pts (6%) received CDK4/6i with endocrine therapy (ET) but without olaparib, 42 (4%) received IO without olaparib, and 282 (28%) received chemotherapy alone; 845 (84%) received ET, with or without other adjuvant therapies. Of 443 pts with TNBC who received adjuvant therapy, 162 (37%) received olaparib (93 received olaparib without IO; of 69 pts who received olaparib with IO, 18, notably, initiated olaparib before/concurrently with IO and 51 initiated olaparib after IO, including 50 within 27 weeks of initiating adjuvant IO); 196 (44%) pts received IO without olaparib and 82 (19%) received chemotherapy alone (including capecitabine in 8 pts). Among pts who received adjuvant therapy, 98/215 (46%) pts with high-risk HR+ eBC and 112/182 (62%) with high-risk TNBC received olaparib. Of 176 pts with TNBC without a pathological complete response (pCR) after neoadjuvant chemotherapy/IO, 90 (51%) did not receive olaparib. Among olaparib-treated pts, 119/217 (55%) with HR+ eBC and 50/162 (31%) with TNBC were not high risk. Conclusions: Real-world analysis showed that >40% of pts with gBRCAm, high-risk HER2− eBC did not receive adjuvant olaparib. Real-world olaparib use often did not align with restrictive criteria outlined in OlympiA, highlighting the need for comprehensive risk assessment to inform adjuvant treatment decisions in pts with gBRCAm HER2− eBC. Although prospective data is lacking, olaparib was frequently used concurrently with or after IO in pts with TNBC.
| HR+ eBC (n=1241) | TNBC (n=713) | High-risk HR+ eBC (n=244) | High-risk TNBC (n=217) | ||||||
| Received ≥1 adjuvant therapy, n | 1010 | 443 | 215 | 182 | |||||
| Adjuvant therapy type, n (%) | |||||||||
| Olaparib | 217 (21) | 162 (37) | 98 (46) | 112 (62) | |||||
| CDK4/6i without olaparib | 61 (6) | 0 | 27 (13) | 0 | |||||
| IO without olaparib | 42 (4) | 196 (44) | 13 (6) | 49 (27) | |||||
| Chemotherapy alone | 282 (28) | 82 (19) | 30 (14) | 20 (11) | |||||
| ET only | 405 (40) | 0 | 47 (22) | 0 | |||||
| Others | 3 (<1) | 3 (<1) | 0 | 1 (<1) | |||||
| Non-high-risk HR+ eBC (n=997) | Non-high-risk TNBC (n=496) | High-risk HR+ eBC (n=244) | High-risk TNBC (n=217) | ||||||
| Received ≥1 adjuvant therapy, n | 795 | 261 | 215 | 182 | |||||
| Total pts who received olaparib, n (%) | 119 (15) | 50 (19) | 98 (46) | 112 (62) | |||||
| Received olaparib, with high-risk defined as non-pCR after neoadjuvant chemotherapy/IO, n (%) | 26 (3) | 32 (12) | 77 (36) | 86 (47) | |||||
| Received olaparib, but did not receive neoadjuvant chemotherapy/IO, with high-risk defined as HR+ eBC with ≥pN2 or TNBC with ≥pN1 or pN0+≥pT2, n (%) | 93 (12) | 18 (7) | 21 (10) | 26 (14) |
Presentation numberPS2-02-04
Real-world treatment patterns and clinical outcomes in patients with emerging estrogen receptor 1 (ESR1)-mutated ER+ metastatic breast cancer in the U.S., 2018-2024
Jane L Meisel, Winship Canter Institute of Emory University, Atlanta, GA
J. L. Meisel1, T. Pham2, C. Chen2, A. Kris3, J. Roose4; 1NA, Winship Canter Institute of Emory University, Atlanta, GA, 2Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 3NA, Flatiron Health, Durham, NC, 4NA, Flatiron Health, New York, NY.
Background: Estrogen receptor alpha 1 (ESR1) mutations (m) are a significant driver of resistance to endocrine therapy (ET) in estrogen receptor-positive (ER+) advanced breast cancer (BC), leading to disease progression. Although the PADA-1 and SERENA-6 clinical studies highlighted the clinical relevance of monitoring for ESR1m emergence during first-line therapy (1L) and showed improvement in outcomes when therapy is changed upon detection, less is known about the real-world practice for patients with emergent ESR1m before progression and their outcomes. This study aims to characterize treatment patterns of patients whose tumors were tested for ESR1m during 1L and assess the impact of ESR1m on clinical outcomes, including real-world overall survival (rwOS) and real-world progression-free survival (rwPFS). Methods: This retrospective, observational cohort study used the US-based, deidentified Flatiron Health Research Database. Adult patients with a confirmed diagnosis of ER+/human epidermal growth factor receptor 2-negative (HER2-) metastatic BC from 1/1/2018 to 6/30/2024 were included. Treatment patterns were descriptively analyzed. rwOS and rwPFS were estimated for those with ESR1m emergence at first ESR1 test during 1L (time zero) using the Kaplan-Meier methodology and compared to those without ESR1 emergence at first ESR1 test using 1:1 propensity score matching on key covariates (time from 1L start to first ESR1 test result, ER expression level, presence of liver metastases, age, duration of prior aromatase inhibitor (AI) therapy, menopausal status). Results: This study included 8581 eligible patients, of whom 7772 (91%) initiated 1L therapy. Tumor ESR1m status was evaluated for 1335 (17%) during 1L, and 1086 were eligible for rwOS and rwPFS analyses. Treatment patterns in 1L were comparable between patients with (n=240) and without (n=1095) ESR1m emerging during 1L, with cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) + AI being the most common 1L treatment for patients with (140/240; 58%) and without (591/1095; 54%) ESR1m. CDK4/6i + fulvestrant was the second most common treatment for patients with (56/240; 23%) and without (248/1095; 23%) ESR1m. Patients not receiving 1L CDK4/6i + AI or CDK4/6i + fulvestrant received either ET monotherapy (9%), chemotherapy (3%), or other therapy (8%). Among patients who initiated 2L, those with an ESR1m detected during 1L compared to those without an ESR1m were more likely to receive 2L CDK4/6i + fulvestrant (49/202; 24% vs 93/573; 16%) or 2L elacestrant (29/202; 14% vs 1/573; 0.2%). Among those with an ESR1m detected during 1L (n = 240), median time to positive ESR1 test result was 18 months (mos) (IQR, 4-33) Median time from ESR1m detection during 1L to progression was 5.9 months.Eligible patients with an ESR1m detected on their earliest ESR1 test during 1L (n = 105) had shorter median rwPFS and median rwOS compared with those without an ESR1m (n = 981, rwPFS 7.7 mos [95% CI: 5.8-11.4] vs. 15.3 mos [95% CI: 14.0-16.8]) and (rwOS 32.2 mos [95% CI: 21.1-Not Reached (NR)] vs 45.6 mos [95% CI: 41.7-54.0]). After propensity score matching, both median rwPFS (7.7 mos [95% CI: 5.7-11.4] vs 13.6 mos [95% CI: 9.9-16.5]; hazard ratio [HR] 0.68 [95% CI: 0.48-0.96]) and median rwOS (32.2 mos [95% CI: 21.1-NR] vs NR [95% CI: 35.4-NR], HR: 0.58 [95% CI: 0.36-0.95]) were shorter in patients with an ESR1m detected during 1L. Conclusion: Patients with metastatic BC whose tumors were found to harbor an ESR1m during 1L had worse clinical outcomes than those who did not have an ESR1m. These findings highlight the potential clinical benefit of surveillance for ESR1m during 1L, and the need for treatment options that delay progression and improve clinical outcomes for patients with emerging ESR1-mutated, ER+ breast cancer.
Presentation numberPS2-02-05
Ibc clear: a decentralized multi-center mrd registry for inflammatory breast cancer
Angela Alexander, University of Texas MD Anderson Cancer Center, Houston, TX
A. Alexander1, M. Kai1, K. A. Abou-Hussein2, S. Ali3, C. Anderson4, S. W. Bahadur5, Y. C. Cheng6, N. T. Ueno7, X. Wang8, A. Lucci9, W. A. Woodward10, S. Saleem11, MDACC Inflammatory Breast Cancer Team; 1Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 2Hematology and Oncology, Cooper MD Anderson Cancer Center, Camden, NJ, 3Breast Medical Oncology, Scripps Health, La Jolla, CA, 4Breast Radiation Oncology, Baptist MD Anderson Cancer Center, Houston, TX, 5Breast Medical Oncology, Mayo Clinic Cancer Centet, Phoenix, AZ, 6Medical Oncology, Medical College of Wisconsin, Milwaukee, WI, 7Medicine, University of Hawaii Cancer Center, Honolulu, HI, 8Surgical Oncology, Rush MD Anderson Cancer Center, Chicago, IL, 9Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 10Breast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 11Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Sugar Land, TX.
IBC Clear: A decentralized multi-center MRD Registry for Inflammatory Breast CancerInflammatory breast cancer is an aggressive subtype of breast cancer with distinct clinical phenotypes, including early recurrence post-tri-modal care (neoadjuvant systemic therapy, surgery, adjuvant radiation). Preliminary data from MDACC have shown a high rate of baseline and serial MRD positivity in IBC patients (35-40% at 6-12 months post-surgery), suggesting that newly diagnosed IBC is an ideal population to test MRD as a standard of care and design adjuvant interventional trials. However, to date, our understanding of the optimal monitoring cadence and interventions following MRD positivity remains limited. IBC Clear is a decentralized protocol developed to collect real-world data from patients treated amongst our IBC Connect network of 19 US-based and three international institutions (7 of whom have expressed interest in participating). Patients who have had, or plan to have Signatera® ordered at the post-chemo/pre-surgery timepoint can self-enroll from the community by emailing IBCClear@mdanderson.org for information on the consent process. Study DesignEligibility criteria include English-speaking patients > 18 years of age with a clinical diagnosis of IBC, with Signatera testing ordered or planned. The post-chemo/pre-surgery timepoint is the only study-mandated timepoint; however, any additional serial testing conducted before or after this timepoint will also be collected. Patients who have at least one Signatera test ordered and plan to have a pre-surgery/post-chemo collection as standard of care will be enrolled in a central REDCap database. Physicians enrolling patients may act on the results according to their clinical judgment, including imaging assessments and treatment changes. These interventions will be captured in the database. An electronic EORTC QLQ C30 survey will also be administered prior to and after receiving Signatera results to gather information about the impact of testing on the patient’s quality of life. A total of 200 patients from all sites will be enrolled over a period of two years. Patients will be followed remotely for up to 2 years from the first Signatera test or until clinical recurrence, whichever comes first.EndpointsData collected will include the feasibility of decentralized data collection studies, as well as Signatera and recurrence information. The goals are to gather real-world data regarding the association of MRD positivity with pathologic response in IBC, patient-reported QOL for worry about recurrence, median lead time between the first MRD-positive result and clinical recurrence, and, if possible, whether the adjuvant therapies decrease the amount of MRD in IBC patients (pending optional longitudinal testing).Status of the study: The study was activated in June 2025. As of abstract submission, 2 patients have been enrolled at the lead site. Collaborating sites are being added. Sponsor: The University of Texas MD Anderson Cancer Center Clinicaltrials.gov Accession number: NCT06966050
Presentation numberPS2-02-06
Benefit of adjuvant chemotherapy for early-stage hormone receptor positive and HER2 negative lobular breast cancer patients: Analysis based on genomic risk scores
Arya Mariam Roy, The Ohio State University, Columbus, OH
A. Roy1, Y. Gokun2, B. Slover2, N. Lopetegui-Lia1, D. Quiroga1, G. Bader1, M. Cherian1, A. Davenport1, K. Johnson1, S. Sardesai1, R. Wesolowski1, S. Myers3, E. Burke3, M. Gatti-Mays1, N. Williams1, D. Stover1; 1Department of Medicine, The Ohio State University, Columbus, OH, 2Center for Biostatistics, The Ohio State University, Columbus, OH, 3Department of Surgery, The Ohio State University, Columbus, OH.
Background: Invasive lobular carcinoma (ILC) accounts for 10-15% of breast cancer (BC) cases and is predominantly hormone receptor-positive (HR+) and HER2-negative (HER2-). Although existing data indicates that ILC is less responsive to chemotherapy (CT) than invasive ductal carcinomas, the benefit of adjuvant CT in early-stage (ES) ILC is incompletely characterized. Further, the utility of the Oncotype DX Recurrence Score (RS), which has been used to predict response to CT in HR+/HER2- BC, has been debated for ILC. To address these issues, we studied the role of adjuvant CT on overall survival (OS) and by RS categories in ES ILC. Methodology: We queried the National Cancer Database for patients (pts) with ES HR+ HER2- ILC who underwent upfront surgical resection between 2010-2021. Pts were categorized to two cohorts based on receipt of adjuvant CT (CT+ and CT-). Subgroup analyses were performed based on RS score categories: low (L) 0-15, intermediate (I) 16-25, high (H) ≥26). Kaplan-Meier curves and log-rank tests determined OS between cohorts. Multivariate Cox proportional hazards models assessed OS, adjusting for potential confounders including age, race, stage, grade, and treatment variables. Results: Of the 162,430 ILC pts identified, 79.6% received adjuvant CT. The CT+ cohort were more likely to be younger pts (median age, IQR: 65 (55, 75) vs 66 (56, 72) years), have fewer comorbidities (Charlson-Deyo score (CDS) 0: 82.5 vs 82.3%, CDS 1: 13.1 vs 12.6%), intermediate grade disease (64.1 vs 62.2%), tumors <2 cm (67.1 vs 50.7%), clinically node negative disease (92.0 vs 77.7%), and receive radiation (63.5 vs 52.3%), and hormonal therapy (98.4 vs 51.4%), (all p<0.001). In the CT+ cohort, more pts had RS L (20.2 vs 8.9%), I (17.1 vs 7.5%), H (3.1 vs 1.7%) compared to CT-cohort (p<0.001). CT receipt conferred higher 5-year and 10-year OS. This survival benefit with adjuvant CT was observed among all RS groups with the highest benefit observed in H RS group (10Y OS: 84.5 vs 63.9%, p<0.001) (Table 1). In multivariate analysis adjusting for relevant confounders, receipt of adjuvant CT was associated with decreased morality in the overall ILC cohort (Hazard ratio (HR), 95% CI: 0.79, 0.76-0.83, p<0.001) and the RS subgroups, though not statistically significant in L RS group (HR, 95% CI: L – 0.77, 0.59 – 1.01, p= 0.06; I – 0.78, 0.62 – 0.98, p= 0.04; H – 0.67, 0.50 – 0.89, p= 0.006). Conclusion: In this study, adjuvant CT was associated with improved OS in ILC pts with RS> 15. These data support use of RS for CT decision-making in appropriate candidates. Further investigations are needed to identify ILC pts who would benefit the most from adjuvant CT.
| Group | Time point (Year) | Received Adjuvant CT OS (%) (95% CI) |
No Adjuvant CT OS (%) (95% CI) |
P-value |
| Overall Cohort | 5 | 91.2 (91.1-91.4) | 82.5 (82.1-83.0) | <0.001 |
| 10 | 76.5 (76.2-76.9) | 61.8 (60.9-62.6) | ||
| Low RS | 5 | 96.5 (96.2-96.7) | 92.7 (91.4-93.8) | <0.001 |
| 10 | 88.1 (87.3-88.8) | 81.5 (78.4-84.2) | ||
| Intermediate RS | 5 | 95.5 (95.2-95.8) | 92.1 (90.6-93.3) | <0.001 |
| 10 | 85.5 (84.7-86.3) | 79.6 (76.3-82.4) | ||
| High RS | 5 | 92.1 (91.1-93.0) | 78.0 (75.6-80.1) | <0.001 |
| 10 | 84.5 (80.4-87.7) | 63.9 (55.1-71.5) |
Presentation numberPS2-02-07
Impact of BRCA homozygous copy loss on clinical outcomes of patients (pts) with metastatic breast cancer (MBC) treated with PARP inhibitors (PARPi)
Adela Rodríguez-Hernández, Dana-Farber Cancer Institute, Boston, MA
A. Rodríguez-Hernández1, J. Quintanilla2, R. Graf2, A. Schrock2, A. Gasco2, B. Bychkovsky1, R. Kitadai1, S. Morganti1, D. Abravanel1, S. Tolaney1, J. Garber1, R. Jeselsohn1, N. Lin1, F. Lynce1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Foundation Medicine, Foundation Medicine, Boston, MA.
Background: PARPi are used to treat MBC, particularly in pts with genomic alterations (GA) in BRCA1/2 by targeting homologous recombination deficiency (HRD). This study explored clinical and genomic factors associated with PARPi sensitivity in a real-world MBC cohort. Methods: This retrospective study included 290 pts with MBC who had genomic testing via FoundationOne/FoundationOne CDx tissue comprehensive genomic profiling (CGP) and prior to treatment with a PARPi. Clinical data were sourced from the US-wide de-identified Flatiron Health-Foundation Medicine clinic genomic database, including ~800 sites across ~280 US cancer clinics (Jan 2011-Dec 2024). HRD alterations (alts) were defined as mutations, homozygous copy loss, or structural rearrangements in any of the following genes: BRCA1/2, ATM, ATR, ATRX, BAP1, BARD1, BRIP1, CHEK1/2, CDK12, FANCA, FANCL, MRE11, RAD51B/C/D, RAD54L or PALB2. HRD signature (HRDsig) status, which is offered as a laboratory professional service, was calculated using genome-wide copy number features, with scores from 0 to 1 and a predefined cutoff of 0.7 to call HRDsig (+). BRCA1/2 germline status was inferred using a validated somatic-germline-zygosity algorithm. Time to next treatment or death (TTNT) on PARPi was analyzed using Cox models adjusted for clinical variables. Results: Among 290 pts, most were female (96.6%, n=280), with a median age of 57 years at PARPi initiation. Of these, 186 (64.1%) had hormone receptor-positive (HR+) disease and 104 (35.9%) had HR- disease. In total, 21 had HER2+ tumors (12 HR+, 9 HR-). PARPi was given +/- endocrine therapy in 236 pts (81.4%) and in combination with other therapies in 54 (18.6%). PARPi was given in 1L (14.1%), 2L (26.9%), 3L (19.3%), and ≥4L (39.7%). Tumors from 199 patients (59.2%) were HRDsig (+): 120 (60.3%) HR+ and 79 (39.7%) HR−. Overall, 237 pts (81.7%) harbored ≥1GA (mutation, homozygous copy loss, or rearrangement) in BRCA1 (n=90), BRCA2 (n=130), or PALB2 (n=19). Nineteen pts had BRCAloss (8 in BRCA1 and 11 in BRCA2). In pre-PARPi CGP samples, HR+ tumors most frequently harbored BRCA2 (52.7%), TP53 (32.8%), BRCA1 (21.0%), while HR- tumors showed higher rates of TP53 (85.6%), BRCA1 (49.0%), BRCA2 (30.8%), and PALB2 (4.8%). Among pts with HR+ disease, those with a BRCAloss vs other BRCA GA detected prior to PARPi exposure had more favorable TTNT (median 15.2 vs. 8.5 months HR:0.40; p=0.012). No differences in TTNT were observed by BRCA1/2 mutation origin (germline (g) vs somatic (s)), type (truncating vs missense), or location (Table 1). Conclusions: Response to PARPi varied across different GA. Among pts with HR+ MBC, those with BRCAloss appeared to derive the greatest benefit. These findings highlight the potential utility of CGP in guiding PARPi treatment decisions.
| HR+MBC | |||
| Comparision | MedianTTNT(months) | HR(95%CI) | p-value |
| OverallHR+(n=186) | 8.0(7.4-9.2) | – | – |
| BRCAloss(n=14)vsotherBRCAalts(n=122) | 15.2vs8.5 | 0.40(0.19-0.82) | 0.012 |
| BRCAalts(n=136)vsnoHRDalts(n=21) | 8.7vs3.5 | 0.39(0.23-0.66) | <0.001 |
| PALB2alts(n=13)vsnoHRDalts(n=21) | 8.6vs3.5 | 0.43(0.20-0.93) | 0.032 |
| HRDsig(+)(n=120)vsHRDsig(-)(n=60) | 8.8vs6.0 | 0.52(0.36-0.76) | <0.001 |
| sBRCA(n=22)vsgBRCA(n=71) | 9.0vs7.9 | 1.28(0.69-2.34) | 0.434 |
| BRCAmuttruncation(n=108)vsotherBRCAalts(n=15) | 8.8vs6.0 | 0.71(0.37-1.36) | 0.301 |
| BRCAmutexon11(n=48)vsothersBRCAalts(n=100) | 9.0vs8.5 | 1.07(0.70-1.63) | 0.767 |
| HR-MBC | |||
| Comparision | MedianTTNT(months) | HR(95%CI) | p-value |
| OverallHR-(n=104) | 7.4(5.2-9.2) | – | – |
| BRCAloss(n=5)vsotherBRCAalts(n=78) | 8.5vs8.1 | 0.55(0.17-1.72) | 0.300 |
| BRCAalts(n=83)vsnoHRDalts(n=11) | 8.5vs3.3 | 0.20(0.09-0.48) | <0.001 |
| PALB2alts(n=5)vsnoHRDalts(n=11) | 7.5vs3.3 | 0.52(0.13-2.03) | 0.344 |
| HRDsig(+)(n=79)vsHRDsig(-)(n=23) | 8.5vs3.6 | 0.43(0.22-0.86) | 0.017 |
| sBRCA(n=11)vsgBRCA(n=40) | 8.2vs7.5 | 1.67(0.57-4.91) | 0.242 |
| BRCAmuttruncation(n=76)vsotherBRCAalts(n=12) | 8.2vs3.4 | 0.66(0.28-1.56) | 0.220 |
| BRCAmutexon11(n=48)vsotherBRCAalts(n=100) | 12.6vs7.4 | 0.98(0.96-1.01) | 0.104 |
Presentation numberPS2-02-08
Real-world PARP inhibitor use and clinical outcomes in US patients with HER2-negative metastatic breast cancer
Siddhartha Yadav, Mayo Clinic, Rochester, MN
S. Yadav1, Q. Li2, Z. Tan3, K. E. Mishkin4, J. F. Hayes5, X. Xu2, F. J. Couch1; 1Department of Oncology, Mayo Clinic, Rochester, MN, 2Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 3Oncology Outcomes Research, AstraZeneca, Mississauga, ON, CANADA, 4Oncology Outcomes Research, Merck & Co., Inc., Rahway, NJ, 5US Medical Affairs Oncology, AstraZeneca, Gaithersburg, MD.
Background: Clinical benefit with PARP inhibitors (PARPi) has been demonstrated in patients (pts) with HER2-negative (HER2−) metastatic breast cancer (mBC) and germline or tumor mutations in BRCA1 or BRCA2 (g/tBRCAm). Understanding real-world PARPi use and its impact on clinical outcomes may improve the treatment of pts with BRCAm HER2− mBC. Methods: This retrospective study captured data from US pts aged ≥18 years diagnosed with HER2− mBC from January 12, 2018, to March 31, 2024, in the deidentified electronic health record-derived Flatiron Health Research Database. PARPi use and outcomes were described by tumor subtype (hormone receptor-positive [HR+]/HER2− mBC or triple-negative mBC [mTNBC]), g/tBRCA status, disease stage at diagnosis (de novo or recurrent), line of therapy (LoT), and prior treatments. Real-world overall survival (rwOS) by PARPi use was estimated using the Kaplan-Meier method. Results: Of 848 pts with gBRCAm mBC, 352 (42%) received PARPi (Table), with consistent PARPi initiation rates of 42%-48% from 2018-2022. PARPi was initiated in 223/566 (39%) pts with gBRCAm HR+/HER2− mBC, 125/275 (45%) pts with gBRCAm mTNBC, and 4/7 (57%) pts with unknown tumor subtype. Among 304 pts with gBRCAm who had received ≥3 LoT by the end of follow-up, 93/225 (41%) pts with HR+/HER2− mBC and 18/78 (23%) pts with mTNBC received PARPi in the third LoT or later (3L+); 50/103 (49%) pts with de novo mBC and 54/184 (29%) pts with recurrent mBC received PARPi in 3L+. PARPi was also initiated in 13/231 (6%) pts with gBRCA variants of uncertain significance (VUS) and 176/662 (27%) pts with tBRCAm and non-gBRCAm/unknown gBRCAm status. Among 539/1573 (34%) pts with g/tBRCAm who received PARPi, 167 had not received prior chemotherapy in any setting; 40 of these pts (39 with HR+/HER2− mBC and 1 with mTNBC) received PARPi in 3L+. With a median follow-up of 17.0 months from initiation of 1L therapy, median rwOS was numerically longer among pts with gBRCAm who received PARPi versus pts who did not (HR+/HER2− mBC: 39.3 months [95% CI: 35.5-46.8], n=208 vs 24.2 months [95% CI: 21.4-33.0], n=259; mTNBC: 27.6 months [95% CI: 18.0-41.7], n=119 vs 16.9 months [95% CI: 14.5-31.9], n=81). Among pts with tBRCAm who were negative for gBRCAm, median rwOS was numerically longer in pts who received PARPi versus pts who did not. Conclusions: Pts with HR+/HER2− mBC had slightly lower rates of PARPi use, and received PARPi later, than pts with mTNBC. Although improved efficacy with PARPi use in earlier LoT has been demonstrated, many pts first received PARPi in 3L+, suggesting unmet potential to ensure timely testing and optimize treatment. Although immortal time bias and other factors should be considered, rwOS was numerically longer in pts with BRCAm HR+/HER2− mBC and with BRCAm mTNBC who initiated PARPi versus pts who did not. Further research into PARPi use and survival trends in pts with BRCAm and/or other actionable biomarkers in mBC is needed.
Presentation numberPS2-02-09
Fr-pop: Real-world data from 7,687 high-risk TNBC patients treated with perioperative pembrolizumab in France
Olivier Tredan, Centre Leon Berard, Cancer Research Center of Lyon (UMR Inserm 1052 – CNRS 5286), Lyon, France
A. de Nonneville1, D. Loirat2, J. Ribeiro3, S. Becourt4, M. Benderra5, N. Benard6, M. Campone7, F. Dalenc8, O. Derbel9, I. Desmoulins10, A. Deleuze11, G. Emile12, A. Goncalves1, J. Grenier13, W. Jacot14, C. Liautard6, L. Mansi15, M. A. Mouret Reynier16, C. Perkins6, B. Pistilli3, J. P. Spano17, A. Vasseur18, F. Penault Llorca19, O. Tredan20; 1Medical Oncology, Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, FRANCE, 2Medical Oncology, Institut Curie- Institut of Women’s Cancers, Paris, FRANCE, 3Breast Unit, Medical Oncology Department, Gustave Roussy Cancer Campus, Villejuif, FRANCE, 4Medical Oncology, Centre Oscar Lambret, Lille, FRANCE, 5Department of Medical Oncology, Institut Universitaire de Cancérologie, Sorbonne University, AP-HP, Tenon Hospital, Paris, FRANCE, 6Medical affairs, MSD France, Puteaux, FRANCE, 7Medical Oncology, Institut de Cancérologie de l’Ouest, Saint Herblain, FRANCE, 8Medical Oncology, Oncopole Claudus Regaud, Toulouse, FRANCE, 9Medical Oncology, Hopital privé Jean Mermoz, Puteaux, FRANCE, 10Medical Oncology, Centre Georges-François Leclerc, Dijon, FRANCE, 11Medical Oncology, Centre Eugene Marquis, Rennes, FRANCE, 12Medical Oncology, François Baclesse Comprehensive Cancer Center, Caen, FRANCE, 13Medical Oncology, Institut Sainte Catherine, Avignon, FRANCE, 14Medical Oncology, Institut du Cancer de Montpellier, Montpellier University, INSERM U1194, Montpellier, FRANCE, 15Medical Oncology, CHRU Jean Minjoz, Besancon, FRANCE, 16Medical Oncology, Centre Jean Perrin, Clermont, FRANCE, 17Medical Oncology, Pitié Salpêtrière AP-HP, Sorbonne Université, INSERM U 1136,, Paris, FRANCE, 18Medical Oncology, Institut Curie, Saint-Cloud, FRANCE, 19Pathology, Centre Jean Perrin, Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont Ferrand, FRANCE, 20Medical Oncology, Centre Leon Berard, Cancer Research Center of Lyon (UMR Inserm 1052 – CNRS 5286), Lyon, FRANCE.
Background: Since the KEYNOTE-522 trial demonstrated improved pathological response and survival outcomes, pembrolizumab in combination with neoadjuvant chemotherapy, followed by adjuvant administration, is the current standard of care for stage II-III triple-negative breast cancer (eTNBC). Objective: This prospective national observational cohort analysis aims to describe real-world data from high-risk eTNBC patients treated with this schema through the French KN522 Early Access Program (EAP) from April 13, 2022, to November 20, 2024. Methods: Eligible patients were adults with confirmed stage II-III TNBC, including ER 1-9% according to French guidelines. Data were collected via physician-completed EAP forms and patient-reported EORTC QLQ-C30 questionnaires. Clinical and safety data were recorded until the EAP concluded or the end of the treatment. Results: A total of 7,687 patients were enrolled during the 31-month EAP across 427 healthcare centers and by 1,259 physicians. Participating centers included comprehensive cancer centers (34%), private clinics (34.1%), and public hospitals (31.8%). Median age of patients was 52 years (range 19-92 years) including 21.8% >65 years old and mean BMI was 26.3 kg/m2 (IQR 22.2 – 29.4). 3002 (39,1%) patients had stage III disease, and 87.5% had an ECOG-PS of 0. Pembrolizumab was administered at the planned dose of 200 mg every three weeks for nearly all patients and 96.1% planned the chemotherapy regimen used in KN522 (carboplatin/paclitaxel -anthracycline/cyclophosphamide). A total of 1105 patients (14.4%) have temporary interrupted pembrolizumab treatment, predominantly during the neoadjuvant phase (74.3%), with 38.1% related to immune-mediated adverse events. The median duration of this interruption was 13 days. 1240 patients (16%) have permanently discontinued pembrolizumab with the main cause being suspected immune-related toxicity (49.1%). The second reported reason for permanent discontinuation was physician decision related to the pathological response (19.3%), whether it was complete (6.3%) or not (13%). The breast-conserving surgery rate was 63,8%, with a median interval of 28 days between the completion of neoadjuvant therapy and surgery. The overall pathological complete response (pCR) rate was 71.6%. For patients who experienced treatment interruption due to immune toxicity (n=737), the pCR rate was 71.2% and was 73.3% for patients younger than 30 years old (n=45). Additional systemic treatments were received by 8.8% of patients in the adjuvant phase (with a higher incidence for patients with residual disease). In most cases, it was chemotherapy, but PARPi and endocrine therapies were also reported. Quality of life data, assessed via the QLQ-C30, showed stable global health scores (mean score of 85.3 at neoadjuvant Cycle one (n=423) vs. 81.3 at adjuvant Cycle nine (n=40)). Pharmacovigilance reported a total of 417 serious treatment-related cases, including eight deaths. Conclusions: Thanks to the substantial participation of physicians and centers across France, FR-POP represents one of the largest real-world cohorts collecting relevant efficacy and safety data on KN522 regimen in clinical practice. With an observed pCR rate exceeding 70% despite the high proportion of stage III tumors, and in a non-selected and older population, these findings clearly point out the efficacy of this treatment strategy and the external validity of KN522 results. However, underreporting of adverse events and the absence of follow up after the end of treatment remain potential limitations of EAP-derived safety data and tolerance of this regimen should continue to be closely monitored.
Presentation numberPS2-02-10
The Benefit and Burden of Adjuvant Endocrine Therapy for Older Individuals with Early Stage, Hormone Receptor Positive Breast Cancer
Danielle Rodin, Princess Margaret Cancer Centre, Toronto, ON, Canada
D. Rodin1, R. Sutradhar1, E. Hahn1, K. Jerzak1, L. Nguyen2, S. Trebinjac3, L. Laszat3, E. Rakovitch3; 1Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 2Institute for Clinical Evaluative Sciences, Sunnybrook Health Sciences Centre, Toronto, ON, CANADA, 3Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON, CANADA.
PurposeTo determine the impact of adjuvant endocrine therapy (ET) among older women diagnosed with hormone positive (HR+), early-stage breast cancer (EBC) on the risks of recurrence and death, and on services utilization. Methods: We identified a population-based cohort of individuals >65 years with T1N0 HR+EBC, treated with breast-conserving surgery (BCS), with or without breast radiotherapy (RT), from Jan 2012- Aug 2021, and followed until December 2023. Exposure to ET was calculated for the 2-year interval following diagnosis, with ET adherence defined as ET use for >65% of the interval, with 1:1 matching performed for cases <65% adherent and those without ET exposure. We calculated the cumulative incidence of ipsilateral local recurrence (LR), contralateral breast cancer (CBC), and breast cancer mortality, adjusting for competing risk of death and stratified by receipt of RT. A difference-in-differences analysis with a Poisson regression was used to compare health service utilization between cases and controls. Results The study cohort included 3486 individuals (median age 71 years, 2,796 received RT); with 1162 in each group (>65% adherent, <65% adherent, and no ET). Among individuals who received RT, >65% adherence to ET was associated with a small absolute reduction in the 10-year cumulative risk of LR compared to no ET (>65% ET: 0.2% (95%CI: 0.05, 0.84); no ET: 3.4% (95%CI: 1.75, 5.93; p=.01)). There was no difference in 10-year risks of CBC or breast cancer mortality. Among individuals who did not receive RT, >65% adherence to ET was associated with a greater reduction in 10-year LR risk (1.8% (95% CI: 0.33, 6.13) vs 7.0% (95%CI: 3.6, 12.0; p=.04)) and a small reduction in breast cancer mortality (0% vs 3.3% (95% CI: 1.2, 7.3; p=0.03)), compared to those who did not receive ET. Health care utilization was significantly increased with ET use and was greatest for individuals >65% adherent, with significantly more diagnostic imaging tests, primary care, and specialist physician visits compared to those who did not receive ET (Table). Individuals <65% adherent had a smaller absolute benefit from ET, but similarly high health service utilization.. Conclusions For older individuals with stage T1N0 HR+EBC, adjuvant ET is associated with a significant increase in health care utilization. For individuals treated with BCS+RT, ET was associated with a small absolute reduction in LR but no difference in CBC or BC mortality, suggesting that the burden of ET may outweigh the benefit in this population.
|
Health Servies Utilization |
ET>65% Adherent |
ET <65% Adherent | ||
| Relative Rate Ratio (95% CI) | P-value | Relative Rate Ratio (95% CI) | P-value | |
| Ambulatory visits | ||||
| Primary care visits | 1.07 (1.04, 1.11) | < 0.001 | 1.04 (1.01, 1.08) | 0.01 |
| Specialist visits | 1.25 (1.19, 1.31) | < 0.001 | 1.08 (1.04, 1.13) | <0.001 |
| Gynecology visits | 1.23 (1.02, 1.48) | 0.03 | 1.27 (1.06, 1.51) | 0.001 |
| Ambulatory visits | 1.23 (1.14, 1.33) | < 0.001 | 1.07 (1, 1.15) | 0.07 |
| Imaging Tests | ||||
| Visits with VTE diagnostic code | 1.65 (1.39, 1.95) | < 0.001 | 1.34 (1.12, 1.59) | <0.001 |
| CT Scans | 1.16 (1.05, 1.29) | < 0.001 | 1.19 (1.07, 1.31) | <0.001 |
| MRI | 1.15 (1.02, 1.31) | 0.02 | 1.16 (1.03, 1.31) | 0.02 |
| Ultrasound | 1.03 (0.97, 1.1) | 0.29 | 1.06 (1, 1.12) | 0.07 |
| Bone mineral density scan | 1.97 (1.78, 2.19) | < 0.001 | 1.42 (1.27, 1.58) | <0.001 |
Presentation numberPS2-02-12
Tolerability of Adjuvant Abemaciclib in Older Adults with High-Risk Early Breast Cancer: Multicentre UK Real-World Data from the CONCISE Study
Olubukola Ayodele, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
H. Abdallah1, O. Ayodele2, A. Ghose3, Y. Owoseni2, A. Maniam4, B. Baraka5, S. Horne6, A. Nazir6, L. Mooney7, F. Nazeer8, N. Atsumi4, L. McAvan9, M. Abraham10, E. Bean11, D. Morgan12, G. Langford12, E. Morris13, R. Muhammad14, E. Daniels15, R. Pravinkumar16, S. Mohammed17, S. Raza18, C. Blair19, R. Khan20, M. Tsalic6, R. Kussaibati6, R. Douglas7, K. Panagiotis21, S. Waters12, J. Smith11, E. Papadimitraki13, C. Michie16, C. Wilson20, A. Konstantis14; 1Oncology Department, Cambridge Clinical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UNITED KINGDOM, 2Oncology Department, University Hospitals of Leicester NHS Trust, Leicester, UNITED KINGDOM, 3Oncology Department, Barts Cancer Centre, Barts Health NHS Trust, London, UNITED KINGDOM, 4Oncology Department, Portsmouth Hospitals University NHS Trust, Portsmouth, UNITED KINGDOM, 5Oncology Department, Nottingham University Hospitals NHS Trust, Nottingham, UNITED KINGDOM, 6Oncology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UNITED KINGDOM, 7Oncology Department, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, UNITED KINGDOM, 8Oncology Department, Royal Surrey Cancer Centre, Royal Surrey NHS Foundation Trust, Surrey, UNITED KINGDOM, 9Oncology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UNITED KINGDOM, 10Oncology Department, Dorset Cancer Centre, University Hospitals Dorset NHS Foundation Trust, Poole, UNITED KINGDOM, 11Oncology Department, Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UNITED KINGDOM, 12Oncology Department, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UNITED KINGDOM, 13Oncology Department, University College London Hospitals NHS Foundation Trust, London, UNITED KINGDOM, 14Oncology Department, Princess Alexandra Hospital NHS Trust, Harlow, UNITED KINGDOM, 15Oncology Department, Dyson Cancer Centre, Royal United Hospitals Bath NHS Foundation Trust, Bath, UNITED KINGDOM, 16Oncology Department, Edinburgh Cancer Centre, Edinburgh, UNITED KINGDOM, 17Oncology Department, Great Western Hospitals NHS Foundation Trust, Swindon, UNITED KINGDOM, 18Oncology Department, Hull University Teaching Hospitals NHS Trust, Cottingham, UNITED KINGDOM, 19Oncology Department, University Hospitals of Bristol and Weston NHS Trust, Bristol, UNITED KINGDOM, 20Oncology Department, The Christie NHS Foundation Trust, Manchester, UNITED KINGDOM, 21Oncology Department, Frimley Health NHS Foundation Trust, Camberley, UNITED KINGDOM.
Background Adjuvant Abemaciclib (AA) combined with endocrine therapy (ET) significantly improves invasive disease-free survival (IDFS) in patients with hormone receptor-positive (HR+), HER2-negative (HER2 -), node-positive early breast cancer (EBC), as demonstrated in the monarchE trial. However, only 15.6% of patients in monarchE were aged ≥65 and no dedicated geriatric safety analysis was performed. Managing older adults with EBC is complex due to comorbidities, reduced physiological reserve and polypharmacy. There is limited real-world evidence on the safety and tolerability of AA in this population. Methods The CONCISE study is a retrospective, multicentre audit of UK practice across 21 centres, including patients treated with AA from July 2022 to April 2025. This analysis focuses on patients aged ≥65 years. Data were collected using the Ledidi platform and analysed using descriptive statistics in Jamovi. Key outcomes include treatment duration, dose modifications and adverse events (AEs), stratified by two subgroups: Cohort 1 (age 65-75) and Cohort 2 (>75). Group comparisons were performed using chi-squared and t-test. Results Of 1,026 patients, 215 (21%) were aged ≥65: 152 in Cohort 1 (median age 69) and 63 in Cohort 2 (median age 78). Most were female (96.3%) and Caucasian (88.4%). Stage II disease was most common (56.7%), with grade 2 tumours (57.8%) and nodal involvement (72.8%). Chemotherapy use differed by age: in Cohort 1, 68% received adjuvant chemotherapy and 11% neoadjuvant chemotherapy; in Cohort 2, only 31% received adjuvant chemotherapy and one patient had neoadjuvant chemotherapy.Median AA duration was 12.6 months in Cohort 1 vs. 8.3 months in Cohort 2 (p=0.028). Discontinuation of the treatment due to toxicity was significantly more common in Cohort 2 (79%) than in Cohort 1 (49%) [p=0.002]. Treatment completion (24 months) was achieved in 47% (Cohort 1) and 21% (Cohort 2). At data cut-off, 53% and 40% of patients remained on treatment in Cohorts 1 and 2, respectively. Disease progression occurred in 4% (Cohort 1) and 0% (Cohort 2).One-level dose reductions were required in 70% (Cohort 1) and 76% (Cohort 2) [p=0.29], while further reductions were needed in 34% and 38% [p=0.55], respectively. The most frequent AEs were diarrhoea (75% vs. 71.4%), anaemia (40% in both), and neutropenia (34.2% vs. 30.2%), with grade ≥2 rates comparable between groups.
| Adverse Event | Cohort 1 (65-75) All Grades / ≥G2 (%) | Cohort 2 (>75) All Grades / ≥G2 (%) | |||
| Diarrhoea | 75.0 / 36.8 | 71.4 / 36.5 | |||
| Anaemia | 40.0 / 6.6 | 40.0 / 8.0 | |||
| Neutropenia | 34.2 / 19.7 | 30.2 / 16.0 | |||
| Thrombocytopenia | 13.8 / 1.3 | 20.6 / 1.6 | |||
| Elevated liver enzymes | 20.4 / 7.2 | 20.6 / 6.4 |
Conclusion In this large UK real-world cohort, older adults with high-risk EBC experienced similar AE profiles to younger counterparts, but with numerically higher discontinuation rates, particularly in those aged >75. Dose reductions were common across both age groups. These findings underscore the importance of careful patient selection, supportive care, and geriatric assessment when offering AA in routine clinical practice.
Presentation numberPS2-02-13
Bridging Diagnostic Gaps in LMICs- Real-World Functionality of a One-Stop Breast Centre in South India
Debashri Shankarraman, Chennai Breast Centre, Chennai, India
D. Shankarraman1, G. Sivaramalingam2, N. Govindarajan3, S. Radhakrishna1; 1Breast Surgery, Chennai Breast Centre, Chennai, INDIA, 2Radiology – Breast Imaging and Interventions, Chennai Breast Centre, Chennai, INDIA, 3Pathology, Chennai Breast Centre, Chennai, INDIA.
Bridging Diagnostic Gaps in LMICs: Real-World Functionality of a One-Stop Breast Centre in South IndiaBackgroundIn low- and middle-income countries (LMICs), fragmented diagnostic pathways often result in delayed breast cancer diagnosis and treatment. Patients are typically referred across departments (surgery, radiology, pathology), leading to multiple visits and potential loss to follow-up. The One-Stop Breast Clinic (OSBC) model, which integrates clinical evaluation, imaging, and biopsy into a single coordinated visit, aims to streamline this process. We present real-world data from a high-volume OSBC in South India to evaluate its diagnostic performance and workflow efficiency.ObjectiveTo assess the feasibility, diagnostic accuracy, and operational efficiency of the OSBC model in a private healthcare setting in India.MethodsThis retrospective audit included all female patients seen at Chennai Breast Centre from January 1 to December 31, 2024. Data were collected on patient demographics, imaging origin and quality, need for repeat evaluations, biopsy results, and diagnostic timelines. New symptomatic patients and follow-up visits were analyzed separately. Key outcome measures included the rate of repeat imaging/biopsy, time to biopsy, and time to treatment planning.ResultsAmong 12,156 patients, 2,777 were new symptomatic cases; 9,379 were follow-ups. New patients ranged in age from 12–93 years (mean: 47.5); Final diagnosis (new patients): Benign: 2,126 (76.5%), Malignant: 627 (22.6%)- Imaging origin: 1,158 (41.7%) had primary imaging at CBC; 1,619 (58.3%) presented with external scans- Repeat imaging required: 1,450 (52.2%), including 550 mammograms and 935 ultrasounds- Biopsies performed: 674 US-guided core biopsies, 2 punch, 8 stereotactic- MRI additionally performed in 70 patientsOf the 495 patients biopsied after repeat imaging, 384 had not been previously advised biopsy externally, suggesting significant diagnostic upgrades. Among 479 patients with prior external biopsies, 120 required a repeat biopsy; of these, 75 (62.5%) were malignant and 45 (37.5%) benign. This highlights a potential for missed malignancies with suboptimal external evaluations and emphasizes the critical need for quality-controlled, structured diagnostic workflows like the OSBC model. Workflow EfficiencyMedian time from patient entry to completion of history, clinical exam, imaging, and biopsy was 125 minutes. Biopsy reports were available within 72 hours. Unlike multispecialty hospitals that require multiple visits for triple assessment, the OSBC model completed this in a single session.Integrated Pathway– Visit 1: Triple assessment and biopsy- Visit 2: Biopsy review; staging and baseline workup initiated for malignancies- Visit 3: Treatment decision finalized with complete reports- Visit 4: Preoperative documentation- Visit 5: Daycare surgeryThis model particularly benefits patients traveling long distances. Staging workup and general health evaluations are performed while awaiting pathology results, ensuring that treatment planning occurs within 2–3 visits. The approach enhances patient compliance, reduces dropouts, and improves system-level efficiency.ConclusionThe OSBC model provides a practical, scalable solution to reduce diagnostic delays in LMICs. In this high-volume South Indian setting, triple assessment was achieved in <2 hours and definitive treatment planning within 3 visits. The model minimizes attrition, improves diagnostic accuracy, and optimizes patient flow. Expansion of such integrated breast care centers may be key to strengthening cancer care infrastructure in resource-limited settings.KeywordsOne-stop breast clinic, LMIC, breast cancer, triple assessment, diagnostic delay, India, integrated care, real-world evidence
Presentation numberPS2-02-14
Improving Identification and Enrollment in HR+/HER2− Breast Cancer Trials Using AI Clinical Trial Patient Matching Tool
Guannan Gong, Yale School of Medicine, New Haven, CT
J. Liu1, G. Gong1, S. Pandya2, J. Xie3, J. Parikh3, N. Fischbach4, P. Kunz4, L. Pusztai1, M. Lustberg1; 1Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 3Yale College, Yale University, New Haven, CT, 4Department of Medicine, Division of Medical Oncology, Yale School of Medicine, New Haven, CT.
Background: Significant enrollment gaps persist in breast cancer clinical trials, particularly among patients with high-risk hormone receptor-positive (HR+), HER2-negative early breast cancer (EBC). Traditional manual methods are challenged by high patient volume, unstructured clinical notes, and limited research staffing. These barriers hinder recruitment efficiency and contribute to limited inclusion of all patient populations. To address these gaps, we deployed a Clinical Trial Patient Matching (CTPM) system powered by artificial intelligence (AI) and natural language processing (NLP) to enhance identification of eligible patients for two multi-site interventional trials: EMBER-4 (NCT05514054) and DARE (NCT04567420). Methods: The CTPM algorithm was deployed across 13 breast oncology sites within the Yale New Haven Health (YNHH) Smilow Cancer Hospital network. From September 2022 to May 2024, the system performed automated weekly pre-screening of all breast cancer patients seen across the network. CTPM extracted both structured and unstructured data from the electronic health record (EHR), applying trial-specific eligibility criteria for two multi-site interventional trials: EMBER-4 and DARE. Identified patient matches underwent manual chart review by a clinical research coordinator (CRC) to confirm eligibility prior to outreach and enrollment. Results: Between September 2022 and May 2024, the CTPM system pre-screened 45,380 unique breast cancer patients across YNHH. Based on trial-specific criteria, the system identified 429 patients as potentially eligible for EMBER-4 and 353 for DARE. Following manual chart review by CRCs, 263 patients were confirmed as eligible for EMBER-4 and 140 for DARE. For the EMBER-4 trial, CTPM identified 263 eligible patients, of whom 2.28% were aged 18-39, 26.6% were >70 years, 26.6% were racial/ethnic minorities, 6.1% NES, 22.8% Medicaid-insured, and 6.1% rural residents. Thirty-seven patients (13.81%) enrolled, with higher representation among racial/ethnic minorities (27.0%), patients > 70 years (13.5%), and Medicaid-insured patients (16.2%). For DARE, the 140 eligible patients identified by CTPM included 5.7% were aged 18-39, 22.1% were >70 years, 25.7% were racial/ethnic minorities, 8.6% NES, 24.3% Medicaid-insured, and 7.1% rural residents. Thirty-one patients (22.1%) enrolled in DARE, with notable representation among patients >70 years (22.6%), racial/ethnic minorities (19.4%), and Medicaid-insured (19.35%) patients. As a result of this enhanced identification and screening process, YNHH was a top enrollment site for patients nationally in both trials. Conclusions: This study presents the first prospective application of an AI-driven trial-matching system for high-risk HR+/HER2- EBC patients. Integration with EHRs enhanced identification of eligible patients, particularly among historically underrepresented groups. The subset of patients deemed ineligible after manual review reflects the difference between the CTPM system’s design— prioritizing sensitivity using a limited set of high-priority eligibility criteria from structured EHR data— and CRCs’ comprehensive reviews including unstructured clinical data not readily extractable through AI/NLP methods. While relaxed algorithm criteria minimized false negatives, we found that a two-tiered approach of broad AI-driven prescreening followed by expert validation supported efficient, scalable trial matching while preserving protocol fidelity. Yet, improved identification did not translate to proportional increases in consent or enrollment rates. To maximize clinical trial participation, AI tools like CTPM must be combined with expert review and targeted interventions that address downstream barriers to patient consent and enrollment.
Presentation numberPS2-02-15
The impact of ASCO/CCO guidelines on the use of adjuvant bone-modifying agents by specialized and general oncologists for elderly patients with breast cancer
Jennifer Caswell-Jin, Stanford University, Stanford, CA
N. Dalal1, H. Tang2, M. Reitsma3, J. Dickerson1, S. Phillips3, B. Staiger4, J. Goldhaber-Fiebert3, J. Caswell-Jin1; 1Division of Oncology, Stanford University, Stanford, CA, 2Center for Population Health Sciences, Stanford University, Stanford, CA, 3Department of Health Policy, Stanford University, Stanford, CA, 4School of Public Health, UC Berkeley, Berkeley, CA.
Background: In 2017, the American Society of Clinical Oncology (ASCO) and Cancer Care Ontario (CCO) jointly published guidelines recommending discussion of adjuvant bisphosphonate therapy for all postmenopausal patients with early-stage breast cancer deemed candidates for systemic therapy. We evaluated the impact of this guideline on the use of adjuvant bone-modifying agents (BMAs) among patients in SEER-Medicare to understand if 1) the guidelines were effective in changing prescribing patterns and 2) there was a difference in the effect of the guidelines on specialized vs generalist oncologists.Methods: We identified women aged 65-85 years with localized or regional breast cancer diagnosed between 2012 and 2018 and continuously enrolled in Medicare fee-for-service for at least 1 year after diagnosis. We excluded patients with missing hormone receptor (HR) or HER2 status, no claims for surgery, radiation, or systemic therapy after diagnosis, death within 6 months of diagnosis, BMA use before breast cancer diagnosis, and those without an identifiable primary oncologist. Primary endpoints were: 1) use of a BMA within 1 year of diagnosis and 2) choice of bisphosphonate vs denosumab. We used multivariate logistic regression to examine the correlation of key factors (age, comorbidity, race/ethnicity, the treating oncologist’s degree of breast cancer specialization) with each endpoint. We report odds ratios (OR) and 95% confidence intervals (CI) from the multivariate analyses. We defined an oncologist’s breast cancer specialization as the percentage of patients with breast cancer they treated, relative to the total SEER-Medicare patients with colon, lung, or breast cancer. We designate oncologists with the highest quartile of breast cancer cases as “specialized” and the other 75% as “generalists.”Results: The cohort included N=76,498 patients. BMA use increased in HR+ disease from 7.4% (95% CI 6.8-8.0%) in 2012 to 12.1% (11.5-12.7%) in 2018, while there was no change in usage in HR- disease (2.2% [1.3-3.1%] in 2012 and 2.2% [1.4-3.0%] in 2018). Specialized oncologists used less BMA overall than generalist oncologists (OR 0.84 [0.80-0.89]). This difference was driven by lower BMA use by specialists in localized HR+ disease (OR 0.79 [0.74–0.84]), with no significant difference in regional or HR- disease (OR 0.97 [0.88–1.08]). There was no clear effect of the guideline publication on the temporal trend of overall BMA use. However, we observed an increase in the selection of bisphosphonate over denosumab after the guideline publication, with the proportion using bisphosphonate increasing from 21.5% (19.8-23.3%) (2015-2016) to 36.1% (34.2-38.0%) (2017-2018). This shift post-guideline was more pronounced in the specialized oncologists (p for interaction = 0.003); the proportion using bisphosphonate (vs denosumab) increased from 20.7% (17.9-23.6%) to 40.7% (37.5-43.8%) in specialized oncologists, compared to an increase from 22.0% (19.8-24.1%) to 33.4% (31.1-35.7%) in generalist oncologists. Overall, BMA use was higher for HR+ vs HR- disease (OR 4.40 [3.82-5.07]), regional vs localized stage (OR 1.17 [1.10-1.24]), older vs younger patients (OR 1.10 [1.04-1.16] for 75-85 vs 65-74), and Asian vs White patients (OR 1.38 [1.23-1.55]), and lower for Black vs White patients (OR 0.63 [0.55-0.71]). Conclusions: Specialized oncologists used adjuvant BMAs less frequently in patients with lower-risk HR+ disease compared to generalist oncologists. They were also more sensitive to the guideline release, demonstrating greater uptake of the recommendation for bisphosphonate over denosumab. Broader alignment with specialist prescribing patterns could promote more efficient resource utilization across the Medicare population.
Presentation numberPS2-02-16
Can NHS PREDICT Guide Oncotype DX Use in ER+/HER2− Early Breast Cancer: A Multicentre Analysis
Sanjit Kumar Agrawal, Tata Medical Center, kolkata, India
S. K. Agrawal1, M. Nadkarni2, R. Ahmed1, R. Sarin3; 1Breast Oncosurgery, Tata Medical Center, kolkata, INDIA, 2Surgical Oncology, Kokilaben Dhirubahi Ambani Hospital and Medical Research Institute, Mumbai, INDIA, 3Surgical Oncology, Apollo Hospitals Indraprastha, Delhi, INDIA.
Background: In early-stage estrogen receptor-positive (ER+), HER2-negative breast cancer, adjuvant chemotherapy decisions are increasingly guided by the Oncotype DX (ODX) Recurrence Score (RS). However, the high cost of ODX limits its use in resource-constrained settings. NHS PREDICT, a validated clinical prognostic tool, is often used to estimate survival benefit from adjuvant therapy. This study aimed to assess the correlation between NHS PREDICT estimates and ODX RS, to evaluate whether NHS PREDICT can help identify patients who would benefit most from ODX testing.Methodology: This multicentric retrospective study included 320 patients with early-stage ER+/HER2− breast cancer who underwent ODX testing between January 2012 and May 2025 across 3 tertiary cancer centers in India. The study was approved by the Institutional Ethics Committee (Waiver No. 2025/TMC/350/IRB7). NHS PREDICT scores were calculated using the online PREDICT tool. Chemotherapy recommendations based on NHS PREDICT were derived via multidisciplinary team (MDT) discussions, while final decisions were based on ODX RS. Clinical data were retrieved from REDCap databases and institutional electronic records. Statistical analysis was performed using SPSS version 25.Results: The mean age was 57.5 ± 10.2 years, mean tumor size 2.57 ± 1.15 cm, Ki-67 24.0 ± 17.6%, ODX RS 17.9 ± 10.8, and NHS PREDICT chemotherapy benefit score 3.29 ± 2.35%. A weak, non-significant correlation was observed between ODX RS and NHS PREDICT benefit score (Pearson r = 0.060, p = 0.285).Concordance Between NHS PREDICT and ODX RS for Chemotherapy Decision-Making:
| Group | Total (N) | Concordant | Upgraded by RS | Downgraded by RS | |||||
| Overall | 320 | 179 (55.9%) | 55 / 231 (23.8%) | 71 / 89 (79.8%) | |||||
| Premenopausal Node-Negative | 66 | 40 (60.6%) | 22 / 60 (36.7%) | 4 / 6 (66.7%) | |||||
| Postmenopausal Node-Negative | 149 | 108 (72.5%) | 32 / 135 (23.7%) | 9 / 14 (64.3%) | |||||
| Postmenopausal Node-Positive | 105 | 46 (43.8%) | 1 / 36 (2.8%) | 58 / 69 (84.1%) |
Clinical risk and Recurrence Score (RS) were concordant in 55.9%. Concordance was highest in postmenopausal node-negative patients (72.5%) and lowest in postmenopausal node-positive patients (43.8%). RS upgraded 23.8% of clinically low-risk patients, highest in premenopausal node-negative cases (36.7%) and lowest in postmenopausal node-positive cases (2.8%). RS downgraded 79.8% of clinically high-risk patients, most notably in postmenopausal node-positive cases (84.1%). On a median follow-up of 29 months (IQR 24-33), there were 8 recurrences (4 local, 4 distant) and 6 deaths (2 cancer-related). The 5-year estimated distant disease-free survival was 96.5%, and cancer-specific survival was 98.9%.Conclusion: In this large multicentric Indian cohort, concordance between NHS PREDICT and RS for adjuvant chemotherapy decision-making was poor. NHS PREDICT frequently overestimated chemotherapy benefit, especially in postmenopausal node-positive patients, where 84% were downgraded by RS. Notably, in postmenopausal node-positive patients with low predicted benefit on NHS PREDICT, only 2.8% were upgraded by RS, suggesting that Oncotype DX testing may be safely omitted in this subgroup. These findings support a selective, cost-effective testing strategy in resource-limited settings
Presentation numberPS2-02-17
The High Unmet Need of Patients with Early Relapse After the KEYNOTE-522 Regimen – Evidence from the Neo-Real/GBECAM-0123 Study
Renata Colombo Bonadio, Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
R. Colombo Bonadio1, L. Lapuchesky2, M. C. Tavares3, N. C. Nunes4, L. Testa1, I. Salas2, M. Winocur2, F. C. Balint3, I. M. de Sousa3, M. O. Andrade5, M. C. Gouveia6, F. Madasi7, J. Bines7, R. P. Ferreira8, D. D. Rosa8, C. L. Santos9, M. R. Monteiro10, Z. S. de Souza11, D. Assad-Suzuki11, C. dos Anjos12, D. M. Gagliato13, A. U. Gomes13, B. M. Zucchetti6, A. Ferrari14, M. L. de Brito15, M. F. Monteiro16, P. A. Signorini17, N. J. Gomes18, G. G. Abuin2, S. Sanches3, M. Tavares19, P. M. Hoff1, M. Estevez-Diz20, R. Barroso-Sousa5; 1Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 2Oncology, SUMA (Grupo Cooperativa Argentino para el estudio y la investigación del Cáncer de Mama), Buenos Aires, ARGENTINA, 3Oncology, A.C.Camargo Cancer Center, São Paulo, BRAZIL, 4Oncology, Grupo Américas, Rio de Janeiro, BRAZIL, 5Oncology, Brasilia Hospital, Oncologia Américas, Brasília, BRAZIL, 6Oncology, Hospital 9 de Julho, Grupo Américas, São Paulo, BRAZIL, 7Oncology, Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, BRAZIL, 8Oncology, Hospital Moinhos de Vento, Porto Alegre, BRAZIL, 9Oncology, Instituto D’Or de Pesquisa e Ensino, Recife, BRAZIL, 10Oncology, Grupo Americas, Hospital Samaritano, São Paulo, BRAZIL, 11Oncology, Hospital Sírio-Libanês, Brasília, BRAZIL, 12Oncology, Hospital Sírio-Libanês, São Paulo, BRAZIL, 13Oncology, Hospital Beneficência Portuguesa, São Paulo, BRAZIL, 14Oncology, Hospital Santa Paula, Grupo Américas, São Paulo, BRAZIL, 15Oncology, Clínica AMO, Salvador, BRAZIL, 16Oncology, Instituto do Câncer do Ceará, Fortaleza, BRAZIL, 17Oncology, Centro Integrado de Pesquisa da Amazônia (CINPAM), Manaus, BRAZIL, 18Oncology, Hospital São Domingos, DASA Oncologia, São Luiz, BRAZIL, 19Oncology, Instituto D’Or de Pesquisa e Ensino, Salvador, BRAZIL, 20Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRAZIL.
The High Unmet Need of Patients with Early Relapse After the KEYNOTE-522 Regimen – Evidence from the Neo-Real/GBECAM-0123 Study Background: Triple-negative breast cancer (TNBC) is an aggressive subtype of tumor characterized by early disease recurrence, with most events occurring within the first two years after definitive treatment. With the adoption of the KEYNOTE-522 regimen—pembrolizumab (P) added to neoadjuvant chemotherapy (NAC)—we aimed to describe the patters and rates of early recurrence events in a real-world population treated with this regimen. Methods: The Neo-Real/GBECAM-0123 study is a real-world data analysis of patients with TNBC treated with P+NAC at sites in Brazil and Argentina since July 2020. The current analysis evaluated recurrence patterns following P+NAC. Early recurrence was defined as recurrence occurring during or within six months after completion of neoadjuvant/adjuvant therapy. Disease progression during NAC that precluded surgery was also classified as early recurrence. Recurrence occurring more than six months after therapy completion was considered late recurrence. Event-free survival (EFS) was calculated from diagnosis to either progression preventing surgery, recurrence, or death from any cause. Overall survival (OS) was calculated from diagnosis to death from any cause. Results: A total of 726 patients were included. The median age was 44 years (range 21-91). Most patients had stage II disease (70.5%), grade 3 tumors (68.9%), and Ki-67 index ≥50% (71.1%); 105 patients (14.5%) had a known pathogenic BRCA1/2 variant.After a median follow-up of 22 months, 74 patients (10.2%) experienced disease recurrence, and 8 patients (1.1%) died without recurrence. The 2-year EFS was 86.9%. Among those with recurrence, 50 (67.6%) were classified as early (including 4 patients with disease progression during P+NAC, which precluded surgery) and 24 (32.4%) as late recurrences.Early recurrence occurred in 2.1% of patients with pCR and in 15.2% of those with residual disease (P < 0.001). From another perspective, among patients with early recurrence after surgery (n = 46), 80.4% (n = 37) had residual disease at surgery, while 19.6% (n = 9) had achieved pCR.Most patients with early recurrence received adjuvant therapy. In patients with pCR, 8 out of 9 (88.9%) received adjuvant P. Among patients with residual disease, 32 out of 37 (86.5%) received adjuvant treatment, including P plus capecitabine in 19 (51.3%), P alone in 6 (16.2%), capecitabine alone in 6 (16.2%), P plus Olaparib in 1 (2.7%). 5 patients (13.5%) received no adjuvant therapy, mainly due to early relapse.Among patients with pCR, the main recurrence sites were the central nervous system (CNS) (33.3%), lymph nodes (22.2%), pleura (22.2%), and locoregional (14.3%). For those with residual disease, the main sites were locoregional (33.3%), lymph nodes (24.3%), CNS (21.6%), bones (21.6%), lung (13.5%), and liver (13.5%).Post-recurrence deaths occurred in 18 patients with early relapse and 7 with late relapse. The 2-year OS rate in the overall cohort was 95.6%. Among patients with recurrence, the 2-year OS rate was 62.3% for those with early relapse and 91.0% for those with late relapse. Discussion: The majority of recurrences after the KEYNOTE-522 regimen occurred early. These patients are resistant to four chemotherapeutic agents and an immune checkpoint inhibitor (ICI) and face markedly poor survival outcomes. Our findings underscore a critical unmet need for effective therapeutic options in this population, which is often excluded from clinical trials investigating first-line regimens, including studies of antibody-drug conjugates combined with ICIs.
Presentation numberPS2-02-18
Real-world epidemiologic and biological characterisation of breast cancer in Algeria: A multicenter real world data study on 6044 patients
Adda Bounedjar, University of Blida 1, Blida, Algeria
A. Bounedjar1, M. A. Melzi1, H. Mahfouf2, A. Dib3, C. Sedkaoui4, H. Djeddi5, E. Kerboua2, W. Benbrahim6, T. Filali7, H. Zidane8, F. Seghier9, D. Yekrou10, F. Bereksi11, A. Bensalem7, B. Larbaoui11, A. Bousahba11, M. Abada12, R. Miloudi12, M. A. Bahi13, H. Fizi14, A. Djellaoui15, F. Benmhidi15, K. Kouidri16, N. Boualaoui17, Z. Benlahreche18, M. Oukkal2; 1Laboratoire de Recherche en Cancérologie, University of Blida 1, Blida, ALGERIA, 2Faculty of medecine, University of Algiers 1, Algiers, ALGERIA, 3Faculty of medecine, University of Setif, Setif, ALGERIA, 4Faculty of medecine, University of Tizi Ouzou, Tizi Ouzou, ALGERIA, 5Faculty of medecine, University of Annaba, Annaba, ALGERIA, 6Faculty of medecine, University of Batna, Batna, ALGERIA, 7Faculty of medecine, University of Constantine, Constantine, ALGERIA, 8Faculty of medecine, University of Mostaganem, Mostaganem, ALGERIA, 9Faculty of medecine, University of Blida 1, Blida, ALGERIA, 10Faculty of medecine, University of Sidi Belabes, Sidi Belabes, ALGERIA, 11Faculty of medecine, University of Oran, Oran, ALGERIA, 12Medical oncology department, Public hospital of Ain Defla, Ain Defla, ALGERIA, 13Medical oncology department, Public hospital of El Oued, Oued Souf, ALGERIA, 14Medical oncology department, Anti cancer center of Ouergla, Ouergla, ALGERIA, 15Medical oncology department, Public hospital of Medea, Medea, ALGERIA, 16Medical oncology department, Anti cancer center of Adrar, Adrar, ALGERIA, 17Medical oncology department, Public hospital of Aflou, Aflou, ALGERIA, 18Faculty of medecine, University of Laghouat, Laghouat, ALGERIA.
Background : Breast cancer is the most frequently diagnosed cancer among women in Algeria, accounting for 46.0% of all female malignancies, with 13,161 new cases reported in 2022 according to the National Network of Cancer Registries. This trend is consistent with global estimates from GLOBOCAN, which also identify breast cancer as the leading cause of cancer among women worldwide. Despite its public health burden, national-level epidemiologic and clinicopathologic data on breast cancer in Algeria remain scarce. Most available studies are single-center or regional, limiting their generalizability. Furthermore, North African populations, including Algerian women, have distinct lifestyle, reproductive, and sociodemographic characteristics compared to Western populations, which may influence disease presentation and biology. To address this gap, we conducted a large, retrospective, multicenter study aiming to describe the demographic, clinical, and pathological characteristics of Algerian breast cancer patients diagnosed in 2024, based on real-world data from major oncology centers across the country. Patients and Methods : We conducted a national, retrospective, multicenter study involving 23 oncology centers across Algeria. Eligible participants were female patients diagnosed and treated for breast cancer between January 1 and December 31, 2024. Data were collected from medical records as part of real-world clinical practice. Variables extracted included age at diagnosis, menopausal status, family history of cancer, clinical stage at presentation, histological type and grade, estrogen receptor (ER), progesterone receptor (PR), HER2 status, Ki67 index, and molecular subtype classification. All data were anonymized and centralized in a secure national database. Descriptive analyses were planned to summarize the demographic, clinical, and pathological characteristics of the study population. Results : A total of 6,044 women with breast cancer diagnosed in 2024 were included from 23 oncology centers across Algeria. The median age at diagnosis was 52 years (range: 20-86), and 60% of patients were postmenopausal. The majority of tumors were detected through self-examination (67.72%), while screening accounted for only 4.17% of cases. The left breast was affected in 48.99% of cases, the right breast in 48.25%, and bilateral involvement was observed in 2.76%. Invasive carcinoma of no special type was the predominant histology (79.90%). Grade III tumors (SBR) accounted for 19.33% of cases. Molecular subtypes were distributed as follows: luminal B (49.33%), luminal A (18.11%), HER2-positive (20.70%), and triple-negative (11.86%). Tumor size distribution showed 13.63% of cases were T1. Advanced tumors (non-inflammatory T4a/b/c) were found in 21.56%, and inflammatory breast cancer (T4d) in 4.74%. Node-negative disease (N0) was present in 36.87%, while N3 involvement was seen in 3.54%. Distant metastases at diagnosis were documented in 15% of patients. The median time to initial treatment was 50.5 days. Conclusion: This large, retrospective multicenter study provides the first national real-world overview of breast cancer in Algeria. The majority of patients presented with luminal subtypes, but a substantial proportion had locally advanced (T4: 26.3%) or metastatic disease (15%) at diagnosis. Most tumors were detected through self-examination, while organized screening remained limited. The observed diagnostic and treatment delays, as well as the frequency of advanced-stage presentation, highlight the urgent need to strengthen early detection programs and promote timely access to care across the country. These findings serve as a foundation for future national strategies aimed at improving breast cancer outcomes in Algeria.
Presentation numberPS2-02-19
Trends and Disparities in Heart Failure Mortality Among US Adult Females with Breast Cancer from 1999 to 2020: A 22-Year Retrospective Study
Mercy C Anyanwu, Icahn School of Medicine at Mount Sinai, New York, NY
M. C. Anyanwu1, A. Raza2, A. Nawaz3, U. Khan4; 1Dubin Breast Cancer, Icahn School of Medicine at Mount Sinai, New York, NY, 2Medical Sciences, Services Institute of Medical Sciences, Lahore, Pakistan, Pakistan, PAKISTAN, 3Medical Sciences, Pakistan Atomic Energy Commission Hospital, Islamabad., Islamabad, PAKISTAN, 4Cardiovascular diseases, University of Maryland School of Medicine, Baltimore, Baltimore,, MD.
Background: Advances in breast cancer treatment have improved survival outcomes, with the 5-year survival rates approximately 99% for early-stage breast cancer patients. Despite these therapeutic achievements, cardiovascular complications, particularly heart failure (HF), have emerged as a leading cause of non-cancer mortality among breast cancer survivors aged 65+, thereby compromising long-term survival and posing a greater mortality risk than cancer itself. We aim to analyze trends in HF-related mortality among women with breast cancer in the U.S. over two decades (1999-2020) and highlight key demographic and regional disparities. Methods: We used the CDC WONDER (Wide-ranging ONline Data for Epidemiologic Research) mortality database to extract age-adjusted mortality rates (AAMRs) per 100,000 US women aged ≥25 years for breast cancer and HF mortality from 1999 to 2020. The Joinpoint Regression Program calculated the average annual percentage change (AAPC) in AAMRs. Results: From 1999 to 2020, HF caused 56,006 deaths among U.S. women with breast cancer, with a decline over time (AAPC: -1.16%; 95% CI: -1.50 to -0.86). The overall AAMR was 1.94 (1.93 – 1.96). Older adults (≥65 years) showed a significantly higher AAMR [8.88 (8.80 – 8.96)] with an AAPC of -1.17% (-1.45 to -0.93) than younger adults (25-64 years) [0.26 (0.25 – 0.27)] with an AAPC of -1.24% (-2.37 to -0.36). Among the racial and ethnic groups, the highest AAMR was observed in non-Hispanic (NH) Blacks [2.37 (2.31 – 2.43)], followed by NH Whites [2.03 (2.01 – 2.05)], Hispanics [0.98 (0.94 – 1.03)], and NH Asians or Pacific Islanders [0.66 (0.61 – 0.72)]. NH Blacks showed the highest mortality burden and a slight increase in AAMRs [AAPC: 0.15% (-0.48 to 0.66)]. NH Asians or Pacific Islanders showed the lowest mortality burden with the highest decline in AAMRs [AAPC: -1.47% (-2.71 to -0.28)], followed by Hispanics or Latinos (-1.27%) and NH Whites (-1.01%). Regionally, the Midwest showed the highest AAMR [2.25 (2.21 – 2.29)], followed by the West [1.95 (1.92 – 1.99)], the Northeast [1.85 (1.81 – 1.88)], and the South [1.81 (1.78 – 1.83)]. The Northeast showed the highest decline in AAMRs [AAPC: -1.48% (-2.06 to -0.98)], and the South showed the lowest [-0.59% (-0.99 to -0.20)]. Women in non-metropolitan areas had higher AAMRs [2.36 (2.32 – 2.41)] than those in metropolitan areas [1.87 (1.85 – 1.88)]. Metro and non-metropolitan regions showed a similar decline in AAMRs over the study period (-1.05% vs. -1.20%). Conclusion: HF-related mortality among U.S. female breast cancer survivors has declined over the past two decades; however, older adults, NH Blacks, patients residing in the Midwest, and non-metropolitan areas experienced higher mortality burdens, highlighting the need for targeted interventions that incorporate risk stratification, integrated cardiovascular surveillance and equity centered interventions into survivorship planning. Ultimately, addressing demographic and regional disparities is essential to improving long-term survival outcomes for breast cancer survivors.
Presentation numberPS2-02-20
Treatment patterns and clinical outcomes following progression on first-line ET + CDK4/6i among patients with HR+/HER2− metastatic breast cancer (mBC) in the US
Virginia Kaklamani, UT Health San Antonio, San Antonio, TX
V. Kaklamani1, S. Valliant2, S. Hunter3, O. Tymejczyk4, A. Yu5, P. Earla6, S. Mehta7, E. Pang8; 1Medicine, UT Health San Antonio, San Antonio, TX, 2Global Oncology HEOR, V&E, Daiichi Sankyo, Inc., Basking Ridge, NJ, 3RWE, Daiichi Sankyo, Inc., Basking Ridge, NJ, 4Global Oncology RWE Generation, Daiichi Sankyo, Inc., Basking Ridge, NJ, 5Global Scientific & Medical Affairs Oncology, Merck & Co., Inc, Rahway, NJ, 6Outcomes Research Value & Implementation (V&I), Merck & Co., Inc., Rahway, NJ, 7HEOR, Daiichi Sankyo, Inc., Basking Ridge, NJ, 8Global Oncology Medical Affairs, Daiichi Sankyo, Inc., Basking Ridge, NJ.
Background Endocrine therapy (ET) in combination with a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) is a preferred first-line (1L) treatment for patients (pts) with HR+/HER2− mBC. However, ET resistance frequently emerges after 1L treatment, with many pts receiving subsequent chemotherapy or other targeted regimens. This study aimed to assess treatment patterns and clinical outcomes in pts with HR+/HER2− mBC that progressed on 1L ET + CDK4/6i in real-world (rw) practice settings. Methods This was a noninterventional, retrospective, observational cohort study using the US-based Flatiron Health Enhanced Datamart. Adult pts with HR+/HER2− mBC (with no other actionable genomic alterations [ESR1, PIK3CA, AKT1, PTEN, or gBRCA]) that progressed on 1L ET + CDK4/6i were included if they initiated a second-line (2L) therapy between Jan 2017 and Sep 2024 with ≥90 d of potential follow-up. Pt demographics, clinical characteristics, and treatment utilization were assessed descriptively. Treatment data were recorded as documented by physicians and may reflect off-label use. The primary (rw progression-free survival [rwPFS]) and secondary (rw overall survival [rwOS], rw time to discontinuation or death [rwTTD/D], and rw time to next treatment or death [rwTTNT/D]) endpoints were assessed using Kaplan-Meier methods. Analyses were performed in all pts, by 2L regimen, and by time to progression on 1L treatment. Results 1415 pts with HR+/HER2− mBC were included (median age, 65 y; 66.2% were White). Of these, 57.3% (n=811) had disease progression on ET + CDK4/6i within 12 mo of 1L initiation (early progression; 33.3% in ≤6 mo); 25.5% had disease progression in 12 to 24 mo and 17.2% within >24 mo. In the 2L, 63.6% of pts received an ET-based regimen, 27.1% received chemotherapy (CT; monotherapy, 23.8%; multiagent, 3.3%), and 9.3% received other regimens (including AKTi, PARPi, and the ADC sacituzumab govitecan or trastuzumab deruxtecan). Pts with early vs later times to disease progression on 1L ET + CDK4/6i used 2L CT more frequently (40.6% [≤6 mo] vs 14.0% [>24 mo]); the opposite was true for 2L ET (52.2% [≤6 mo] vs 75.3% [>24 mo]). With a median follow-up of 14.6 mo, the 2L median rwPFS ranged from 4 to 8 mo and median rwOS ranged from 14 to 32 mo; the shortest estimates were among pts on 2L CT and with early disease progression on 1L ET + CDK4/6i (Table). Conclusions In this study of pts with HR+/HER2− mBC that progressed on 1L ET + CDK4/6i, over half had 1L disease progression within 12 mo and about one-third received CT in a subsequent line. The median rwPFS on 2L treatments was shorter among those who were using CT in 2L or had early progression (<12 months) on 1L ET + CDK4/6i, highlighting the emergence of ET resistance and a need for more effective therapeutic alternatives to improve pt outcomes.
Presentation numberPS2-02-21
Real-world outcomes of CDK4/6 inhibitors in patients with metastatic invasive lobular carcinoma: a retrospective single-center study
Anastasia Danilova, Moscow City Oncology Hospital 62, Stepanovskoe, Russian Federation
A. Danilova1, P. Shilo2, I. Nigmatullina1, D. Stroyakovskiy1; 1Medical oncology, Moscow City Oncology Hospital 62, Stepanovskoe, RUSSIAN FEDERATION, 2Medical oncology, Lahta clinic, Saint Petersburg, RUSSIAN FEDERATION.
Background: Invasive lobular carcinoma (ILC) is a distinct histological subtype of hormone receptor-positive/HER2-negative (HR+/HER2−) breast cancer, accounting for up to 15% of cases. Despite its unique clinical behavior, patients with ILC have been underrepresented in pivotal CDK4/6 inhibitor trials, and real-world data on treatment outcomes in this population remain limited.Methods: We conducted a retrospective study of patients with metastatic ILC treated with CDK4/6 inhibitors at City Clinical Hospital No. 62 (Moscow) from 2015 to 2023. Among 803 patients with HR+/HER2− metastatic breast cancer, 141 had confirmed ILC. Demographic, clinical, and treatment-related data were extracted from medical records. Survival outcomes were estimated using the Kaplan-Meier method, and comparisons between subgroups were performed using log-rank tests.Results: The median age was 60 years, and the median follow-up was 46 months. Most patients had grade 2 tumors (82.9%) and high ER expression (Allred 8 in 72.3%). At the time of CDK4/6 inhibitor initiation, 64.7% received treatment in the first line. The most frequently used agents were palbociclib (49.3%) and ribociclib (45.7%), while abemaciclib was prescribed in 5% of cases. The median overall survival (OS) in the full ILC cohort was 49 months (95% CI: 46.1-51.9). When stratified by CDK4/6 inhibitor, the median OS was 50 months for palbociclib (95% CI: 46.4-53.6), 44 months for ribociclib (95% CI: 32.9-55.1), and not reached for abemaciclib, with no statistically significant differences between groups (log-rank p=0.276). The median progression-free survival (PFS) was 30 months overall (95% CI: 22.2-37.8), with 32 months for palbociclib, 30 months for ribociclib, and 17 months for abemaciclib (p=0.156). Subgroup analyses showed no significant differences in OS by age (<60 vs ≥60; p=0.620), presence of bone (p=0.316), liver (p=0.854), or lung metastases (p=0.410). The type of endocrine backbone (aromatase inhibitor, fulvestrant, or exemestane) was not associated with differences in OS (p=0.778). Patients treated in the first line had longer OS (49 months) compared to those treated after progression on prior therapies (30 months), though the difference did not reach statistical significance.Conclusions: In this real-world cohort of patients with metastatic ILC, CDK4/6 inhibitors demonstrated clinically meaningful survival outcomes consistent with previously reported results in broader HR+/HER2− populations. No significant differences in OS or PFS were observed between individual CDK4/6 inhibitors or across clinical subgroups. These findings support the use of CDK4/6 inhibitors in ILC, while highlighting the need for prospective studies to better define optimal treatment strategies in this distinct population.
Presentation numberPS2-02-22
Impact of Body Mass Index on the Efficacy and Safety of Adjuvant Abemaciclib in a Real-World Brazilian Cohort of Patients With High-Risk Early Breast Cancer
Monique Celeleste Tavares, Hospital A.C Camargo Cancer Center, São Paulo, Brazil
C. Pereira dos Santos, M. Celeleste Tavares, C. Campanholo Marques, S. Moraes Sanches; Oncology, Hospital A.C Camargo Cancer Center, São Paulo, BRAZIL.
Title:Impact of Body Mass Index on the Efficacy and Safety of Adjuvant Abemaciclib in a Real-World Brazilian Cohort of Patients With High-Risk Early Breast CancerBackground:Obesity is an increasing concern in breast cancer, influencing both prognosis and treatment tolerance. While abemaciclib has demonstrated efficacy in high-risk HR (hormone receptor) +/HER2- patients, real-world evidence regarding its safety and efficacy across BMI (body mass index) categories remains limited.Methods:We conducted a retrospective and prospective, observational study involving 146 patients who received adjuvant abemaciclib combined with endocrine therapy between January 2021 to June 2025 at a comprehensive cancer center. Patients were stratified by BMI (kg/m²) as obese (≥30), overweight (25 ≤ BMI < 30), and non-overweight (<25). We assessed treatment-related toxicities, dose reductions, pathological response to neoadjuvant chemotherapy (RCB), recurrence, and disease-free survival. Statistical analyses included chi-square tests, Spearman correlation, Kaplan-Meier survival curves, and Cox regression.Results:Obese patients experienced higher rates of dose reduction (72%) compared to non-obese (47%) and overweight patients (29%), although the difference did not reach statistical significance (p = 0.075). Regarding neoadjuvant response, non-overweight patients achieved more RCB-0 (complete response), while 30.8% of obese patients had RCB-III (high residual disease). Although no significant association was found between BMI and RCB (p = 0.135), a clinical trend was observed. Larger tumors (>5 cm) were more frequent in obese patients (23.1%) compared to non-obese (14.3%).Anemia showed a positive correlation with BMI (Spearman p = 0.043), with a higher frequency in obese patients (27.3%) versus non-overweight (15.2%). No statistically significant associations were observed between BMI and other toxicities (diarrhea, neutropenia, transaminase elevation, arthralgia).Four recurrences were documented: three in obese and one in a non-overweight patient. Kaplan-Meier curves suggested a trend toward longer disease-free survival among obese patients (median: 641 days) compared to non-overweight patients (237 days). However, this finding did not reach statistical significance (Cox regression HR = 2.6; p = 0.235). ANOVA for time to recurrence showed a significant difference between BMI groups (p = 0.003), favoring delayed recurrence in obese patients.Conclusion:In this real-world Brazilian cohort, obesity was associated with higher rates of dose reduction due to toxicity, lower pathological response to neoadjuvant chemotherapy, and larger tumors. Paradoxically, obese patients demonstrated longer disease-free survival, a finding consistent with the “obesity paradox” described in the literature. These results highlight the need for further research into the complex role of body composition in HR+/HER2- breast cancer treated with CDK4/6 inhibitors.
Presentation numberPS2-02-23
Clinicopathological profile and outcomes in young Indian patients with breast cancer – A single-center experience
Tanay Shah, HCG Aastha Cancer Center, Ahmedabad, India
M. Shah, S. Patel, T. Shah, D. G. Vijay, S. Alurkar; Medical oncology, HCG Aastha Cancer Center, Ahmedabad, INDIA.
Background:Breast cancer has emerged as the most prevalent malignancy among women in India. Its epidemiological pattern in the Indian subcontinent differs significantly from that seen in Western countries. One of the most notable differences is the earlier age of onset in Indian women. In young women, breast cancer poses unique challenges due to its aggressive biological behaviour, concerns regarding fertility, and worse outcomes. This study aims to evaluate the clinicopathological characteristics, treatment approaches, and clinical outcomes of patients aged 40 years or younger, treated at a tertiary cancer center.Methods:We retrospectively analyzed the records of 102 breast cancer patients aged ≤40 years, presented between January 2023 and February 2025 at HCG Aastha cancer center Ahmedabad. Data were collected on demographics, family history, genetics, tumor histology, staging, receptor status, treatment modalities, and recurrence. Results:The median age was 36 years (range 21 to 40 years). 4.9% of the patients were nulliparous, and 75.5% had no family history of malignancy. Genetic testing was done in 40 patients (39.2%). Among them, results were positive in 19 patients (18.6%), negative in 16 patients (15.7%), and showed variants of uncertain significance (VUS) in 5 patients (4.9%). Among these, BRCA mutations were predominant. In 51% of patients, genetic testing was advised but, they did not undergo the testing. Histology revealed Invasive Ductal Carcinoma (IDC) in 97 (95.1%) patients. Imaging for diagnosis primarily included bilateral mammography (79.4%). MRI and PET-CT were used in selected cases. Stage II was the most common stage at diagnosis, observed in 51 patients (50%), followed by Stage IV in 20 patients (19.6%). Hormone receptor positive, HER2neu negative was observed in 42 patients (41.1%), while both hormone receptor and HER2neu positivity were seen in 31 patients (30.4%). HER2neu positivity alone was noted in 9 patients (8.8%), triple-negative breast cancer (TNBC) was found in 15 patients (14.7%). Mammogram showed mainly dense breasts, well circumscribed and multicentric lesions. Surgical management was offered to the majority of patients who did not present with Stage IV disease. Among them, breast-conserving surgery (BCS) was performed in 34 patients (33.3%), while modified radical mastectomy (MRM) was performed in 33 patients (32.4%), indicating a nearly equal distribution between the two surgical approaches. Neoadjuvant chemotherapy (NACT) was given to 28 patients (27.4%), of whom 5 achieved a pathological complete response (pCR). Most patients remained on follow-up, with 10.8% experiencing disease recurrence. Among these, sites of recurrence included bone-only (1.9%), brain-only (1.9%), visceral-only (1.9%), and combined bone and visceral metastases (4.9%). Nearly half of the patients (47.1%) had no recurrence, while 40.2% were lost to follow-up. Recurrences were predominantly seen in those initially diagnosed with Stage II and III disease.Conclusion:Young women (</= 40 years) with breast cancer present with stage II-III disease, multicentric lesions that can be picked up on digital mammography most of the times. Most common subtype is hormone positive breast cancer, and 18% were deteced with germline mutations, BRCA being the most common. Recurrence was observed even in early-stage cases, underscoring the need for vigilant follow-up. Multidisciplinary care, including routine genetic counselling, remains essential. Future research should prioritize fertility preservation, psychosocial support, and long-term outcomes in this high-risk population.
Presentation numberPS2-02-24
Comparative Effects of Semaglutide vs Tirzepatide on Weight Change and Cardiovascular Risk in Breast Cancer Survivors
Jasmin Hundal, Cleveland Clinic, Clevelan, OH
J. Hundal1, D. Parekh2, A. Harisingani3, A. Abushamma4, Omer Ashruf, J. Sukumar5; 1Taussig Cancer Institute, Cleveland Clinic, Clevelan, OH, 2Internal Medicine, SUNY Upstate Medical Univeristy, Syracuse, NY, 3Internal Medicine, Loyola Medicine MacNeal Hospital, Berwyn, IL, 4Internal Medicine, Akron General Medical Ctr, Akron, OH, 5Hematology and Oncology, MD Anderson Cancer Institute, Houston, TX.
Background:Obesity and cardiometabolic dysfunction are common in breast cancer survivors and linked to higher treatment toxicity, cardiovascular morbidity, and worse outcomes. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), including semaglutide and Tirzepatide, show cardiometabolic benefits in non-cancer populations, but their impact in breast cancer survivors remains unclear. This study compared the effects of semaglutide and Tirzepatide versus metformin on weight trajectory and major adverse cardiovascular events (MACE) in a real-world oncology cohort.Methods:We conducted a retrospective cohort study using the TriNetX Analytics Research Network, a federated, de-identified electronic health record database. Female breast cancer patients with obesity and/or type 2 diabetes mellitus who were prescribed semaglutide (n=3,009) or Tirzepatide (n=1,652) from January 1, 2018, to June 1, 2025, were identified and matched 1:1 to metformin users without GLP-1 RA exposure using propensity scores. Baseline characteristics included demographics, comorbidities, cancer therapies, and cardiometabolic risk factors. Matching was performed using age, sex, race, BMI, hemoglobin A1c, and comorbidities. Outcomes included weight change at 12 and 24 months, and incidence of MACE, defined as myocardial infarction, stroke, or cardiovascular death. Cox proportional hazards models estimated hazard ratios (HRs) and 95% confidence intervals (CIs).Results: At 12 and 24 months, both semaglutide and Tirzepatide groups demonstrated significantly greater weight loss compared to matched metformin users (Table 1). Semaglutide was associated with a significant reduction in MACE versus metformin (HR 0.78, p=0.032), including MI (HR 0.68, p=0.045), with favorable trends in cardiovascular death (HR 0.75, p=0.07) and stroke (HR 0.81, p=0.18). Tirzepatide demonstrated more pronounced cardioprotective effects, reducing MACE (HR 0.63, p=0.001), MI (HR 0.52, p=0.004), and cardiovascular death (HR 0.61, p=0.033), with a trend toward stroke reduction (HR 0.72, p=0.09). Conclusions:In this real-world analysis of breast cancer patients with cardiometabolic comorbidities, both semaglutide and Tirzepatide led to clinically meaningful and sustained weight loss at 12 and 24 months and reduced cardiovascular risk, with Tirzepatide showing the greatest benefit. Our findings underscore the potential of GLP-1 receptor agonists to improve cardiometabolic health, a key modifiable risk factor in breast cancer survivors, by enhancing weight and cardiovascular outcomes. These results support the role of GLP-1 receptor agonists as adjuncts to lifestyle interventions in survivorship care. Prospective trials in breast oncology populations are needed to confirm these findings and guide the integration of strategies for cardiovascular, metabolic, and cancer risk reduction.
| Treatment | Baseline Weight (lbs) Mean +/-SD | 12-Month Weight (lbs) Mean +/-SD | 24-Month Weight (lbs) Mean +/-SD | Weight Loss at 12 months (lbs) | Weight Loss at 24 months (lbs) |
| Semaglutide | 204 ± 45.4 | 195.2 ± 45.0 | 199.4 ± 45.2 | 8.8 | 10.7 |
| Matched Metformin (Semaglutide_ | 198 ± 45.7 | 195.2 ± 45.0 | 192.2 ± 44.6 | 4.6 | 5.8 |
| Tirzepatide | 211 ± 44.4 | 200.3 ± 45.1 | 198.8 ± 45.6 | 10.7 | 12.2 |
| Matched Metformin (Tirzepatide) | 203 ± 46.1 | 199.4 ± 45.2 | 197.9 ± 44.9 | 3.6 | 5.1 |
Presentation numberPS2-02-25
Centering the Patient Experience within TNBC Care and Fostering Equity in the Hispanic and Latina Populations
Tariqa Ackbarali, Medlive, Lake Worth, MA
T. Ackbarali1, L. Braithwaite2, C. Tellez3, S. Tolaney4; 1Hematology/Oncology Team, Medlive, Lake Worth, MA, 2Cancer Center, University of Wisconsin-Carbone Cancer Center, Madison, WI, 3Hispanic Breast Cancer Clinic, Northwestern University, Chicago, IL, 4Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
BACKGROUND: In recent years, TNBC has transformed from a difficult-to-treat malignancy with few effective treatment options into an active area of research, where new treatments are emerging each year. As novel treatments are established in advanced disease settings, and the focus of research turns to improving options in earlier stages of disease, it is important that oncologists remain up to date on the current and future care of patients with TNBC. This is particularly important for Hispanic and Latina patients, who face numerous disparities in treatment access and quality of care. A tethered educational initiative was designed to ensure that clinicians and patients/caregivers stay up-to-date on current and emerging TNBC treatments, disparities in Hispanic/Latina populations, and strategies to improve quality of care. METHODS: Two, interactive, video-based programs were designed for patients/caregivers, and clinicians in collaboration with Surviving Breast Cancer and Salud America. The programs were hosted on Medlive.com from April 2024 to June 2025. Patients were interviewed and their real-world stories embedded within the program. We report on an analysis of patient outcomes and the intersection of patient and clinician perspectives, including behavioral impact, and qualitative insights. RESULTS: To date, more than 2,273 clinicians have engaged with the education program and over 70,000 views via social media of the patient/caregiver program. Of participating patients/caregivers, 80% were non-White (50% Hispanic/Latina) and 78% were people diagnosed with breast cancer, 22% of whom had TNBC. Following the program, 82% felt confident discussing a treatment plan with their healthcare team, and 71% were likely to consider participating in a clinical trial. Qualitative data elucidating specific intended changes and patient experiences will be shared. Patients/caregivers prioritized community-centric resources more than HCPs recognized. Ethnicity-specific barriers were felt more strongly by patients. Of the participants in the clinician program, 80% were physicians, 76% of whom noted their specialty as oncology. Increased recognition (42%) of ADCs as the preferred second-line option for biomarker-negative TNBC, reflected by a rise in ADC selection post-activity. Improved understanding (47%) of immune checkpoint inhibitor toxicities, though some learners may still underestimate long-term endocrine-related side effects. Modest gains in awareness of racial disparities in TNBC, suggesting clinicians recognize inequities but may need further education on actionable solutions. CONCLUSIONS: The disparities highlight the importance of culturally attuned approaches that extend education and support to familial/social networks, which are often critical for patients from Hispanic and Latina populations. Ethnicity-specific barriers highlights a disconnect in recognizing the importance of peer representation and cultural similarity within support networks, which can greatly affect patient comfort and outcomes. These findings underscore a notable disconnect between what patients/caregivers prioritize and what clinicians perceive as their challenges. Patients tend to emphasize the importance of community and cultural representation in support systems, while HCPs focus on logistical barriers, such as language services. Bridging this gap requires a dual focus: addressing systemic barriers while fostering culturally tailored support networks that resonate with patients’ lived experiences. Support was provided by an independent educational grant from Gilead Sciences, Inc.
Presentation numberPS2-02-26
Treatment patterns and prognosis of patients with HER2-negative early breast cancer and germline pathogenic variants in BRCA1, BRCA2, ATM, CHEK2 and PALB2
Vishnu Prasath, The Ohio State University College of Medicine, Columbus, OH
V. Prasath1, A. N. Riner2, J. Kim3, A. Clark2, B. Slover3, R. Wesolowski1, J. C. Kai1, S. Jhawar4, C. Spears5, B. A. Oppong2, S. D. Sardesai1; 1Medical Oncology, The Ohio State University College of Medicine, Columbus, OH, 2Surgery, The Ohio State University College of Medicine, Columbus, OH, 3Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, 4Radiation Oncology, The Ohio State University College of Medicine, Columbus, OH, 5Medical Genetics, The Ohio State University College of Medicine, Columbus, OH.
Introduction: There is conflicting evidence on the prognosis of patients with HER2-negative early breast cancer (HER2- eBC) and germline pathogenic variants (PV) including BRCA1, BRCA2, PALB2, ATM, and CHEK2. Studies are limited by marked heterogeneity and a small number of evaluable patients. Here, we present treatment patterns and prognosis in hereditary breast cancer(BC) susceptibility gene mutation carriers with HER2- eBC from CancerLinQ, a health technology platform comprised of de-identified real-world data from over 2.5 million patients across the United States. Methods: Patients with stage I-III HER2- eBC diagnosed between 2002-2023 were identified in CancerLinQ. Those with germline PVs in BRCA1, BRCA2, PALB2, ATM, and CHEK2 constituted the mutation carrier (MC) group while those with negative genetic testing made up the non-MC group. Demographics, pathology findings, genomic testing, and cancer treatment data were extracted. Patients with metastases within 9 months of diagnosis, and patients without stage information, and those without germline genetic testing were excluded. The primary outcome was recurrence free survival (RFS). Secondary objectives included overall survival (OS) and utilization rate of contralateral prophylactic mastectomy (CPM) in each group. RFS and OS were presented with Kaplan-Meier curves and were analyzed with the Cox proportional hazard regression method. Results: 1105 patients with germline PVs in BRCA1 (360,32.6%), BRCA2 (461,41.7%), PALB2 (76,6.9%), ATM (67,6.1%), and CHEK2 (91,8.2%) constituted the MC cohort; the non-MC cohort included 2129 patients. The MC cohort had a younger age at diagnosis and a higher rate of triple negative and nodal disease (p<0.05). The MC group also received neoadjuvant chemotherapy more frequently and had a higher pathologic complete response rate. The adjusted RFS was improved for the non-MC group (HR 0.84,p=0.019) in comparison to the MC cohort; however, there was no statistically significant difference in OS between groups or by gene mutation. CPM use was more common in patients younger than 50 years and differed by gene mutation (p<0.05). CPM vs breast conserving therapy or therapeutic mastectomy showed improved adjusted RFS (p=0.04) and OS (p=0.007) in the MC cohort. Black patients demonstrated worse OS (HR 1.3,p=0.005) regardless of MC status. Conclusions: This is one of the largest real-world analyses of hereditary BC susceptibility gene MCs with HER2- eBC. The MC group reported worse RFS. CPM use was more common in younger patients and in those with PVs in BRCA1/2. CPM use was independently associated with improved OS in MC group. This highlights the need for timely genetic testing and identification of at-risk patients who will benefit from personalized treatment approaches. Table 1:
| Characteristic | Overall(N = 3,234) | Mutation Carriers (N = 1,105) | Non-Mutation Carriers(N = 2,129) | p-value | |||||
| Median Age(Min, Max) | 50 (24, 96) | 49 (24, 88) | 51 (25, 96) | <0.001 | |||||
| Race | 0.002 | ||||||||
| Black or African American | 318(9.8%) | 121(11.0%) | 197(9.3%) | ||||||
| Other | 393(12.2%) | 127(11.5%) | 266(12.5%) | ||||||
| Unknown | 462(14.3%) | 125(11.3%) | 337(15.8%) | ||||||
| White | 2,061(63.7%) | 732(66.2%) | 1,329(62.4%) | ||||||
| N stage | <0.001 | ||||||||
| N0 | 1,898(58.7%) | 614(55.6%) | 1,284(60.3%) | ||||||
| N1 | 747(23.1%) | 303(27.4%) | 444(20.9%) | ||||||
| N2 | 172(5.3%) | 71(6.4%) | 101(4.7%) | ||||||
| N3 | 73(2.3%) | 25(2.3%) | 48(2.3%) | ||||||
| Unknown | 344(10.6%) | 92(8.3%) | 252(11.8%) | ||||||
| Stage | <0.001 | ||||||||
| Stage1 | 1,518(46.9%) | 433(39.2%) | 1,085(51.0%) | ||||||
| Stage2 | 1,211(37.4%) | 453(41.0%) | 758(35.6%) | ||||||
| Stage3 | 505(15.6%) | 219(19.8%) | 286(13.4%) | ||||||
| Surgical Procedure | <0.001 | ||||||||
| BCT | 1,652(60.0%) | 413(48.3%) | 1,239(65.3%) | ||||||
| CPM | 510(18.5%) | 262(30.6%) | 248(13.1%) | ||||||
| Therapeutic Mastectomy Only | 590(21.4%) | 180(21.1%) | 410(21.6%) |
Presentation numberPS2-02-27
Validation of a Novel Prognostic Staging System for Breast Cancer Incorporating Age at Diagnosis
Helen M Johnson, The University of Texas MD Anderson Cancer Center, Houston, TX
H. M. Johnson1, W. Dong1, Y. Shen1, W. Irish2, J. H. Wong2, N. A. Vohra2, N. Tamirisa1; 1Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Surgery, East Carolina University, Greenville, NC.
Background: Accumulating evidence supports age at diagnosis as an independent prognostic variable for breast cancer-specific and overall survival (OS). Using SEER data, we previously developed a novel prognostic staging system that incorporates age at diagnosis and demonstrated more refined risk stratification compared with the current American Joint Committee on Cancer (AJCC) staging schema. We now aim to externally validate this novel breast cancer staging system. Methods: The National Cancer Database was used to identify adult females diagnosed with invasive breast cancer from 2010-2015. Women with prior history of malignancy, unknown vital status, or unknown AJCC clinical prognostic stage (CPS) variables were excluded. Serial multivariable Cox’s proportional hazards models were used to evaluate associations between OS and CPS +/- age +/- effect modification between CPS and age. Models were evaluated using the Akaike information criterion (AIC). Results: Among 663,659 patients, the median age was 60 years (IQR 51-70) and the median follow-up was 91.3 months (95% CI 91.2-91.4). At last follow-up, 133,273 patients (20.1%) were dead. The model that incorporated both age and effect modification had the lowest AIC (3320681, versus 3400173 for the model excluding age and 3326910 for the model including age without effect modification), indicative of the best fit. This model was used to predict OS at various timepoints, stratified by CPS. Within each stage group, differential survival was seen across the age spectrum, consistent with the initial model developed in SEER (Table). Within each stage group from IB-IIIC, women diagnosed at age 40 had the best survival, whereas women at the extremes of age had inferior survival. For example, 3-year OS for patients with stage IIIA disease was 0.648 across all ages, highest (0.831) for patients diagnosed at age 40, lower (0.714) for age 20, and lowest (0.127) for age 90. Conclusion: This analysis provides external validation of our novel prognostic staging system for breast cancer. Including age at diagnosis as a prognostic variable results in more refined risk stratification across all AJCC stage groups. Future editions of AJCC should consider utilization of not only anatomic and pathologic variables, but also age at diagnosis, to achieve more accurate survival predictions. While poorer OS is expected for older women due to age-related competing mortality risks, further research is necessary to understand why younger women with breast cancer have higher morality than those diagnosed closer to age 40.
Presentation numberPS2-02-28
Prognostic Value of a Machine Learning Tool for Recurrence Risk Stratification in HR+/HER2- Early Breast Cancer: A Brazilian Cohort Analysis
Rubem Moreira, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
R. Moreira1, D. Huo2, A. Pearson3, I. Small4, O. Olopade3, J. Bines1, F. Howard3; 1Oncology, Instituto Nacional de Cancer, Rio de Janeiro, BRAZIL, 2Department of Public Health Sciences, University Chicago Medicine, Chicago, IL, 3Oncology, University Chicago Medicine, Chicago, IL, 4Data Management and Analysis, Instituto Nacional de Cancer, Rio de Janeiro, BRAZIL.
Purpose Genomic assays are unavailable in Brazil’s public health system and are not covered in the private sector, limiting risk stratification in early stage HR+/HER2- breast cancer. Previous studies have developed machine learning tools to predict the 21-gene recurrence score directly from quantitative immunohistochemistry results and clinical factors. We evaluated this approach in a Brazilian cohort to determine the ability to predict prognosis and guide chemotherapy decisions in a resource‑constrained setting without genomic testing. Patients and methods We retrospectively analyzed HR+/HER2- breast cancer patients (stage I-III, ≤3 positive nodes) treated at the Instituto Nacional de Cancer, in Brazil (INCA) from 2016 – 2018. Age, grade, histologic subtype, and quantitative ER, PR, and Ki-67 expression were used to predict the 21-gene recurrence score with the previously published machine learning tool. Patients were separated into low-risk group and a high-risk group using a predefined threshold that achieves 90% sensitivity for recurrence score > 25. Disease free interval (DFI) and overall survival (OS) were estimated by Kaplan-Meier curves; hazard ratios were derived from Cox models to determine the prognostic difference of the low / high risk groups. Results We identified a sample of 299 cases for this analysis, with an average age of 59. 92 (31%) were PR negative, 212 (70%) had tumor sizer ≥ 2 cm, 163 (55%) had Ki-67 expression of 20% or higher, and 181 (61%) received chemotherapy. Median follow‑up was 73 months (95% CI, 71-75). Using the 90% sensitivity threshold, 15.4% of patients were low risk. Recurrence rates were 2.2% in low‑risk versus 19.4% in high‑risk patients. Five‑years DFI at 90% sensitivity was significantly better for low‑risk versus high‑risk (97.4% vs. 81.8%; long-rank p=0.0054). No significant differences in 5‑year OS were observed. In multivariate analysis, stage III disease and Ki‑67 ≥ 20% were also independent predictors of recurrence. Although chemotherapy was not associated with a reduction in disease free interval in either group, the excellent outcomes in the low risk cohort with (recurrence rate 6.3%) or without (recurrent rate 0%) chemotherapy, suggest this subgroup can safely forgo such treatment without genomic testing. Conclusion This machine learning prognostic tool demonstrated strong prognostic performance in a Brazilian cohort of HR+/HER2- early stage breast cancer patients, identifying a low‑risk subgroup with excellent DFI and no apparent benefit from chemotherapy, and is available for use online (rsncdb.cri.uchicago.edu). These findings support its use for therapy de‑escalation when genomic assays are unavailable. Further prospective studies are required to validate these findings.
Presentation numberPS2-02-29
Real world evidence of systemic therapy in hormone receptor positive advanced breast cancer (HR+ ABC) in Australia – ARORA Registry
Michelle Li, Walter Eliza Hall Institute of Medical Research, Victoria, Australia
M. Li1, V. Wong1, S. Baron-Hay2, R. de Boer3, F. Boyle4, A. Campbell1, I. M. Collins5, K. Cuff6, L. Gately7, C. Georgiou8, S. Greenberg9, E. Jude10, B. Karki11, K. Mok12, C. Morton1, L. Nott13, M. Nottage14, N. Rainey15, J. Torres16, I. Tung17, P. Gibbs1, S. Lok1; 1Gibbs Lab, Walter Eliza Hall Institute of Medical Research, Victoria, AUSTRALIA, 2Medical Oncology, Northern Cancer Institute, New South Wales, AUSTRALIA, 3Medical Oncology, St Vincent’s Private Hospital, Victoria, AUSTRALIA, 4Medical Oncology, Mater North Sydney Hospital, New South Wales, AUSTRALIA, 5Medical Oncology, South West Healthcare, Victoria, AUSTRALIA, 6Medical Oncology, Princess Alexandra Hospital, Queensland, AUSTRALIA, 7Medical Oncology, Alfred Health, Victoria, AUSTRALIA, 8Medical Oncology, Bendigo Health, Victoria, AUSTRALIA, 9Medical Oncology, Western Health, Victoria, AUSTRALIA, 10Medical Oncology, Austin Health, Victoria, AUSTRALIA, 11Medical Oncology, Toowoomba Hospital, Queensland, AUSTRALIA, 12Medical Oncology, Liverpool Hospital, New South Wales, AUSTRALIA, 13Medical Oncology, Royal Hobart Hospital, Tasmania, AUSTRALIA, 14Medical Oncology, Royal Brisbane and Women’s Hospital, Queensland, AUSTRALIA, 15Medical Oncology, Cairns Hospital, Queensland, AUSTRALIA, 16Medical Oncology, Goulburn Valley Health, Victoria, AUSTRALIA, 17Medical Oncology, Eastern Health, Victoria, AUSTRALIA.
BACKGROUND: The incorporation of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) into first line (1L) systemic treatment for patients with HR+ ABC is now standard practice and leads to improved survival. However, there is a paucity of data on uptake of standard therapies and their associated toxicities in the real-world setting. Here, we present updated results from the ARORA registry. METHODS: ARORA is a secondary data use study of Australian patients aged ≥18 years and diagnosed with HR+ ABC after 1 Jan 2020. Prospective data is captured on patient characteristics, disease course, systemic therapy sequencing and treatment outcomes in routine clinical practice. Database lock for analysis was 19 May 2025. RESULTS: Data was analysed from 457 patients at 17 sites, with a median follow-up of 38.8 months. Median age was 64 years (24% ≥75 years). 65.6% were ECOG 0-1 and 2.8% had a BRCA1/2 pathogenic variant. 44.2% had liver or lung metastases, and 19% had bone-only disease. 60.4% had relapsed metastatic disease. Of these, 9.1% and 46.7% had received neoadjuvant and adjuvant chemotherapy respectively following early stage disease, and 75.2% had received adjuvant endocrine therapy (ET). 44.5% relapsed within 12 months of stopping adjuvant ET. 9 patients (2%) did not receive any systemic therapy for ABC. Of the 448 patients on 1L therapy, 335 (74.8%) received CDK4/6i + ET, 67 (15%) received ET alone and 41 (9.1%) received chemotherapy. Clinician preference for 1L CDK4/6i was palbociclib (32.6%), followed by ribociclib (27.2%) and abemaciclib (11.2%). Patients who received 1L ET alone were older (median 79 vs 63 years, 62.7% vs 18.5% ≥75 years), with poorer performance status (44.8% vs 8.7% ECOG ≥2) and more co-morbidities (29.8% vs 10.5% Charlson index ≥2). Conversely, those who received 1L chemotherapy were younger (median 58 years, 7.3% ≥75 years), with better performance status (46.3% vs 37.9% ECOG 0) and fewer co-morbidities (92.7% vs 88.9% Charlson index <2). They were more likely to have visceral metastases compared to those on CDK4/6i + ET (58.5% vs 45.7%), particularly liver metastases (46.3% vs 19.1%). Median 1L progression-free survival with 1L CDK4/6i + ET was 33.9 months, consistent with the upper end reported in trials (23.8–33.6 months). Common reported toxicities with CDK4/6i were diarrhoea (62.2% abemaciclib vs 10% ribociclib vs 6.6% palbociclib), neutropenia (44.9% palbociclib vs 40.8% ribociclib vs 22.2% abemaciclib) and nausea/vomiting (28.3% ribociclib vs 22.2% abemaciclib vs 17.6% palbociclib). Hepatotoxicity occurred in 5.8% ribociclib vs 4.6% abemaciclib vs 0.7% palbociclib. 2.5% of patients on ribociclib had QTc prolongation. Pneumonitis occurred in 1 patient on each CDK4/6i. Treatment was stopped due to toxicity for 20.5% ribociclib, 18% abemaciclib and 12.3% palbociclib. At the time of database lock, 117 of 335 patients (34.9%) who received 1L CDK4/6i + ET had moved to second line (2L). 60 (51.3%) received chemotherapy alone, 39 (33.3%) received ET alone and 5 (4.3%) received targeted therapies (e.g. mTOR or PIK3CA inhibitors). Capecitabine was the most common chemotherapy regimen (44/60, 73.3%). Median 2L duration of treatment was longest with targeted therapies (6.8 months), followed by chemotherapy (4.8 months) and ET alone (3.3 months). Overall survival data is immature. CONCLUSION: While CDK4/6i + ET is recommended as 1L treatment of HR+ ABC, three-quarters of routinely treated patients received this combination. A substantial minority received ET alone or chemotherapy. Age, ECOG, co-morbidity burden and presence of visceral disease remain major determinants of treatment choice. CDK4/6i toxicity profiles are consistent with trials. In 2L, chemotherapy is preferred in over half of patients, but clinical outcomes trend better if targeted therapies are available.
Presentation numberPS2-02-30
Five-year survival outcomes after systemic therapy in older patients with early-stage triple negative breast cancer at memorial sloan kettering cancer center.
Jasmeet Singh, Memorial Sloan Kettering, New York, NY
J. Singh1, A. Kulkarni2, F. Ehrich1, C. White1, Y. Chen1, K. Alexander1, A. Shahrokni1, M. Robson1, L. Norton1, D. Lake1; 1Medicine, Memorial Sloan Kettering, New York, NY, 2Medicine, Columbia University Medical Center, New York, NY.
INTRODUCTION Approximately 15% cases of Triple Negative Breast Cancer (TNBC) are diagnosed in individuals over the age of 70. Older patients remain underrepresented in clinical trials. We conducted a retrospective review of patients to describe the clinical practice patterns and outcomes in patients aged ≥70 years with stage I-III TNBC who were treated at Memorial Sloan Kettering Cancer Center (MSKCC). MATERIAL AND METHODS Medical records of patients with patients aged ≥ 70 years diagnosed with stage I-III triple negative breast cancer from January 1st, 2015, through December 31st, 2019, were abstracted. Charts were analyzed for patient demographics, cancer stage, pathology reports, surgical and systemic therapy data. Chemotherapy (C) and non-chemotherapy (NC) groups were compared based on baseline characteristics, toxicities, and outcomes. We compared patient characteristics by chemotherapy type using Wilcoxon rank sum test, Pearson’s Chi-squared test, and Fisher’s exact test. Overall Survival (OS), Breast Cancer Specific Survival (BCSS), and Recurrence Free BCSS (RF-BCSS) were examined using Cox proportional hazards models. All statistical analyses were conducted using R. RESULTS Total 144 patients were included in our data analysis. The median age of participants was 75 years (range 70-96). The number of patients with stage I, II, and III disease was 61 (42%), 66 (46%), and 17 (12%), respectively. NC group was associated with older median age (81 years vs. 74 years (p<0.001)) and a higher rate of referral to geriatric medicine (31% in NC vs. 15% in C, p=0.045). The NC group had significantly higher difficulties in ADLs such as walking (27% vs. 12%, p=0.041), dressing (19% vs. 4.4%, p=0.014), and bathing (19% vs. 4.4%, p=0.014) compared to the C group. NC group also had significantly higher difficulties in IADLs such as meal preparation (27% vs. 3.3%, p0.9) and RF-BCSS (HR 0.68, 95% CI 0.34-1.37, p=0.3) between C- and NC- groups. Doxorubicin-Cyclophosphamide-Paclitaxel (ACT) and Cyclophosphamide-Methotrexate-5-Fluorouracil (CMF) were the most common chemotherapy regimens administered. Compared to ACT, CMF was associated with greater median age (median age 75 vs. 71 years, p0.9), BCSS (HR 1.26, 95% CI 0.40-3.97; p=0.7), and RF-BCSS (HR 1.25, 95% CI 0.44-3.51; p=0.7) in patients receiving ACT versus CMF. CONCLUSIONS In our single institutional experience, we observed that two-thirds of older patients with early-stage TNBC received systemic polychemotherapy. C-group had an improved OS but not BCSS and RF-BCSS, a finding likely driven by non-oncological factors, with NC being a frailer group of patients with other competing causes of mortality. ACT and CMF were the most common regimens used. Use of CMF was associated with age ≥75, stage I disease, ADL/IADL difficulties, and adjuvant setting. We observed no difference in survival outcomes between the two chemotherapy regimens. CMF remains an efficacious option and a potentially viable alternative to ACT in TNBC, especially in the setting of early stage and advanced age.
Presentation numberPS2-04-02
Real-world evidence of immunohistochemical discordance in breast cancer brain metastases
Jessé Silva, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
G. Carvalho1, F. Rodrigues2, P. Fernandes2, I. Small1, J. Silva1, L. Araujo1; 1Clinical Research and Technological Development Division, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL, 2Pathology Department, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL.
Background This study assessed immunohistochemical (IHC) discordance between primary breast cancer (BC) and brain metastases (BCBM) in a Brazilian population. While 20-40% of BC patients develop BCBM, particularly those with HER2-positive or triple-negative subtypes, phenotypic changes may occur during disease progression, driven by tumor heterogeneity and alterations in the tumor microenvironment (TME). Due to the limited availability of BCBM samples, data on IHC discordance remain scarce. Methods This retrospective cohort study included women who underwent neurosurgical resection for BCBM between January 2015 and December 2022 at the Brazilian National Cancer Institute (INCA). Clinical and pathological data were extracted from medical records. IHC markers ─ estrogen receptor (ER), progesterone receptor (PR), HER2, and Ki67 ─ were assessed in BCBM samples and compared with the corresponding archival primary breast cancer specimens. An alluvial diagram was used to visually represent patterns of IHC discordance. The primary objective was to describe the frequency of IHC discordance between primary BC and corresponding BCBM. Secondary objectives included evaluation of associations between clinicopathological and sociodemographic features and IHC discordance, as well as its impact on survival outcomes. Results Among the 105 eligible patients, 64% (n = 67 patients) were Black or mixed-race, and 41% (n = 42) had not completed primary education. A significant proportion were obese (43%, n = 45), and 42% (n = 44) had hypertension. The mean age was 49 years (SD ±10), and 58% (n = 61) were premenopausal. Most tumors were ductal (87%, n = 92), and grade II (45%, n = 47) or grade III (46%, n = 48), with the HER2-positive subtype being the most frequent (48%, n = 51). Overall, 69% (n = 52) of patients had locally advanced disease, with a mean of 5 (SD ±6) affected lymph nodes. Among those who received neoadjuvant chemotherapy, 16% (n = 13) achieved a pathological complete response. About 14% (n = 15) presented with de novo metastatic disease. At the time of BCBM diagnosis, 91% (n = 95) exhibited neurological symptoms, most commonly headache (62%, n = 59) and dizziness (37%, n = 35). A single brain lesion was observed in 67% (n = 70) of cases. IHC discordance between primary BC and BCBM was found in 44% (n = 46) of patients, but this was not associated with differences in survival outcomes, including overall survival (OS) (p = 0.96), recurrence-free survival after BCBM diagnosis (post-BCBM RFS) (p = 0.81), or OS after BCBM diagnosis (post-BCBM OS) (p = 0.53). The most common phenotypic changes were loss of ER/PR expression (76%, n = 35) and enrichment of HER2 expression (15%, n = 7). Loss of ER/PR expression was not associated with post-BCBM risk of death (p = 0.52). Conclusion A notably high frequency of IHC discordance between primary breast tumors and brain metastases was observed in this real-world Brazilian study, exceeding rates previously reported in the literature. To our knowledge, this is the first Brazilian study to investigate IHC changes in this context, conducted in a population with a high proportion of Black and mixed-race individuals. These results underscore the biological heterogeneity of breast cancer in underrepresented populations and emphasize the need for more precise, individualized management strategies for patients with BCBM.
Presentation numberPS2-04-03
Herpes zoster vaccination and risk of breast cancer in women undergoing mammographic screening: a trinetx real-world analysis
Olga Serra, IRST IRCCS Dino Amadori, Meldola, Italy
O. Serra1, A. Farolfi1, F. Merloni1, C. Gianni1, G. Miserocchi1, N. Gentili1, M. Mariotti1, G. Di Menna1, C. Casadei1, F. Rusconi2, A. Andalò1, S. Sabbioni1, A. Carlini1, D. Montanari1, M. Sirico1, R. Maltoni1, L. Cecconetto1, S. Sarti1, M. Palleschi1, A. Musolino1; 1Medical Oncology, Breast & GYN Unit, IRST IRCCS Dino Amadori, Meldola, ITALY, 2TrineTX, TrineTX, Cambridge, MA, USA, MA.
Herpes Zoster Vaccination and Risk of Breast Cancer in Women Undergoing Mammographic Screening: A TriNetX Real-World AnalysisBackgroundHerpes Zoster vaccination is routinely recommended for adults aged ≥50 years to prevent shingles and its complications. To date, no evidence exists that herpes zoster vaccination reduces breast cancer (BC) risk. Observed associations between vaccinations and lower cancer incidence in some real-world studies may reflect differences in healthcare behavior, immune status, or unmeasured confounding rather than a true protective effect. We investigated whether vaccination with Shingrix®, a recombinant, adjuvanted herpes zoster vaccine, might influence the risk of subsequent BC diagnosis in women undergoing routine mammographic screening.MethodsWe performed a retrospective real-world study using the TriNetX Global Collaborative Network. We included women aged ≥50 years undergoing mammographic screening. The initial cohorts included 5,696 vaccinated women and 1,116,138 unvaccinated women. The vaccinated cohort comprised women who received Shingrix® after screening, while the unvaccinated cohort included women who underwent screening but were never vaccinated against herpes zoster. Patients with a prior diagnosis of BC before the observation period were excluded. Propensity score matching (PSM) was performed based on age, race, ethnicity, Charlson Comorbidity Index, BMI, smoking, alcohol-related disorders, genetic susceptibility to malignancy, and systemic corticosteroid use, resulting in 5,680 women per group. Additionally, multivariable Cox proportional hazards models were applied, adjusting for age, genetic susceptibility, and alcohol-related disorders, with a separate analysis restricting the unvaccinated group to academic medical centers. The observation period extended to 20 years in the TriNetX network. The outcome was the incidence of BC, analyzed as time-to-event from the index date of vaccination or screening.ResultsAfter matching, 5,680 vaccinated women were compared to an equal number of unvaccinated women. The cumulative incidence of BC was significantly lower among vaccinated patients, with 6.4% developing BC compared to 10.7% in the unvaccinated group (risk difference −4.2%, 95% CI −5.2% to −3.2%; p<0.001). Cox regression in the unmatched cohorts confirmed this association, yielding a hazard ratio of 0.617 (95% CI, 0.557–0.684; p<0.0001) when comparing vaccinated patients to the overall unvaccinated group. When restricting the analysis to unvaccinated women treated at academic centers, the association appeared even stronger, with a hazard ratio of 0.447 (95% CI, 0.404–0.496; p<0.0001). However, the absolute differences in incidence rates remained modest. ConclusionsIn this large real-world analysis, vaccination with Shingrix® was associated with a significantly reduced risk of BC diagnosis among women undergoing mammographic screening. However, the observed 40–45% risk reduction is difficult to explain biologically and may result from healthy user bias, residual confounding related to factors such as family history or hormonal therapy use, or surveillance bias leading to earlier detection in vaccinated women. These findings should be interpreted as hypothesis-generating and require confirmation in prospective studies.Keywordsbreast cancer, herpes zoster vaccination, Shingrix, real-world data, TriNetX, cancer risk, immunization, screening.
Presentation numberPS2-04-04
First Real-World Evidence Linking Anxiety to Poorer Treatment Response and Survival in Early-Stage Breast Cancer
Yulong Chen, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China, Guangzhou, China
Y. Chen, J. Zhao, L. Ding, Q. Li, J. Chai, Y. Wang; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China, Guangzhou, CHINA.
First Real-World Evidence Linking Anxiety to Poorer Treatment Response and Survival in Early-Stage Breast Cancer Introduction:Anxiety is a prevalent but often underrecognized comorbidity in breast cancer patients. Despite advances in systemic therapies that have improved the cure rates of early-stage disease, the psychological burden associated with diagnosis, treatment-related toxicity, and uncertainty about prognosis remains substantial. Emerging evidence suggests that anxiety may not only impair quality of life but also negatively impact treatment efficacy and survival. However, robust data on this association are lacking. This study represents the first large-scale, real-world investigation evaluating the impact of anxiety on pathological complete response (pCR) and disease-free survival (DFS) in early-stage breast cancer. The association between anxiety and pCR is reported here for the first time. Methods:In this single-center, real-world cohort study, 2,113 female patients with early-stage breast cancer treated between June 2023 and June 2025 were enrolled. Anxiety was assessed at baseline using the Zung Self-Rating Anxiety Scale (SAS), with scores ≥50 indicating clinically significant anxiety. Clinical and demographic data were collected, including tumor stage, treatment type, and reimbursement status. pCR was evaluated post-neoadjuvant therapy. DFS was defined as the interval from surgery to documented recurrence or last follow-up. Logistic regression and Cox proportional hazards models were used for multivariate analysis. Results:Of the 2,113 patients, 700 (33.1%) exhibited clinically significant anxiety, including 58 (8.3% of anxious patients) with severe anxiety. Higher anxiety prevalence was associated with age ≤50 years (P < 0.001), more advanced tumor stage (P < 0.001), and lower medical insurance coverage (P < 0.05). BMI, marital status, and socioeconomic status were not significantly correlated. Patients with anxiety had significantly lower pCR rates (OR = 2.58; 95% CI: 1.82-3.71; P < 0.001), a finding that remained significant after adjusting for potential confounders, including molecular subtype, tumor burden, and other clinical variables (adjusted OR = 2.83; 95% CI: 1.87-4.33; P < 0.001). In survival analysis, the median DFS was 36.7 months in the anxiety group vs. 50.0 months in the non-anxiety group (P < 0.001). Anxiety was an independent prognostic factor for inferior DFS in multivariate Cox regression (HR = 2.56; 95% CI: 1.51-4.32; P < 0.001). Conclusion:This real-world study provides the first evidence that anxiety is an independent predictor of both lower pCR rates and shorter DFS in early-stage breast cancer. These findings underscore the clinical importance of routine psychological screening and timely intervention. Addressing anxiety may not only improve patient well-being but also enhance treatment response and survival outcomes.
Presentation numberPS2-04-05
Long-term outcomes and toxicities of doxorubicin-cyclophosphamide chemotherapy in breast cancer: a McGill University Health Centre experience, 100-month interim analysis
Eliana Rohr, McGill University, Montréal, QC, Canada
E. Rohr1, T. Alotaibi2, F. Moria3, N. Bouganim4; 1Department of Internal Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, CANADA, 2Department of Medical Oncology, The Medical Services of the Ministry of Interior, Riyadh, SAUDI ARABIA, 3Faculty of Medicine, King Abdulaziz University, Jeddah, SAUDI ARABIA, 4Department of Medical Oncology, McGill University Health Centre, Montréal, QC, CANADA.
Background:Doxorubicin-cyclophosphamide (AC) has remained a cornerstone of breast cancer therapy for over four decades.¹,² Despite its widespread use in clinical trials, long-term data and toxicity data remain limited in non-clinical trial patients. Chronic complications such as chemotherapy-induced cardiomyopathy (CIMP), defined by left ventricular dysfunction, and chemotherapy-induced leukemia carry important quality-of-life consequences.4-6 In this study, we assessed the efficacy and toxicity of AC chemotherapy in a real-world cohort of breast cancer patients with a median follow-up of 8.1 years. Methods:We conducted a descriptive analysis of a prospectively maintained institutional database, including breast cancer patients treated with AC chemotherapy (doxorubicin 60 mg/m² and cyclophosphamide 600 mg/m²) in the adjuvant or neoadjuvant setting at the McGill University Health Centre from 2016 to 2025, with a median follow-up time of 8.1 years. Primary outcomes included metastatic recurrence (rate and site), all-cause mortality, cardiotoxicity, and hematological malignancies. Subgroup analyses were performed based on age, stage, and tumor subtype. Statistical analyses included descriptive statistics for baseline characteristics, Fisher’s exact tests for categorical comparisons, Kaplan-Meier estimates for survival outcomes, and multivariable Cox proportional hazards models. All analyses were conducted using R (v4.5.1). Results:A total of 98 patients were included in the final analysis. All patients were women, with a mean age of 60.7 ± 12.0 years. Most were diagnosed with invasive ductal carcinoma (87.8%) and had stage II disease (56.1%). No patients were lost to follow-up, with a median follow-up duration of 97 months. Excluding those with de novo metastatic disease (7 patients), recurrence occurred in 13 of 91 patients (14.3%), with a median time to recurrence of 40.1 months. Metastatic rates were significantly higher in stage III compared to stage I-II disease (42.1% vs. 6.9%, p < 0.001). No significant difference in recurrence was observed across tumor subtypes (p = 0.919). The most common metastatic sites were bone (84.6%) and liver (76.9%). Overall mortality was 18.4%, significantly higher in advanced-stage (stage III-IV) patients compared to early-stage (stage I-II) disease (50.0% vs. 6.9%, p < 0.001). Triple-negative breast cancer had the highest subtype-specific mortality (29.4%). Five-year overall survival was 88.7%, with advanced stage at diagnosis emerging as the only statistically significant independent predictor of mortality (HR 8.96, 95% CI 3.19-25.18; p < 0.001). The cardiotoxicity rate was 1.0%, with one patient developing CIMP-defined heart failure (LVEF 10-15%) and requiring coronary care unit admission. No secondary hematologic malignancies were observed. ConclusionThis eight-year, prospectively collected real-world dataset confirms outcomes reported in randomized clinical trials. The favourable long-term survival and low rates of metastasis, as well as an acceptable long-term safety profile, reinforce the use of doxorubicin-cyclophosphamide (AC) chemotherapy in high-risk patients. CIMP was an uncommon but important toxicity.
Presentation numberPS2-04-06
Real-world outcomes of trastuzumab deruxtecan with or without endocrine therapy in her2-low, hormone receptor-positive metastatic breast cancer: a propensity-matched analysis
Michela Palleschi, IRST IRCCS Dino Amadori, Meldola, Italy
M. Palleschi1, A. Farolfi1, C. Gianni1, G. Miserocchi1, M. Corianò2, N. Gentili1, M. Mariotti1, G. Di Menna1, O. Serra1, C. Casadei1, F. Rusconi3, A. Andalò1, S. Sabbioni1, A. Carlini1, D. Montanari1, M. Sirico1, R. Maltoni1, L. Cecconetto1, S. Sarti1, F. Merloni1, A. Musolino1; 1Medical Oncology, Breast & GYN Unit, IRST IRCCS Dino Amadori, Meldola, ITALY, 2Medical Oncology and Breast Unit,, Department of Medicine and Surgery, University Hospital of Parma, Parma, ITALY, 3TriNetX, LLC, TriNetX, Cambridge, MA.
Real-World Outcomes of Trastuzumab Deruxtecan with or without Endocrine Therapy in HER2-Low, Hormone Receptor-Positive Metastatic Breast Cancer: A Propensity-Matched AnalysisAuthors (max 50): 21Michela Palleschia, Alberto Farolfia, Caterina Giannia, Giulia Miserocchia, Matilde Corianòb, Nicola Gentilic, Marita Mariottia, Giandomenico Di Mennaa, Olga Serraa, Chiara Casadeia, Francesca Rusconid, Alice Andalòc, Simone Sabbionia, Andrea Carlinia, Daniela Montanaria, Marianna Siricoa, Roberta Maltonia, Lorenzo Cecconettoa, Samanta Sartia, Filippo Merlonia , Antonino MusolinoaAffiliation:a Medical Oncology, Breast & GYN Unit IRCCS Istituto Romagnolo per lo Studio Dei Tumori (IRST) “Dino Amadori”b Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy.c Data Unit, IRCCS Istituto Romagnolo per lo studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italyd TriNetX, LLC, Cambridge, MA, USA Background:Combination therapy involving anastrozole or fulvestrant with fam-trastuzumab deruxtecan-nxki (TDX-d) has demonstrated promising antitumour activity in chemotherapy-naïve, HER2-low, hormone receptor-positive (HR+) metastatic breast cancer (mBC) patients, as evidenced by the phase 1b DESTINY-Breast08 trial (NCT04556773). Although this regimen is not currently established as a standard of care, it is being utilized in several oncology centers. However, real-world evidence supporting this strategy remains limited.Methods:Applying the TriNetX Global Collaborative Network, we retrospectively identified two cohorts comprising 658 mBC patients, stratified by receipt of TDX-d monotherapy or TDX-d in combination with endocrine therapy (ET). Propensity score matching (1:1) was performed to balance the cohorts (TDX-d plus ET, n=85; TDX-d alone, n=85) for age, race, and body mass index, all of whom had prior exposure to CDK4/6 inhibitors and chemotherapy. Hazard ratios (HRs) were calculated to compare overall survival (OS) between groups.Results:Following matching, the median age was 57.3 years (±13.9) in the TDX-d plus ET group and 56.7 years (±14.1) in the TDX-d monotherapy group. Median follow-up was 87.9 months for the combination cohort and 46.1 months for the monotherapy cohort. The combination group demonstrated a trend towards improved overall survival compared to TDX-d alone (HR=0.79; 95% CI, 0.465-1.344; p=0.384), although statistical significance was not achieved.Conclusions:This represents the first large-scale real-world cohort analysis evaluating the addition of endocrine therapy to TDX-d in patients with HER2-low, HR+ mBC. While these preliminary findings suggest a potential survival benefit with the combination approach, the limited follow-up duration preclude definitive conclusions. Further prospective studies are warranted to elucidate the clinical utility of combining TDX-d with ET in this patient population. Our research aims to perform ongoing follow-up of these patients to generate more robust data, thereby further clarifying the clinical benefit of combining TDX-d with ET in this population.Key-words (max 5): Trastuzumab deruxtecan; metastatic breast cancer; endocrine therapies; TrineTX; real world evidence
Presentation numberPS2-04-07
A large-scale, US-based study evaluating real-world medical cost savings and reduced healthcare resource utilization associated with longer first-line treatment for advanced breast cancer
Clara Chen, AstraZeneca, Gaithersburg, MD
C. Chen1, A. Ghosh2, R. Potluri3; 1Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 2-, Putnam Associates, Gurugram, INDIA, 3-, Putnam Associates, New York, NY.
Background Cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) + aromatase inhibitor (AI) is the current standard of care for first-line (1L) treatment of hormone receptor (HR)+/human epidermal growth factor receptor (HER2−) advanced breast cancer (ABC). However, treatment resistance is common and affects 1L treatment duration, which may lead to higher healthcare expenditure due to disease progression and health deterioration. To better understand the association between medical costs and the duration of 1L treatment, this study investigated the real-world costs and healthcare resource utilization (HCRU) for patients with ABC who received 1L CDK4/6i + AI, stratified by the duration of 1L treatment (DoT). Methods This retrospective, observational cohort study extracted data from a large US claims database for commercially insured and Medicare Advantage patients diagnosed with ABC between April 2017 and August 2024. Eligible patients received CDK4/6i + AI as 1L treatment and had a follow-up of ≥6 months (m). Patients were grouped into two cohorts: ≤12 m DoT and >12 m DoT until cessation of both CDK4/6i + AI. Baseline characteristics were summarized using descriptive statistics. For patients with ≥12 m of follow-up (or less if due to death), medical costs and HCRU for the first 12 m from the start of 1L treatment were captured and calculated per patient per month (PPPM). Generalized linear models were used to analyze the relationship between DoT and medical costs/HCRU, adjusting for age, year of ABC diagnosis, comorbidities, and insurance type. Cumulative medical costs per patient were calculated in monthly intervals for the total study population according to DoT, from the start of 1L treatment to a maximum of 24 m. Results Among the 6464 patients in the study sample (mean age: 60.9 years, 65.0% commercially insured), 4546 had ≥12 m of follow-up (or less if due to death). Compared to patients with DoT >12 m (n=3830), those with DoT of ≤12 m (n=716) had higher mean PPPM costs ($6,424 vs $3,645, p12 m versus DoT ≤12 m at 12 m of follow-up, and $46,479 lower at 24 m of follow-up. Key factors driving the medical cost savings in patients with DoT >12 m included reduced inpatient/outpatient care and fewer office visits. Conclusions Longer duration of 1L treatment was associated with reduced HCRU and substantial medical cost savings. Adoption of treatment approaches that prolong 1L DoT could meaningfully reduce the economic burden of cancer care. Table. Medical costs and HCRU for patients, based on the duration of 1L treatment
| DoT ≤12 m (n=716) | DoT >12 m (n=3830) | ||||
| Medical costs (adjusted), $, mean PPPM [95% confidence intervals] | |||||
| Inpatient stays | 1,933 [1,862-2,008] | 730 [708-754] | |||
| Office visits | 1,908 [1,838-1,982] | 1,246 [1,207-1,286] | |||
| Outpatient care | 1,523 [1,466-1,582] | 953 [923-984] | |||
| Emergency room visits/ ambulatory care | 215 [207-223] | 75 [72-77] | |||
| Other | 499 [480-518] | 315 [304-325] | |||
| Total medical costs | 6,424 [6,185-6,672] | 3,645 [3,531-3,763] | |||
| Healthcare resource utilization, mean per 100 patients per month [95% confidence intervals] | |||||
| Inpatient visits | 12 [11-12] | 4 [4-4] | |||
| Inpatient stays (days) | 109 [105-113] | 42 [41-43] | |||
| Office visits | 478 [475-481] | 374 [372-376] | |||
| Outpatient care | 204 [202-206] | 154 [152-155] | |||
| Emergency room visits/ ambulatory care | 26 [25-26] | 13 [12-13] |
Presentation numberPS2-04-08
Incidence of second primary tumors and second primary breast cancers events in patients with metastatic breast cancer: results from the Austrian AGMT_MBC-Registry
Vanessa Castagnaviz, Paracelsus Medical University Salzburg, Salzburg, Austria
V. Castagnaviz1, S. P. Gampenrieder1, A. Pichler2, W. Herz3, R. Pusch4, C. Dormann4, C. Suppan5, 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, M. Hubalek17, M. Knauer18, C. F. Singer19, G. Rinnerthaler5, R. Greil20; 1Department of Internal Medicine III, Paracelsus Medical University Salzburg, Salzburg, AUSTRIA, 2Internal Medicine – Department for Haemato-Oncology, LKH Hochsteiermark, Leoben, AUSTRIA, 3Department of Surgery, Breast Health Center, LKH Hochsteiermark, Leoben, AUSTRIA, 4Internal Medicine I, Ordensklinikum Linz Barmherzige Schwestern – Elisabethinen, Linz, AUSTRIA, 5Division of Oncology, Department for Internal Medicine, Medical University Graz, Graz, 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: Second primary tumors (SPTs) occur in approximately 10% of patients following an initial cancer diagnosis and are associated with increased cancer-related mortality. Ongoing advancements in systemic therapies have led to a growing population of both breast cancer (BC) survivors and patients living with metastatic disease. Consequently, individuals with BC face a heightened risk of developing SPTs and second primary breast cancers (SPBC), with a reported 25-year cumulative incidence of contralateral BC of about 10%. Here we present real-world data on SPTs and SPBC in patients with metastatic breast cancer (MBC) derived from the Austrian Study Group of Medical Tumor Therapy (AGMT) MBC-Registry. Patients and methods: The AGMT_MBC-Registry is a multicenter, nationwide, ongoing retrospective and prospective registry capturing data from MBC patients across Austria. For this analysis, only patients with documented information on the presence or absence of SPTs were included. The analysis of SPBC was limited to patients with known tumor location and diagnosis date. To avoid misclassification, no distinction was made between local recurrences and ipsilateral SPBC. Bilateral BC was defined as cancer diagnosed in both breasts within 90 days. Results: As of June 26, 2024, the AGMT_MBC-Registry included 2,850 patients. Among 2,630 evaluable patients, 225 (8.6%) were diagnosed with a SPT, with 12% (27/225) of these having more than one SPT. Most SPTs were diagnosed after the initial BC diagnosis but prior to the development of metastatic disease. A total of 260 malignancies were reported: 88% were solid tumors and 12% hematologic. The most frequent solid tumors were colorectal cancer, melanoma, and lung cancer, while non-Hodgkin lymphoma and acute leukemia were the most common hematologic malignancies (Table 1). Among 2,576 evaluable patients, 533 (20.7%) experienced a SPBC: 145/533 (27.2%) contralateral, 287/533 (53.8%) ipsilateral, and 101/533 (18.9%) initially presented with bilateral BC. Conclusion: In this real-world analysis of patients with MBC, 9% were diagnosed with SPT and 21% experienced a SPBC. These findings highlight the critical role of histopathological verification of suspicious lesions and metastases and underscore the importance of continued awareness for SPTs – not only among BC survivors but also in patients with metastatic disease Table 1:
| Solid tumors (n = 229) | n (%) | Hematologic malignancies (n = 31) | n (%) |
| Colorectal cancer | 31 (13.5 %) | Non-Hodgkin Lymphoma (NHL) | 7 (22.6 %) |
| Melanoma | 29 (12.7 %) | Acute leukemia | 4 (12.9 %) |
| Lung cancer | 22 (9.6 %) | Hodgkin lymphoma (HL) | 4 (12.9 %) |
| Renal cell carcinoma | 20 (8.7 %) | Myeloproliferative neoplasms (MPNs) | 3 (9.7 %) |
| Ovarian cancer | 16 (7 %) | Chronic lymphocytic leukemia (CLL) | 2 (6.5 %) |
| Thyroid cancer | 14 (6.1 %) | Myelodysplastic syndrome (MDS) | 2 (6.5 %) |
| Cervical cancer | 11 (4.8 %) | Unknown | 2 (6.5 %) |
| Bladder cancer | 8 (3.5 %) | Other | 7 (22.6%) |
| Endometrial cancer | 7 (3.1 %) | Total | 31 (100%) |
| Gastric cancer | 7 (3.1 %) | ||
| Pancreatic cancer | 6 (2.6 %) | ||
| Urothelial carcinoma | 4 (1.7 %) | ||
| Vulvar cancer | 4 (1.7 %) | ||
| Head and neck cancers | 3 (1.3 %) | ||
| Neuroendocrine tumors | 3 (1.3 %) | ||
| Prostate cancer | 3 (1.3 %) | ||
| Unknown | 3 (1.3 %) | ||
| Other | 38 (16.6%) | ||
| Total | 229 (100%) |
Presentation numberPS2-04-09
Impact of prior treatment exposure and clinical-pathological factors on response and survival with Sacituzumab govitecan in advanced breast cancer: a consecutive real-world series
Carla Gullotta, Hospital Clinic Barcelona, Barcelona, Spain
C. Gullotta1, S. Cobo1, B. Walbaum1, M. Bergamino1, R. Gómez-Bravo1, O. Martínez-Sáez1, F. Schettini1, E. Segui1, I. García-Fructuoso1, T. Pascual1, N. Chic1, M. González-Rodríguez1, G. Riu2, E. Carcelero2, A. Rodríguez-Hernández1, E. Sanfeliu3, P. Galván1, M. Muñoz1, M. Vidal1, A. Prat1, F. Braso-Maristany1, B. Adamo1; 1IDIBAPS, Hospital Clinic Barcelona, Barcelona, SPAIN, 2Department of Pharmacy, Hospital Clinic Barcelona, Barcelona, SPAIN, 3Pathology Department, Hospital Clinic Barcelona, Barcelona, SPAIN.
Title: Impact of prior treatment exposure and clinical-pathological factors on response and survival with sacituzumab-govitecan in advanced breast cancer: a consecutive real-world series Background: Sacituzumab govitecan (SG) is an antibody-drug conjugate (ADC) approved for the treatment of advanced triple negative (TNBC) and estrogen receptor positive-HER2 negative (ER+/HER2-) breast cancer (BC). Real-world data on response determinants are limited. We evaluated clinical-pathological factors associated with objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) in a consecutive patient series treated in routine practice. Methods: This retrospective study included 74 consecutive female patients treated with SG at the Hospital Clinic of Barcelona between April 2022-April 2025. Clinical-pathological variables included tumor subtype, ECOG status, number of metastatic sites, visceral metastases, prior therapies, and Ki-67 index (pre-treatment or most recent metastatic biopsy). No patients had received prior ADC containing a TOP1 inhibitor payload. ORR was assessed radiologically according to RECIST version 1.1. ORR associations were tested by univariate logistic regression; survival by Kaplan-Meier and Cox models. Results: Median age was 57 years; 41 patients (54.9%) had TNBC and 33 (45.1%) ER+/HER2- BC. 61 patients had visceral disease (86%), 10 patients had brain metastasis (14%), 42 were ECOG status>1 (59.2%), and 43 had >3 metastatic sites (60.6%). The median number of previous chemotherapy lines in metastatic setting was 2. The ORR was 34.3% (2.9% complete and 31.4% partial). Stable disease and progressive disease occurred in 22.9% and 42.8%, respectively. ORR was 44.4% among the 31 patients who had received fewer than three prior lines of chemotherapy, compared to 27.9% in the 43 patients treated in the third-line setting or beyond. In univariate logistic regression, having received ≥3 chemotherapy lines, compared to 3 metastatic sites (HR=1.77, p=0.048) were independently associated with shorter PFS. No statistically significant associations were observed between PFS and Ki-67 (p=0.650). TNBC subtype (HR=4.46, p=0.006), visceral disease (HR=7.85, p=0.046), and ECOG status > 1 (HR=10.17, p<0.001) predicted for shorter OS. In contrast, ER+/HER2- (HR=0.24, p=0.010) and prior endocrine therapy (HR=0.32, p=0.004) were associated with longer OS. No statistically significant associations were observed between OS and Ki-67 index (p=0.153). Conclusions: In this real-world cohort, SG demonstrated limited antitumor activity across both TNBC and ER+/HER2- breast cancer. Clinical benefits were greater in patients with limited prior treatment exposure, preserved ECOG status, and lower disease burden. These findings support the earlier use of SG.
Presentation numberPS2-04-10
Adherence to endocrine prevention in patients with atypical hyperplasia and lobular carcinoma in situ: promising trends from real-world use of low dose tamoxifen.
Natasha Stonebanks Cuillerier, Jewish General Hospital, Montreal, QC, Canada
N. Stonebanks Cuillerier1, H. Almarzooqi2, V. Villareal-Corpuz1, S. Saboobheh2, I. Prakash2, M. Basik2, J. Boileau2, K. Martel2, S. Meterissian2, M. Pollak3, S. M. Wong2; 1Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, CANADA, 2Department of Surgery, McGill University, Montreal, QC, CANADA, 3Department of Oncology, McGill University, Montreal, QC, CANADA.
INTRODUCTION: Endocrine prevention is a well-established preventative strategy for women with high-risk lesions (HRL). The objective of the study was to evaluate trends and factors associated with endocrine prevention adherence in this patient population. METHODS: We performed a retrospective cohort study of all women referred for high-risk evaluation due to diagnosed atypical ductal hyperplasia (ADH), atypical lobular hyperplasia (ALH), and lobular carcinoma in situ (LCIS) between 2019-2025. Data pertaining to eligibility, initiation, and adherence to endocrine prevention were extracted from the medical record. Univariate analyses were performed using Chi-squared and Fisher’s exact test to evaluate factors associated with adherence. RESULTS: Of 216 female HRL patients 188 met criteria and were included in the analytic cohort including 107 (56.9%) with ADH, 66 (35.1%) with ALH, and 15 (8%) with LCIS. The median age was 55 years (interquartile range [IQR]: 50-62). Overall, 106 (56.4%) HRL patients accepted a prescription of endocrine prevention. Low-dose tamoxifen was the most common regimen prescribed (64.2%), compared to regular dose tamoxifen (11.3%), raloxifene (19.8%), and anastrozole (4.7%). At a median follow up of 28 months (IQR 11-46 months), 71 women (67% of those who accepted a prescription) initiated endocrine prevention and 65 (61.3%) remained adherent. Lack of prior hormone replacement therapy use (65.6% vs 30.8%, p=0.02) and family history of breast cancer (77.8% vs 55.7%, p=0.04) were significantly associated with adherence to endocrine prevention. Adherence was not significantly different by regimen prescribed (p=0.66), nor was it significantly associated with age (p=0.08) or prior surgical excision of their HRL (p=0.52). Of the 42 patients who initiated tamoxifen 5 mg, 39 (92.9%) remained adherent during follow up, and of 12 women (23%) who had completed at least 3 years of therapy, 10 (83.3%) elected to continue or complete a total 5-year course of low dose tamoxifen.CONCLUSION: Adherence to endocrine prevention is high in women who initiate medication with few discontinuing due to side effects. Many women who initiate low dose tamoxifen remain adherent, with some electing to continue the regimen for 5 years total.
Presentation numberPS2-04-11
Treatment Sequencing in HR+ HER2- Metastatic Breast Cancer and Associated Real World Outcomes
Nicholas P McAndrew, UCLA, Santa Monica, CA
N. P. McAndrew1, E. T. Corcoran2, S. Alkassis1, S. Franch-Expósito2, K. Roche2, S. Islam2, C. Igartua2, J. Mercer2, P. A. Fields2, A. Bardia1, E. Cohen2; 1Medicine (Hematology/Oncology), UCLA, Santa Monica, CA, 2AI, Tempus AI, Chicago, IL.
Background: Hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (mBC) represents a significant clinical challenge with evolving treatment strategies. Cyclin-dependent Kinase 4/6 inhibitors (CDK4/6i) and taxanes are commonly used to treat HR+ HER2- mBC. This study investigates the impact of first- and second-line therapy selections on real-world progression-free survival (rwPFS) and real-world overall survival (rwOS) in patients with HR+ HER2- mBC. Methods: The study cohort included 13,064 patients with HR+ HER2- mBC from the de-identified Tempus real-world multimodal database who received either first- (1L) or second-line (2L) CDK4/6i, taxane, or other therapy. Kaplan-Meier survival analysis and multivariate Cox regression accounting for main epidemiological confounding variables (age at diagnosis, presence of visceral metastases, and recurrent vs. de novo treatment status) were used to assess rwPFS and rwOS. Progression events included death, noted progression event or initiation of a new line of therapy. DNA sequencing data from the Tempus xT assay were analyzed to evaluate mutation landscapes and genetic alterations associated with different treatment sequences. Results: Patients with HR+ HER2- mBC (N = 13,046) who received 1L CDK4/6i + endocrine therapy (ET) had significantly improved rwPFS and rwOS relative to those receiving a taxane. We observed a trend towards faster progression in patients who received taxane as a 2L treatment after 1L CDK4/6i as opposed to other treatments (HR=1.33, P=0.052). Of patients who received 1L CDK4/6i, 2L rwPFS was longest in patients who were re-challenged with CDK4/6i (predominantly Palbociclib+Letrozole to Palbociclib+Fulvestrant). Patients who received 2L CDK4/6i following 1L taxane had longer 2L rwPFS than those who received a non-CDK4/6i 2L treatment (median PFS=5.9 months 2L CDK4/6i vs 3.6mo non-CDK4/6i). Across all patients who received 2L CDK4/6i, there was a trend toward better 2L rwPFS for those with 1L taxane compared to those who received 1L CDK4/6i (median PFS 5.9 mo vs median PFS=5.1mo). This result of best 2L response in patients receiving CDK4/6i following taxane in 1L remains consistent when fitting a multivariate model including treatment order, age at diagnosis, presence of visceral metastases, treatment recurrence, and prevalent mutations (BRCA, RB1, FGFR2, AKT1, ESR1, PIK3 (HR=0.80, p=0.109). Conclusion: This study demonstrates that the sequence of taxane and CDK4/6i treatments might impact survival outcomes in patients with HR+ HER2- mBC. While CDK4/6i+ET outperforms taxane in 1L, the findings suggest that taxane may cause sensitization to CDK4/6i, conferring better survival outcomes in a subset of patients where 1L taxane is advised, or a potential advantage to chemo before CDK4/6i re-challenge. These results need validation in additional datasets and underscore the importance of treatment sequencing in optimizing clinical outcomes for patients with HR+ mBC. Note: NPM and ETC contributed equally to this work
Presentation numberPS2-04-12
Real-world overall survival with palbociclib plus an aromatase inhibitor (AI) in patients with HR+/HER2− metastatic breast cancer (MBC) who are overweight/obese
Adam Brufsky, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA
A. Brufsky1, N. Iyengar2, X. Liu3, B. Li4, D. Makari3, L. McRoy3, A. Cohen5, M. Estevez6, V. Abramson7, R. Layman8, G. Curigliano9; 1Division of Hematology/Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, 2Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 3US Oncology Medical Affairs, Pfizer Inc., New York, NY, 4Global Biometrics & Data Management, Pfizer Inc., New York, NY, 5Research Oncology, Flatiron Health Inc., New York, NY, 6Research Sciences, Flatiron Health Inc., New York, NY, 7Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, 8Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 9Department of Oncology and Hemato-Oncology, University of Milano, European Institute of Oncology, IRCCS, Milano, ITALY.
Background A cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) combined with endocrine therapy (ET) has been the standard of care (SOC) as first-line (1L) treatment for hormone receptor-positive/HER2-negative (HR+/HER2−) MBC for the past decade. Palbociclib (PAL), the first CDK4/6i approved, received accelerated US approval in 2015 for HR+/HER2− MBC based on its demonstrated efficacy and safety profile. An increasing number of real-world (RW) studies have shown that 1L PAL+ET significantly improves progression-free survival, tumor response, and overall survival (OS) compared with ET alone across diverse HR+/HER2− MBC populations. Overweight and obesity are common conditions associated with an increased risk of breast cancer and can influence cancer treatment outcomes. However, little is known about the effectiveness of CDK4/6i+ET, specifically in patients (pts) with HR+/HER2− MBC who are overweight/obese in routine US clinical practice. Methods This was a retrospective analysis of deidentified longitudinal data from the Flatiron Health Research Database. Pts were included if they were overweight or obese (BMI ≥25 kg/m2), had HR+/HER2− MBC, and initiated 1L PAL+AI or AI between Feb 2015−Jul 2024. OS was defined as the number of months from start of PAL+AI or AI alone to death. Date of death was a consensus mortality endpoint based on electric health records, Social Security Death Index, and obituary data, validated against the National Death Index. Pts were retrospectively followed until death, last medical activity, or January 2025 (data cut-off), whichever came first. Stabilized inverse probability of treatment weighting (sIPTW) was used to balance pt characteristics. Cox proportional hazards models were used to estimate the relative effectiveness of PAL+AI vs AI alone. Results A total of 8076 pts were eligible for analysis, including 5009 (62.0%) treated with PAL+AI and 3067 (38.0%) treated with AI alone. Compared with AI-alone pts, those treated with PAL+AI were younger and more likely to have ≥2 metastatic sites and lung/liver involvement but less likely to have ECOG score ≥2. After sIPTW, pt characteristics were generally balanced. Unadjusted median OS (95% confidence interval [CI]) was 51.4 (49.3-53.4) months with PAL+AI and 41.1 (38.9-43.3) months with AI (hazard ratio [HR]= 0.75 (0.71-0.80), P<0.0001). After sIPTW, median OS (95% CI) was 50.7 (48.7-53.1) months with PAL+AI and 42.4 (39.8-44.7) months with AI (HR=0.79 [0.74-0.85], P<0.0001). Consistent results were observed with 1:1 propensity score matching as a sensitivity analysis. See Table for baseline pt characteristics and OS results. Conclusions Compared with AI alone, PAL+AI is associated with improved OS in pts with HR+/HER2- MBC who are overweight/obese in US real-world setting, supporting 1L PAL+AI as a SOC for pts with HR+/HER2- MBC who are overweight/obese.
Presentation numberPS2-04-13
Treatment Outcomes Among African American and Non-Hispanic White Breast Cancer Patients Treated with KEYNOTE-522
La-Urshalar Brock, Winship Cancer Institute at Emory University and Nell Hodgson Woodruff School of Nursing, Atlanta, GA
L. Brock1, K. Freeman2, A. A. Ashley McCook-Veal3, A. Labatut4, L. Lei5, S. Selimovic6, J. M. Switchenko7, N. A. Giordano6, M. Bhave8; 1Medical Oncology, Winship Cancer Institute at Emory University and Nell Hodgson Woodruff School of Nursing, Atlanta, GA, 2Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, 3Biostatistics Shared Resource, Winship Cancer Institute at Emory University, Atlanta, GA, 4Oncology Clinical Pharmacy – Breast, Winship Cancer Institute at Emory University, Atlanta, GA, 5Hematology and Medical Oncology, Emory University – School of Medicine, Atlanta, GA, 6Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, 7Biostatistics & Bioinformatics, Rollins School of Public Health and Winship Cancer Institute at Emory University, Atlanta, GA, 8Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA.
Background: Pembrolizumab is used with neoadjuvant chemotherapy in high-risk early-stage triple-negative breast cancer (TNBC).1 Results from the 2021, KEYNOTE-522 trial (KN-522) show improved pathological complete response (pCR) and event-free survival (EFS).1,2 Although TNBC accounts for ~15% of breast cancers, African American (AA) women experience higher incidence and worse outcomes than Non-Hispanic White women (NHW).3,4 Despite this, AA representation in immunotherapy clinical trials for breast cancer (e.g., KEYNOTE-522, IMPASSION-130, KEYNOTE-355) was <6%1,5,6, highlighting the need for real-world evaluations of treatment outcomes across racial groups. This retrospective study examines real-world toxicities and treatment outcomes of pembrolizumab combined with neoadjuvant chemotherapy in early-stage (I-III) TNBC, based on the KN-522 regimen, with a focus on racial differences between AA and NHW patients. Methods: We abstracted data from early-stage TNBC patients treated with the KN-522 regimen between 2021 and 2024 from an academic comprehensive cancer center. Descriptive analyses were conducted to compare clinical and safety outcomes among AA and NHW patients using ANOVA, Chi-square, or Fisher’s exact tests.7 Univariate and multivariable models evaluated associations between race and toxicity/treatment outcomes. Results: A total of 124 patients were identified: 80 Black (64.5%) and 44 NHW (35.5%), with a median age of 56.5 years (IQR: 42.5-66). Most had T2 tumors (54%), high-grade histology (84.6%), and at least one comorbidity (66.1%). Fifty percent achieved a pCR, while 5.6% experienced disease progression on KN-522, and 8.9% had local/distant recurrence. Significant racial differences were observed in age at treatment initiation (median: 59 years for AA vs. 51.5 years for NHW; p=0.018), BMI (mean: 32.2 vs. 29.1; p=0.026), and marital status (p<0.003). A higher proportion of NHW patients experienced anxiety (31.8% vs. 7.5%; p<0.001) and thyroid disorders (22.7% vs. 7.5%), while hypertension was more common in AA patients (57.5% vs. 29.5%; p=0.003). AA patients had a greater proportion of high-grade tumors (63.4% vs. 36.5%; p=0.576) and HER2-low expression (67.2% vs. 32.7%; p=0.233). pCR was achieved more among AA compared to NHW patients (65% vs. 35%). Although there were no statistically significant differences in overall outcomes across treatment or immune-related adverse events (irAEs), AA patients more frequently experienced treatment-related toxicities, including anemia (63.4% vs. 36.3%), grade 3/4 neutropenia (41.2% vs. 31.8%), and chemotherapy-induced peripheral neuropathy (60% vs. 43.1%). Additionally, treatment modifications related to the KN-522 regimen (p=0.965) and pembrolizumab (p=0.528) were more common among AA patients at 64.3% and 70.9% compared to 35.6% and 29.0% among NHW patients, respectively. Progression-free survival did not significantly differ between AA and NHW patients (p = 0.7562). Both groups exhibited high PFS rates at 12, 24, and 36 months. At 36 months post-treatment, overall survival probabilities for AA patients were 89.7% (95% CI: 76.6%-95.6%) and 97.3% (95% CI: 82.3%-99.6%) among NHW patients. Conclusion: This real-world analysis revealed that AA patients receiving KN-522 experienced higher rates of treatment-related toxicities, irAEs, and regimen modifications compared to NHW patients. Despite this, treatment effectiveness was comparable. These findings underscore the need for larger studies to validate outcomes and investigate factors influencing racial differences in treatment response.
Presentation numberPS2-04-14
Safety of Ribociclib for patients with hormone receptor-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer: the Royal Marsden experience
Abigail Pomeranc, Royal Marsden Hospital, London, United Kingdom
A. Pomeranc, N. Chukwuma, E. Crewe, C. Courtney, J. Din, W. Hodgson, R. Binoy, P. Mone, S. Shrestha, R. Smith, S. Butt, A. Chung, O. Cornwall, M. Aboulela, Z. Campbell, S. McGrath, S. Redana, A. Ring, S. Johnston, N. M. Battisti; Medical Oncology, Royal Marsden Hospital, London, UNITED KINGDOM.
Background: Ribociclib is approved in combination with an aromatase inhibitor (AI) or fulvestrant for patients with hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer, and more recently has been approved as an adjuvant treatment in combination with an AI for patients with HR-positive, HER2-negative early breast cancer at higher risk of recurrence. Therefore, evaluating its safety in a less selected patient population in the real world is key. We aimed to evaluate it in patients treated at our Institution. Methods: We retrospectively identified patients with HR-positive, HER2-negative breast cancer who received Ribociclib between December 2017 and February 2024 at our institution. Demographics, disease characteristics, blood tests and toxicities were recorded for 90 patients. Simple statistics were used as appropriate. Results: Ninety patients were included in the analysis with a median age of 58 years (range: 30-86). Out of these, 27 (24.3%) were <50 years old and 50 (55.6%) were white, 78 (92.9%) had an ECOG performance status of 0-1 and 26 (29.2%) were premenopausal. Thirteen patients (14.4%) were receiving Ribociclib in the adjuvant setting, while 77 (85.5%) with palliative intent. Out of those with advanced disease, 46 patients (59.7%) had visceral metastases and 74 (96.1%) bony metastases. Forty-five (54.2%) had grade 3 tumours. Among patients with complete safety data, 72 (82.8%) had any grade of neutropenia (grade 3-4 in 48 [55.2%]), thirty-one (36.0%) had any grade of deranged liver function (grade 3-4 in 7 [8.1%]), 40 (45.5%) had any grade of anaemia, 20 (22.7%) had any grade of nausea, 30 (34.0%) had any grade of fatigue, 13 (14.7%) had any grade of skin rash and 1 (1.1%) had any grade of interstitial lung disease; seven patients (9.1%) had QTc prolongation (grade 1 in all patients). Ribociclib was dose reduced in 35 patients (41.7%) and discontinued in 54 patients (62.8%): due to progression in 33 (61.1%), hepatotoxicity in 4 (7.4%), haematological toxicity in 3 (5.6%) and patient preference in 2 (3.7%). Conclusions: Our analysis of the safety profile of ribociclib in this population confirms a similar neutropenia and dose reduction rate compared with published trials findings. However, rates of anaemia and deranged liver function were higher in our patient population. These findings are key to inform decision-making in a less selected patient population in the real world, especially ahead of the increasing use of ribociclib in the curative setting.
| Characteristic | Category | n (%) | Characteristic | Category | n (%) |
| Age (n=90) | <50 | 27 (24.3%) | Grade (n=83 | Well differentiated | 4(4.8%) |
| 50 & above | 63 (75.7%) | Moderately differentiated | 34 (41.0%) | ||
| Sex (n=90) | Female | 90 (100%) | Poorly differentiated | 45 (54.2%) | |
| Male | 0 (0%) | Treatment intent (n=90) | Palliative | 77 (85.5%) | |
| Ethnicity (n= 90) | White | 50 (55.6%) | Curative | 13 (14.4%) | |
| Black | 6 (6.7%) | Endocrine Agent (n=90) | Tamoxifen | 1 (1.1%) | |
| Asian | 14 (15.6%) | Aromatase inhibitor | 75 (83.3%) | ||
| Mixed | 3 (3.3%) | Fulvestrant | 14 (15.6%) | ||
| Other | 17 (18.8%) | Ovarian function suppression (n=90) | No | 60(66.7%) | |
| ECOG PS (n=84) | 0 | 47 (55.9%) | Yes | 30 (33.3%) | |
| 1 | 31 (36.9%) | Disease Progression (n=85) | No | 46 (54.1%) | |
| 2 | 5 (6.0%) | Yes | 39 (45.9%) | ||
| 3 | 1 (1.2%) | Treatment after ribociclib (n=58) | Chemotherapy | 30(51.7%) | |
| Menopausal Status (n=89) | Pre-menopausal | 26(29.2%) | Endocrine | 21(36.2%) | |
| Peri-menopausal | 7 (7.9%) | Trial | 2 (3.5%) | ||
| Post-menopausal | 56 (62.9%) | No further treatment | 5 (8.6%) | ||
| Tumour Characteristics (n=77) | Visceral metastasis | 46 (59.7%) | |||
| Bony metastasis | 74 (96.1%) | ||||
| Nodal metastasis | 56 (72.7%) | ||||
| Chest wall metastasis | 9 (11.7%) |
Presentation numberPS2-04-15
Real-world outcomes of sacituzumab govitecan in patients with pretreated triple-negative breast cancer and brain metastases: data from a central European cohort
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
J. Żubrowska1, M. Kubeczko2, M. Pieniążek3, A. Polakiewicz-Gilowska2, I. Kolářová4, M. Malejčíková5, L. Rusinova6, M. Holánek7, R. Soumarová8, K. Winsko-Szczęsnowicz9, A. Konieczna10, A. Młodzińska10, D. Krejčí11, I. Danielewicz12, M. Szymanik-Resko12, T. Ciszewski13, M. Lisik-Habib14, A. Pękala14, H. Študentová15, J. Šustr16, A. Rudzińska17, B. Czartoryska-Arłukowicz9, A. Łacko3, M. Jarząb2, R. Pacholczak-Madej18, Z. Bielčiková19, M. Püsküllüoğlu20; 1Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, POLAND, 2Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 31. Department of Oncology 2. Breast Unit Clinical Oncology Day Care Department, 1. Wrocław Medical University, 2. Lower Silesian Comprehensive Cancer Center, Wrocław, POLAND, 4Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Králové, CZECH REPUBLIC, 5Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 6Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 7Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, and Masaryk Memorial Cancer Institute, Brno, CZECH REPUBLIC, 8Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 9Department of Clinical Oncology, M. Skłodowska-Curie Bialystok Oncology Center, Białystok, POLAND, 10Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, POLAND, 11Department of Oncology, First Faculty of Medicine, Charles University in Prague, and Bulovka University Hospital, Prague, CZECH REPUBLIC, 12Oddział Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 13Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 14Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 15Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 16Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University, and University Hospital Pilsen, Plzen, CZECH REPUBLIC, 17Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków, POLAND, 18Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND, 19Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 20Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND.
Background Patients with metastatic triple-negative breast cancer (mTNBC) and brain metastases (BM) constitute a subgroup with poor prognosis and limited treatment options. In the pivotal ASCENT trial, sacituzumab govitecan (SG) demonstrated clinical benefit in pretreated patients with mTNBC. An exploratory subgroup analysis of ASCENT trial included 61 patients with radiologically stable BM (32 in the SG arm), showing a modest improvement in progression-free survival (PFS) with SG versus chemotherapy (2.8 vs. 1.6 months), but no overall survival (OS) advantage (6.8 vs. 7.5 months). While suggestive of intracranial activity, these findings were derived from a small, highly selected population. Given the limited evidence in this setting, we aimed to evaluate the effectiveness and safety of SG in a real-world cohort of patients with active or treated BM Materials and Methods This retrospective multicenter study included female patients with mTNBC and radiologically confirmed BM who initiated SG between August 2021 and May 2025 across 13 oncology centers in the Czech Republic, Poland and Slovakia. Clinical data were extracted from patients’ medical records. Collected variables included baseline characteristics, number and size of BM lesions, prior neurosurgical resection, use and timing of central nervous system (CNS)-directed radiotherapy, treatment response, and adverse events (AEs) (graded per CTCAE v5.0). Radiologic responses were assessed using RECIST 1.1. Overall response rate (ORR), PFS, and OS were evaluated. Survival outcomes were estimated using the Kaplan-Meier method. Univariable Cox proportional hazards models were used to assess the association between BM burden and survival. Statistical analyses were performed in R (v4.3.3); p < 0.05 was set as significant. Results A total of 29 patients with mTNBC and confirmed BM at SG initiation were included. The median number of BM was 3 with median size of the biggest lesion 16.5 mm. Neurosurgical resection had been performed in 4 (14.3%) and CNS-directed radiotherapy in 28 (89.3%) patients prior to SG initiation. The median number of prior palliative systemic therapy lines was 2 (range: 1-3). The median follow-up was 6.05 months (IQR: 3.66-11.06; range: 0.79-27.47). At the time of analysis, 22 patients had died and 26 had the treatment with SG discontinued. The median OS was 8.9 months. Estimated OS rates at 3, 6, 9, and 12 months were 85.2%, 60.3%, 42.3%, and 28.2%, respectively. Median PFS was 3.09 months, with 3-, 6-, 9-, and 12-month PFS rates of 53.6%, 31.4%, 22.5%, and 12.0%, respectively. ORR was observed in 8 of 26 evaluable patients (30.8%). CNS progression was documented in 6 patients (23.1%). Neither the number nor the size of CNS lesions showed a statistically significant association with OS or PFS. SG administration required dose delays due to AEs in 15 patients (51.7%), and dose reductions in 11 (37.9%). Two cases (7.7%) of treatment discontinuation due to toxicity were reported. No unexpected new safety signals were observed. Conclusions SG demonstrated clinically meaningful activity and a manageable safety profile in this real-world cohort of patients with mTNBC and BM. Median PFS and OS outcomes appeared comparable or numerically favorable to those reported in the ASCENT BM subgroup analysis. Neither the number nor size of BM lesions significantly impacted outcomes, suggesting SG may be effective irrespective of intracranial disease burden. These data may support broader consideration of SG in routine management of CNS-involved TNBC.
Presentation numberPS2-04-16
Single institution retrospective analysis of antibody-drug conjugate (ADC) sequencing in HR+/HER2-low and HR-/HER2-low metastatic breast cancer.
Austin Kordic, City of Hope, Duarte, CA
A. Kordic, N. Ruel, N. Kethireddy; Medical Oncology, City of Hope, Duarte, CA.
Purpose: Antibody-drug conjugates (ADC) have made a profound impact on the treatment landscape of HR+/HER2-low and HR-/HER2-low metastatic breast cancer. Sacituzumab govitecan (Trodelvy) and trastuzumab deruxtecan (Enhertu) are ADC with topoisomerase I payloads bound to anti-Trop-2 and anti-HER2 monoclonal antibodies, respectively. Enhertu received FDA approval in 2022 based on the results of the DESTINY-Breast04 study as did Trodelvy in 2023 per the TROPiCS-02 study. The optimal sequencing of these agents (ET versus TE) remains a topic of debate and investigation in this patient population.Methods: Our retrospective, single center study evaluated 100 patients with HR+/HER2-low and HR-/HER2-low metastatic breast cancer who were treated sequentially with Enhertu and Trodelvy (ET vs TE) at the City of Hope Comprehensive Cancer Center from 01/2019-12/2023. Both groups had received at least one prior line of therapy in the metastatic setting. The primary objective was to estimate the median combined time to progression following ET vs TE (PFS1+ PFS2) to determine if the PFS for the 2nd ADC declined significantly based on the sequence. PFS data was censored per the end-time point of the second treatment. Secondary endpoints included response rates (RR), treatment-related AE, and overall survival (OS). Patient demographics, treatment history, presence of visceral disease, and comorbidities were evaluated. Adverse event grading and investigator-assessed radiographic treatment response was determined using CTCAE v5.0 and RECIST 1.1 definitions, respectively. High-grade AE (hg-AE) was defined as Grade 3 or higher. HR and HER2 IHC status were assessed based on the most recent biopsy sample before each line of therapy. HR+ status included patients with 10% or greater ER/PR positivity and HER2-low included patients with IHC 1+/2+ with a negative FISH.Results: Most patients received ET (70/100) vs TE (30/100). The primary endpoint of PFS1 + PFS2 was comparable between the sequences (ET: 11.1 months vs TE: 8.2 months; log-rank p=0.7). The median PFS for Enhertu (ET: 6.8 months vs TE: 2.3 months; p < 0.0001) and Trodelvy (TE: 5.6 months vs ET: 2.5 months; p = 0.0006) was significantly increased when used first in the sequence. A similar trend was noted with RR for Enhertu (ET 65.7% vs TE 33.3%; p = 0.003) and Trodelvy (63.3% vs 25.7%; p = 0.0004) when given first in the sequence. Median OS was comparable between ET vs TE (22.9 months vs 17.5 months; p = 0.7). Neutropenia and GI toxicities were the most commonly reported AE with a similar rate of cumulative hg-AE in patients who received ET vs TE. In the ET group, there were significantly lower rates of hg-AE while on Enhertu compared to Trodelvy (28.5% vs 51.4%; p = 0.006), whereas rates were comparable (23.3% vs 40%; p = 0.2) in the TE group. The presence of baseline lung metastases was significantly higher in the ET group with 10 reported ILD events in the ET group vs 0 in the TE group. There were significantly more HR+ patients in the ET group, while HER2 IHC status was similar between groups (1+: 61% vs 2+: 28%).Conclusion: Patients with HR+/HER2-low and HR-/HER2-low metastatic breast cancer who received sequential ADC therapy with Enhertu and Trodelvy (ET or TE) had comparable PFS 1 + PFS2 and OS results. There were significant increases in PFS and RR with each ADC when used first compared to second in the treatment sequence. There were similar rates of cumulative hg-AE between the ET and TE groups with less hg-AE events in patients who received Enhertu with ET vs TE. Limitations of this study include the retrospective analysis and exclusion of patients who did not receive both drugs sequentially in the context of varied timelines of FDA approval. Prospective studies are needed to further evaluate the impact of ADC sequencing on treatment outcomes and toxicities.
Presentation numberPS2-04-17
Clinical and pathological characteristics of early hormone receptor-positive, HER2 negative breast cancer patients in Panama
Cristiane Martin, Instituto Oncológico Nacional, panama, Panama
C. Martin, o. castillo; Medical Oncology, Instituto Oncológico Nacional, panama, PANAMA.
Clinical and pathological characteristics of early hormone receptor-positive, HER2-negative breast cancer patients in Panama.Cristiane Martin-Palacios, Miguel Manzano, María Lim, Jonathan Quintero, José Amador, José Pinto, Taysser Sowley, Yaribeth Muñoz, Omar Castillo-Fernandez.Background: Breast cancer is the most common malignancy and a leading cause of mortality among women. In Latin America, clinical characteristics and treatment outcomes vary by region. Improving early detection and treatment has enhanced prognosis. In Panama, limited data exists, so gathering information on early hormone receptor-positive and HER2-negative breast cancer patients can help develop better treatment strategies.Methods: This retrospective study aims to describe the clinical and pathological characteristics of early hormone receptor-positive, HER2-negative breast cancer patients treated at the Instituto Oncologico Nacional between January 2017 and December 2019. Data was summarized using frequencies and percentages. We utilized Kaplan-Meier curves to analyze survival and recurrence rates. Recurrence factors were assessed through univariate analysis with the log-rank test and multivariate analysis using Cox regression.Results: We analyzed 368 patients, primarily women (98.9%, n = 365). The median age was 58 years. Most were postmenopausal (68.3%), and 55% had N0 disease. Staging showed 32.5% in stage I, 45.8% in stage II, and 21.1% in stage III. Regarding grade, 83% were grade 1 or 2, while 17% were grade 3. The median tumor size was 2.5 cm. Estrogen receptor positivity was 90%, and progesterone receptor positivity was 60%. In adjuvant treatment, 24.9% received tamoxifen, 56.8% aromatase inhibitors, and 17.6% sequential therapy.At the follow-up on May 31, 2025, 15.5% of patients had local or distant recurrence, and 10.3% had died; the median time to recurrence-free survival was not reached. Univariate analysis identified several risk factors for recurrence: premenopausal status (HR 1.76 CI 1.04-2.97; p=0.032), tumor size >2 cm (HR 2.40 CI 1.33-4.33; p=0.03), positive lymph nodes (HR 2.40 CI 1.33-4.33; p=0.03), grade 3 tumors (HR 2.08 CI 1.16-3.70; p=0.01), high nodal rate (HR 2.97 CI 1.59-5.54; p=0.001), stage III (HR 4.2), and use of aromatase inhibitors versus tamoxifen (HR 1.89). Multivariate analysis showed only stage III disease (HR 2.91 CI 2.49-7.04; p<0.001) and adjuvant tamoxifen use versus aromatase inhibitors (HR 2.27 CI 1.14-4.54; p = 0.02) were significantly associated with recurrence, while grade 3 tumors showed a trend toward significance (HR 1.84 CI 0.98-3.44; p = 0.055 ).Conclusion: Stage III, high-grade tumors, along with the use of adjuvant tamoxifen, are associated with a higher risk of recurrence. This underscores the need for strategies such as using GnRH analogs in premenopausal patients and implementing adjuvant CDK4/6 inhibitors for those at high risk within our institution.
Presentation numberPS2-04-18
Long-term safety in breast cancer patients receiving doxorubicin-cyclophosphamide chemotherapy with secondary G-CSF prophylaxis: A single-centre prospective study
Eliana Rohr, McGill University, Montréal, QC, Canada
E. Rohr1, T. Alotaibi2, F. Moria3, N. Bouganim4; 1Department of Internal Medicine, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, CANADA, 2Department of Medical Oncology, The Medical Services of the Ministry of Interior, Riyadh, SAUDI ARABIA, 3Faculty of Medicine, King Abdulaziz University, Jeddah, SAUDI ARABIA, 4Department of Medical Oncology, McGill University Health Centre, Montréal, QC, CANADA.
Introduction: Granulocyte colony-stimulating factors (G-CSF) are essential supportive agents for patients receiving myelosuppressive chemotherapy, reducing febrile neutropenia (FN) and preserving chemotherapy dose intensity, especially in high-risk or dose-dense regimens.¹⁻³ Guidelines recommend “primary prophylaxis” for patients at high risk for FN, while “secondary prophylaxis” is reserved for those with FN or borderline neutrophil counts during earlier cycles. While primary G-CSF benefits are well-documented, data on secondary prophylaxis, particularly in intermediate-risk regimens such as adjuvant doxorubicin-cyclophosphamide (AC) chemotherapy, are limited. Some studies suggest secondary G-CSF may reduce hospitalizations and improve treatment delivery, but robust, real-world, long-term safety data are lacking.⁵,⁶ A possible signal between G-CSF and secondary malignancies have been suggested.⁷ Accordingly, evaluation of the long-term safety of secondary prophylaxis is warranted. This study prospectively evaluated long-term safety and outcomes of secondary G-CSF prophylaxis in breast cancer patients treated with q3-weekly AC chemotherapy at the McGill University Health Centre. Methods: We conducted an interim analysis of a prospectively maintained database at the McGill University Health Centre (2016-2025). Ninety-eight patients who received q3-weekly AC chemotherapy (doxorubicin 60 mg/m² and cyclophosphamide 600 mg/m²) in the adjuvant or neoadjuvant setting, were included. Patients treated with dose-dense regimens or meeting criteria for primary G-CSF prophylaxis were excluded. Secondary G-CSF (filgrastim or pegfilgrastim) was administered at physician discretion to prevent dose reductions or delays. Key outcomes, including metastatic recurrence, mortality, overall survival, and follow-up time, were assessed using multivariable logistic regression and Cox proportional hazards models, adjusting for age, stage at diagnosis, and tumour subtype. All statistical analyses were conducted using R (v4.5.1). Results:Of 98 patients, 44 (45%) received secondary G-CSF. Fifty-eight percent of patients received G-CSF at cycle 2. Baseline characteristics were balanced between groups. No statistically significant difference in the incidence of new metastases was observed between the G-CSF and non-G-CSF groups (7.5% vs. 19.6%; adjusted OR 0.25, 95% CI 0.04-1.09; p = 0.09). No significant difference in all-cause mortality was observed (25% vs. 13%; adjusted OR 2.09, 95% CI 0.61-7.70; p = 0.25). Survival outcomes were comparable (5-year OS: 86.4% vs. 90.7%; adjusted HR 1.92, 95% CI 0.71-5.18; p = 0.20). Median follow-up among survivors was similar (96.8 vs. 94.6 months), with five-year follow-up exceeding 84% in both cohorts. No secondary hematologic malignancies occurred in either group. Conclusion: In this real-world cohort, secondary G-CSF prophylaxis during q3-weekly AC chemotherapy was not associated with significant differences in all-cause mortality between groups. No secondary hematological malignancies were observed. These results support the liberal use of secondary G-CSF in curative-intent regimens to prevent delays or reductions in dose intensity. Continued follow-up of this cohort is warranted to confirm long-term safety of liberal G-CSF use.
Presentation numberPS2-04-19
10-year survival outcomes of early-staged breast cancer patients with intermediate- and high- risk hormone receptor-positive her2-negative disease
Winnie Yeo, Chinese University of Hong Kong, Hong Kong, Hong Kong
W. Yeo1, L. Yuen2, C. Kwok3, I. Soong4, J. Chiu5, T. Ng6, S. Chan7, T. Wong2, C. Yolanda8, L. Lawrence9, C. Yau9, W. Hung9, C. Polly9; 1Clinical Oncology, Chinese University of Hong Kong, Hong Kong, HONG KONG, 2Surgery, Hong Kong Breast Cancer Foundation, Hong Kong, HONG KONG, 3Clinical Oncology, Princess Margaret Hospotal, Hong Kong, HONG KONG, 4Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong, HONG KONG, 5Clinical Oncology, University of Hong Kong, Hong Kong, HONG KONG, 6Clinical Oncology, Tuen Mun Hospital, Hong Kong, HONG KONG, 7Surgery, United Christian Hospital, Hong Kong, HONG KONG, 8Clinical Oncology, CUHK Medical Centre, Hong Kong, HONG KONG, 9Clinical Oncology, Hong Kong Breast Cancer Foundation,, Hong Kong, HONG KONG.
Background:Adjuvant endocrine therapy (ET) therapy combining with CDK4/6 inhibitors have improved outcomes of early-staged breast cancer (EBC) patients (pts) with intermediate- or high- risk HR-positive HER2-negative disease based on MonarchE and NATALEE studies. The objective of this study was to assess the invasive disease-free survival (iDFS) of HR-positive HER2-negative EBC patients prior to the use of adjuvant CDK4/6 inhibitors. Methods: This is a retrospective study based on data extraction from the Hong Kong Breast Cancer Registry. Pts from 11 public and 4 private hospitals/clinics were consented for data collection. The study protocol was approved by the regional ethics committee of individual units. Pts with HR-positive HER2-negative EBC who underwent definitive surgery between January 2006 and December 2011 were studied. The inclusion criteria of NATALEE were applied. Pts were divided into two groups: group 1, those who satisfied NATALEE criteria (considered to have intermediate- or high- risk); group 2, those not eligible for NATALEE and had stage 1 cancers. Pts background characteristics were collected. iDFS was evaluated using the Kaplan-Meier method. Descriptive statistics were used to report the clinical outcomes between the two groups. Results: A total of 3512 pts were included; 1885 (53.7%) in group 1 and 1627 (46.3%) in group 2. The median age was 50.2 years (range: 24.2-101.4). 1624 (46.2%) were premenopausal, 1670 (47.6%) were postmenopausal while 218 (6.2%) had unknown menopausal status at diagnosis. 883 (25.2%) had grade 3 cancers. High tumour Ki67 (defined as >/=20%) was reported in 790 (25.2%) patients. 2459 (70.0%) received adjuvant radiotherapy, 2230 (63.5%) underwent adjuvant chemotherapy. 3341 patients received adjuvant ET (95.1%). For group 1 vs group 2, the 10-year iDFS were 77.1% vs 89.6% respectively. iDFS data according to age, menopausal status, grading, Ki-67, use of adjuvant chemotherapy and radiotherapy, are listed in Tabel 1. Conclusions: The present study confirmed that in the era prior to adjuvant ET plus CDK4/6 inhibitor, patients who had intermediate- or high- risk HR-positive HER2-negative EBC had worse prognosis. Among those who would have satisfied NATALEE criteria (group 1), older age and postmenopausal at diagnosis had significantly worse iDFS, while use of adjuvant chemotherapy was associated with better iDFS. For those with stage 1 EBC (group 2), older age and high tumour grade had significantly worse iDFS, while having receiving adjuvant radiotherapy was associated with better iDFS. Real-world studies such as the present one provide informative data and further studies may enable better determination of appropriate patient population for the use of adjuvant CDK4/6 inhibitor treatment
| Subgroups | Group 1 | Group 2 | |
| Age | <65 years | 90.4% | 78.1% |
| >/=65 years | 83.0% | 69.8% | |
| p-value | 0.004 | 0.016 | |
| Menopausal status | Premenopausal | 90.4% | 79.8% |
| Postmenopausal | 89.2% | 74.6% | |
| p-value | >0.05 | 0.011 | |
| Tumour grade | Grade 1-2 | 90.2% | 78.0% |
| Grade 3 | 84.9% | 75.1% | |
| p-value | 0.006 | >0.05 | |
| Ki-67 | Low | 91.1% | 87.7% |
| High | 78.9% | 76.2% | |
| p-value | >0.05 | >0.05 | |
| Use of adjuvant chemotherapy | Yes | 90.1% | 78.5% |
| No | 89.3% | 63.9% | |
| p-value | >0.05 | <0.001 | |
| Use of adjuvant radiotherapy | Yes | 92.5% | 77.6% |
| No | 85.9% | 75.1% | |
| p-value | <0.001 | >0.05 |
Presentation numberPS2-04-20
Real-world efficacy of everolimus in ER+/HER2- metastatic breast cancer following CDK4/6 inhibitors: a multicenter registry-based study
Hikari Kiyohara, Tokai University School of Medicine, Isehara, Japan
H. Kiyohara1, T. Hanamura1, Y. Sagara2, M. Miyashita3, S. Saji4, N. Niikura1; 1Depertment of Breast Oncology, Tokai University School of Medicine, Isehara, JAPAN, 2Department of Breast and Thyroid Surgical Oncology, Social Medical Corporation Hakuaikai Sagara Hospital, Kagoshima, Japan., Kagosima, JAPAN, 3Department of Breast and Endocrine Surgical Oncology, Tohoku University School of Medicine, Sendai, JAPAN, 4Department of Medical Oncology, Fukushima Medical University, Fukushima, JAPAN.
Objective: Everolimus combined with endocrine therapy is one of the standard treatment options for patients who develop resistance to CDK4/6 inhibitors. However, prospective studies and real-world evidence regarding the efficacy of everolimus in patients previously treated with CDK4/6 inhibitors remain limited. This retrospective study evaluated the clinical outcomes of everolimus in such patients and aimed to identify factors associated with treatment response. Methods: We conducted a retrospective, registry-based analysis using data from the Advanced Breast Cancer Database (ABCD) Project, coordinated by the Japan Breast Cancer Research Group. Of 184 patients treated with everolimus plus endocrine therapy between January 2019 and March 2025, 169 ER+/HER2- cases with prior CDK4/6 inhibitor exposure and complete data were included. Collected variables included age, performance status, treatment line, reason for treatment discontinuation, best response, menopausal status, recurrence type, BRCA1/2 mutation status, histology, ER/PgR/HER2 expression, Ki-67 index, prior chemotherapy, antibody-drug conjugate (ADC) use, and type and duration of prior CDK4/6 inhibitor therapy. Sites and number of metastatic organs were also assessed. Time to treatment failure (TTF) and overall survival (OS) were measured from everolimus initiation. Prognostic factors for TTF were analyzed using univariate and multivariate Cox proportional hazards models. Kaplan-Meier methods and log-rank tests were used for survival comparisons. Results: The median age was 63 years, and 73% of patients had a performance status of 0. Treatment was discontinued due to disease progression in 64%. The objective response rate (CR + PR) was 9%. Most patients were postmenopausal (96%), with 30% presenting with de novo stage IV disease and 69% experiencing post-operative recurrence. Pathogenic BRCA1/2 variants were observed in 6%. Invasive ductal carcinoma accounted for 88% of cases. ER was ≥10% in 98%, PgR ≥10% in 62%, and HER2-low breast cancer (IHC score 1+ or 2+ with ISH-negative) was identified in 63%. The median Ki-67 index was 28%. Everolimus was administered within the third line in 62% of patients. CDK4/6 inhibitor therapy lasted less than one year in 51%; palbociclib only was used in 56%, abemaciclib only in 24%, and both agents in 20%. Prior chemotherapy and ADC therapy were given in 31% and 1%, respectively. Metastatic sites (not mutually exclusive) included bone (64%), liver (41%), lung (34%), and brain (2%), while distant lymph node and pleural involvement were observed in 28% and 17%, respectively. Bone-only metastasis was reported in 12%. The number of metastatic organs was one in 19%, two in 50%, and three or more in 30%. The median TTF was 4.1 months, and the median OS was 28.7 months. Univariate analysis identified several factors significantly associated with shorter TTF: non-ductal histology, PgR <10%, prior CDK4/6 TTF <1year, previous chemotherapy, prior ADC use, and ≥4 metastatic sites (all p < 0.05). In multivariate analysis, non-ductal histology (HR 2.34), prior chemotherapy (HR 2.28), and ≥4 metastatic sites (HR 1.83) remained significant predictors of inferior TTF. A progressive decline in TTF was observed with increasing numbers of risk factors (log-rank p < 0.05). Conclusion: This large-scale real-world study provides clinically meaningful insight into the effectiveness of everolimus following CDK4/6 inhibitors in ER+/HER2- metastatic breast cancer. Several clinicopathologic factors independently predicted reduced treatment durability, which may inform post-CDK4/6 treatment decision-making. Importantly, to our knowledge, this represents the largest real-world cohort reported to date in this setting, supporting the strength and generalizability of the findings.
Presentation numberPS2-04-21
Impact of Pre-Neoadjuvant Radioactive Iodine Seed Marking on Pathological Complete Response and Survival in Breast Cancer Patients
Yihao Geng, Shandong Provincial Tumor Hospital, Shan Dong, China
Y. Geng, Q. Zhang, Z. Shi, Z. Bi, Y. Wang, P. Qiu; Breast Surgery Ward I, Shandong Provincial Tumor Hospital, Shan Dong, CHINA.
Background:Pre-neoadjuvant marking of the primary tumor and biopsy-confirmed metastatic axillary lymph nodes is critical for enabling de-escalation of surgery, facilitating the retrieval of the lesion and assessing treatment response. Radioactive iodine-125 (125I) seeds are commonly used for localization. However, potential radiobiological effects from 125I seeds could confound pathological assessment of treatment response, possibly leading to suboptimal adjuvant therapy decisions and compromised long-term prognosis. We evaluated the pathological and prognostic impact of 125I seed marking before Neoadjuvant therapy (NAT) and employed peripheral blood profiling to explore immune-related mechanisms.Methods:We conducted a retrospective analysis of 1,594 cT1-4N1-3M0 breast cancer patients who received NAT treatment at Shandong Cancer Hospital and Institute during 2017 to 2024. 216 patients with and 432 without 125I seed marking were included after a 1:2 propensity score match. The primary endpoint was pCR; the secondary was invasive disease-free survival (iDFS). To investigate immune modulation, peripheral blood samples from triple-negative breast cancer (TNBC) patients were collected at baseline as well as after second NAT cycle for flow cytometry (CD3⁺/CD4⁺/CD8⁺/CD16⁺/CD19⁺).Results:The 125I marked group achieved significantly higher pCR rates than the without 125I seed marked group: total pCR (55% vs. 46%, P=0.030), breast pCR (63% vs. 53%, P=0.016), and axillary pCR (71% vs. 60%, P=0.009). Subgroup analyses highlighted the greatest benefits in TNBC (tpCR:59% vs. 39%, P=0.033) and hormone receptor-positive/HER2-negative (HR+/HER2−) subtypes (tpCR:26% vs. 8%, P=0.028). After a median follow-up of 36.7 months, 50 iDFS events were recorded. the 3-year iDFS was significantly higher in the 125I seed marked group (96% vs. 90%; P = 0.018). Kaplan-Meier survival analysis based on pCR achievement showed that patients achieved pCR in both groups had a lower risk of recurrence compared to non-pCR patients (125I seed marked group: 99% vs. 83%, P = 0.003; without 125I seed marked group: 96% vs. 85%, P = 0.021).Importantly, within the pCR patients, Both groups exhibited similarly favorable prognosis, regardless of 125I seed marked (99% vs. 96%, P = 0.875).Flow cytometry showed increased CD8⁺ T cells and CD16⁺ NK cells, and reduced CD4⁺ T cells in 125I marked patients. Conclusions:125I seed marking before NAT improves pCR rates, especially in TNBC and HR+/HER2− subtypes. As pCR is strongly associated with improved iDFS, this increase in pCR likely translates into survival benefit. Flow cytometry suggests that localized radiation at the 125I seed marking may promote CD4⁺ T cell trafficking and Treg exhaustion, thereby enhancing effector and cytotoxic immune responses. These findings support the immunologic relevance of seed localization and provide a rationale for future immunotherapy combinations.
Presentation numberPS2-04-22
Real world data of primary resistance to cyclin-dependent kinase 4/6 inhibitors treatment in hormone-receptor positive metastatic breast cancer
Alicia Carrasco Navas, Consorci Corporació Sanitària Parc Taulí, Sabadell, Spain
O. Cano i Cano1, M. Fragío Gil1, A. Carrasco Navas1, M. Busquets Godall1, A. Ribas Bravo1, O. Civit Figueras1, C. Marin1, J. Sandoval Alves1, I. Gallego Bernardo1, M. Monroe Rivera1, S. Karzazi Casin1, J. Arqué Garrido1, M. Sierra Boada1, S. Soriano Sánchez1, J. Giner Joaquin1, P. Ribera Fernandez1, L. Vilà Martínez1, R. Querol Niñerola1, T. Bonfill Abella1, E. Gallardo Diaz1, L. Medina Ortega1, B. Calvo Martinez1, M. Rosich Peris1, L. Soriano Gutierrez2, S. Redondo Capafons2, N. Gaztelumendi Corcoles3, P. Andreu Cobo1, G. Llort Pursals1, E. Dalmau Portulas1, M. Seguí Palmer1, L. Fernández-Morales1; 1Medical Oncology, Consorci Corporació Sanitària Parc Taulí, Sabadell, SPAIN, 2Medical Pharmacology, Consorci Corporació Sanitària Parc Taulí, Sabadell, SPAIN, 3Clinical Trials Unit, Consorci Corporació Sanitària Parc Taulí, Sabadell, SPAIN.
Background: Around 30% of patients (pts) with breast cancer develops metastases and 70 % of pts with metastatic breast cancer (MBC) are hormone-receptor (HR)-positive/HER2-negative. Endocrine therapy (ET) combined with cyclin-dependent kinase 4/6 inhibitors (CDKi) has become the standard of care in this setting, except in cases of life-threatening disease. However, a major challenge is to overcome endocrine resistance, either primary or secondary, which limits the long-term efficacy of ET and worsens prognosis. Methods: Here, we analysed the clinical characteristics of pts at our hospital who develop primary resistance to CDKi plus ET for MBC. In a cohort of 180 pts who received ET plus CDKi for MBC between 2017-2024 in Hospital Parc Taulí, we identified 46 pts with primary resistance in the metastatic setting so we conducted a retrospective observational study to analyse clinical, treatment, efficacy and survival variables on this group of pts. We defined primary resistance as a relapse within 2 years of adjuvant ET or disease progression during the first 6 months of first-line ET for MBC and secondary resistance as a relapse that occurs after at least 2 years of adjuvant ET or within the first year after its ending. Results: 46 patients (pts) were included, median age was 52 years, 52 % were postmenopausal and 44 % had high-proliferation disease. At the diagnosis 80% had early disease and among these, 33 % received adjuvant chemotherapy and 84 % received adjuvant ET. In this group who received adjuvant treatment, 5 pts (16%) had primary resistance, 24 (77%) secondary resistance and only 2 pts had ET sensitivity. Only 7 pts (15%) in the cohort were treatment naïve. As CDKi 65% received Palbociclib, 26% Abemaciclib an 9% Ribociclib. As ET 2% received Anastrozol, 2% Exemestano, 56% Fulvestrant and 40% Letrozol. ET plus CDKi was received as first line in 41% of the pts and in 59% as second line or beyond. The majority of patients received CDKi as second-line therapy since, at the beginning of this study, they were not yet approved for first-line use.Among those treated in second line or beyond, 48 % had ET and 52 % chemotherapy as first-line treatment. Overall median progression free survival was 3 months, 4 months in pts with primary resistance in the adjuvant setting, 3 months in pts with secondary resistance, and 2 months in hormone sensivity pts (p=0.231). Median overall survival (OS) was 9 months. In the subgroup analysis, median OS was 13 months for previous primary resistance, 8 months for secondary resistance, and 12 months for hormone sensivity pts (p=0.276). Of note, OS was 8 months in pts who received CDKi as first-line and 11 months in the second line or beyond (p=0.579). This may be due to the fact that there were no patients with previous secondary resistance in the second-line or beyond group. Conclusions: Findings from this cohort suggest that pts with primary resistance in the metastatic setting has poor outcomes and supports the data that in this context pts with previous primary resistance has better results that pts with secondary resistance in the adjuvant setting, similar to the findings in the MONARCH 2 trial. One limitation of this real world data is that this is a small group of pts and the lack of PIK3CA mutation determination which is not currently analysed routinely prior to starting first-line treatment.
Presentation numberPS2-04-23
Poor response to systemic therapy upon progression on cyclin dependent kinase 4/6 inhibitors in HR+ Inflammatory Breast Cancer
Azadeh Nasrazadani, MD Anderson Cancer Center, Houston, TX
A. Nasrazadani1, M. Kai2, R. Tidwell3, B. Lim1, V. Valero1, R. Layman1, The MDACC Inflammatory Breast Cancer Team, W. Woodward4; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, MD Anderson Cancer Center, Houston, TX, 3Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, 4Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX.
BACKGROUND Inflammatory breast cancer (IBC) is an aggressive subtype of breast cancer, which exhibits an aggressive clinical course regardless of stage at diagnosis or molecular subtype. We have previously reported on the suboptimal outcomes and high recurrence rates of patients with hormone receptor positive (HR+) IBC, and notably have reported short duration of response to standard of care CDK4/6 inhibitors (CDKI) in the first line metastatic setting. Herein, we report time on treatment (ToT) on systemic therapies in the 2nd line metastatic setting for patients with HR+ IBC. METHODS Patients with HR+ HER2- IBC for which CDKI was administered in the first line metastatic setting were identified from the IBC registry at MD Anderson Cancer Center (N = 36). Patients unable to tolerate CDKI therapy were excluded from this analysis (N = 3). Therapies utilized and ToT (months) in the 2nd line setting was evaluated as reported by the treating physician. RESULTS Among N = 33 patients evaluated, upon progression on standard of care CDKI and endocrine directed therapy (ET) combinations, patients received the following therapies with median ToT (months (min-max ToT)), respectively: capecitabine (N = 7; 3 (2-5)), everolimus/ET (N = 6, 3 (1-4)), Abraxane (N = 3, 3(2-10)), Eribulin (N = 2, 2.5(2-3)), capivasertib/fulvestrant (N = 1, 6(6)), taxol (N = 1, 5(5)), elacestrant (N = 1, 3(3)), fulvestrant (N = 1, 3(3)), tamoxifen (N = 1, 1(1)), intrathecal topotecan (N = 1, 1(1)), non standard/clinical trial (N = 4, 2.5(1-4)). Notably, 5 patients (15%) succumbed to death while on first line CDKI therapy due to progression of disease. CONCLUSION Patients with metastatic HR+ IBC demonstrate poor response to ET and cytotoxic chemotherapies upon progression on first line standard CDKI/ET, with a disproportionately high rate of death occurring in the first line setting due to advanced disease. Given previously reported low ToT on CDKI/ET in the first line and high incidence of brain relapse in this population, clinical trial design utilizing targeted therapies and more novel approaches in earlier settings are worthy of consideration.
Presentation numberPS2-04-25
Efficacy of maintenance endocrine therapy combined with trastuzumab + pertuzumab following taxane induction in HR+/HER2+ metastatic breast cancer: A real-world reproduction of the PATINA trial control arm.
Laurent Mathiot, Institut de Cancérologie de l’Ouest, Saint-Herblain, France
L. Mathiot1, A. Lusque2, T. Grinda3, T. Petit4, A. Mailliez5, T. Bachelot6, M. Arnedos7, C. Lévy8, E. Brain9, V. Massard10, T. De La Motte Rouge11, A. Goncalves12, I. Desmoulins13, A. Savoye14, M. Leheurteur15, L. Bosquet16, F. Dalenc17, W. Jacot18, J. Frenel1; 1Department of Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 2Biostatistics & Health Data Science Unit, Oncopole Claudius Regaud, IUCT, Toulouse, FRANCE, 3Department of Cancer Medicine, Gustave Roussy, Villejuif, FRANCE, 4Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, ICANS, Strasbourg, FRANCE, 5Department of Medical Oncology, Centre Oscar Lambret, Lille, FRANCE, 6Department of Medical Oncology, Centre Léon Bérard, Lyon, FRANCE, 7Department of Medical Oncology, Institut Bergonié, Bordeaux, FRANCE, 8Department of Medical Oncology, Centre François Baclesse, Caen, FRANCE, 9Department of Medical Oncology, Institut Curie, Saint-Cloud, FRANCE, 10Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-lès-Nancy, FRANCE, 11Department of Medical Oncology, Centre Eugène-Marquis, Rennes, FRANCE, 12Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, FRANCE, 13Department of Medical Oncology, Centre Georges François Leclerc, Dijon, FRANCE, 14Department of Medical Oncology, Institut Godinot, Reims, FRANCE, 15Department of Medical Oncology, Centre Henri Becquerel, Rouen, FRANCE, 16Health Data and Partnerships Department, Unicancer, Paris, FRANCE, 17Department of Medical Oncology, Oncopole Claudius Regaud, IUCT, Toulouse, FRANCE, 18Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, FRANCE.
Background: The PATINA trial demonstrated that adding the CDK4/6 inhibitor palbociclib to maintenance endocrine therapy (ET) and trastuzumab ± pertuzumab significantly prolonged median progression-free survival (PFS) by approximately 15 months (HR 0.74 [95% CI: 0.58-0.94]). Given its potential to redefine treatment standards, our objective was to replicate and validate the control arm outcomes of the PATINA trial using real-world data. Methods: We conducted a retrospective analysis using data from the multicenter, real-world Epidemio-Strategy and Medical Economics (ESME) database, encompassing 18 French Comprehensive Cancer Centers. Eligible patients had hormone receptors-and HER2-positive (HR+)/HER2+ metastatic breast cancer (MBC), initiated first-line treatment with a taxane plus trastuzumab + pertuzumab, who did not progress after completion of chemotherapy and subsequently received maintenance therapy with trastuzumab + pertuzumab combined with endocrine therapy. Real world progression free survival (PFS) and overall survival (OS) were estimated from maintenance ET initiation in the overall cohort and in subgroups of patients. Multivariable analyses were performed including age, tumor grade, de novo MBC status, number of metastatic sites, presence of visceral metastases, and hormone receptor status (ER+/PR+ or ER+/PR-). Results: Between January 2013 and December 2023, 368 patients with HR+/HER2+ MBC who completed induction chemotherapy and transitioned to maintenance therapy with trastuzumab + pertuzumab and ET were included. The median age was 56 years (range 20-88) and most of patients had de novo MBC (67%). Prior chemotherapy +/- anti-HER2 therapy in the (neo)adjuvant setting was reported in 17.1% of patients. The median duration of taxane induction for metastatic disease was 3.9 months (range 3-7.6) and 85.6% received an aromatase inhibitor as endocrine therapy combined with trastuzumab and pertuzumab. Best response to induction therapy, a stratification factor in PATINA trial, was not recorded. After a median follow-up of 57.8 months [95% CI: 53.1-63.6], the median PFS was 28.8 months [95% CI: 22.0-33.0], with a 5-year PFS rate of 35.4% [95% CI: 29.9-40.9]. The median overall survival (OS) was not reached, with 3- and 5-year OS rates of 85.2% [95% CI: 80.8-88.7] and 70.3% [95% CI: 64.3-75.5], respectively. A higher number of metastatic sites (≥3) was linked to poorer PFS [adjusted HR, 95% CI: 2.07 (1.50-2.85)], whereas de novo metastatic disease was associated with improved PFS [adjusted HR, 95% CI: 0.75 (0.56-1.00)]. A sensitivity analysis performed in patients treated before 2018 (32%), i.e. before PATINA was opened to accrual, showed consistent results a median PFS of 28.1 months [95% CI: 14.9-38.7] and a 5-year OS rate of 66.6% [95% CI: 57.3-74.4]. A notable difference compared to the PATINA control arm was the high rate de novo MBC in our cohort.Conclusion: In this real-world analysis, patients with HR+/HER2+ MBC treated with first-line metastatic induction chemotherapy followed by maintenance trastuzumab + pertuzumab and ET achieved outcomes in line with those reported in the control arm of the PATINA trial.
Presentation numberPS2-04-26
A comparison of clinical features and treatment outcomes in breast cancer leptomeningeal metastasis between African American and Caucasian populations: a single institution experience
Bipin Ghimire, Henry Ford Health, Detroit, MI
B. Ghimire1, L. Rogers2, M. Girgis1; 1Hematology-Oncology, Henry Ford Health, Detroit, MI, 2Neurosurgery, Henry Ford Health, Detroit, MI.
Title: A comparison of clinical features and treatment outcomes in breast cancer leptomeningeal metastasis between African American and Caucasian populations: a single institution experience Background: Leptomeningeal disease (LMD) is a rare but devastating complication of malignancies, affecting up to 5% of breast cancer patients. Survival is poor, typically 3-4 months, despite aggressive treatment. There is very limited literature regarding the clinical variables of breast cancer and LMD in African Americans (AAs) and how treatment outcomes differ from non-AAs. Due to the unique demographics of the patient population at the Henry Ford Health Cancer, including a high percentage of AAs, we sought to analyze these factors as compared with Caucasian patients. Methods: We retrospectively reviewed breast cancer patients diagnosed with LMD at Henry Ford Health from August 2014 to August 2024. LMD diagnosis followed the European Society of Medical Oncology (ESMO) criteria, and treatment responses were assessed using modified Response Assessment in Neuro-Oncology (RANO)-LM criteria. Results: Forty-one patients were identified (18 Caucasian, 17 AA, 6 others), with a similar median age at diagnosis in AAs and Caucasians (52 vs 51 years). AAs had an equal number of hormone receptor (HR)-positive and triple-negative tumors (35%), followed by HER2-positive (29%), which is in contrast to Caucasians who had more HER2-positive (44%), followed by HR-positive (39%), and triple-negative (17%). Although the most common histology was invasive ductal in both AAs (71%) and Caucasians (65%), invasive lobular was more commonly seen in Caucasians (35%) vs only 12% in AAs. More AA patients had stage I disease at diagnosis (41%) vs only 6% of Caucasians, whose majority had de novo stage IV disease (67% vs 24% in AAs). 77% of AAs (13/17) had parenchymal brain metastases in addition to LMD, compared to only 7 of 18 in Caucasians. A higher proportion of AAs had better performance status (ECOG 1) than Caucasians (44% vs 6%). Among AAs, 47% received systemic therapy after LMD diagnosis, and 41% had CNS radiation, which was similar to Caucasians, among whom 44% received radiation and 39% received systemic therapy. Twenty-nine percent of AAs received intrathecal (IT) therapy (median treatments: 5) compared to 39% of Caucasians (median treatments: 9). Methotrexate was the most common IT drug in both groups. Among evaluable patients, there was a trend towards improved overall response rates in AAs (44% vs 25% in Caucasians), with similar disease control rates (78% vs 75%). Event-free survival (EFS) and overall survival (OS) were similar in both AAs (mEFS: 2.2 months; mOS: 2.2 months) and Caucasians (mEFS: 1.7 months; mOS: 1.6 months). On univariate analysis, poor performance status (ECOG) predicted worse EFS and OS in AAs, but this difference was not seen in Caucasians. Other factors, including receptor subtypes, histology, grade, stage at diagnosis, time to onset of LMD, and metastatic burden, showed no significant impact on EFS and OS in both populations. Conclusion: Though these numbers are small, our experience identified clinical differences between the AA and Caucasian populations, particularly in receptor status, performance status, stage at diagnosis, histology, and presence of parenchymal metastases. Although AAs had numerically better response rates, survival outcomes were similar in both groups. The variables and outcomes we identified will require larger population-based studies to generalize the findings.
Presentation numberPS2-04-27
Real-world Clinical Outcomes of Trastuzumab Deruxtecan Among HER2+ Metastatic Breast Cancer Patients with and without Brain Metastases: Data from U.S Community Oncology Practices
Sibel Blau, ONCare Alliance, Tacoma, WA
S. Mehta1, H. Neuhalfen2, A. Peevyhouse2, D. Adhikari2, C. Williams2, M. Garretson1, S. Blau2, M. Walker2; 1HEOR, Daiichi-Sankyo, Basking Ridge, NJ, 2RWD, ONCare Alliance, Tacoma, WA.
Background: Trastuzumab deruxtecan (T-DXd) was initially approved by the FDA for treatment of patients with HER2+ unresectable or metastatic breast cancer (mBC) with two or more prior anti-HER2 regimens (3L+) in 12/2019 and subsequently approved among patients with one prior anti-HER2 regimen (2L) in 5/2022. Clinical trials including DESTINY-Breast12 have demonstrated substantial and durable activity of T-DXd overall, and within the central nervous system, in patients with active and stable brain metastasis (BM). However, limited data exists regarding the intracranial activity and real-world clinical benefit associated with T-DXd in this patient population. This study examined clinical outcomes among patients with HER2+ mBC, with and without active or stable BMs, who were treated with T-DXd in the community oncology setting. Methods: This retrospective cohort study used deeply-curated electronic medical record data from practices affiliated with ONCare Alliance. A total of 300 adult patients with mBC diagnosis, confirmed HER2+ status, who received T-DXd in the 2L or later setting anytime from 01/2020 to 08/2024 were selected for chart abstraction. Patient demographic characteristics, clinical characteristics, and comorbidities were assessed descriptively, and compared with chi-square or Fisher exact tests. Clinical outcomes, including real-world time to treatment failure (rwTTF) and real-world progression-free survival (rwPFS), were assessed using Kaplan-Meier median and confidence interval estimates. All analyses were stratified by BM vs. no BM status and active (new or worsening) vs. stable (unchanged or improving) BM status. Results: Of 300 patients, 95 patients (68 active BMs and 27 stable BMs) had BMs at initiation of T-DXd. Overall, the median follow-up period was 17.0 months, mean age was 59.3 years (±12.6) and a majority were White (82.3%), post-menopausal (76.3%) and had an ECOG score of 0-1 (80.3%). The baseline patient demographic and clinical characteristics were generally similar among patients with vs. without BM, except for age (mean 54.0 vs. 61.7 years, p < 0.001), HR status (HR+ 55.8% vs. 68.3%, p = 0.0355) and comorbidity index (mean 0.28 vs. 0.68, p < 0.001). No statistically significant differences in baseline demographics and clinical characteristics were observed between active vs. stable BM patients. No significant differences in clinical outcomes were observed between BM vs. no BM cohorts (median rwTTF was 10.0 [95% CI 8.1 – 12.5] vs. 11.1 months [9.0 – 12.6], p = 0.3507; and median rwPFS was 12.1 [8.6 – 14.2] vs. 13.5 months [11.8- 14.91], p = 0.2483). Similarly, no significant differences in clinical outcomes were observed between active vs. stable BM cohorts (median rwTTF was 11.3 [8.3 – 12.9] vs. 8.1 months [4.3 – 14.6], p = 0.9679; and median rwPFS was 12.1 [8.9 – 14.2] vs. 10.6 months [5.5 – 23.7], p = 0.4140). Conclusion: This study is among the largest real-world data studies characterizing the clinical effectiveness of T-DXd among HER2+ mBC patients with vs. without brain metastasis and among patient segments with active vs. stable status. Results support the effectiveness of T-DXd in patients with brain metastasis, and showed no difference in outcomes based on active vs. stable status. Findings from this study reinforce the value of utilizing T-DXd to improve long-term outcomes of all patients with HER2+ mBC, irrespective of the presence of BM or active vs. stable brain metastasis.
Presentation numberPS2-04-28
Symptom trajectories using remote symptom monitoring across treatment type and stage
Jeffrey A Franks, University of Alabama at Birmingham, Birmingham, AL
J. A. Franks1, L. Deng1, S. Olisakwe1, C. Williams2, A. Azuero3, A. Falcao1, E. Ortiz1, I. Starks1, C. Hogea4, G. Rocque1; 1Hematology & Oncology, University of Alabama at Birmingham, Birmingham, AL, 2Preventative Medicine, University of Alabama at Birmingham, Birmingham, AL, 3School of Nursing, University of Alabama at Birmingham, Birmingham, AL, 4Gilead Sciences, Gilead Sciences, Foster City, CA.
Background: Remote symptom monitoring (RSM) using patient reported outcomes allow real-time reporting of symptoms from patients to their healthcare team. This provides a unique opportunity to understand symptom trajectories across stage and treatment.Methods: This retrospective cohort study is a secondary analysis of data from a hybrid, type 2 single-arm trial of a navigator-supported RSM program from May 2021-May 2024. Upon treatment initiation, patients self-reported pain, gastrointestinal, respiratory, and urinary symptoms via weekly surveys. Symptoms were categorized as moderate or severe using the PRO-CTACAE grading tool. Patients with at least one survey per month for three months were included. Generalized linear mixed-effect models estimated associations between 3-month symptom trajectories and stage (early [EBC] vs. metastatic [MBC]) or treatment (chemotherapy vs. targeted or immunotherapy) using odds ratios [OR] and 95% confidence intervals [CI]. Models were adjusted for age at treatment initiation, race, and comorbidity status, and included an interaction term for time and treatment or stage.Results: Of 292 patients, the median age was 55; most patients were white and had 2-3 comorbidities. A quarter of patients received targeted or immunotherapy and had MBC. Although results did not reach statistical significance, symptoms trajectories differed by treatment type and stage, as well as between specific symptoms (Table). Compared to those receiving chemotherapy, patients receiving targeted or immunotherapy had 36% higher odds of GI symptoms in month one (M1), with lower odds in M2, but higher odds in M3. The odds of respiratory symptoms were 54% lower for patients receiving targeted or immunotherapy initially (M1), and remained low in M2, but by M3 were similar to those receiving chemotherapy. The odds of urinary symptoms were 64% lower for patients receiving targeted or immunotherapy in M1 but became 83% higher in M2 and had two-fold higher odds in M3 compared to those receiving chemotherapy. The odds of pain for patients with targeted or immunotherapy vs. chemotherapy were only higher in M1. When comparing symptom trajectories by stage, patients with MBC had lower odds of GI symptoms in M1 and M2, but similar odds in M3 to those with EBC. The opposite trend was seen for respiratory disease where those with MBC saw higher odds in M1, M2, and M3 compared to EBC, though becoming more similar across time. The odds of urinary symptoms were similar across stage over time. Finally, the odds of pain were 51% higher in MBC vs. EBC patients in M1, 25% lower in M2, and similar in M3.Conclusion: Symptom trajectories in the first three months of treatment varied by treatment type and stage. Further evaluation is needed to understand how patterns may reflect initial acute vs. later cumulative toxicities occurring over time. Future studies utilizing a larger sample are needed to better generalize these findings.
Presentation numberPS2-04-29
Parp inhibitors use in patients in germline palb2 or somatic brca1/2 mutations carriers with metastatic breast cancer: real life data from the esme database
Pauline ROTTIER, Centre François Baclesse, Caen, France
P. ROTTIER1, L. CHALTIEL2, Z. NEVIERE1, W. JACOT3, A. MAILLIEZ4, F. DALENC5, T. BACHELOT6, E. BRAIN7, V. MASSARD8, B. SAUTEREY9, T. GRINDA10, C. BAILLEUX11, M. ARDENOS12, L. BOSQUET13, G. EMILE1; 1Medical Oncology, Centre François Baclesse, Caen, FRANCE, 2Biostatistics Department IUCT, Oncopole Claudius Regaud, Toulouse, FRANCE, 3Medical Oncology, Institut régional du Cancer Montpellier, Montpellier, FRANCE, 4Medical Oncology, Centre Oscar Lambret, Lille, FRANCE, 5Medical Oncology, Oncopole Claudius Regaud, Toulouse, FRANCE, 6Medical Oncology, Centre Léon Bérard, Lyon, FRANCE, 7Medical Oncology, Institut Curie, Paris, FRANCE, 8Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-les-Nancy, FRANCE, 9Medical Oncology, Institut de Cancérologie de l’Ouest, Centre Paul Papin, Angers, FRANCE, 10Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 11Medical Oncology, Centre Antoine Lacassagne, Nice, FRANCE, 12Medical Oncology, Institut Bergonié, Bordeaux, FRANCE, 13Health data and partnerships department, Unicancer, Paris, FRANCE.
Background: Poly (ADP-ribose) polymerase inhibitors (PARPi) are approved for the treatment of HER2-negative metastatic breast cancer (MBC) in germline (g)BRCA1/2 pathogenic alteration (m) carriers. Olaparib and talazoparib showed efficacy in MBC patients with somatic (s)BRCA1/2m and/or gPALB2m in phase 2 trials. We aimed to investigate the effectiveness of PARPi in this setting in the real-life ESME cohort. Methods: ESME-MBC, a nationwide observational cohort, gathers data on MBC patients treated in 18 French Cancer Centers from 2008 on. We selected all patients treated with PARPi and who had either sBRCA1/2m or gPALB2m MBC. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS) from treatment initiation, PFS and OS according to type of mutation, type of PARPi and line of treatment. The Kaplan-Meier method was used to assess survival. Results: Among 35,687 patients included in the ESME database from 2008 to 2022, 57 were eligible for the present analysis (46 with sBRCA1/2m;11 gPALB2m). Median age at treatment was 54 years [31-83]). 39% were triple negative MBC (17 sBRCA1/2m and 5 gPALB2m), 60% HR-positive/HER2-negative (28 sBRCA1/2m and 6 gPALB2m). The median number of treatment lines prior PARPi, including endocrinotherapy, was two [0-9]. PARPi was initiated in first- or second line for 24 patients, representing 64% of triple-negative patients and 42% of HR-positive/HER2-negative patients. 32 patients (56%) received olaparib, 22 talazoparib (39%, all with sBRCA1/2m) and 3 another PARPi. A clinical trial was the context for 36.8% of prescriptions. In the whole population, median PFS and OS were 5.4 [95%CI: 4.3; 8.3] and 13.2 months [11.2; 19.7] respectively. For patients bearing sBRCA1/2m and gPALB2m, median PFS were 4.9 [3.0; 8.2] and 8.3 [3.0; not achieved (NA)] months respectively, and median OS 12.1 [10.4; 19.7] and 17.6 [2.4; NA] months respectively. Median PFS was 4.9 [2.8; 6.7] and 8.2 months [3.9; 13.1] for talazoparib and olaparib respectively; and median OS 12.1 [7.2; 24.3] and 15.0 months [10.1; 26.6]. Conclusion: In this multicenter real-life cohort of MBC patients with a sBRCA1/2 or a gPALB2 mutation, effectiveness of PARPi appeared in line with phase II trials. These data further support the use of olaparib or talazoparib in gPALB2m, and possibly in sBRCA1/2m.
Presentation numberPS2-04-30
Enrolment of Patients with Metastatic Lobular Breast Cancer in Clinical Trials in the Multicenter ESME Cohort
Eleonora De Maio, IUCT ONCOPOLE, Toulouse Cedex 9, France
E. De Maio1, L. Chaltiel1, V. Massard2, A. Mailliez3, T. Grinda4, J. Frenel5, E. Brain6, T. Bachelot7, A. Gonçalves8, B. Sauterey9, D. Pasquier10, T. Petit11, L. Bosquet12, S. Guiu13, F. Dalenc1; 1Medical Oncology, IUCT ONCOPOLE, Toulouse Cedex 9, FRANCE, 2Medical Oncology, Institut de Cancérologie de Lorraine, Vandœuvre-lès-Nancy, FRANCE, 3Medical Oncology, Centre Oscar Lambret, Lille, FRANCE, 4Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 5Medical Oncology, Institut de Cancérologie de l’Ouest, Centre René Gauducheau,, Saint-Herblain,, FRANCE, 6Medical Oncology, Institut Curie, Paris, FRANCE, 7Medical Oncology, Centre Léon Bérard, Lyon, FRANCE, 8Medical Oncology, Institut Paoli Calmettes, Marseille, FRANCE, 9Medical Oncology, Institut de Cancérologie de l’Ouest, Angers, FRANCE, 10Radiotherapy, Centre Oscar Lambret, Lille, FRANCE, 11Medical Oncology, Centre Paul Strauss ICANS, Strasbourg, FRANCE, 12RWD, Unicancer,, Paris, FRANCE, 13Medical Oncology, Institut du Cancer de Montpellier, Montpellier, FRANCE.
Background: Invasive lobular carcinoma (ILC) represents about 15% of breast cancers and is the second most common histological subtype after invasive carcinoma of no special type (NST). ILC differs from NST in clinical presentation, histological features, and molecular profile. Nevertheless, ILC-specific characteristics are often overlooked when making treatment decisions in clinical practice. Available evidence suggests that patients (pts) with ILC are underrepresented in clinical trials, possibly due to features such as predominant bone involvement, which are often non-measurable by RECIST 1.1 criteria, leading to exclusion from trials. This study aimed to evaluate clinical trial enrolment among pts with metastatic breast cancer (MBC) and ILC subtype. Methods: The ESME MBC (Epidemiological Strategy and Medical Economics – Metastatic Breast Cancer) cohort is a national registry collecting individual-level patient data from 18 French Comprehensive Cancer Centers (NCT03275311). This analysis included all pts treated between 2008 and 2023 who had a histological classification of ILC, NST, or mixed ILC+NST. We assessed clinical trial enrolment rates according to histological subtype, considering trials initiated at any treatment line and specifically at first line. Trial characteristics and enrolment distribution across treatment periods (2018) were described by histological subtype. A subpopulation of pts with ILC considered potentially eligible for clinical trials (female, ECOG 0-1, aged less than 70 years, no CNS metastases, no recent other malignancy) was defined to study clinical characteristics, treatments, and predictors of trial participation using multivariable logistic regression. Results: Out of 38,285 pts in the ESME database, 32,722 were eligible: 27,493 (84.0%) NST, 4,897 (15.0%) ILC, and 332 (1.0%) mixed ILC+NST. A total of 5,124 pts (15.7%) were enrolled in at least one clinical trial. Trial inclusion rates at any treatment line were 12.6% for ILC and 16.2% for NST (p<0.0001). It was 13.0% for mixed subtypes. Overall, trial participation at first line was low (9.1%). ILC pts had lower enrolment in first-line trials (7.4%) compared to NST (9.5%) and mixed (7.5%). Enrolment peaked between 2012 and 2018 whatever the subtype: 59.1% for ILC, 54.6% for NST and 48.0 for mixed. Pts with ILC were more often enrolled in phase III trials (58.1%) than pts with NST (50.0%), but less than those with mixed subtype (63.6%). Conversely, pts with ILC and mixed were underrepresented in phase I trials (9.0% and 4, 5% respectively) compared to 14.5% for pts with NST. Considering first-line trials, pts with ILC were more likely to receive endocrine therapy based regimen (61.7%) compared to pts with NST (33.6%) or mixed (48.0%). Conversely, they were less likely to receive chemotherapy (27.5%, 50.0% and 44.0% for pts with ILC, NST and mixed respectively) and immunotherapy (1.9%, 7.2% and 4.0% respectively). Among the 4,897 pts with ILC, 1,168 (23.9%) met eligibility criteria for clinical trials and 179 (15.3%) of these were enrolled in first line trials. Grade I tumors were less frequent among trial participants (p = 0.030), and enrolment peaked during 2012-2018 but declined thereafter (p < 0.0001). No other significant differences were observed. Multivariable logistic regression identified only histological grade II as significantly associated with clinical trial enrolment (OR 2.07(95% CI [1.13; 3.77]), p = 0.018). Conclusion: In this large real-life series, pts with metastatic ILC had lower enrolment in clinical trials than those with NST, suggesting access disparities. However, multivariable analysis did not clearly identify factors influencing first line trial enrolment, highlighting the need for further investigation. Additional analyses will be shared later this year.
Presentation numberPS2-06-01
Real World Outcomes of the Keynote 522 Regimen for Triple-Negative Breast Cancer
Katherine E Myers, Case Western Reserve University/University Hospitals, Cleveland, OH
K. E. Myers, H. Dirawi, M. Lubarsky, M. Stojsavljevic, N. Stabellini, I. Sheng, M. M. Mirsky, A. J. Montero; Internal Medicine, Case Western Reserve University/University Hospitals, Cleveland, OH.
Introduction: The Keynote 522 study established the use of perioperative chemoimmunotherapy as the standard of care in the treatment of early stage, triple-negative breast cancer, showing benefit in both pathological complete response (pCR) and event-free survival (EFS) when compared to chemotherapy plus placebo. However, the original study did not comment on the racial, ethnic, or socioeconomic characteristics of its participants. This is despite well-documented disparities in breast cancer outcomes for women of color and those of lower socioeconomic status (SES) [1,2]. The purpose of this study was to evaluate real-world outcomes of the Keynote 522 regimen in an NCI-designated, comprehensive cancer center for any heterogeneity in clinical outcomes. Methods: Our study consisted of a single-institution, retrospective analysis of 207 patients with resectable, stage II-III triple negative breast cancer who received care at a combined academic/community hospital network in Northeastern Ohio from 2020-2025, and who received neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab per the Keynote 522 protocol. Patient age, race, ethnicity, insurance provider, poverty index, ECOG score, comorbidity index, menopausal status, primary tumor classification, CPS score, carboplatin schedule (weekly vs q3weeks), LDH level, and immunotherapy-related adverse events were obtained via IRB-approved chart review. The primary outcomes included pCR, EFS, and rates of grade III or higher irAEs. Analysis of pCR and EFS was performed using both univariate and multivariable Cox regression analysis, with multivariable analysis controlling for race, ethnicity, insurance provider, and irAE occurrence and grade. Results: At our institution we identified 207 patients who received KN-522 regimen: 65% self-identified as White, 33% as Black, and 2.1% as Hispanic or Latino. The median poverty index was 16, with 75.4% of patients living above the average national poverty line. Across the entire cohort, a pCR (defined as ypT0/Tis and ypN0) was observed in 60% of patients, compared to 64.8% reported in the Keynote 522 study. Multivariable analysis was performed. Of the evaluated factors, age at diagnosis was the only clinical/demographic factor that significantly correlated with pCR—younger age was associated with higher associated chances of pCR (OR 0.97, 95% CI 0.95-0.99, p-value 0.013). Two-year overall EFS was 82% (95% CI 76-89%). No statistically significant difference in EFS was observed across age, race, ethnicity, poverty index, or insurance provider. Pre-menopausal patients were found to have a significantly higher associated risk of disease recurrence (HR 2.91, 95% CI 1.3-6.51, p-value 0.009), whereas patients who experienced any irAEs were found to have lower associated risk of recurrence (HR 0.37, 95% CI 0.16-0.85, p-value 0.019), though this only held true for patients with grade I/II irAEs vs grade III/IV on subgroup analysis (HR for grade I/II 0.36, 95% CI 0.15-0.89, p-value 0.027; HR for grade III/IV 0.41, 95% CI 0.09-1.78, p-value 0.233). Conclusions: We did not find any socioeconomic or racial heterogeneity with respect to pCR rates in TNBC patients who received neoadjuvant chemo-immunotherapy with the KN-522 regimen. We found that the onset of low-grade irAEs was associated with lower disease recurrence rates, consistent with prior studies showing overall improved PFS and OS in patients who experience irAEs, regardless of cancer type [3]. Overall, this suggests that the data presented in the original Keynote 522 study, which did not comment upon race or other social determinants of health of its participants, appears to reflect observed pCR rates in a real-world setting among TNBC patients. References: 1. doi: 10.1056/NEJMp2200244. 2. doi: 10.1177/107327482513415203. doi: 10.3389/fonc.2021.633032
Presentation numberPS2-06-02
Real-world outcomes comparing zoledronic acid and pamidronate in patients with metastatic breast cancer with bone metastases: A propensity score matched analysis from a global federated health research network
Yajur Arya, Mayo Clinic, Jacksonville, FL
Y. Arya1, A. Syal1, 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 Hospital, Philadelphia, PA.
Introduction: Breast cancer most commonly metastasizes to the bones, with bone pain and other skeletal related events (SREs) having a limiting effect on quality of life. Bisphosphonates are widely used in breast cancer patients to reduce SREs and bone loss, particularly in those with bone metastases or receiving endocrine therapy. We aim to study the real world comparative data between two widely used bisphosphonates in clinical practice, zoledronic acid and pamidronate, 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, and 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 zoledronic acid cohort and 69.1 in the pamidronate cohort. In the zoledronic acid cohort, the ethnicity distribution was 74.68 % White, 11.55% African American, and 6.36% Hispanic. In contrast, the pamidronate group had an ethnicity distribution of 63.04% White, 20.64% African American, and 8.16% 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 (Hazard Ratio [HR] 0.593, 95% CI: 0.509-0.691, p-value <0.001), osteoporotic fracture (Risk difference: -5.28%, 95% CI: -7.65 to -2.90), p-value < 0.001), hypercalcemia [HR: 0.262, 95% CI: 0.212 – 0.323, p-value =0.001), pathological fracture (Risk difference: -6.88%, 95% CI: -10.69 to -3.07), p-value < 0.001). However, the zoledronic acid group had higher risk of falls [Risk difference: 3.52%, 95% CI: 0.518 to 6.522), p-value =0.021). There was no statistically significant difference in the occurrence of osteonecrosis between the two cohorts. 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, osteoporotic fracture, hypercalcemia, and pathological fracture compared to those receiving pamidronate. Additional longitudinal cohort studies are imperative to explore the outcomes between these agents and inform clinical practice guidelines.
Presentation numberPS2-06-03
Sacituzumab Govitecan Dosing and Neutropenia Risk in Patients with HER2-Negative Metastatic Breast Cancer
Alexandra Bili Newman, MD Anderson Cancer Center, Houston, TX
A. B. Newman1, A. Raghavendra2, E. Grannan2, F. Meric-Bernstam3, D. Tripathy2, J. Litton2, S. Damodaran2; 1Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 2Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX.
Background: Sacituzumab govitecan (SG), a TROP2-directed antibody drug conjugate with topoisomerase I inhibitor payload, has demonstrated improved efficacy over standard chemotherapy in HR-positive and triple-negative breast cancer based on the TROPiCS-02 and ASCENT trials, respectively. However, neutropenia represents a significant dose-limiting toxicity that may necessitate dose modifications or hospitalization. Given increasing real-world use and associated adverse effects, varying dosing and supportive care strategies have emerged. Methods: This single-center retrospective cohort study analyzed HER2-negative metastatic breast cancer patients receiving SG (April 2020-December 2024) evaluating patient demographics, disease characteristics, treatment patterns, safety outcomes, and G-CSF use. Inverse probability weighting (IPW) based on propensity scores was applied to control for confounding variables. Results: Among 366 patients, most initiated SG at 10 mg/kg (70%), with fewer starting at 7.5 mg/kg (22%) or 5 mg/kg (8%) (Table 1). Dose reductions were more common when starting at 10 mg/kg (42%) compared with 7.5 mg/kg (16%) and were primarily due to neutropenia (66%) followed by diarrhea (22%) and fatigue (17%). Three patients discontinued treatment due to toxicity. After adjustment for age, prior treatment lines and prophylactic G-CSF use, patients starting at 10 mg/kg had 2.8-fold higher odds of grade 3-4 neutropenia compared with 5 mg/kg (OR 2.77, 95% CI 1.29-6.27, p=0.011), while the duration of treatment was shorter at 5 mg/kg. Age was not associated with neutropenia risk or hospitalization rate after adjusting for starting dose, prophylactic G-CSF use, and prior treatment lines. Neither starting dose nor prior treatment lines significantly affected hospitalization rates. Prophylactic G-CSF was significantly associated with less grade 3-4 neutropenia (OR 0.12, 95% CI 0.07-0.18, p<0.001) when controlling for age, prior treatment lines, and starting dose and numerically fewer hospitalizations. Prophylactic G-CSF was most frequently used in patients who started treatment at reduced doses, but utilization rates did not differ by age. Conclusions: This real-world analysis demonstrates a clear dose-dependent relationship with severe neutropenia, with 10 mg/kg dosing conferring a significantly higher risk compared to 5 mg/kg. Age was not associated with increased risk of neutropenia or hospitalization. These findings do not support empiric dose reduction based solely on advanced age. Primary prophylactic G-CSF provides substantial protection against severe neutropenia regardless of starting dose or age.
Presentation numberPS2-06-04
Invasive lobular breast cancer: Ten-year follow-up data from a single center
Ivica Ratosa, Institute of Oncology Ljubljana, Ljubljana, Slovenia
I. Ratosa1, D. Stefanovski2, M. Sojar1, V. Jeric Horvat2, R. Cencelj Arnez3, B. Seruga2, D. Ribnikar2; 1Division of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 2Division of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 3Division of Surgery, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA.
Background: Invasive lobular carcinoma (ILC) is recognized as a distinct disease from breast carcinoma of no special type. Studies have shown that breast-conserving surgery (BCS) followed by radiotherapy (RT) gives local control and overall survival (OS) rates that are similar to those of mastectomy. There is also some evidence that BCS plus radiotherapy improves survival and local control in early-stage breast cancer, including ILC, compared to mastectomy alone. Methods: Individual clinical records provided demographic, histological, systemic, and local therapy data. We compared clinical and tumor characteristics between groups using independent-sample t-tests for continuous variables and Pearson’s χ2 tests for categorical data. Continuous variable medians with sample ranges were used. The log-rank test compared the two groups, while the Kaplan-Meier method computed survival curves. Cox’s proportional hazards models—univariate and multivariate—were used to assess survival effects. Results: The study cohort consisted of 372 patients with early-stage ILC diagnosed between 2003 and 2008 and treated with either BCS (n=138, 37.1%) or mastectomy (n=234, 62.9%). Median age of patients was 62 years (range, 33-90). Median follow-up was 10.7 years (0.1-21.2). Patient and treatment characteristics are presented in a table. A total of 33 patients (8.9%) in the entire group developed loco-regional recurrence. In terms of surgical type, locoregional relapse was observed in 8 (5.8%) patients of the BCS group and 25 (10.7%) patients of the mastectomy group (p=0.132). Compared to patients receiving RT higher rate of locoregional recurrence was observed among patients who did not receive RT (n=11; 6.4% vs n=22; 11.0%); however, the difference was not statistically different (p=0.144).The 5-year and 10-year freedom from locoregional recurrence were 98% and 95%, respectively. Eighty-two (22.0%) patients experienced distant relapse. Compared to BCS, more patients in the mastectomy group experienced distant relapse (n=61; 26.2% vs n=21; 15.3; p=0.019). During the follow-up period, 163 (43.8%) patients died. The 5-year and 10-year survival rates for the whole cohort were 85% and 64%, respectively. The OS for the entire cohort was 14.1 years (13.7-16.8), for BCS it was 15.1 years (15.6-19.7), and for mastectomy it was 13.4 years (12.2-15.6), respectively (p=0.020). OS differed according to the specific ILC subtype, being the longest for tubulolobular and the shortest for the solid subtype (p=0.016). In a multivariate analysis, lower T stage (p<0.001) and premenopausal status (p<0.01) were the only two factors that correlated with improved OS. Conclusions: In our single-center 10-year follow-up data of ILC, we have found that ILC had very high 5- and 10-year locoregional control, regardless of the type of local treatment. However, ILC is associated with a high risk of late distant recurrence.
| Parameter | All patients N=372 (%) | Breast conserving surgery, N=138 (%) | Mastectomy N=234 (%) | p value | |||||
| Tumor size [mm], range | 21 (4-150) | 16 (5-55) | 25 (4-150) | <0.001 | |||||
| Tumor multifocality | 28 (7.5) | 11 (8.0) | 17 (7.3) | 0.0234 | |||||
| Tumor multicentricity | 53 (14.2) | 0 (0) | 53 (22.6) | <0.001 | |||||
| Number of positive lymph nodes, range | 0 (0-42) | 0 (0-23) | 0 (0-42) | <0.001 | |||||
| Hormone receptor positive status | 359 (96.5) | 135 (97.8) | 224 (95.7) | 0.387 | |||||
| HER2 positive | 21 (5.6) | 6 (4.3) | 15 (6.4) | 0.490 | |||||
| Lympho-vascular invasion | 24 (6.5) | 3 (2.2) | 21 (9.0) | 0.009 | |||||
| Perineural invasion | 47 (12.6) | 15 (10.9) | 32 (13.7) | 0.519 | |||||
| Post-menopausal status | 271 (72.8) | 102 (73.9) | 169 (72.2) | 0.713 | |||||
| Adjuvant chemotherapy only | 13 (3.5) | 3 (2.2) | 10 (4.3) | 0.159 | |||||
| Adjuvant chemotherapy and endocrine therapy | 93 (25.0) | 19 (13.8) | 74 (31.6) | <0.001 | |||||
| Endocrine therapy only | 254 (68.3) | 112 (81.2) | 142 (60.7) | <0.001 | |||||
| No systemic therapy | 12 (3.2) | 4 (2.9) | 8 (3.4) | 0.326 | |||||
| Radiation therapy | 172 (46.2) | 110 (79.7) | 62 (26.5) | <0.001 |
Presentation numberPS2-06-06
Real-world utilization of tissue-free ctDNA monitoring in patients with breast cancer
Douglas K Marks, NYU Langone Health, Mineola, NY
D. K. Marks1, C. B. Tambaoan2, H. Tukachinsky2, L. W. Pasquina2, A. Gasco2, M. Childress2; 1Perlmutter Cancer Center, NYU Langone Health, Mineola, NY, 2Clinical Development, Foundation Medicine Inc, Boston, MA.
Background: Leveraging circulating tumor DNA (ctDNA) to assess treatment response and emergence of resistance alterations is coming to the forefront in breast cancer, particularly for patients receiving endocrine therapy. However, little is known about real-world clinical utilization. Here we describe the real-world clinical use and results of a tissue-free ctDNA monitoring assay in patients with breast cancer. Methods: The clinical laboratory developed test, FoundationOne®Monitor (F1Monitor), was available in a limited pilot. Enrollment was rolling. The timing and number of tests were determined by each physician as appropriate for their practice. Therapy abstraction was requested but not required. ctDNA was detected and quantified by ctDNA tumor fraction (TF); variants were tracked with variant allele frequency (VAF) and the origin predicted algorithmically. Results: F1Monitor was ordered by 26 physicians for 193 patients (239 tests) with breast cancer between 9/30/2024-04/30/2025. Median turnaround time was 8 days. Therapies were abstracted for 39 patients, including 16 (41%) on endocrine therapy ± CDK4/6 inhibitor. At data cutoff, the median follow-up was 71 days. 33 patients had ≥2 tests. The median interval between the first two tests was 42.0 days (Interquartile Range [IQR]: 34.0, 60.7). The median cadence of serial testing (3+ tests, 7 patients) was 57 days (IQR: 46.8, 64.5). CtDNA TF was detected in 35% (68/193) of patients overall and 41% (16/39) known to be on therapy. Physicians were more likely to pursue serial monitoring if the first test had ctDNA TF detected. The median and maximum ctDNA TF was 2.8% and 84%. The lowest ctDNA TF and reportable variant VAF values quantified were 0.19% and 0.08%, respectively. ctDNA TF dynamic changes were observed in 55% (18/33) of patients during their course of testing (timepoint results: newly detected: 0; any increase: 9; <50% decrease: 4; 50-89% decrease: 4; 90-99% decrease: 5; clearance: 4). A median of 3 reportable alterations were detected per test with 97% (91/94) of tests with a ctDNA TF detected result having ≥1 copy number variant or predicted tumor somatic short variant and 71% (67/94) having ≥1 predicted clonal hematopoiesis (CH) alteration. Of tests with ctDNA TF not detected, 91% (132/145) had no variants or only alterations predicted to be CH or germline reported. Clinically actionable variants in breast cancer were detected in 31% (60/193) of patients overall and 70% (48/68) with ctDNA TF detected, with VAFs down to 0.1% for each gene: PIK3CA n=32; ESR1 n=18; BRCA1 n=9; BRCA2 n=9; PTEN n=6; ERBB2 n=6; AKT1 n=4. In patients with ≥2 tests, mutations in clinically actionable genes in breast cancer were persistent in 45% (15/33), cleared in 15% (5), and newly emerging in 15% (5). Conclusion: In real-world tissue-free ctDNA-based monitoring in breast cancer, F1Monitor demonstrated high sensitivity, detecting ctDNA TF as low as 0.19% and VAF as low as 0.08% including alterations in clinically actionable genes such as ESR1 down to 0.1%. Further, ctDNA TF demonstrated high specificity, with 91% of “ctDNA TF not detected” results having no alterations or only CH or germline predicted alterations. Importantly, ctDNA TF was detected in 41% of patients on known active therapy with observed dynamic changes for 55% of patients with longitudinal testing, indicating utility for monitoring treatment response and emerging resistance. In the limited follow up time, physicians chose to monitor patients approximately every 6-8 weeks with a tendency for repeat testing when ctDNA TF was detected at the first test.
Presentation numberPS2-06-08
Interim results: Real world study of treatment discontinuations and modifications for patients with HER2-low metastatic breast cancer on trastuzumab deruxtecan on a Canadian patient support program
Christine Brezden-Masley, Mount Sinai Hospital, Toronto, ON, Canada
C. Brezden-Masley1, S. Shokar2, A. Nam2, R. A. Qadeer2, Z. Senhaji Mouhri3, B. Salvo4, N. Bonar4, B. Suero4; 1Medicine, Mount Sinai Hospital, Toronto, ON, CANADA, 2Scientific Affairs, AstraZeneca Canada, Mississauga, ON, CANADA, 3Oncology Medical Affairs, AstraZeneca Canada, Mississauga, ON, CANADA, 4Value and Evidence, EVERSANA, Burlington, ON, CANADA.
Background: Trastuzumab deruxtecan (T-DXd) is a HER2-targeted antibody drug conjugate approved for the treatment of HER2+ and HER2-low metastatic breast cancer (mBC). Currently, there is limited evidence assessing how T-DXd is used in a Canadian real-world (RW) setting. The aim of this observational study was to describe RW treatment-related outcomes using data collected from patients with HER2-low mBC who enrolled in AstraZeneca Canada’s patient support program (PSP) following regulatory approval based on the DESTINY-Breast04 (DB-04) trial, and who were among the earliest HER2-low mBC recipients of T-DXd in Canada. Methods: This interim analysis included patients who enrolled in the PSP from January 2023 to March 2024 and initiated T-DXd therapy. Index date was the date of treatment initiation with follow-up until treatment discontinuation or close of the PSP. Primary objectives included rates of treatment discontinuation and dose modifications (dose reduction or discontinuation or dose interruptions >1 cycle length). Secondary objectives included time to treatment discontinuation (TTD), reasons for discontinuation (reported by physicians), and duration of treatment (excluding cumulative length of dose interruptions). Time-to-event outcomes were analyzed using the Kaplan Meier method; other outcomes were summarized descriptively. Results: A total of 860 patients were included with a mean age of 61.8 years (standard deviation [SD], 11.3; range, 29-94); 1.3% (n=11) had no prior lines of chemotherapy in the metastatic setting, 40% (n=345) had 1, 27% (n=233) had 2, and 31% (n=270) had 3+; 93% (n=793) were hormone receptor-positive (HR+) and 7% (n=63) were hormone receptor-negative (HR-). Median follow-up was 4.6 months (range 0.2-22.8). Among patients that started at a dose of 5.4 mg/kg (77.9%, n=670), 4.4 mg/kg (9.8%, n=84), or 3.2 mg/kg (1.4%, n=12), 28% (n=212) had a dose reduction, 54% (n=417) had a discontinuation, 67% (n=516) had a dose reduction or discontinuation, and 54% (n=410) had a dose interruption. Some patients (n=94, 10.9%) received a dose not specified in the label (neither 5.4, 4.4, or 3.2 mg/kg). The cumulative probabilities of treatment discontinuation at 3, 6 and 9 months were 21.7% (95% confidence interval [CI], 18.8-24.5), 44.1% (95% CI, 40.3-47.6), and 60.0% (95% CI, 55.8-63.8), respectively. Median overall TTD was 6.9 months (95% CI, 6.2-7.9); median TTD was 7.3 months (95% CI, 6.7-8.2) among HR+ patients and 3.4 months (95% CI, 3.2-4.6) among HR- patients. Median duration of treatment (excluding dose interruptions) was 6.3 months (95% CI, 5.6-7.1). Of the total observed discontinuation (54%, n=462), progression was the most common reason (52%, n=241) followed by prescriber decision (16%, n=73), death (15%, n=68), patient decision (6.3%, n=29), adverse event (3.5%, n=16), other (2.6%, n=12), or unknown/missing data (5.0%, n=23). Conclusion: Early RW use of T-DXd in the Canadian PSP, designed for patients eligible under the regulatory authorization based on DB-04, reflected treatment patterns in a more heavily pretreated population. Due to the short duration of follow-up, TTD may be underestimated. During the PSP enrollment period, T-DXd use was still in its early stage of experience in Canada and as such, understanding of optimal therapy management may have still been evolving.
Presentation numberPS2-06-09
Vacuum-assisted biopsy in the era of low-risk ductal carcinoma in situ active monitoring: real world data and implications.
Henrique Lima Couto, Redimama-Redimasto, Belo Horizonte, Brazil
H. L. Couto1, C. N. Valadares2, A. N. Soares3, B. T. Silva4, B. F. Ricardo5, P. H. Toppa6, B. A. Coelho4, V. Resende7, E. C. Pessoa8, F. M. Reis9, T. C. Oliveira4, D. d. Pires4, S. d. Ferreira4, L. B. Oliveira4, P. M. Soares4, T. P. Moraes4, A. C. Mendonça4, R. G. Saliba4, L. L. Dominguez4, B. A. Pires4, C. A. Padua10, G. F. Cunha Júnior10, N. Sharma11, M. S. Castilho12, J. S. Oliveira4, A. C. Oliveira4, D. R. Siqueira4, A. Mattar13, M. Antonini14, P. Clark15, H. S. Bartels15, H. L. Amorim16, G. d. Silva Junior17, A. d. Amorim13; 1CEO, Redimama-Redimasto, Belo Horizonte, BRAZIL, 2Mastology, Paulistano Hospital, São Paulo, São Paulo, Brazil., São Paulo, BRAZIL, 3Pos Graduation Department, Santa Casa of Belo Horizonte College, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 4Breast Cancer Center, Redimama-Redimasto, Belo Horizonte, BRAZIL, 5Breast Pathology, Anatomia BH Pathology, Belo Horizonte, BRAZIL, 6Breast Pathology, Analys Pathology, Belo Horizonte, BRAZIL, 7Surgery, Faculty of Medicne of The Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 8Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil., Botucatu, BRAZIL, 9Department of Gynecology and Obstetrics, Faculty of Medicne of The Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 10Cetus Oncology, Belo Horizonte, Minas Gerais, Brazil., MIRA – Medical Group, Belo Horizonte, BRAZIL, 11Breast Unit, Leeds Teaching Hospital NHS Trust St James Hospital., Redimama-Redimasto, Leeds, BRAZIL, 12Radio-oncology, Radiocare, Belo Horizonte, Minas Gerais, Brazil., Belo Horizonte, BRAZIL, 13Mastology, Women Hospital, São Paulo, São Paulo, Brazil., São Paulo, BRAZIL, 14Department of Gynecology and Obstetrics, Hospital of the State Public Servant of São Paulo, Sao Paulo, Sao Paulo Brazil., Belo Horizonte, BRAZIL, 15Mastology, Military Police Hospital of Minas Gerais, Belo Horizonte, BRAZIL, 16Breast Unit, UD Clinic, Belo Horizonte, BRAZIL, 17Mastology, Orizonti Hospital, Belo Horizonte, BRAZIL.
AbstractBackground: The short-term oncological safe of active monitoring for ductal carcinoma in situ (DCIS) with low risk (LR-DCIS) of progression to invasive cancers (IC) has been demonstrated. This study evaluates vacuum assisted biopsy (VAB) as diagnostic test for LR-DCIS active monitoring (AM) in real-world clinical practice.Methods: Database analysis of 116 cancers [both invasive breast cancers (IC) and ductal carcinoma in situ (DCIS)] diagnosed by VAB submitted to standard surgical treatment with complete histological data from VAB and surgery from 04/13/2017 to 11/28/2020. The VAB results matched the surgical pathology, considered the gold standard, and AM criteria. The pathological diagnoses were grouped into malignancies requiring guideline surgical treatment [DCIS with high risk (HR-DCIS) of progression to IC or IC] versus those eligible to alternative active monitoring (LR-DCIS). HR-DCIS/IC were considered positive while LR-DCIS negative results. VAB sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and accuracy were obtained.Results: Mean age 55.6 [± 12.27]; mean IC size 7.14 [± 5.17]mm and 12.6 [± 11.63]mm for DCIS. Out of 116 malignancies diagnosed by VAB, 15 (12.9%) resulted LR-DCIS in the biopsy, 10 (8.6%) confirmed LR-DCIS in surgery, and 5 (4.3%) upgraded to HR-DCIS/IC in surgery. VAB showed 95.28% sensitivity, 100% specificity, PPV was 100%, and NPV 66.67%. VAB LR-DCIS AM was 6% (7/116) and underdiagnoses 1.7% (2 pT1a-bN0 hormone receptor positive).Conclusion: VAB LR-DCIS AM would lead to a moderate (6%) overall reduction of short-term breast cancer surgical overtreatment counterbalanced by few cases of underdiagnosed invasive cancers potentially treatable by adjuvant hormone therapy.Trial registration: retrospectively registered on 10/06/20219 in the Ethics Committee of Santa Casa of Belo Horizonte under the number 25761019.8.0000.5138
Presentation numberPS2-06-10
Pathologic Complete Response by Breast Cancer Molecular Subtype in a Colombian Cohort: Real-World Evidence from Latin America
Monica A Solis Velasco, Universidad del Valle, Cali, Colombia
M. A. Solis Velasco1, V. S. Muñoz-Anacona2, D. F. Penagos-Cabrera1, L. A. Olave-Asprilla1, A. D. Fernandez-Osorio3, U. O. Cardona-Nuñez1, M. Zuluaga-Zuluaga1, V. L. Roman-Vasquez4, A. J. Guerrero-Villota2; 1Department of Surgical Oncology, Universidad del Valle, Cali, COLOMBIA, 2Department of Hematology and Oncology, Clinica Imbanaco, Cali, COLOMBIA, 3Research Centre, Clinica Imbanaco, Cali, COLOMBIA, 4Department of Breast Surgery, Clinica Imbanaco, Cali, COLOMBIA.
Background:Pathologic complete response (pCR) after neoadjuvant therapy (NAT) is a validated surrogate for long-term outcomes in breast cancer. Real-world data from Latin America are limited. We aimed to evaluate the pCR rate across molecular subtypes and its association with survival outcomes. Methods: We conducted a retrospective study of 488 patients with stage I–III breast cancer who received NAT and surgery (2019-2024) at a tertiary center in Cali, Colombia. Data were extracted from electronic records. pCR was defined as ypT0/Tis ypN0. Molecular subtypes were assigned by immunohistochemistry and FISH; staging followed AJCC 8th edition. Most patients received standard NAT according to institutional protocols: AC–T in luminal and AC–TC more often in triple-negative cases. HER2-positive patients received chemotherapy with either single-agent trastuzumab or dual blockade. Non-standard regimens were used based on clinical judgment or comorbidities. Primary outcome was pCR; secondary outcomes included recurrence, disease-free survival (DFS), and overall survival (OS). Associations were assessed using chi-square, Wilcoxon rank-sum, and multivariable logistic regression. Kaplan–Meier and log-rank tests were used for survival analyses. Results: Median age was 52 years (IQR: 44-61). Most tumors were invasive ductal carcinoma (93%) and grade III. Molecular subtypes were: Luminal A (13%), Luminal B (30%), HER2-Luminal B (17%), HER2-enriched (14%), and triple-negative (26%). Clinical stage was II–III in 97%, 56% T1–T2 and 59% node-positive. The overall pCR rate was 34.6% (169/488). Rates varied significantly across molecular subtypes: 60.3% in HER2-enriched, 54.9% in HER2-Luminal B, 44.2% in triple-negative, 17% in Luminal B, and 1.6% in Luminal A (p < 0.001). pCR was significantly more frequent in ER-negative (50.0% vs. 24.5%; p < 0.001) and PR-negative tumors (49.1% vs. 21.0%; p 10%) (60.0% vs. 22.7%, p = 0.001; and 37.1% vs. 18.8%, p = 0.016, respectively). HER2-positive tumors showed greater pCR than HER2-negative (57.3% vs. 24.6%; p 20% (39.0% vs. 13.3%; p < 0.001) and grade III histology (p < 0.001). T1–T2 tumors had higher pCR than T3–T4 (39.5% vs. 28.5%; p = 0.012). No significant associations were found with nodal status (p = 0.690) or clinical stage (p = 0.484). pCR rates did not differ significantly by regimen within molecular subtypes. In triple-negative tumors, adding carboplatin showed no benefit (AC–TC vs. AC–T: 43.6% vs. 47.7%, p = 0.351). In HER2-positive disease, dual vs. single-agent anti-HER2 therapy yielded similar pCR (52.8% vs. 59.2%, p = 0.782). Recurrence was lower in the pCR group (4.7% vs. 12.9%, p = 0.005), particularly for distant metastases (2.96% vs. 9.1%, p = 0.011). Patients who achieved pCR had significantly improved DFS compared to those who did not (log-rank p = 0.047). OS showed a numerical but non-significant difference (1.2% vs. 3.1% mortality, log-rank p = 0.35). Conclusion: In this real-world Latin American cohort, pCR rates and their prognostic significance were consistent with international evidence. HER2-positive and triple-negative tumors showed the highest response, while Luminal subtypes had limited benefit from standard NAT. pCR was independently associated with more favorable DFS. These findings support its value as a surrogate endpoint and highlight the need for subtype-adapted strategies in the neoadjuvant setting, such as immunotherapy in triple-negative disease.
Presentation numberPS2-06-11
Real World Experience of PIK3CA, AKT inhibitors, and oral SERD in patients with hormone receptor positive metastatic breast cancer
Yeonjoo Choi, Cedars-Sinai Medical Center, Los Angeles, CA
Y. Choi1, D. Lin1, P. McAndrew2, M. El-Masry2, D. Park2, N. Banerjee3, C. T. Chung4, D. M. Hoffman2, J. Bitar1, Y. Yuan1; 1Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 2Tower Oncology, Cedars-Sinai Medical Center, Beverly Hills, CA, 3Department of Medical Oncology, Cedars-Sinai Medical Center, Tarzana, CA, 4Department of Medical Oncology, The Angeles Clinic and Research Institute, Santa Monica, CA.
Background PIK3CA/AKT/PTEN pathway alteration is the most common cancer driver pathway in hormone-positive (HR+) metastatic breast cancer(MBC), and multiple PIK3CA/AKT targeted therapies were recently approved for routine clinical use. In addition, oral selected estrogen receptor downregulator (SERD) elacestrant is currently approved for ESR1-mutated HR+ MBC as a single agent only. Approximately 15% of HR+MBC harbor PIK3CA and ESR1 co-mutation. Restricted by available clinical trial eligibilities, there is no clear guideline for rational sequencing of these agents in post-CDK4/6 inhibitor setting. This report is aimed to describe the real-world experience of the PIK3CA pathway inhibitor and oral SERD use in patients with HR+ MBC. Method Patients with HR+/HER- breast cancer at the Cedars-Sinai Medical Center who received PIK3CA pathway inhibitors including alpelisib, inavolisib, or capivaserib were included and dated from January 2019 to June 2025. The retrospective study was conducted through an IRB-approved protocol. Genomic Alterations were identified via standard-of-care commercial NGS platforms. PFS was evaluated as the time to start treatment to disease progression or death from any cause. PFS was calculated by survival analysis and described with Kaplan-Meier curve. Result A total of 107 patients who had PIK3CA pathway alteration and received PI3CK/AKT inhibitors were identified: fulvestrant alpelisib (N=51), fulvestrant capivasertib (N=35), inavolisib (N=2), 2 or more PIK3CA/AKT inhibitors (N=11), and PIK3CA/AKT inhibitors and Elacestrant (N=8). The median age was 66 (34-86). Race: White, 77; African American,11; Asian, 9; other, 6; unknown, 3; declined, 1. 96 out of 107 patients had invasive ductal cancer; 11 patients had invasive lobular carcinoma. Median prior lines of therapy were 2 (0-9). 105 patients received prior CDK 4/6 inhibitors. Median PFS was: alpelisib 3.2 months (95% CI 2.6- 4.5) and 5.4months (CI 3.1-10.9) for capivasertib-treated patients by survival analysis. In 11 patients receiving 2 PIK3CA/AKT inhibitors, mPFS1 was 10.4 months (95% CI 5.8-24.4) and mPFS2 was 9.1 months (95% CI 2.2-N/A). 20 out of 107 patients had PIK3CA and ESR1 co-mutations. Among 20 patients, 8 patients received SERD and PIK3CA/AKT inhibitors sequentially . 3 patients started elacestrant first, and 5 received PIK3CA/AKT inhibitors first. The median PFSs of 3 patients who received 1st line elacestrant followed by AKT inhibitors were 1.6, 7.8, and 20.1 months, respectively. Otherwise, the PFS of 1st line PIK3CA/AKT inhibitor patients were 4.6, 6.7, 9.7, and 12.0 months. One patient with alpelisib experienced treatment interruption and dose reduction due to skin toxicity, while Elacestrant was well-tolerated without major adverse effects. Regarding other co-mutations, the most frequent alteration is CDH1, which is known to be a hallmark in ILC. ATM mutation was the 2nd common alteration (4 of 107), and BRCA1/CHEK2 mutation was observed in 2 patients. Conclusion In this retrospective analysis, 15% of patients with PIK3CA/AKT mutation reported ESR1 co-mutation. Our study is limited by its small sample size, and we observed most patients with ESR1 and PIK3CA pathway co-mutation were able to benefit from sequential therapy of oral SERD or PIK3CA pathway inhibitors, whichever treatment comes first, and showed a favorable response to either treatment. Future effort for a multi-institutional study is planned.
Presentation numberPS2-06-12
Quantifying the Minimum Acceptable Benefit for a Novel Oral Adjuvant Therapy in HR+/HER2− Early Breast Cancer: A Probabilistic Threshold Technique Study in Italy
Alberto Zambelli, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
A. Zambelli1, R. Berardi2, B. Canali3, R. Mercati3, A. Messi4, M. Suter5, I. Viganò5, D. Valsecchi6, G. Arpino7; 1Oncology Unit, ASST Papa Giovanni XXIII Hospital, Bergamo, ITALY, 2Department of Medical Oncology, Università Politecnica delle Marche, AOU delle Marche, Ancona, ITALY, 3Real World Solutions, IQVIA Solutions Italy S.r.l., Milan, ITALY, 4Therapeutic area communications and advocacy, Novartis Farma S.P.A., Milan, ITALY, 5Medical Oncology, Novartis Farma S.P.A., Milan, ITALY, 6Evidence Generation, Novartis Farma S.P.A., Milan, ITALY, 7Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, ITALY.
Background: Hormone receptor positive (HR+), human epidermal growth factor receptor 2 negative (HER2-) breast cancer (BC) is the most frequently diagnosed BC subtype. In HR+/HER2- early breast cancer (eBC), adjuvant endocrine therapy (ET) – administered after surgery, with or without radiotherapy or chemotherapy – is the standard of care to reduce recurrence risk. Recently, combining ET with cyclin-dependent kinase 4/6 inhibitors has emerged as a new adjuvant strategy for patients with HR+/HER2− eBC. As adjuvant therapy aims to prevent recurrence rather than cure, understanding patients and clinicians’ preferences regarding risk-benefit trade-offs in this setting is critical. This study uses a probabilistic threshold technique (PTT) to estimate the minimum additional benefit (MAB) in 5-year invasive disease-free survival (iDFS) required by Italian patients and clinicians to prefer a novel oral adjuvant therapy over the current standard of care. Methods: The PTT questionnaire, simplified in wording for patients, was structured around a reference and a target option described by six attributes, whose levels were based on published data. The reference option reflected the profile of endocrine therapy (ET) alone, while the target option of ribociclib plus ET (Table 1). Respondents were blinded to the real-world identity of these treatments, labeled in the questionnaire as Treatment A and B. The key attribute (5-year iDFS) was initially set at 72% for both alternatives based on the efficacy of the reference option and its value varied iteratively for the target option at each question, until the respondent changed their initial choice or was indifferent between the two alternatives. Descriptive statistics were used to analyze responses. Results: A total of 112 clinicians and 99 patients were included in the final analysis. Clinicians required an average increase in iDFS of 5.17 percentage points (p.p.) (SD=6.26), while patients required 6.40 p.p. (SD=7.98) to prefer the target over the reference; the difference between the two groups was not statistically significant (t-test, p=0.21). Furthermore, no significant differences were observed within each group based on relevant personal characteristics. Median MAB was 4.50 p.p. for clinicians and 4.00 p.p. for patients. Patient preferences were more polarized: 33.3% accepted a MAB ≤1 p.p., while 25.3% required >10 p.p., compared to 25.0% and 12.5% of clinicians, respectively. Conclusion: The analysis provides quantitative insights into how Italian patients and clinicians evaluate risk-benefit trade-offs of novel adjuvant therapies for HR+/ HER2- eBC. While average MAB are similar, patient preferences are more polarized, ranging from very low to very high efficacy thresholds. This heterogeneity reinforces the need for shared decision-making that incorporates individual patient values.
| Attribute | Reference option | Target option |
| 5-year iDFS | 72% | 72% |
| Grade 3+ neutropenia | 1% | 44% |
| Grade 3+ diarrhea | 0% | 1% |
| Treatment schedule and duration | One drug, oral, daily for 5 years |
Two drugs: 1) Oral, daily for 5 years; 2)Oral, daily for 21 days followed by 7 days off for 3 years |
| Potential for dose modifications | No | Yes |
| Impact on sexual health | Negative impact | Negative impact |
Presentation numberPS2-06-13
Implementation of Liquid Biopsy in a Real-World Clinical Setting: Guiding Treatment Decisions in Metastatic Breast Cancer through Genomic Profiling
Alvaro Garcia-Bauto, Institut Català d’Oncologia, Barcelona, Spain
A. Garcia-Bauto1, D. Azuara Garcia2, M. Gil-Gil1, J. Cruz Velez1, A. Vethencourt Casado1, B. Fullana Griimalt1, A. Fernández Ortega1, P. Sabat Viltro1, S. Vázquez Fernández1, C. Ejarque Martínez1, S. Recalde Penabad1, A. Stradella1, V. Obadia Gil1, R. Villanueva Vázquez1, S. Pernas Simón1, C. Lázaro3, C. Falo1; 1Medical Oncology, Institut Català d’Oncologia, Barcelona, SPAIN, 2Bellvitge Biomedical Research Institute, Idibell, Barcelona, SPAIN, 3Bellvitge Biomedical Research Institute, IDIBELL, Barcelona, SPAIN.
Introduction: Metastatic breast cancer (mBC) exhibits tumor evolution and genomic heterogeneity with somatic alterations linked to progression and therapeutic resistance. We conducted a prospective Real-World Data study to evaluate ctDNA-detected mutations (MUT) via LB and their association with clinical outcomes. Methods: Prospective, single-center study of 98 mBC pts undergoing pre-treatment ctDNA analysis (mainly 1st/2nd line). Plasma was collected in Streck® tubes and analyzed with the Plasma-SeqSensei™ BC panel (Sysmex®), targeting PIK3CA, ESR1, ERBB2, AKT1, and TP53. Progression free survival to 1st-line metastatic therapy (PFS1) was estimated by Kaplan-Meier and log-rank test according to prevalent mutations; prognostic impact assessed by multivariate (MVA) Cox models (p≤0.05). Results: Of 98 mBC pts, 85 were evaluable and included in the final analysis. Baseline characteristics are summarized in Table 1. ctDNA identified ≥1 mutation in 52 pts (61%) of whom 81% had actionable mutations: PIK3CA (30%), ESR1 (18%), ERBB2 (7%), AKT1 (2%). Most frequent PIK3CA variants were E545K (28%), H1047R (21%), and E726K (15%); ESR1 variants included D538G (38%) and Y537S (15%). TP53 was enriched in grade III tumors (30% vs. 10%; p=0.04), ERBB2 in lobular histology (75% vs. 25%; p=0.01), ESR1 in pretreated pts (21% vs. 12%, p=0.39), and PIK3CA in younger pts (<65y: 80% vs. 56%; p=0.03). In the overall cohort, TP53 MUT were associated with shorter PFS1 vs wild-type (WT) (17.7 vs. 23.9 months (mo); p=0.02); PIK3CA and ESR1 MUT showed no significant impact. In CDK4/6 inhibitors-naïve pts (23%), TP53, PIK3CA, and ESR1 MUT were significantly associated with shorter PFS1 (10 vs. 64; 19 vs. 56.6; 17 vs. 41.5 mo; respectively p=0.001/0.01/0.05); no such associations were observed in pretreated patients. MUT-negative patients had significantly longer PFS1 than those harboring ≥1 mutation (66.5 vs. 28.4 mo; p=0.003). In pts with de novo stage IV, relapse during or after adjuvant endocrine therapy (ET), or first-line hormonal treatment, PIK3CA-MUT tumors showed shorter PFS1 compared to WT on both fulvestrant + CDK4/6i (3 vs. 17 mo; p=0.03) and aromatase inhibitor (AI) monotherapy (52 vs. 128 mo; p=0.046). ESR1-MUT conferred worse PFS1 vs. WT on AI – alone (21 vs. 113 mo; p=0.025) and AI+ CDK4/6i (27.5 vs. 59.5 mo; p=0.038) – but not with fulvestrant + CDK4/6i (15 vs. 8 mo; p=0.8). MVA identified TP53 MUT (HR 2.1; p=0.03) and prior ET (HR 3.6; p=0.016) as independent prognostic factors. Conclusion: ctDNA analysis in mBC identifies mutations with prognostic and treatment-specific relevance. These findings support the clinical utility of LB to refine risk stratification and optimize endocrine-based therapy.
| All patients (N=85) | PIK3CA Mutant | PIK3CA Wild-type | P Value | ESR1 Mutant | ESR1 Wild-Type | P Value | |||||||||||||||||||||||||
| Age – median (range) | 61 (51-69) | 57 (50- 66 | 62 (50-73) | 0.078 | 64 (56-72) | 59 (48-70) | 0.101 | ||||||||||||||||||||||||
| Sex (male/female) | 1(1) / 86 (99) | ||||||||||||||||||||||||||||||
| Histology | 0.678 | 0.359 | |||||||||||||||||||||||||||||
| Ductal | 69 (81) | 22 (85) | 46 (80) | 14 (93) | 54 (77) | ||||||||||||||||||||||||||
| Lobular | 16 (19) | 4 (15) | 12 (20) | 1 (7) | 15 (23) | ||||||||||||||||||||||||||
| Grade | 0.857 | 0.704 | |||||||||||||||||||||||||||||
| I-II | 66 (77) | 22 (84) | 46 (80) | 12 (80) | 54 (784) | ||||||||||||||||||||||||||
| III | 13 (15) | 4(15) | 9 (17) | 3 (20) | 10 (16) | ||||||||||||||||||||||||||
| Treatment scenario at the time of LB | 0.396 | 0.181 | |||||||||||||||||||||||||||||
| No previous treatment | 29 (34) | 11 (42) | 18 (30) | 3 (20) | 26 (37) | ||||||||||||||||||||||||||
| 1st line or progression disease during adjuvant treatment | 44 (51) | 13 (50) | 31 (53) | 11 (73) | 33 (47) | ||||||||||||||||||||||||||
| 2nd or more lines | 12 (14) | 2 (8) | 10 (17) | 1 (7) | 11 (16) | ||||||||||||||||||||||||||
| Previous treatment | 0.227 | 0.773 | |||||||||||||||||||||||||||||
| Yes | 53 (70) | 13 (50) | 20 (34) | 10 (67) | 42 (60) | ||||||||||||||||||||||||||
| No | 34 (40) | 13 (50) | 39 (66) | 4 (31) | 28 (40) | ||||||||||||||||||||||||||
| Subtype | 0.519 | 0.125 | |||||||||||||||||||||||||||||
| Luminal A-like | 38 (46) | 15 (58) | 23 (41) | 6 (43) | 32 (47) | ||||||||||||||||||||||||||
| Luminal B-like | 39 (48) | 10 (38) | 29 (52) | 39 (50) | 32 (47) | ||||||||||||||||||||||||||
| HER2 | 4 (5) | 1 (4) | 3 (5) | 0 (0) | 4 (6) | ||||||||||||||||||||||||||
| Triple negative | 1 (1) | 0 (0) | 1 (2) | 1 (7) | 0 (0) | ||||||||||||||||||||||||||
| Site | 0.067 | 0.772 | |||||||||||||||||||||||||||||
| Non-visceral | 51 (60) | 12 (47) | 39 (66) | 10 (67) | 41 (59) | ||||||||||||||||||||||||||
| Visceral | 34 (40) | 14 (54) | 20 (34) | 5 (33) | 29 (41) | ||||||||||||||||||||||||||
Presentation numberPS2-06-14
Breast Cancer (BC) in Younger Women: A Multicentre Cohort Study of Clinical and Pathological Features
Karen Desouza, University College London Hospital NHS, London, United Kingdom
R. Boopathy1, R. Javed2, B. Gwyther3, B. Kirresh2, J. Islam3, A. Hallam1, E. Papadimitraki2, D. Betal2, K. Desouza2; 1Medical Oncology, University College London, London, UNITED KINGDOM, 2Medical Oncology, University College London Hospital NHS, London, UNITED KINGDOM, 3Medical Oncology, Whittington Health NHS, London, UNITED KINGDOM.
Background Younger women with BC have more aggressive disease and poorer survival than older women.¹ Incidence is rising faster in this group (1.4% in <50y vs 1% overall), yet they remain underrepresented in trials, limiting insights into their distinct tumour biology and risk profiles.²˒³ Aim To examine risk factors, clinicopathological features, age-related trends, and treatment responses in women <50y with BC. Methods We conducted a multicentre retrospective study of women <50y (n=337) diagnosed with invasive BC. All cases were discussed at the University College London Hospital (UCLH) and Whittington Health (WH) Breast Multidisciplinary Team meetings (May 2019-February 2025). Patients were stratified into five age groups: ≤30, 31-35, 36-40, 41-45, 46-49y. Variables included age at diagnosis, risk factors, tumour characteristics, recurrence, and treatment data. Categorical variables were compared with Chi-squared/Fisher’s exact tests; continuous variables with Mann-Whitney/Kruskal-Wallis tests. Logistic regressions were performed to assess associations. Results Patients aged ≤30 had more node-positive disease at diagnosis than patients aged 41-49 (OR 3.085; p=0.022). Younger age was associated with a higher-grade (≤30y: 73.7% G3; p=0.002) and a higher Ki-67 (p=0.028). The youngest women had lower BMI (p=0.026). A higher BMI was associated with increased odds of G3 tumours (p=0.024) and TNBC (p=0.007). The full <50 cohort showed higher rates of G3 (42.7%) and node-positive tumours (44.8%) than general BC population datasets.4,5 Most BC diagnoses in parous women occurred within 10 years postpartum (57.5%), supporting a postpartum risk window. Genetic mutations were more frequent in younger women (p=0.020). 47.2% of mutation carriers had no family history of BC. Neoadjuvant systemic therapy was administered to 26.1% of patients, achieving a pathological complete response in 64.7% of TNBC and 61.1% of HER2-positive cases. At the time of analysis, 15 patients (4.5%) had documented recurrence and 6 (1.8%) had died. Conclusions: BC in women <50y, particularly those ≤30y, is characterised by higher grade, elevated Ki-67, and more frequent nodal positivity. Clinicians must maintain a high index of suspicion when evaluating breast symptoms in young women, and perform careful nodal assessment. Our findings also challenge the reliance on family history for genetic testing, indicating that the current UK testing criteria should be expanded to a wider patient cohort to avoid missing pathogenic mutations. Future work must prioritise survivorship, addressing impacts on fertility, premature menopause, and psychological wellbeing in this population. A key limitation of this study is its small retrospective design. To strengthen the evidence base, a prospective registry capturing clinical, pathological, and survivorship data in young women with BC should be considered.
Presentation numberPS2-06-15
Population-based outcomes and patterns of care for patients with stage I-III, HER2-negative breast cancer, by estrogen receptor expression levels: a high-resolution study.
Fabio Girardi, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
F. Girardi1, S. Guzzinati2, M. V. Dieci3, P. Napolitano3, L. Dal Maso4, F. Puglisi5, V. Guarneri3, M. Zorzi2; 1Division of Medical Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, ITALY, 2Epidemiological Department, Azienda Zero, Padua, ITALY, 3Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, ITALY, 4Cancer Epidemiology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, ITALY, 5Department of Medicine, University of Udine, Udine, ITALY.
Background HER2-negative breast cancer (BC) encompasses a wide range of estrogen receptor (ER) expression levels. Currently, tumors are classified as ER-positive if at least 1% of tumor cells express ER. However, outcomes vary significantly across this spectrum. Notably, ER-low tumors (ER 1-9%) have been associated with outcomes as poor as those observed in triple-negative breast cancer (TNBC, ER 0%). Despite this heterogeneity, few population-based studies have systematically assessed outcomes across the continuum of ER expression, while accounting for treatment details and tumour characteristics. To address this gap, we analyzed a large, population-based cohort of women diagnosed with stage I-III, HER2-negative BC. Methods We used a high-resolution dataset of individual-level records from the Veneto Tumour Registry, a large population-based cancer registry in Italy. Information on stage, ER levels, grade, and Ki67 was collected. Patient-level data on surgery, chemotherapy (CT), and endocrine therapy (ET) were retrieved from population-based prescription repositories and claims. Patients were stratified by ER level expression: 0% (TNBC), 1-9% (ER-low), 10-49%, and ≥50%. Eligible CT treatments were those given within 12 months before or after surgery. Eligible endocrine treatments included letrozole, anastrozole, exemestane, or tamoxifen following surgery. We estimated cancer-specific survival by ER category, up to six years after diagnosis, using validated cause of death data. Results The study population included 3,771 patients 18 years of age or older, diagnosed with stage I-III, HER2-negative BC between 2017 and 2021, with follow-up until December 31, 2024. The majority of patients (77.8%) were aged 50 years or older. Stage I tumours accounted for 60.9% of cases, while stage II and III tumours made up 31.4% and 7.6%, respectively. ER expression levels were as follows: 6.9% of patients had TNBC, 1% had ER-low, 1.6% had ER 10-49%, and 90.5% had ER ≥50%. Most tumors were grade 1 or 2 (75.4%), and 63.2% had a Ki67 index below 20%. Among patients with an ER level ≥1%, ET was given to 23.7% of those with ER-low, 79% with ER 10-49%, and 89.7% with ER ≥50%. ET discontinuation within the first year remained below 10% across all groups. CT was received by 79.9% of patients with TNBC, 81.6% with ER-low BC, 59.7% with ER 10-49%, and 29.5% with ER ≥50%. Three-year cancer-specific survival was 90.2% for TNBC, 90.5% for ER-low, 94.6% for ER 10-49%, and 98.4% for ER ≥50%. At six years post-diagnosis, survival declined to 85% for patients with TNBC, stabilized around 90% for both ER-low and ER 10-49% groups, and remained above 95% in the ER ≥50% group. Conclusion To our knowledge, this is one of the largest population-based studies to report outcomes and cancer care patterns in patients with HER2-negative early BC across the full spectrum of ER expression, treated in real-world clinical practice. Our findings highlight that survival varies by ER expression level and over time. Patients with TNBC or ER-low BC showed similarly poor short-term outcomes. Over the longer term, survival in the TNBC group declined steadily, whereas trajectories for the other subgroups, including ER-low, were less steep. The 5% absolute difference in six-year cancer-specific survival between TNBC and ER-low BC may suggest a modest benefit associated with ET, which was received by up to one-quarter of patients in the ER-low group. However, the small sample size in this subgroup limits definitive conclusions. Larger, high-resolution, population-based studies with long follow-up are needed to validate the impact of real-world treatment practices and inform cancer care.
Presentation numberPS2-06-16
Two cohorts of young women (<41y at diagnosis) with invasive breast cancer: Single center data on how demographics, tumor characteristics, therapies and 5-year survival differ when treated 2000-2005 and 2014-2019
Brechje Goots, University Hospitals Leuven, Leuven, Belgium
B. Goots1, A. Laenen2, S. Han1, M. Van Houdt1, Y. Van Herck3, F. Derouane3, C. Weltens4, H. Janssen4, A. Baten4, J. Verhoeven4, G. Floris5, C. Desmedt6, K. Van Baelen1, A. Smeets7, I. Nevelsteen7, C. Van Ongeval5, M. Keupers5, V. Celis5, A. Coessens5, R. Prevos5, H. De Boodt5, H. Wildiers3, P. Neven1; 1Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, BELGIUM, 2Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, BELGIUM, 3Department of General Medical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 4Department of Radiotherapy, University Hospitals Leuven, Leuven, BELGIUM, 5Department of Imaging and Pathology, University Hospitals Leuven, Leuven, BELGIUM, 6Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, BELGIUM, 7Department of Surgical Oncology, University Hospitals Leuven, Leuven, BELGIUM.
Background: Over the past two decades, breast cancer characteristics and outcomes have improved, largely due to earlier detection through increased awareness and screening, as well as improvements in (neo-)adjuvant therapies. However, real-world data on breast cancer in women up to age 40 remain rare.Methods: This retrospective study compares demographics, tumor characteristics, therapies and 5-year survival between two cohorts of female patients with invasive breast cancer diagnosed before the age of 41 at UZ Leuven, during the periods 2000-2005 (cohort 1) and 2014-2019 (cohort 2). The Mann-Whitney U test was used for continuous variables and Fisher’s exact test for categoric variables. Significance was defined if p<0.05. Results: Cohort 1 consist of 233 patients, with a mean age of 35.5 years at diagnosis. Cohort 2 consists of 237 patients, with a mean age of 34.9 years at diagnosis. A germline mutation was detected in 36/108 (33.3%) of patients in cohort 1 and in 51/206 (24.8%) of patients in cohort 2; this did not significantly differ between the two cohorts. See table: At diagnosis, patients in cohort 2 were significantly more likely to have been detected through screening, to have a second-degree relative with breast cancer, to be pregnant, to have had their first full-term pregnancy at an older age, to have breastfed and to have used an intrauterine device. They were significantly less likely to have used oral contraceptives at diagnosis. In addition, tumors were significantly more likely multifocal and ER>1% HER2 amplified, but significantly less likely lobular, grade 3, and ER <1% HER2 amplified. Significant more patients in cohort 2 were metastatic at diagnosis, received neoadjuvant and anti-HER2 therapy. At year 5, significantly more patients in cohort 2 were disease free, distant relapse free and alive.Conclusion: A comparison of women under 41 years diagnosed with invasive breast cancer across two time periods revealed differences in demographics, tumor characteristics, systemic treatments, and substantial disparities in outcomes. Mean age at first full-term pregnancy significantly increased with more than 1 year, the number of multifocal tumors doubled, the number of lobular tumor decreased from nearly 8% to nearly 1%, the number of patients that received neoadjuvant therapy nearly tripled and 5-year distance relapse decreased from nearly 20% to nearly 6% when cohort 1 was compared to cohort 2.
| Variable | Cohort1 | Cohort2 | p-value |
| Screening | 15/225(6.7%) | 30/235(12.8%) | 0.029 |
| Pregnantatdiagnosis | 5/230(2.2%) | 18/237(7.6%) | 0.009 |
| Meanageatfirstfull-termpregnancy | 26.8yrs | 27.9yrs | 0.027 |
| Everbreastfeed | 81/123(65.9%) | 113/146(77.4%) | 0.041 |
| Intrauterinedeviceatdiagnosis | 29/219(13.2%) | 57/223(25.6%) | 0.001 |
| Oralcontraceptiveatdiagnosis | 92/219(42%) | 71/223(31.8%) | 0.030 |
| Multifocal | 32/228(14%) | 70/223(31.4%) | <0.001 |
| Grade1 | 10/232(4.3%) | 18/234(7.7%) | 0.125 |
| Grade2 | 75/232(32.3%) | 94/234(40.2%) | 0.078 |
| Grade3 | 147/232(63.4%) | 122/234(52.1%) | 0.014 |
| Lobular | 18/231(7.8%) | 4/236(1.7%) | 0.002 |
| Ductal | 208/231(90%) | 228/236(96.6%) | 0.005 |
| Mixed | 5/231(2.2%) | 4/236(1.7%) | 0.749 |
| LuminalA-like | 71/227(31.3%) | 76/237(32.1%) | 0.921 |
| LuminalB-like | 53/227(23.4%) | 47/237(19.8%) | 0.368 |
| ER+/HER2+ | 26/227(11.5%) | 48/237(20.3%) | 0.011 |
| ER–/HER2+ | 26/227(11.5%) | 14/237(5.9%) | 0.046 |
| TNBC | 51/227(22.5%) | 52/237(22%) | 0.911 |
| anti-HER2therapy | 12/233(2.2%) | 57/237(24.1%) | <0.001 |
| Neoadjuvanttherapy | 36/233(15.5%) | 103/237(43.5%) | <0.001 |
| Metastaticatdiagnosis | 5/233(2.2%) | 14/237(5.9%) | 0.038 |
| Diseasefreeafter5y | 176/232(75.9%) | 196/226(86.7%) | 0.004 |
| 5-yearmortality | 32/232(13.8%) | 14/226(6.2%) | 0.008 |
| 5-yeardistantrelapse | 46/232(19.8%) | 13/226(5.8%) | 0.006 |
Presentation numberPS2-06-19
Treatment Trends in Early-Stage Breast Cancer: A Population-Based Study of Medicare Fee-For-Service Enrollees in Texas
Laura B Amin, The University of Texas MD Anderson Cancer Center, Houston, TX
L. B. Amin1, C. Shaefer2, G. Cecilia2, D. Allen-Benson2, E. Caballero2, M. Chavez-MacGregor3; 1Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 2UTHealth Houston School of Public Health, UTHealth Houston School of Public Health, Houston, TX, 3Breast Medical Oncology and Health Services Research Departments, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Early-stage breast cancer is typically treated with a multi-modality approach. Among older patients, concerns exist about suboptimal cancer care delivery and the possibility of greater risk of experiencing treatment delays. Since treatment delays are associated with adverse outcomes, we sought to describe patterns of care among older patients with breast cancer in Texas and estimate delays in chemotherapy administration.Methods: Using Medicare Fee-For-Service parts A, B, and D claims data, we identified individuals in Texas who underwent unilateral or bilateral mastectomy or lumpectomy from 2017 through 2020, had a breast cancer diagnosis code and had 1 year of continuous enrollment before and after surgery. Individuals with metastatic disease and malignancies of non-breast origin were excluded. Using CPT, HCPCS, and NDC codes, we developed an algorithm to identify breast cancer subtype according to treatment received. A total of 5,988 patients were identified and categorized. Among the 1,562 patients treated with chemotherapy the number of days from diagnosis to first dose of chemotherapy was estimated for patients receiving neoadjuvant chemotherapy and the number of days between surgery to chemotherapy for those treated in the adjuvant setting. Treatment delay was defined as >60 days. Descriptive statistics were reported and logistic regression models were performed to predict the odds of delaying chemotherapy.Results: Of the 5,988 individuals identified with early-stage breast cancer (HR-positive 81.5%, TNBC 11.4, HER2-positive 7.1%), median age was 72 years old [QR 69-77]. Majority (91.8%) were White and 17.6% resided in rural counties. Among those receiving chemotherapy, 32.3% received it in the neoadjuvant setting (35.9% TNBC, 34.6% HER2-positive, and 22.9% HR-positive). The median time from diagnosis to neoadjuvant chemotherapy was 34 days. For patients that received adjuvant chemotherapy, the median number of days from surgery to first dose of chemotherapy was 48 days. Overall, 24.8% of patients receiving chemotherapy experienced a delay. Among patients who received neoadjuvant chemotherapy, those living in counties with higher high-school graduation rates were significantly less likely to have a chemotherapy delay (OR=0.41, 95%CI 0.19-0.87), more comorbidities (OR=1.19 95%CI 0.99-1.41) had a borderline association with more delays. For those who received adjuvant chemotherapy, older age (OR=1.2, 95%CI 1.04-1.38), and receiving MammaPrint or Oncotype testing (OR=1.71, 95%CI 1.24-2.36), was associated with a higher likelihood of chemotherapy delay. Estimates for patients treated for TNBC and HER2-positive breast cancer suggested a lower likelihood of experiencing delays compared to those with HR-positive tumors.Conclusions: Among older individuals with early-stage breast cancer enrolled in Medicare Fee-For-Service in Texas, 24.8% of patients receiving chemotherapy experienced a delay > 60 days from either diagnosis to chemotherapy or surgery to chemotherapy. Understanding the patterns of care among older patients can provide important insights into treatment patterns and identify vulnerable patients.
Presentation numberPS2-06-20
Fr-popea: French real-world-data of peri-operative pembrolizumab KN522 regimen in extreme ages subgroups (<30 y.o and >70 y.o) with high-risk TNBC.
Jean Sebastien Frenel, Institut de Cancérologie de l’Ouest, Saint Herblain, France
M. Robert1, A. de Nonneville2, E. Volant1, C. Liautard3, C. Perkins3, F. Penault Llorca4, O. Tredan5, J. S. Frenel1; 1Medical Oncology, Institut de Cancérologie de l’Ouest, Saint Herblain, FRANCE, 2Medical Oncology, Aix-Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, FRANCE, 3Medical affairs, MSD France, Puteaux, FRANCE, 4Pathology, Centre Jean Perrin, Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont Ferrand, FRANCE, 5Medical Oncology, Centre Leon Berard, Cancer Research Center of Lyon (UMR Inserm 1052 – CNRS 5286), Lyon, FRANCE.
Background: The current standard of care for early triple-negative breast cancer (eTNBC) patients is peri-operative pembrolizumab, as demonstrated by the improved pathological response and survival outcomes in the KEYNOTE-522 trial. However, the youngest and oldest patients are underrepresented in randomized clinical trials. Methods: The FR-POPEA study is a prospective, observational, subgroup cohort analysis of high-risk TNBC patients who were treated with perioperative pembrolizumab in the French KN522 Early Access Program (EAP) from April 2022 to November 2024. Eligible patients were adults with confirmed stage II-III TNBC (including ER 1-9% according to the French guidelines). Real-world data were collected through EAP forms completed by physicians. Clinical and safety data were collected until the end of the EAP or the end of the treatment. The oldest group was ≥70 years old (the control group was <70 years old), and the youngest group was <30 years old (the control group was ≥30 years old). Results: Over the 31-month duration of the EAP, a total of 7,687 patients were included from 427 different French healthcare centers. The oldest population represented 12.5% (n=962 patients) of the whole population (with a cutoff of ≥65y it represents 21,8% (n=1,678)). Notably there were 106 octogenarians and three nonagenarians. Compared to the control group, the oldest patients (≥65y) had higher rates of comorbidities such as cardiovascular disease (21.6% vs. 6.5%) and diabetes (12.6% vs. 4.7%), a greater proportion of stage III disease (43.6% vs. 38.4%), and a higher proportion of ECOG ≥1 (37.1% vs. 11.1%). Pembrolizumab was combined with a different chemotherapy regimen than KN522 trial in 5% of cases, compared to 1.2% in the control group. A total of 187 interruptions for pembrolizumab treatment were reported and 28.3% of them were due to an adverse reaction suspected to be immune-related (irAE) compared to 36.3% for the control group. Permanent discontinuations of pembrolizumab were documented in 178 patients (18.5%), with 41.6% due to an irAE (vs. 50.1% in the control group). A lower rate of breast-conserving surgery (60.6% vs. 64.2%) was observed. The pathological complete response (pCR) rate was lower among the oldest group (64.9% vs 72.4%). The youngest population represented 2% (n=153 patients) of the total cohort, increasing to 17.8% (n=1,370) when using an age cutoff under 40 years. The under 30y group exhibited a higher rate of family history of breast cancer (40.5% vs. 33.6%), a similar proportion of stage III disease (40.5% vs 39%) and a lower rate of ECOG≥1 (7.2% vs. 13.4%). Twenty-three interruptions for pembrolizumab were reported and 52.2% of them were due to an irAE (35.2% for the control group). Furthermore, pembrolizumab was permanently discontinued in 21 patients (13.7%) with 61.9% of them were due to an irAE (64.8% in the control group). Clinical outcomes indicated a higher rate of breast-conserving surgery (71.1% vs. 63.7%) and a slightly higher pCR rate (73.3% vs. 71.4%) compared to the control group. Conclusions: The FR-POPEA study is the largest real-world assessment of the KN522 regimen in both the oldest and youngest subgroups. These results offer valuable real-world confirmation of the feasibility, efficacy, and safety of peri-operative pembrolizumab combined with chemotherapy in early-stage TNBC. Although there is a lack of extensive data on BRCA mutation status and formal geriatric assessments, these outcomes are reassuring and complement evidence from randomized trials.
Presentation numberPS2-06-21
Real-word utilization of the 21-gene assay for guiding treatment decisions in patients with HR+/HER2- early breast cancer in the US
Gebra Cuyun Carter, Exact Sciences, Madison, WI
G. C. Carter1, J. Racz2, N. Gu1, J. Bennett3, Q. A. Le1, M. Kohli4, J. Rodriguez-Silva4, Y. Barhoush5, L. Batchu5, Y. Abdou6; 1Health Economics and Outcomes Research, Exact Sciences, Madison, WI, 2Medical Affairs, Exact Sciences, Redwood City, CA, 3Biostatistics, Exact Sciences, Redwood City, CA, 4Research/RWE, STATinMED, Dallas, TX, 5Programing/Analytics, STATinMED, Dallas, TX, 6Department of Medicine, Division of Oncology, UNC School of Medicine, Chapel Hill, NC.
Objective: The 21-gene assay is both prognostic of distant recurrence and predictive of chemotherapy benefit and has been validated by evidence from randomized controlled trials in hormone receptor positive (HR+), human epidermal growth factor receptor-2 negative (HER2-) early-stage breast cancer (BC). This study explored real-world utilization of the 21-gene assay and subsequent treatment patterns in the US population. Methods: A retrospective study was conducted using de-identified data from Exact Science’s commercial database linked to STATinMED RWD Insights, an all-payer medical and pharmacy claims dataset. Female patients aged ≥18 years with ≥1 inpatient or ≥2 outpatient BC claims between January 1, 2015, and November 30, 2023, were included. First BC diagnosis was defined as the index date with patients required to have data 12 months pre- and 30 days post-index date. Patients were excluded if the index date occurred after the testing date, were pregnant, had another cancer diagnosis, or treated for cancer during the baseline period. Variables of interest included Recurrence Score® (RS) result and date, nodal, HR, and HER2 gene expression status and chemotherapy (CT) use (defined as any CT use within 12 months of index date). Descriptive analyses were conducted in the overall population by nodal status, by RS result category, and by race and ethnicity (Non-Hispanic Black (NHB), Non-Hispanic White (NHW), Asian, and Hispanic).Results: Among 119,383 early-stage HR+/HER2- BC patients in this analysis who underwent 21-gene assay testing, 21,706 (18.2%) had node-positive (N1), and 97,677 (81.8%) had node-negative (N0) disease. Among patients with N0 disease and available data in the specified groups, majority were NHW (56.5%), followed by Hispanic (9.0%), NHB (6.1%), and Asian (2.0%); a similar distribution was seen among patients with N1 disease. In the N0 cohort, most patients had low (0-10; 26.0%) or intermediate (11-25; 61.0%) RS results, while 12.9% had high (26-100) RS results. The distribution based on RS result was similar in the N1 cohort (86.1% low/intermediate risk (0-25), and 13.9% high risk (26-100)). The proportion of patients with a high RS result appeared numerically higher among NHB patients in both N0 (14.9%) and N1 (16.5%) compared with NHW (12.7%, 13.8%), Asian (12.5%, 14.3%), and Hispanic (12.9%, 14.2%) patients. Across both N0 and N1 disease, CT use increased with higher RS category overall and within each race and ethnic group. However, there were no apparent differences in CT use by race or ethnicity. Conclusions: This large real-world analysis of patients with early-stage HR+/HER2- BC shows that CT utilization increases appropriately with higher RS results, consistent with guideline-based care. Importantly, CT use was generally consistent across racial and ethnic groups, suggesting equitable application of RS-guided treatment recommendations.
Presentation numberPS2-06-22
Genomic Insights in Metastatic Invasive Lobular Carcinoma: A Real-World Study
Pegah Farrokhi, University of Minnesota, College of Pharmacy, Minneapolis, MN
P. Farrokhi1, D. Stenehjem2; 1Department of Pharmaceutical Care and Health Systems, University of Minnesota, College of Pharmacy, Minneapolis, MN, 2Department of Pharmacy Practice and Pharmaceutical Sciences, University of Minnesota, College of Pharmacy, Duluth, MN.
Background: Despite recognition that invasive lobular carcinoma (ILC) harbors distinct genomic alterations that bypass estrogen and CDK4/6 dependence, systematic outcome data linking specific alterations to therapeutic response remain limited. This represents a critical research gap, as ILC comprises 10-15% of breast cancers yet exhibits unique biological behavior and treatment resistance patterns compared to invasive ductal carcinoma. This study aimed to identify how individual genomic alterations affect clinical outcomes in patients with metastatic ILC. Methods: This observational cohort study used the Flatiron Health-Foundation Medicine clinicogenomics database to identify adults with hormone receptor-positive, HER2-negative metastatic ILC who underwent genomic profiling between 90 days before metastatic diagnosis and 90 days after initiation of first-line therapy (January 2015-September 2022). Patients enrolled in clinical trials or with less than six months of follow-up were excluded. We evaluated alterations in PIK3CA, RB1, ERBB2, ERBB3, NF1, AKT1, PTEN, FGFR1, FGFR2, FGFR3, ARID1A, TBX3, MDM4, ATM, MYC, and GATA3—assessing ERBB2 only in tumors with HER2-negative status confirmed by IHC and FISH. Overall survival (OS), time to treatment discontinuation (TTD), and time to next treatment (TTNT) were compared using Kaplan-Meier analysis and multivariable Cox proportional hazards models adjusted for age, tumor grade, menopausal status, ECOG performance status, and co-alterations. A subgroup analysis focused on patients receiving first-line CDK4/6 inhibitors plus endocrine therapy (CDK4/6i + ET). Results: A total of 256 patients were included (median age 65 years), with 141 patients (55%) receiving first-line CDK4/6i + ET. NF1 alterations (16%), often co-occurring with PIK3CA alterations, were associated with significantly worse TTNT (6.8 vs 11.8 mo; HR=2.2, p<0.01) and TTD (5.6 vs 8.5 mo; HR=1.8, p<0.01) in the overall cohort, and shorter TTD (5.4 vs 12.5 months; HR=2.6, p<0.01) and TTNT (6.9 vs 15.6 mo; HR=3.2, p<0.01) in the CDK4/6i+ET subgroup. RB1 alterations (7%), with frequent co-occurring PIK3CA and PTEN mutations, were associated with significantly shorter TTNT (6.3 vs 10.6 mo; HR=1.9, p=0.04), TTD (4.7 vs 8.3 mo, HR= 1.8, p=0.05), and numerically shorter OS (20.9 vs 29.4 months; HR=1.2, p=0.6) in the overall cohort. ARID1A (16%) was associated with significantly shorter overall OS (20.1 vs 31.8 mo; HR=1.7, p=0.03). Among patients treated with CDK4/6i + ET with this alteration, TTNT was numerically shorter. GATA3 alterations (7%) were significantly associated with shorter TTD (4.4 vs 8.2 mo; HR=2.5, p=0.01) in the CDK4/6i + ET group. In the overall cohort, GATA3 alterations were associated with a trend toward shorter OS and TTD. Additional alterations, including ERBB2 (19%), PTEN (11%), ATM (12%), PIK3CA (55%), ERBB3 (8%), TBX3 (14%), MDM4 (9%), FGFR1 (14%), FGFR2 (6%), FGFR3 (3%), AKT1 (3%), and MYC (5%), did not demonstrate a statistically significant impact on survival outcomes in this analysis. Conclusions: These findings represent a large-scale systematic analysis linking specific genomic alterations to treatment resistance patterns in metastatic invasive lobular carcinoma, identifying NF1, RB1, GATA3, and ARID1A as potential predictors of reduced benefit from standard therapies. While requiring validation in independent cohorts, these associations suggest that patients with these alterations may benefit from consideration of alternative therapeutic approaches targeting these pathways. Implementation of routine genomic screening in clinical practice is essential to guide personalized therapeutic decisions and improve patient outcomes. Future research should focus on developing targeted approaches to overcome the resistance observed with these genomic alterations.
Presentation numberPS2-06-23
Impact of somatic PIK3CA Mutations on clinical outcomes in HER2-Positive Breast Cancer
Nickolas Stabellini, Case Western Reserve University, Cleveland, OH
N. Stabellini1, S. D. de Oliveira2, T. Mizukami2, A. J. Montero2; 1Department of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, 2Department of Hematology/Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH.
Background: The phosphatidylinositol-3-kinase (PI3K) pathway, driven by the PIK3CA gene, is crucial for tumor initiation, growth, proliferation and therapy resistance. PIK3CA mutations (PIK3CAm) occur in ~35% of all breast cancers (BC), most commonly in estrogen receptor positive (ER+) and HER2-negative BC. In HER2-positive (HER2+) BC, these mutations are found in ~25-30% of cases. Although PIK3CAm are linked to lower rates of pathological complete response (pCR) in the neoadjuvant setting for HER2+ patients (pts), the prognostic significance of somatic PIK3CAm remains unclear. In the recently presented phase 3 Destiny Breast09 trial, the presence of a PIK3CAm was identified as a marker of early progression in HER2+ metastatic BC (mBC) on first line therapy with trastuzumab/pertuzumab monoclonal antibody therapy. We, therefore, aimed to evaluate the impact of PIK3CAm status on clinical outcomes in HER2+ BC pts receiving first- and second-line therapy. Methods: We analyzed clinico-genomic data from the BC cohort of the American Association for Cancer Research (AACR) Project Genomics Evidence Neoplasia Information Exchange (GENIE) Biopharma Collaborative (version 1.2), which includes pts diagnosed between 18-56 years of age who underwent tumor sequencing between 2013 and 2020. We included adult HER2+ BC pts who had received at least one line of therapy. The primary outcomes were overall survival (OS) and progression-free survival (PFS). We used Kaplan-Meier curves to estimate and compare OS and PFS. Univariable and multivariable Cox proportional hazard regressions were used to compare outcomes between patient groups, accounting for line of therapy. Subgroup analysis was performed according to TNM stage and PIK3CA wild type (wt) vs PIK3CAm. Results: We included 212 pts, of which 58 (27.3%) had a documented PIK3CAm. Of the 212, 62 were mBC, of which 15 (26%) had PIK3CAm. The median age at diagnosis was 43. Most patients were White (77%), Non-Hispanic (95%), and initial TNM stage II BC diagnosis (33%). Statistically significant differences between PIK3CAm and PIK3CAwt were observed in use of endocrine therapy (78% vs 62%, p=0.04) as well as the median time to distant metastasis (38 vs 22 months, p= 0.01). The observed median OS for the overall cohort starting from the date of first-line therapy, was 121.8 months. Median OS between pts with PIK3CAm and PIK3CAwt did not significantly differ (121.8 vs. 125.1 months, p=0.42). In the multivariable Cox regression, line of therapy was not significantly associated with an increased risk of death. The observed median PFS for the overall cohort from start of first-line therapy was 14.2 months. These results showed a trend by PIK3CAm status: 20.3 months for PIK3CAwt and 6.9 months for PIK3CAm (p=0.4). Multivariable Cox regression also revealed a higher associated risk of progression for patients with PIK3CAm on their second line of therapy (HR = 2.65, 95% CI 1.02-6.94). These findings were consistent in the subgroup analysis by TNM stage (Stages I-III and Stage IV). Conclusion(s): In this real-world cohort of HER2+ BC patients, PIK3CA mutations were not associated with worse median OS but demonstrated a trend toward shorter median PSF survival in early lines of therapy. Multivariable analysis revealed that patients with PIK3CAm receiving second-line therapy had a statistically significant 2.6-fold increased associated-risk of disease progression. These findings suggest the presence of a PIK3CAm may serve as a negative predictive biomarker for treatment duration in HER2+ disease. Given that approximately 30% of HER2+ BC harbor PIK3CAm, our results support prioritizing clinical investigation of PI3K inhibitors in combination with anti-HER2 therapy in the first-line metastatic setting for HER2+/PIK3CA-mutant BC to optimize treatment sequencing and potentially delay disease progression.
Presentation numberPS2-06-24
Global Prevalence of Prophylactic Mastectomy in Women: A Systematic Reviews and Meta-Analysis
Huynh Yen Phi, Taipei Medical University, Taipei City, Taiwan
H. Y. Phi1, T. V. Binh1, E. W. Loh2, K. W. Tam3; 1International PhD Program in Medicine, Taipei Medical University, Taipei City, TAIWAN, 2Division of General Surgery, Department of Surgery, Taipei Municipal Wan Fang Hospital, Taipei City, TAIWAN, 3Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei City, TAIWAN.
IMPORTANCE The trend of prophylactic mastectomy is increasing globally, despite being an aggressive surgery with unclear effectiveness in cancer prevention. OBJECTIVE To estimate the global prevalence of prophylactic mastectomy in women to assess the predictive factors influence women’s decisions. DATA SOURCES PubMed, Embase, Web of Science, Scopus, Cochrane Library STUDY SELECTION Studies that (1) provided real-world data on the number of women who underwent prophylactic mastectomy in relation to the total population; (2) used the national or hospital database. DATA EXTRACTION AND SYNTHESIS The Restricted Maximum Likelihood (REML) estimator random-effects model with Freeman-Turkey double arcsine transformation was used for data analysis. Sensitivity analysis, meta-regression, and subgroup analysis were performed. MAIN OUTCOMES AND MEASURES The prevalence of prophylactic mastectomy was reported using data from the national databases and hospital sources. RESULTS Data from 10.7 million women across 23 countries were included. The overall prevalence of prophylactic mastectomy was 5.04% (95% CI, 5.03-5.06). Prevalence was highest in America (5.19%) and lowest in Asia (1.58%), highlighting differences in healthcare systems, cultural perceptions, and the significant influence of the “Angelina Jolie effect” in Western countries. Substantial differences in the prevalence of prophylactic mastectomy were noted when stratified by 28 predictive factors. Across all regions, genetic testing, healthcare accessibility, insurance support, and psychosocial factors were associated with increased prevalence of prophylactic mastectomy. CONCLUSION AND RELEVANCE Our findings indicate that the global prevalence of prophylactic mastectomy continues to increase. The notably higher rates in America, compared to other countries, reflect disparities in healthcare systems and inconsistent clinical practices worldwide. Global efforts, including standardized guidelines, established genetic testing criteria, and uniform counseling, are key to providing women with reliable information and support for informed decision-making.
Presentation numberPS2-06-25
Impact of RB1 Mutations on Clinical Outcomes in Metastatic Breast Cancer: A Real-World Subtype-Dependent Survival Analysis
Pegah Farrokhi, University of Minnesota, College of Pharmacy, Minneapolis, MN
P. Farrokhi1, D. Stenehjem2; 1Department of Pharmaceutical Care and Health Systems, University of Minnesota, College of Pharmacy, Minneapolis, MN, 2Department of Pharmacy Practice and Pharmaceutical Sciences, University of Minnesota, College of Pharmacy, Duluth, MN.
Background: The tumor-suppressor gene RB1 acts as a master regulator of the cell cycle by controlling the critical G1-S transition and also modulates hormone-receptor signaling and tumor immunogenicity. However, whether the loss of RB1 uniformly influences patient prognosis across different metastatic breast cancer subtypes has been unclear. Therefore, this study aimed to contrast the clinical impact of RB1 mutations in metastatic hormone-receptor-positive/HER2-negative (HR+/HER2-) and triple-negative breast cancer (TNBC). Methods: This observational study analyzed data from the Flatiron Health-Foundation Medicine clinicogenomics database (~280 US cancer clinics). Adults ≥18 years with metastatic HR+/HER2- and TNBC breast cancer who underwent genomic profiling between 90 days before metastatic diagnosis and 90 days after first-line therapy initiation (January 2015-September 2022) were included. Patients (pts) were excluded if they had participated in a clinical trial or had a follow-up period of less than six months. Demographic and clinical characteristics were summarized using descriptive statistics, while overall survival (OS) and time-to-treatment-discontinuation (TTD), were evaluated using the Kaplan-Meier method and Cox regression analyses. Results: We analyzed cohorts of 286 TNBC pts and 1,713 HR+/HER2- pts, stratified by RB1 mutation status. In the TNBC cohort, RB1-mutated pts comprised 16% (n = 46) of the cohort. The median age at metastatic diagnosis was younger for pts with RB1-mutant TNBC at 54 years compared to 61 years for those with wild-type tumors. Additionally, all RB1-mutant tumors exhibited TP53 co-mutation. Median OS in the TNBC cohort was similar for RB1-mutant versus wild-type disease (18.9 vs 12.9 mo; log-rank = 0.7, adjusted HR for wild-type vs mutant(aHR) = 1.1, 95% CI: 0.7-1.7). In patients treated in the first -line (1L) setting with chemotherapy, the median OS (14.2 vs 11.5 mo; log-rank= 0.7, aHR 1.2, 95% CI 0.6-2.3) and TTD (4.6 vs 3.6 mo; log-rank = 0.4, a HR 1.2, 95% CI 0.7-2.1) were also similar. Patients with RB1 mutations who received anti-PD-1/PD-L1 immunotherapy in the first-line setting had comparable median OS (12.3 vs 12.6 months; log-rank p = 0.6, aHR ≈ 1.1, 95% CI: 0.4-2.2) and a trend toward worse TTD (2.8 vs 3.6 months; log-rank = 0.1, aHR ≈ 1.3, 95% CI: 0.6-2.8). In HR+/HER2- patients, RB1 mutations occurred in 5% of the pts (n=83). RB1-mutated patients were slightly older with a median age of 62 years versus 61 years for wild-type tumors. RB1-mutant tumors exhibited greater enrichment for endocrine-resistance drivers including PTEN, TP53, and ESR1. Median OS was significantly shorter in RB1-mutant pts (21.4 vs 40.1 mo; log-rank p < 0.01, aHR for wild-type vs mutant = 0.8, 95% CI: 0.5-1.2). RB1 loss also shortened TTD (4.3 vs 8.3 mo; log-rank <0.01; aHR = 0.8, 95% CI: 0.6-1.1). In the CDK4/6 inhibitor treatment cohort, RB1 deficiency was associated with significantly shorter median TTD (10.9 vs 13.7 mo; log-rank = 0.03, aHR = 0.9, 95% CI: 0.61-1.26), suggesting potential therapeutic resistance. Conclusions: This real-world analysis suggests that the clinical impact of RB1 mutations may be subtype dependent. In TNBC, where RB1 mutations were more common (16% vs 5%), RB1 loss had no meaningful impact on chemotherapy or immunotherapy effectiveness or overall survival. In HR+/HER2- breast cancer, RB1 mutations represent a marker of poor prognosis, with shorter OS overall and TTD with CDK4/6 inhibitors, where RB1 loss may contribute to therapeutic resistance. The co-occurrence with other resistance drivers including PTEN and ESR1 further compounds treatment resistance. These findings potentially have implications for precision medicine approaches, with RB1 mutation status potentially guiding therapeutic strategies in HR+/HER2- disease.
Presentation numberPS2-06-26
Sociodemographic and Clinical Factors Associated with Late-Stage Breast Cancer in Young Brazilian Women: A 23-Year Nationwide Study of 52,734 Patients
Ana Carolina Pires, Oncoclinicas & CO, Rio de Janeiro, Brazil
A. Pires1, A. Bergmann1, C. Resende2, L. Thuler3, M. Bello4, G. Bretas1, A. Gonçalves1; 1Instituto OC, Oncoclinicas & CO, Rio de Janeiro, BRAZIL, 2Instituto OC, Oncoclinicas & CO, Brasília, BRAZIL, 3Epidemiologia Clínica, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL, 4Hospital do Câncer III – INCA, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL.
Introduction: Cancer in adolescents and young adults (AYAs), aged 15 to 39 years, constitutes a major global health challenge, with over 1.2 million new cases diagnosed each year. As a consequence, AYAs are frequently diagnosed at more advanced stages and experience worse clinical outcomes compared to other age groups. In young women, breast cancer (BC) represents a major contributor to cancer-related illness and death.Objective: To determine the frequency and identify sociodemographic and clinical factors associated with late-stage BC diagnosis among young women treated within Brazil’s public healthcare system.Methods: A secondary database study was conducted, including women aged 15 to 39 years who were diagnosed and treated for BC (ICD-10 C50) within Brazil’s public healthcare system, as recorded in the national Hospital Cancer Registry between 2000 and 2023. Non-analytic or lacking clinical stage information cases were excluded. The primary end point was late-stage diagnosis, defined as clinical stage III or IV at presentation. Sociodemographic and clinical variables were collected for analysis. Descriptive analysis was performed, and multivariable logistic regression using the Stepwise Forward method was applied to identify factors associated with the outcome. Variables with a p-value < 0.05 were retained in the final model.Results: Among the 52,734 eligible patients, 0.1% were aged 15-19 years (n = 39), 12.2% were aged 20-29 years (n = 6,431), and 87.7% were aged 30-39 years (n = 46,264), with a median age at diagnosis of 35 years (SD ±3.93). The majority self-identified as Black or mixed-race (55.3%), 54.3% reported being in a partnered relationship, and 59.5% had ≥9 years of formal education. Half of the cases were reported from the Southeast region (50.7%). At the time of diagnosis, 8.4% of patients reported alcohol consumption, and 6.1% were current smokers. The predominant histological subtype was invasive carcinoma of no special type (80.6%). Most patients (78.7%) were referred for treatment via the public healthcare system. In multivariable logistic regression analysis, independent factors associated with late-stage diagnosis included younger age (OR 0.97; 95% CI, 0.96-0.98; p < 0.001), Black or mixed-race ethnicity (OR 1.34; 95% CI, 1.24-1.42; p < 0.001), lower educational attainment (OR 1.27; 95% CI, 1.20-1.34; p < 0.001), unpartnered status (OR 1.12; 95% CI, 1.06-1.18; p < 0.001), invasive carcinoma of no special type (OR 1.62; 95% CI, 1.48-1.76; p < 0.001), and referral through the public health system (OR 1.34; 95% CI, 1.25-1.43; p < 0.001). Alcohol consumption, tobacco use, and geographic macroregion were not significantly associated with stage at diagnosis.Conclusion: This nationwide cohort study of over 50,000 young Brazilian women with breast cancer—one of the largest to date—shows that late-stage diagnosis remains common, affecting nearly 40% of patients. Advanced-stage disease was independently associated with Black or mixed-race ethnicity, low educational level, unpartnered status, invasive carcinoma of no special type, and referral through the public health system. These findings underscore the impact of social vulnerability on cancer staging and persistent inequities in timely access to care. Targeted public health strategies addressing social determinants, early symptom recognition, and equitable access to services are critical to reduce disparities and improve outcomes in this high-risk group.
Presentation numberPS2-06-27
Early Mortality in Young Women with Breast Cancer: Insights from 66,000 Cases in the Brazilian Public Health System (2000-2023)
Ana Carolina Pires, Oncoclinicas & CO, Rio de Janeiro, Brazil
A. Pires1, A. Gonçalves1, C. Resende2, L. Thuler3, M. Bello4, A. Bergmann1, G. Bretas1; 1Instituto OC, Oncoclinicas & CO, Rio de Janeiro, BRAZIL, 2Instituto OC, Oncoclinicas & CO, Brasília, BRAZIL, 3Epidemiologia Clínica, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL, 4Hospital do Câncer III – INCA, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL.
Introduction: Breast cancer (BC) in adolescents and young adults (AYAs), defined as individuals aged 15-39 years, poses distinct clinical and public health challenges as underrepresentation in guidelines or screening exclusion. It is the most common solid tumor in young women, contributing substantially to morbidity and mortality. AYAs are more likely to experience diagnostic delays, aggressive subtypes, and worse outcomes due to unique biological and psychosocial vulnerabilities. In Brazil, where ethnic and socioeconomic disparities are pronounced, understanding the drivers of BC mortality in young women treated in the public healthcare system is essential. This study investigates factors associated with mortality in a nationwide cohort of Brazilian women aged 15-39 years diagnosed with BC over a 23-year period. Methods: A secondary database study was conducted, including women aged 15 to 39 years who were diagnosed and treated for BC (ICD-10 C50) within Brazil’s public healthcare system, as recorded in the national Hospital Cancer Registry between 2000 and 2023. Non-analytic cases were excluded. The primary end point was death occurring after diagnosis and during the initial course of proposed treatment. Sociodemographic, clinical, and tumor-related variables were collected. Descriptive analysis was performed, and multivariable logistic regression using the Stepwise Forward method was applied to identify factors associated with the outcome. Variables with a p-value < 0.05 were retained in the final model. Results: Of 66,726 eligible women, most were aged 30-39 (87.3%), self-identified as Black or mixed-race (55.4%), had ≥9 years of education (59.0%), and lived with a partner (54.3%). The majority were from the Southeast (44.8%) and Northeast (26.9%). Invasive carcinoma of no special type was the most common subtype (86.8%). At diagnosis, 40.3% were stage II and 36.7% stage III. Treatment initiation exceeded 60 days in 44.6% of cases. Neoadjuvant chemotherapy was administered to 39.5%, surgery to 49.3%, radiotherapy to 8.3%, and endocrine therapy to 2.0%. A total of 2,337 women (3.5%) died before or during the initial course of treatment. In adjusted analyses, younger age was associated with higher mortality, with a 2% reduction in odds of death per year of age (OR 0.98; 95% CI: 0.96-0.99; p = 0.003). Black or mixed-race women had a 40% higher risk of death (OR 1.40; 95% CI: 1.23-1.61), and those with lower education, a 36% increase (OR 1.36; 95% CI: 1.21-1.52). Living in the North, Northeast, or Central-West was associated with 30% higher mortality (OR 1.30; 95% CI: 1.14-1.47), and absence of a partner increased the risk by 23% (OR 1.23; 95% CI: 1.10-1.38). Advanced stage (III/IV) was the strongest predictor of death (OR 7.54; 95% CI: 6.37-8.93; p < 0.001). Conclusion: This nationwide study demonstrates a high burden of early mortality among young Brazilian women with breast cancer, particularly those from marginalized racial, educational, and geographic backgrounds. Non-white ethnicity, low educational attainment, absence of a partner, and residence in underserved regions were independently associated with increased risk of death. Late-stage diagnosis was the most significant predictor. These findings underscore the urgent need for targeted public health interventions to promote earlier diagnosis, reduce treatment delays, and improve equity in breast cancer care across Brazil.
Presentation numberPS2-06-28
Clinicogenomic and Immune Profiles of Male Breast Cancer by Race: Insights from a Large Real-World Cohort
Yara Abdou, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC
Y. Abdou1, E. Jaeger2, S. Fragkogianni2, E. Williams2, M. Ciampricotti2, K. Reeder-Hayes1, P. Spanheimer3; 1UNC Breast Center, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 2Medical Affairs, Tempus AI, Chicago, IL, 3Division of Surgical Oncology, University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC.
BackgroundMale breast cancer (MBC) is a rare disease with limited data on potential racial differences in clinical and molecular characteristics, despite the fact that such differences are well-recognized in females. Understanding differences in the immune microenvironment and clinicogenomic features of MBC by race could inform future treatment strategies and improve health equity. This study leveraged the largest real-world (rw) cohort of MBC patients (pts) to date to characterize these features in Black/African American and White men. MethodsMale pts with primary breast cancer who underwent xF or xT testing in the Tempus multi-modal database (Tempus AI, Inc., Chicago, IL) were identified using Tempus Lens. Statistical comparisons used Kruskal-Wallis and Chi-squared/Fisher’ exact tests where appropriate. Pts with xR testing (RNA) were classified into PAM50 subtypes and immune infiltration was quantified using quantiseq. Hormone receptor status was extracted from clinical documentation within 60 days of sample collection. Tumor mutational burden (TMB; mutations/megabase) and microsatellite instability were assessed by DNA sequencing. Real World overall survival (rwOS) was defined from the time of sample collection to death, last follow-up, or a study cutoff of 7 years, and was estimated using Kaplan-Meier methodology; comparisons used the log-rank test. ResultsThe study cohort included 291 MBC pts: 128 White (44%) and 36 Black (12%). Among pts with available ethnicity, 93% were not Hispanic or Latino. Clinical subtypes were HR+, HER2- (39%), HER2+ (7.6%), triple-negative (7%) and 50% unknown or not otherwise specified. Median age at diagnosis was 63 years, and 87% of staged pts presented with stage IV disease. PAM50 subtyping identified Luminal A (62% of White and 68% of Black) and Luminal B (23% of White and 14% of Black) as the most common subtypes, with no significant racial differences in molecular subtypes (p=0.6). TMB was low (<10 mt/mB) and all tumors were microsatellite stable. AR (41%), PIK3CA (27%), TP53 (26%), ESR1(12%), TERT (11%) and GATA3 (11%) were amongst the most frequent somatic mutations found in the cohort overall, with no racial differences identified. BRCA2 was the most frequent germline mutation identified overall. PD-L1 negativity (<1% CPS) was more frequent in Black vs. White pts (71% vs. 50%, p=0.5). Notably, M2 macrophage infiltration was significantly lower in Black vs. White pts (median 4.58 vs 5.62, p=0.036); CD8+ T cell infiltration was also not statistically significantly lower in Black vs White pts (median 0.03 vs. 0.15, p=0.3). Median rwOS was numerically lower in Black vs White pts, though the difference was not statistically significant (19.1 vs 23.1 months; p=0.5). ConclusionsThis real world analysis of MBC pts revealed largely similar clinicogenomic profiles between Black and White pts, with the notable exceptions of significantly lower M2 macrophage infiltration and numerically lower CD8+ T cell infiltration and higher rates of PD-L1 negativity in Black pts. These immune microenvironment differences may have implications for immunotherapy response, underscoring the need for further research into the biological underpinnings and potential clinical impact of immune differences in racially diverse MBC populations.
Presentation numberPS2-06-30
Real-world uptake of gBRCA testing as a companion diagnostic for olaparib in patients with HER2-negative recurrent high-risk early breast cancer in Japan: a cross-sectional multicenter study (BRCAwareness)
Tomoe Taji, Kansai Medical University, Osaka, Japan
T. Taji1, Y. Uemura2, Y. Kimura3, N. Maeda4, A. Ito5, H. Seki6, D. Takabatake7, M. Harao8, S. Nakamoto9, R. Matsunuma10, C. Koganezawa11, T. Iwamoto12, H. Mukai13, Y. Kikawa1; 1Breast Sugery, Kansai Medical University, Osaka, JAPAN, 2Center for Clinical Science, Japan Institute for Health Security, Tokyo, JAPAN, 3Breast Surgical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 4Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, JAPAN, 5Breast Sugery, Akita Red Cross Hospital, Akita, JAPAN, 6Breast Sugery, Kyorin University Hospital, Tokyo, JAPAN, 7Breast Sugery, National Hospital Organization Shikoku Cancer Center, Ehime, JAPAN, 8Breast Oncology, Jichi Medical University, Tochigi, JAPAN, 9Breast and Endocrine Surgery, Okayama University Hospital, Okayama, JAPAN, 10Breast Sugery, Shizuoka General Hospital, Shizuoka, JAPAN, 11Breast Oncology, Hokkaido Cancer Center, Hokkaido, JAPAN, 12Breast and Thyroid Sugery, Kawasaki Medical School, Okayama, JAPAN, 13Medical Oncology, National Cancer Center Hospital East, Chiba, JAPAN.
Background In patients (pts) with HER2-negative, high-risk early breast cancer (BC), adjuvant (adj) olaparib (OLA) significantly improves invasive disease-free survival and overall survival in those carrying germline BRCA1 or BRCA2 (gBRCA) pathogenic variants (PVs) 1). To optimize clinical outcomes, timely gBRCA testing as a companion diagnostic is essential. However, the real-world uptake of gBRCA testing in this population remains unclear. Methods We enrolled newly diagnosed pts with invasive HER2-negative BC who underwent curative surgery from January 1, 2023 to December 31, 2023 in Japan. Patient selection was based on the inclusion criteria from the OlympiA trial1). Specifically, ER-positive pts required either ≥ 4 positive lymph nodes after surgery or non-pathological complete response (pCR) with a CPS+EG score ≥ 3 following neoadjuvant chemotherapy (NAC). ER-negative pts required an invasive tumor >2 cm or ≥ 1 nodal metastasis after surgery, or non-pCR after NAC. Furthermore, we collected both patient-level and institutional data. The primary outcome was the proportion of pts who underwent gBRCA testing. Secondary outcomes included: (1) factors linked to no testing; (2) proportion of PVs among those tested; (3) proportion of pts with gBRCA PVs who started OLA; and (4) other treatments used in pts with gBRCA PVs not receiving OLA. Based on a previous U.S. report in which 87% of young women with BC underwent gBRCA testing2), we hypothesized that at least 88% of eligible pts should be tested. Assuming an expected testing rate of 88%, a sample size of 688 pts was required to maintain a 95% confidence interval (CI) width of 0.05. The target sample size was set at 700 to account for potential exclusions. Results Of 824 pts recruited from 46 facilities during the primary enrollment, 700 were randomly selected for secondary registration. After excluding 9 ineligible pts, a total of 691 were analyzed. The median age was 62 [20-96] years, 99% were female, 52 had bilateral BC, 157 had a family history of BC, and 18 had dementia. ER status was positive in 51%. NAC was administered to 362 pts (52%), including 148 (21%) with ER-positive BC, while 199 (29%) ER-positive BC pts had upfront surgery. Regarding facility characteristics, the median number of breast cancer specialists per facility was 3 [1-17], and genetic counselors were available in 27 facilities (59%). gBRCA testing was performed in 437 of 691 pts (63.2%; 95% CI, 59.5-66.9). Of 254 untested patients, 168 (66%) were not informed, mainly due to physician oversight (57%) or perceived ineligibility related to age, comorbidities, or treatment refusal (40%). Among 69 pts who were informed but declined testing, the reasons for no testing were psychological distress (46%), cost of testing (35%), and concerns about genetic implications for family members (12%). Among 42 pts (9.6%) with gBRCA PVs (10 pts were ER-positive), 32 received OLA. Of the remaining 10, all but 3 had perioperative chemotherapy, and none received abemaciclib. Results of our multivariate analysis on missed testing will be presented at the conference. Conclusions Despite its clinical importance as a companion diagnostic for adj OLA, timely gBRCA testing was underutilized in the real world. Two-thirds of untested pts were not informed of the testing option, highlighting a potential gap between clinical trial evidence and practice. To ensure equitable access to adj OLA, greater efforts are needed to improve physician awareness and streamline institutional workflows supporting genetic testing. References 1) Geyer CE et al. Ann Oncol. 2022;33(12):1250-1268. 2) Rosenberg SM et al. JAMA Oncol. 2016;2(6):730-736.
Presentation numberPS2-07-01
Cemiplimab in High-risk or Locally Advanced Luminal and Triple Negative Breast Cancer (CemiHALT)
Lubna N Chaudhary, Medical College of Wisconsin, Milwaukee, WI
L. N. Chaudhary1, H. Abid1, J. M. Jorns2, A. Szabo3, Y. Sun4, S. Kamaraju1, Y. Cheng1, A. Kong5, T. W. Yen5, C. Patten5, C. S. Cortina5, E. Weil1, I. Chervoneva4, C. R. Chitambar1, H. Rui4; 1Internal Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, 2Pathology, Medical College of Wisconsin, Milwaukee, WI, 3Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, 4Pharmacology, Physiology & Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 5Surgery, Division of Surgical Oncology, Medical College of Wisconsin, Milwaukee, WI.
Introduction: PD-1 inhibitors (PD-1i) are associated with significantly higher pathological complete responses (pCR) in patients (pts) with early-stage triple-negative breast cancer (TNBC) as well as luminal/estrogen receptor-positive (ER+) breast cancer (BC) with higher pCR rates reported in PD-L1-positive pts. However, some PD-L1-negative pts respond as well. Most clinical trials of PD-1i have thus far focused on PD-L1 expression for pt eligibility. The alternative PD-1 ligand, PD-L2, with reported ~3-fold greater affinity for PD-1, remains understudied. We recently reported that high levels of PD-L2 in pts with treatment-naïve ER+ BC were an independent predictor of shorter progression-free survival. We therefore sought to assess the correlation of PD-L1 and PD-L2 with pCR in TNBC and ER+/HER2-negative BC treated with PD-1i and neoadjuvant chemotherapy (NACT). Methods: CemiHALT (NCT04243616) is a single-arm, open-label Phase II study of PD-1i cemiplimab (Regeneron Pharmaceuticals) and NACT in pts with high-risk TNBC or locally advanced ER+/HER2-negative BC with positive PD-L1 and/or PD-L2 tumor expression. Baseline PD-L1 or PD-L2 expression in ≥1% of cancer cells or stromal immune cells in diagnostic core biopsies was considered positive. Pts were treated with standard taxane and anthracycline-based NACT (weekly paclitaxel x 12 with carboplatin added for TNBC only, followed by adriamycin and cyclophosphamide x 4 cycles). The study was designed to administer two doses of cemiplimab (350mg standard dose) with 1st dose as monotherapy to prime the immune microenvironment and 2nd dose concurrently with NACT start. The trial was later amended to incorporate cemiplimab concurrently throughout NACT for TNBC pts after FDA approval of neoadjuvant pembrolizumab per KEYNOTE-522 in July 2021. Radiographic responses were assessed by RECIST criteria. Wilcoxon paired analyses and Spearman correlation were used to correlate PD-L1 and PD-L2 protein expression with pCR rates and radiographic responses respectively. Fisher’s test and McNemar’s tests were used for comparison of variables and PD-L1/PD-L2 expression between ER+ and TNBC subtypes. All analyses were planned with 80% power and a significance level of 0.05. Results: Thirty-six pts were enrolled between June 2020 and January 2025, with 23 ER+/HER2-negative and 13 TNBC pts (5 with 2 doses, 8 with 8 doses). Median age at diagnosis was 49yrs (range 26-70) with 81% White, 14% African American, and 5.6% Hispanic pts. TNBC pts had high baseline PD-L1 and PD-L2 protein expression (92% and 100% respectively), whereas ER+ pts had a higher PD-L2 expression at 83% vs 61% for PD-L1. Of the evaluable 31 pts, the pCR rate was 23% in the overall cohort with a significantly higher pCR seen in TNBC compared to ER+/HER2-negative pts (50% vs 5.3%, p=0.007) and no differences seen by doses of cemiplimab in TNBC pts. Higher proportion of TNBC pts had complete and partial responses radiographically (25% and 58.3% respectively) while ER+/HER2- pts had more stable disease (41.6%). Baseline PD-L1 and/or PD-L2 expression was not significantly associated with pCR rates or radiographic responses. Treatment-related adverse events were not higher than expected. Conclusion: Addition of cemiplimab to NACT resulted in 23% pCR rate in the overall cohort with a significantly higher pCR (50%) seen in TNBC pts. Baseline PD-L1 and/or PD-L2 expression was not associated with pCR rates or radiographic responses. This study incorporated only 2 doses of cemiplimab for ER+ pts which could explain the low pCR rates in this cohort. Given the discordance in PD-L1 and PD-L2 expression with a higher prevalence of PD-L2 in ER+ pts, further studies exploring PD-L1/PD-L2 as a combined marker for PD-1i use are warranted. To our knowledge, this is the first clinical trial with PD-L2 status as an eligibility criterion for PD-1i treatment.
Presentation numberPS2-07-02
Tissue-free epigenomic circulating tumor DNA (ctDNA) analysis pre- and post-surgery in early breast cancer: clinical features and prognostic utility
Arielle J Medford, Mass General Cancer Center/Harvard Medical School, Boston, MA
A. J. Medford1, J. Knape1, D. Dustin2, A. Dedeoglu1, O. Rieur1, H. Zhang2, M. Shivahamy3, I. Kuter1, R. O. Abelman1, J. M. Peppercorn1, S. A. Wander1, N. Vidula1, S. J. Isakoff1, B. Moy1, L. W. Ellisen1, D. Juric1, L. M. Spring1, A. Bardia4; 1Oncology, Mass General Cancer Center/Harvard Medical School, Boston, MA, 2Oncology, Guardant Health, Palo Alto, CA, 3Oncology, University of Massachusetts Medical School, Worcester, MA, 4Oncology, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA.
Introduction The development of new highly sensitive assays allows for measurement of ctDNA in early breast cancer (eBC). These new tests have much lower limits of detection, which increases the potential for recurrence monitoring in the curative-intent setting. Promising applications include tracking response to neoadjuvant therapy (NAT), determining prognosis after NAT, and predicting relapse. The tissue-agnostic approach to ctDNA testing has a faster initial turnaround time (7-10 days) and is not limited by sequencing errors and tumor quantity/quality, which makes the utility of this assay type in the neoadjuvant context compelling. This study evaluates the use of a tissue-free epigenomic ctDNA assay from patients (pts) with high-risk eBC who received NAT, to determine detectability of ctDNA across subtypes and stages, and assess its use as a prognostic tool. Methods Pts with eBC consented to have serial plasma collected throughout their courses of treatment. Samples were collected at clinically relevant time points, which included prior to NAT initiation (baseline/pre-NAT), during NAT (on-NAT), after completion of NAT/prior to surgery (post-NAT/pre-op), following surgery (post-op), and throughout the surveillance period. ctDNA analysis was performed using the Guardant Reveal assay, a tissue-free, epigenomic assay, which provides a binary ctDNA detection result (present vs absent) and quantifies ctDNA via tumor fraction. Fisher’s exact test was performed to evaluate the relationship between ctDNA detectability and categorical clinical variables, and the relationship of ctDNA detection status to survival outcomes was evaluated using univariate Cox proportional hazards models and log-rank tests. Results A total of 446 samples from 85 pts with eBC treated at the Massachusetts General Hospital were evaluable, with a 97% (446/461) quality control pass rate. Baseline ctDNA was detectable in 76% (35/46) of patients, with detection rates consistent across breast cancer subtypes and clinical stages: 70% (7/10) in ER+/HER2-, 80% (16/20) in HER2+, and 70% (12/16) in TNBC; 50% (2/4) in stage I, 79% (23/29) in stage II, and 77% (10/13) in stage III disease. There were a total of 12 recurrences (14.1%) in the evaluable cohort. At the pre-NAT time point, ctDNA was detectable in all pts who later developed distant recurrence. At a median follow-up of 51.3 months (range 2.2-99.6 months), stratification by median tumor fraction (TF) revealed a significant association between higher TF and worse D-RFI (p = 0.0052). ctDNA detection at pre-NAT (p > 0.99) or post-NAT/pre-op (p = 0.50) was not significantly associated with the likelihood of residual disease after NAT, however ctDNA detection at the post-NAT/pre-op time point was strongly associated with worse D-RFI (p < 0.0001). In the post-op setting, ctDNA detection was associated with a 15-fold increased risk of distant recurrence (p < 0.0001). Conclusion ctDNA was detected via a tissue agnostic, methylation-based ctDNA analysis across eBC subtypes. All pts with recurrence had baseline detectable ctDNA, and detectable ctDNA post-operatively was associated with a very high risk of recurrence, with higher TF associated with poorer D-RFI. Furthermore, while post-NAT/pre-op ctDNA did not predict residual disease, detectability was strongly associated with worse D-RFI. This analysis is among the first data across all breast cancer subtypes utilizing a tissue-free assay for prognostic validation in both the NAT and post-operative setting. Additional analyses are ongoing to determine sensitivity, specificity, and lead time to recurrence, as well as the association of other clinical interventions (e.g. radiation, adjuvant therapy) on ctDNA detectability and dynamics. Updated results from these analyses will be presented at the meeting.
Presentation numberPS2-07-03
Improved 3-year IDFS with anthracycline-based therapy for patients with 70-gene signature High 2, Luminal B, HR+HER2- early-stage breast cancer
Joyce O’Shaughnessy, Baylor University Medical Center, Texas Oncology, Dallas, TX and Sarah Cannon Research Institute, Dallas, TX
J. O’Shaughnessy1, A. Brufsky2, C. L. Graham3, C. R. Osborne4, R. L. Rahman5, A. Elkhanany6, E. A. Brown7, L. P. Gold8, N. M. Johnson9, D. Giffoni10, J. Alberty-Oller11, R. L. Mahtani12, H. Ramaswamy13, N. Stivers13, A. Menicucci13, W. Audeh13; 1Medical Oncology, Baylor University Medical Center, Texas Oncology, Dallas, TX and Sarah Cannon Research Institute, Dallas, TX, 2Medical Center, University of Pittsburgh, Pittsburgh, PA, 3Breast Surgical Oncology, Piedmont, Cartersville, GA, 4Medical Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX, 5Sugery, Texas Tech University Health Sciences Center, Lubbock,, TX, 6Medical Oncology, Lester & Sue Smith Breast Center Dan L Duncan Comprehensive Cancer Center Baylor College of Medicine, Houston, TX, 7Medical Oncology, Comprehensive Breast Care, Troy, MI, 8Surgical Oncology, Comprehensive Breast Care, Dallas, MI, 9Surgical Oncology, Legacy Health, Portland, OR, 10Medical Oncology, Verspeeten Family Cancer Centre / London Health Sciences Centre, London, ON, CANADA, 11Medical Oncology, Kings County Hospital Center, Brooklyn, NY, 12Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Plantation, FL, 13Medical Affairs, Agendia, Inc., Irvine, CA.
Background: The ABC trials evaluated the efficacy of taxane with cyclophosphamide (TC) vs anthracycline and taxane-based chemotherapy (AC-T) in patients (pts) with clinically high risk, HER2-negative (HER2-) breast cancer (BC), and observed no significant difference between these treatment regimens among pts with hormone receptor-positive (HR+) BC. The MammaPrint® (MP), 70-gene signature, identifies pts with early BC (EBC) who derive (neo)adjuvant chemotherapy (CT) benefit. To examine the utility of MP in identifying pts likely to benefit from AC-T, we provide an updated analysis of data presented at ASCO 2024 and evaluated 3-year (yr) outcomes among propensity-score matched (PSM) pts with MP High Risk, HR+HER2- BC who received adjuvant TC or AC-T on the prospective observational FLEX Study. Methods: The FLEX Study (NCT03053193) enrolled EBC pts who received standard of care MP with BluePrint® (BP) intrinsic subtyping. 1261 pts had MP High Risk, BP Luminal B, HR+HER2- EBC and received either adjuvant TC or AC-T (non-randomized) with 3.2 yrs median follow-up. High Risk was further stratified into High Risk 1 (H1) and High Risk 2 (H2). PSM was performed to balance differences in age, tumor size and nodal status between the TC and AC-T-treated pts for the H1 and H2 groups, separately. The 3-yr invasive disease-free survival (IDFS), as defined by STEEP 2.0, was compared within H1 and H2 groups using Kaplan-Meier analysis and log-rank tests, grouped by TC vs AC-T. Cox proportional hazards models were used to evaluate the effect of CT regimen and clinical features on survival within each group. Results: Among 1261 pts, 1107 had H1 and 154 had H2 HR+ HER2- BC. Pts who received TC (H1, n=818; H2, n=103) were PSM with pts who received AC-T (H1, n=289; H2, n=51), yielding a cohort of 578 pts with H1 and 102 with H2 BCs with no significant differences in clinical/pathologic features between the two CT groups within each of the H1 and H2 cohorts. For pts with H1 BC, no significant difference in 3-yr IDFS was observed between AC-T (95.6%) and TC (94.6%) treatment (p = 0.98; Table). In contrast, H2 pts treated with TC had a significantly worse IDFS of 89.3% compared with 100% for AC-T-treated pts, with an absolute benefit of 10.7% (p = 0.048). Multivariate Cox regression analysis within the H1 group showed no association with improved IDFS with AC-T, while the use of AC-T in H2 pts showed a trend towards improved IDFS compared to TC, but did not reach significance likely due to sample size. Conclusions: In this PSM analysis of non-randomized, real-world FLEX Study data with 3.2 yrs median follow-up, pts with H2 HR+HER2- EBC had significantly improved IDFS with AC-T compared to TC. In contrast, pts with H1 disease did not benefit more from AC-T vs TC. These findings further support the utility of MammaPrint in informing prognosis and CT selection in pts with HR+HER2- EBC.
| – | – | – | – | – | – | Cox Proportional Hazards Model for IDFS (ACT vs TC)* | |||||
| – | – | Kaplan Meier and Log-Rank tests for 3-yr IDFS | – | – | – | Univariate | Multivariate | ||||
| MP Risk | CT Regimen | 3-yr IDFS | IDFS Difference | 95% CI | P-value | HR | 95% CI | P-value | HR | 95% CI | P-value |
| H1 |
TC (n = 289) |
94.6% | – | 92.0-97.3 | – | reference | – | – | reference | – | – |
| H1 |
AC-T (n = 289) |
95.6% | 1.0% | 93.1-98.1 | p = 0.980 | 1.01 | 0.57-1.80 | p = 0.980 | 0.99 | 0.52-1.67 | p = 0.812 |
| H2 |
TC (n = 51) |
89.3% | – | 80.8-98.7 | – | reference | – | – | reference | – | – |
| H2 |
AC-T (n = 51) |
100% | 10.7% | 100-100 | p = 0.048 | 0.16 | 0.02-1.29 | p = 0.085 | 0.18 | 0.02-1.57 | p = 0.120 |
| *Only CT regimen results included in table. For H1, T3/T4 was associated with worse IDFS compared to T1/T2 (Univariate: HR = 4.43, 95% CI: 1.98-9.95, p<0.001; Multivariate: HR = 4.05, 95% CI: 1.74–9.43, p=0.001) accounting for age, T and N status, grade, and CT regimen |
Presentation numberPS2-07-04
Time-to-progression patterns during first and second-line endocrine therapy across clinical features and liquid biopsy-defined mutational profiles in HR+/HER2- metastatic breast cancer
Lorenzo Gerratana, IRCCS CRO Aviano National Cancer Institute / University of Udine, Aviano, Italy
L. Gerratana1, A. A. Davis2, C. Reduzzi3, A. J. Medford4, L. Foffano1, E. Podany2, M. Velimirovic5, K. Clifton2, B. Pastò3, L. Pontolillo3, R. Occhiogrosso Abelman4, C. Gianni6, S. Tapiavala2, E. Molteni7, M. Lipsyc-Sharf8, E. Nicolò3, E. Andreopoulou3, W. J. Gradishar9, F. Puglisi1, C. X. Ma2, A. Bardia8, M. Cristofanilli3; 1Department of Medical Oncology, IRCCS CRO Aviano National Cancer Institute / University of Udine, Aviano, ITALY, 2Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, 3Department of Medicine, Weill Cornell Medicine, New York, NY, 4Cancer Center, Massachusetts General Hospital, Boston, MA, 5Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 6Struttura Complessa di Oncologia Medica a Direzione Universitaria, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, Meldola, ITALY, 7Department of Medicine, University of Udine, Udine, ITALY, 8Division of Hematology/Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 9Department of Medicine, Robert H. Lurie Comprehensive Cancer Center of Northwestern University Feinberg School of Medicine, Chicago, IL.
Background: Monitoring strategies for HR+/HER2- metastatic breast cancer (MBC) remain largely based on fixed, trial-derived, schedules. Emerging evidence from the SERENA-6 and INAVO120 studies underscore the limitations of this approach and the need for a risk-adjusted, biomarker-guided strategy incorporating adaptive treatment escalation to optimize treatment sequencing. This study investigated the impact of clinical and liquid biopsy-based features on time-to-progression during first and second-line endocrine therapy (ET) in HR+/HER2- MBC. Methods: We retrospectively analyzed a cohort of 909 patients with HR+/HER2- MBC who underwent circulating tumor DNA (ctDNA) profiling through the Guardant360 next-generation sequencing (NGS) panel within the multi-institutional “Precision Medicine Action for Cancer” (PMAC) academic consortium. Single nucleotide (SNVs) and copy number variations (CNVs), as well as clinical variables were evaluated. Oncogenic pathways were defined according to Sanchez-Vega et al. (Cell, 2018). Prognostic impact on progression-free survival (PFS), measured from the time of ctDNA collection, was investigated through log-rank test. Associations were evaluated using chi-square with false discovery rate correction (q value). Temporal patterns of disease progression were modeled through smoothed hazard function estimated up to 30 months. Results: A subset of 264 patients treated with 1st or 2nd-line ET was selected from the overall PMAC cohort of 909 HR+/HER2- MBC cases. The majority of patients were treated in 1st line (n=157, 59.5%), while 107 (40.5%) were treated in 2nd line. De novo MBC was present in 54 patients (20.5%). The majority received ET combined with CDK4/6 inhibitors (n=187, 70.8%), with visceral involvement in 115 cases (43.7%) and bone metastases in 208 (79.1%).Time-dependent hazard estimates revealed distinct temporal patterns of progression across baseline genomic alterations. SNVs in ESR1, PIK3CA, and TP53 were associated with early hazard peaks. ESR1 mutations displayed the steepest initial peak of PFS events at 3.6 months, followed by a plateau and a second peak at 15.3 months. PIK3CA had an early peak at 2.1 months and remained stable until 16 months. TP53 peaked at 4.8 months and steadily declined thereafter. Delayed hazard peaks were observed for SNVs in GATA3 (8.4 months), ARID1A (8.4 months), and RB1 (11.4 months), as well as for CNVs in FGFR1 (16.8 months) and PIK3CA (8.1 months), suggesting a delayed risk of progression.A significantly higher frequency of cell cycle pathway SNVs was observed in patients within the early ESR1 hazard peak (16% vs. 2.1%; p=0.003, q=0.013). RTK pathway CNVs were also significantly enriched in this subgroup (32% vs. 15%; p=0.013, q=0.044). No significant differences were found for the PI3K, TP53, RAS, or MYC pathways.PFS was significantly worse in patients with ESR1 and PIK3CA co-mutations, while double wild-type cases having the best outcomes (p < 0.0001). ESR1 SNVs were associated with early progression, irrespective of PIK3CA status. Conclusions: The temporal analysis of progression for patients with HR+/HER2- MBC revealed distinct patterns associated with specific genomic alterations. Early peaks in PIK3CA and TP53 mutations suggest intrinsic resistance, while delayed peaks in RB1, ARID1A, and FGFR1 indicate acquired mechanisms to ET +/- CDK4/6i therapy. The bimodal pattern observed in ESR1-mutant tumors, along with the enrichment of cell cycle and RTK pathway alterations in patients with early progression, further supports their role in endocrine resistance. If validated, these data may have important implications for both disease monitoring using ctDNA and rationale clinical trial design to overcome intrinsic and acquired resistance.
Presentation numberPS2-07-05
Methylation-based ctDNA Dynamics as a Biomarker for Treatment Response and Prognosis in Patients on the plasmaMATCH trial
Iseult M Browne, The Institute of Cancer Research, London, United Kingdom
I. M. Browne1, R. J. Cutts1, A. Amenssag1, I. Garcia-Murillas1, C. S. Swift1, C. Loader1, S. Hrebien1, A. Pearson1, J. Pascual1, A. M. Wardley2, M. Esteban-Garcia1, L. S. Kilburn3, I. R. MacPherson4, R. D. Baird5, R. Roylance6, J. M. Bliss3, A. E. Ring7, N. C. Turner1; 1The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UNITED KINGDOM, 2Medical Oncology, The Christie NHS Foundation Trust, Manchester, UNITED KINGDOM, 3ICR Clinical Trials and Statistics Unit (ICR-CTSU), The Institute of Cancer Research, London, UNITED KINGDOM, 4Medical Oncology, The Beatson Cancer Centre, Glasgow, UNITED KINGDOM, 5Medical Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridgeshire, UNITED KINGDOM, 6Medical Oncology, University College London Hospitals NHS Foundation Trust, London, UNITED KINGDOM, 7Breast Unit, Royal Marsden Hospital, London, UNITED KINGDOM.
Methylation-based ctDNA Dynamics as a Biomarker for Treatment Response and Prognosis in Patients on the plasmaMATCH trial Background: Early assessment of circulating tumour DNA (ctDNA) dynamic changes on treatment predict response to therapy. Previous studies have tracked mutations in plasma, which requires prior knowledge of the tumour mutations and is also at risk of false results such as tracking mutations arising from clonal haematopoesis. We have developed a robust tumour agnostic ctDNA assay that tracks the level of aberrant breast cancer specific DNA methylation in plasma. We assessed the potential of baseline and early on-treatment changes in breast cancer ctDNA methylation levels, to predict progression-free survival (PFS) and objective response rate (ORR) to therapy. Methods: Patients enrolled on the plasmaMATCH trial with matched cycle 1-day1 (C1D1) and cycle 2-day 1 (C2D1) plasma samples available for analysis were included. PlasmaMATCH was a phase 2 platform trial which assessed the accuracy of ctDNA mutation testing in advanced breast cancer, and the ability of ctDNA testing to select patients for mutation-directed therapy. A capture based enzymatic ctDNA based methylation assay targeting 1297 differentially methylated regions (DMR), as well as specific promoters such as BRCA1, was used to distinguish the presence of breast cancer and the level of ctDNA. Tumour ctDNA levels were inferred by estimating the Methylation Read Percent (MPCT) based on the top 60 DMRs ranked on high entropy or high subtype methylation, with the circulating methylation ratio (CMR) calculated as the ratio of C2D1 MPCT / C1D1 MPCT. Patients with low CMR indicated lower ctDNA levels on treatment. We evaluated the association of baseline MPCT, and median change in CMR, with PFS and ORR. Cohorts A-D were analyzed together (ER-positive breast cancer on targeted therapy), with cohort E analysed separately (TNBC on olaparib PARP and ceralasertib ATR inhibitor combination). Results: We analysed C1D1/C2D1 matched samples from 74 patients for ctDNA methylation (30 from cohorts A-D and 44 from cohort E). All samples were successfully sequenced to a mean CpG coverage of 364x (range 89-975x). Splitting patients by median baseline MPCT was prognostic for PFS for cohort E: mPFS low MPCT 9 months v high 4 months (HR 0.52, 95% CI 0.26-1.02; p=0.016), and ORR 36% (8/22) v 18% (4/22) (OR 2.56, 95% CI 1.36-4.96, p=0.006). There was no association in Cohorts A-D: PFS HR 1.27, 95% CI 0.59-2.71, p = 0.463; ORR OR 1, 95% CI 0.49-2, p>0.99. In cohort A-D, splitting patients by median CMR dynamics was predictive of PFS; mPFS low CMR 6 months vs high CMR 2 months (HR 0.43, 95% CI 0.19-0.96; p=0.008), and ORR 33% (5/15) vs 7% (1/15) (OR 6.54, 95% CI 2.85-16.57, p <0.0001). In cohort E splitting patients by median CMR was also highly predictive of PFS; mPFS low CMR 9 months vs high CMR 3 months (HR 0.47, 95% CI 0.24-0.94, p=0.01), and ORR 45% (10/22) vs 9% (2/22) (OR 8.27, 95% CI 3.80-18.52, p <0.0001). In cohort E, 36% of patients (16/44) had detectable BRCA1 methylation at C1D1. There was no association between BRCA1 methylation and PFS (HR 0.92, 95% CI 0.47-1.82, p=0.79), nor ORR (OR 0.86, 95% CI 0.43-1.50, p 0.633). Conclusions: Using a tumour agnostic breast cancer ctDNA methylation assay, we show that on treatment ctDNA methylation dynamics are predictive of response to targeted therapy. There was no association between BRCA1 methylation in ctDNA and outcomes, suggesting the sensitivity to PARP and ATR inhibition combination of cohort E was not mediated by BRCA1 methylation. Subject to further independent validation, ctDNA methylation dynamics may present a simple way to identify early patients who are responding to novel therapy combinations.
Presentation numberPS2-07-06
Prevalence and dynamics of circulating tumor DNA among patients with triple-negative breast cancer undergoing preoperative systemic therapy with or without immunotherapy
Tae-Kyung Robyn Yoo, Asan Medical Center, Seoul, Korea, Republic of
T. R. Yoo1, S. Morganti2, Q. Jin3, A. Patel2, C. Snow2, K. Santos2, C. Stever2, O. Cunningham2, T. Rahman2, J. Tejeda-Zanudo2, J. Baginska2, J. Bsat2, M. Luo2, I. G. Martino2, B. M. Drummey2, S. A. Virani2, K. Howarth4, C. Rushton5, S. Birkeälv4, S. Woodhouse5, G. Putcha4, E. Christoforou6, S. Kirschner6, A. Moeini7, N. Ahmad8, N. Tung9, N. Sinclair2, M. Constantinou10, M. R. Faggen2, S. Sinclair11, S. Lo12, J. L. Meisel13, E. Winer14, E. A. Mittendorf15, N. Tayob3, U. N. Lin2, S. M. Tolaney2, A. Garrido-Castro2, H. A. Parsons2; 1Surgery, Asan Medical Center, Seoul, KOREA, REPUBLIC OF, 2Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 3Data Science, Dana-Farber Cancer Institute, Boston, MA, 4Clinical, SAGA Diagnostics, Morrisville, NC, 5R&D, SAGA Diagnostics, Morrisville, NC, 6Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 7Translational Medicine, Oncology R&D, AstraZeneca, Barcelona, SPAIN, 8Medical Oncology, Phelps Cancer Center, Pittsfield, MA, 9Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, MA, 10Medical Oncology, Cancer Institute, Rhode Island Hospital, Providence, RI, 11Medical Oncology, Northern Light Eastern Maine Medical Center, Bangor, ME, 12Medical Oncology, Stamford Hospital, Stamford, CT, 13Medical Oncology, Glenn Family Breast Center, Winship Cancer Institute, Emory University, Atlanta, GA, 14Medical Oncology, Yale Cancer Center, New Haven, CT, 15Surgery, Brigham and Women’s Hospital, Boston, MA.
Background: Preoperative systemic therapy (PST) with immunotherapy (IO) is the current standard-of-care for patients (pts) with early-stage triple-negative breast cancer (TNBC). Circulating tumor DNA (ctDNA) persistence in the (neo)adjuvant setting is associated with residual disease at surgery and a higher risk of recurrence. However, studies of ctDNA in TNBC have not evaluated treatment in the modern, IO-containing era. We tested the performance of a highly sensitive assay to evaluate the prevalence and dynamics of ctDNA in the neoadjuvant TNBC setting with and without IO.Methods: We prospectively enrolled and retrospectively identified patients with TNBC receiving PST in an IRB-approved, multi-center registry. Plasma samples were collected at diagnosis (baseline), 4-7 weeks after start of PST (on-treatment), at completion of PST and before surgery (preop), within 3 months after surgery (postop), and every 6 months during follow-up. ctDNA was measured using the Pathlight assay (SAGA Dx), a tumor-informed assay based on structural variants identified from 15X whole-genome sequencing of tumor/normal samples, to detect and quantify minimal residual disease. The primary endpoint was ctDNA detection rate during and after PST.Results: We evaluated 444 plasma samples from 80/97 (82.5%) pts with >1 plasma sample, sufficient tumor tissue and available buffy coat. Pts were a median of 50.4 years old, and 34 (42.5%) had stage III disease. Pts were enrolled between 2019 and 2024, and due to a change in standard treatment, 31 (38.8%) pts received cytotoxic chemotherapy (CT) alone, while 49 (61.2%) pts received an IO-containing regimen. At the time of surgery, 49 (61.3%) pts had pathologic complete response (pCR).Personalized ctDNA assays were designed targeting 5-16 structural variants (median 12). ctDNA was detected at baseline in 71/76 (93.4%; 95% CI 85.3 – 97.8) pts, on-treatment in 24/70 (34.3%; 95% CI 23.3-46.6) pts, preop in 7/53 (13.2%; 95% CI 5.5-25.3) pts, and postop in 8/59 (13.6%; 95% CI 6.0-25.0) pts. Among pts who were ctDNA-positive (ctDNA+) at baseline with a paired on-treatment sample (n=62), 37 (59.7%) pts cleared ctDNA by the on-treatment timepoint. All pts who cleared on-treatment remained ctDNA-negative at the preop timepoint, when available (n=26). Among pts who were ctDNA+ at baseline with a paired preop sample (n=46), 39 (84.8%) pts cleared ctDNA at completion of PST. The negative predictive value (NPV) of ctDNA clearance for pCR by the on-treatment timepoint was 78.4% (29/37; 95% CI 61.8-90.2), while the NPV at the preop timepoint was 71.8% (28/39; 95% CI 55.1-85.0). On IO- vs non-IO containing regimens, the NPV of ctDNA clearance at the on-treatment timepoint was 85.7% (18/21; 95% CI 63.7-97.0) for IO-containing regimens and 68.8% (11/16; 95% CI 41.3-89.0) for pts undergoing cytotoxic CT alone.Among pts with residual disease at surgery (n=31), 19 (61.3 %) underwent adjuvant therapy including IO. 6/19 pts remained ctDNA+ after surgery, with persistent ctDNA in all subsequent samples until clinical recurrence. Median postoperative follow-up was 26.2 (IQR 18.6, 37.3) months. Of 8 pts who remained ctDNA+ after surgery, 7 (87.5%) had a clinical recurrence with a median lead time of 10.9 (IQR 6.1, 16.4) months. All (n=10) pts experiencing distant recurrence were ctDNA+ prior to the event. Follow-up is ongoing.Conclusions: ctDNA detection rate was high among pts with TNBC receiving PST. In pts with residual disease, ctDNA detected after surgery persisted despite adjuvant therapy; all such pts developed disease recurrence. This study highlights ctDNA as a promising biomarker for personalized risk assessment and systemic treatment guidance in pts with early-stage TNBC receiving modern preoperative systemic therapy.
Presentation numberPS2-07-07
Relationship of early ctDNA dynamics with response to inavolisib alone or in combination with endocrine therapy +/- CDK4/6 inhibitor in PIK3CA-mutated HR+, HER2- metastatic breast cancer from the first-in-human phI/Ib trial
Stephanie Hilz, Genentech, South San Francisco, CA
S. Hilz1, M. K. Accordino2, P. L. Bedard3, A. Cervantes4, V. Gambardella4, E. P. Hamilton5, A. Italiano6, K. L. Jhaveri7, D. Juric8, K. Kalinsky9, I. E. Krop10, M. Oliveira11, C. Saura11, P. Schmid12, N. C. Turner13, E. S. Sokol14, M. Childress15, R. S. Huang15, Z. J. Assaf16, J. Aimi16, S. Royer-Joo17, J. L. Schutzman17, K. E. Hutchinson16; 1Computational Sciences, Genentech, South San Francisco, CA, 2Division of Hematology and Oncology, Department of Medicine, Columbia University Medical Center, New York, NY, 3Division of Medical Oncology & Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, CANADA, 4Medical Oncology Dept., Hospital Clinico Universitario, INCLIVA, Biomedical Research Institute, University of Valencia, Valencia, SPAIN, 5Breast Cancer and Gynecologic Cancer Research, Sarah Cannon Research Institute, Nashville, TN, 6Early Phase Trials and Sarcoma Units, Institut Bergonie and University of Bordeaux, Bordeaux, FRANCE, 7Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 8Department of Medicine, Mass General Cancer Center, Boston, NY, 9Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 10Medical Oncology, Yale Cancer Center, New Haven, CT, 11Medical Oncology Department, Vall d’Hebron University Hospital and Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 12Centre of Experimental Medicine, Barts Cancer Institute, Queen Mary University London, London, UNITED KINGDOM, 13Institute of Cancer Research, Royal Marsden Hospital London, London, UNITED KINGDOM, 14Cancer Genomics Research, Foundation Medicine Inc., Cambridge, MA, 15Clinical Development, Foundation Medicine, Inc., Boston, MA, 16Translational Medicine, Genentech, South San Francisco, CA, 17Oncology, Genentech, South San Francisco, CA.
Background: Rapid determination of patient response to cancer therapy is crucial for timely treatment decisions, and may enable faster clinical trial decision-making to accelerate drug development. Traditional response assessments require imaging over several weeks. Across multiple indications and therapies, circulating tumor (ct)DNA dynamics have demonstrated the potential to serve as earlier response endpoints. Here, we evaluate the relationship between patient response and changes in ctDNA after 15 days of treatment in the first-in-human PhI/Ib study (NCT03006172) of the recently FDA-approved p110α-inhibitor, inavolisib. Methods: ctDNA was isolated from plasma samples collected from patients immediately prior to treatment (baseline) and at Cycle 1 Day 15 (C1D15) of treatment. Sequencing was performed with the Foundation Assay for Circulating Tumor DNA (FoundationACTTM) or FoundationOne® Liquid CDx. Responders were defined as those patients with confirmed complete or partial response per RECIST v1.1; non-responders had progressive disease. Logistic regression modeling was used to explore associations between ctDNA metrics and response. Maximally selected rank statistics were used for determining the optimal progression-free survival (PFS) cutpoints. Key ctDNA metrics including total number of alterations (totalAlts) and maximum (max) or sum of PIK3CA variant allele frequency (VAF) were computed for the subset of responder/nonresponder samples at baseline (n = 128) and C1D15 (n = 98). For the 38 responder/nonresponder patients with paired baseline:C1D15 samples, log2 and % change were also calculated. Finally, PFS stratification was evaluated for the 100 patients who had both paired baseline:C1D15 samples and PFS data. Results: Univariate logistic regression to test the association between response/non-response and ctDNA metrics identified % change in PIK3CA VAF (p = 0.006 for PIK3CA sumVAFs; p = 0.007 for PIK3CA maxVAF) and C1D15 totalAlts (p = 0.034) as associated with response. The median % change between baseline and C1D15 PIK3CA sumVAF was -58.41% for responders, and 0% for non-responders. Percent change in PIK3CA maxVAF and sumVAFs were highly correlated with one another (rho = 0.996 , p < 2.2e-16), so the % change in PIK3CA sumVAFs was prioritized moving forward. Further analyses showed C1D15 totalAlts did not significantly correlate with PIK3CA sumVAFs (rho = 0.186, p = 0.063), and C1D15 totalAlts did not remain significantly correlated with response in a multivariate model (p = 0.058). We tested the ability of % change in PIK3CA sumVAFs to stratify patients by PFS and identified 0% change to be the optimal cutpoint that effectively stratified patients (HR=10.4, 95% CI, 4.6-23.6), with 10 patients showing >0% change (median 43.74%) vs. 90 patients with ≤ 0% change (median -61.18%). Stratification by quartile or median changes did not effectively stratify patients by PFS. Conclusions: Together, our results suggest that early changes in ctDNA, specifically the % change in PIK3CA VAF between baseline and C1D15, are associated with objective response to inavolisib-containing regimens and may enable early stratification of patient response that is ultimately predictive of PFS. Additional research with larger cohorts and encompassing the evaluation of tumor fraction (not available for this dataset) are warranted to confirm these findings and to understand the applicability to other targeted therapies for HR+, HER2- breast cancer.
Presentation numberPS2-07-08
Longitudinal ctDNA Tracking in Early and Recurrent Breast Cancer Using an Ultrasensitive Structural Variant-Based Assay: An Updated Analysis from the TRACER Study
Mitchell J Elliott, Princess Margaret Cancer Centre, Toronto, ON, Canada
M. J. Elliott1, J. Roh1, T. Bird1, M. Li1, M. Nadler1, E. Amir1, V. Kumar1, C. Yu2, K. Howarth3, G. Putcha3, S. Woodhouse3, L. Siu1, P. Bedard1, H. Berman1, D. W. Cescon1; 1Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 2Cancer Genomics Program, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 3SAGA Dx, SAGA Dx, Morrisville, NC.
Background: We previously reported the performance of an ultrasensitive structural variant (SV) and digital PCR (dPCR)-based circulating tumor DNA (ctDNA) assay in a cohort of 100 patients with early breast cancer (EBC) treated with neoadjuvant therapy, demonstrating high sensitivity, specificity, and a long lead time to distant relapse. While the personalized SV ctDNA fingerprint used in this assay holds promise for monitoring ctDNA dynamics in recurrent metastatic disease, it has not yet been evaluated in this setting. Here, we report findings from an expanded EBC cohort with extended clinical follow-up and retrospective ctDNA analysis in the metastatic setting. Methods: We conducted a retrospective analysis of patients with EBC treated with neoadjuvant therapy at the Princess Margaret Cancer Centre using an ultrasensitive, SV-based, tumor-informed ctDNA assay. Plasma samples were collected at baseline, during treatment, perioperatively, and throughout follow-up. Several patients who experienced disease recurrence consented to continued prospective plasma collection following metastatic relapse. For these patients, plasma was collected at the time of metastatic recurrence (metastatic baseline) and at subsequent restaging time points during treatment. ctDNA dynamics in the metastatic setting were evaluated retrospectively and compared with clinical and radiographic response assessments. Clinical outcomes were last updated on June 1, 2025. Results: A total of 121 patients were included (36 triple-negative breast cancer [TNBC], 44 HER2-positive [HER2+], and 41 ER-positive/HER2-negative [ER+/HER2-]). Twenty-seven patients (22%) experienced recurrence (1 local, 26 distant; 11 TNBC, 3 HER2+, 13 ER+/HER2-) with a median follow-up of 3.7 years (range: 0.4-8.0). All patients with detectable ctDNA in the postoperative or adjuvant setting developed metastatic recurrence (sensitivity: 100%, specificity: 100%), including one case of isolated intracranial relapse. The median lead time from ctDNA detection to metastatic recurrence among evaluable patients was 381 days (range: 4-1931 days). Ten patients (6 TNBC, 1 HER2+, 3 ER+/HER2-) with recurrence underwent ongoing plasma collection in the metastatic setting, spanning a median of one line of therapy (range: 1-3 lines), including treatment with endocrine therapy plus a CDK4/6 inhibitor, chemotherapy, chemo-immunotherapy, and antibody-drug conjugates (ADCs). The median variant allele fraction (VAF) at metastatic baseline was 2.7% (range: 0.01-37.8%). ctDNA remained detectable in all but one patient, who had a complete radiographic response to an ADC plus immunotherapy (median VAF: 0.3%; range: 0-37.8%), with 3 of 26 timepoints falling below a VAF of 0.01%. Using radiographic response as the clinical reference standard, ctDNA monitoring, defined as any decrease for response and any subsequent rise for progression, demonstrated strong concordance with radiographic outcomes (sensitivity: 100%; negative predictive value: 100%). In all cases, a rise in ctDNA (ie. molecular progression) preceded radiographic progression. SVs identified at baseline were reproducibly detected across longitudinal timepoints in both early-stage and metastatic settings, supporting the stability of this tumor-informed approach to ctDNA detection. Conclusion: These results reaffirm the performance of an ultrasensitive ctDNA assay using an SV and dPCR-based approach. Preliminary results support the feasibility of this assay for ctDNA monitoring in the recurrent metastatic setting, across a range of standard therapies. Additional metastatic monitoring data in a larger cohort of metastatic breast cancer will be presented. Clinical Trial Information: NCT03702309
Presentation numberPS2-07-10
A Single Cell Peripheral Blood Immune Cell Atlas of Breast Cancer Disease States by Race reveals Differences in Peripheral Blood Immune Cell Composition and Functional Pathways
Fangyuan Chen, Dana-Farber Cancer Institute, Boston, MA
F. Chen1, E. R. Ogayo1, T. Rahman1, A. Recko1, A. M. Parsons1, P. van Galen2, S. S. McAllister2, E. A. Mittendorf1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Hematology, Brigham and Women’s Hospital, Boston, MA.
Background: Racial disparities in breast cancer (BC) are well documented— Non-Hispanic Black (NHB) women are more likely to develop BC at a younger age and experience worse survival outcomes than Non-Hispanic White (NHW) women. Previous studies using pan-cancer cohorts have suggested that changes in peripheral immune cells accompany tumor development. However, in breast cancer, whether the trajectory of these immune changes differs by race across key stages of disease progression, remains poorly understood. To address this gap, we systematically characterized and compared the circulating immune landscape of NHB and NHW women across the continuum of breast cancer progression, from high-risk lesions (HRL) to ductal carcinoma in situ (DCIS) to invasive breast cancer (BC). Methods: PBMCs were isolated from age-matched NHB (HRL:10, DCIS:6, BC:13) and NHW women (HRL:10, DCIS:4, BC:13). scRNA-seq was performed using the 10x Genomics 5’ v2 platform, with data processing by CellRanger (v7.1.0) and Seurat (v5.1.0). Cell types were identified via reference-guided annotation with Azimuth (v0.5.0) and canonical markers. Differentially expressed genes (DEGs) between NWB and NHW were detected using edgeR (v4.4.2, by FDR<0.05) after pseudobulking by cell type in each patient, the number of DEGs was used to quantify PBMC transcriptomic differences by race and across disease stages. Pathway enrichment of DEGs was performed with Gene Set Enrichment Analysis (GSEA). Results: We analyzed 436,077 high-quality single PBMCs classified into 36 cell types. The proportions of various PBMC cell types differed between NHB and NHW women across all disease stages. For example, the ratio of classical:non-classical monocytes was lower in NHB women than NHW women at all disease stages. We observed a markedly higher number of DEGs between BC and HRL than between DCIS and HRL, suggesting a greater transcriptomic change with the transition to invasive breast cancer. These differences were mostly reflected in naïve and non-switched memory B cells, naïve and central memory CD4+ T cells, monocytes, and mature NK cells. In BC patients, for monocytes, NHB race was related to abundance of non-classical monocytes and IFN pathway in all monocytes. In CD4+ T cells, NHB showed higher PD-1 and lower TIM-3 levels, potentially suggesting an early exhaustion state towards immune suppression, which may affect immunotherapy outcome. TNF-α, apoptosis, IL2-STAT5, and TGF-β signaling pathways were consistently up-regulated in multiple cell types in NHB PBMC, and oxidative phosphorylation was constantly down-regulated. Conclusion: This study is one of the largest single cell transcriptomic profiling of circulating immune system comparing race across disease states of breast cancer. Results showed that BC gene expression (GE) diverge from HRL much more than DCIS. GE difference by race was also largest in BC, featuring increased IFN in monocytes and an early exhaustion in CD4 T cells in NHB patients. Further investigation is warranted to determine how these differences in systemic immune cell quality in NHB breast cancer patients relate to response to immunotherapy and survival.
Presentation numberPS2-07-11
The improved long-term outcome in denosumab-treated postmenopausal women with early luminal breast cancer in Abcsg 18 is driven by progesterone receptor-positive tumors.
Christian F Singer, Medical University of Vienna, Vienna, Austria
C. Singer1, D. Hlauschek2, D. Egle3, A. Reiner4, G. G. Steger5, G. Huber6, R. Greil7, G. Rinnerthaler8, F. Fitzal9, C. Brunner3, C. Suppan8, G. Pfeiler1, S. P. Gampenrieder7, M. Seifert1, S. Kacerovsky-Strobl10, C. Deutschmann1, K. Wimmer11, M. Balic12, R. Jakesz5, C. Fesl2, H. Fohler13, M. Gnant5; 1Department of Gynecology and Obstetrics, Medical University of Vienna, Vienna, AUSTRIA, 2Statistics Department, Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, AUSTRIA, 3Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, AUSTRIA, 4Institut für Klinische Pathologie, Molekularpathologie und Mikrobiologie, Klinik Donaustadt, Vienna, AUSTRIA, 5Comprehensive Cancer Center, Medical University of Vienna, Vienna, AUSTRIA, 6Ordination Dr. Huber, Breast Center Carinthia, St. Veit/Glan, AUSTRIA, 7Department of Internal Medicine III, Oncologic Center, Salzburg Cancer Research Institute, Paracelsus Medical University, Salzburg, AUSTRIA, 8Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, AUSTRIA, 9Department of Surgery and Breast Health Center, Hanusch Hospital, Vienna, AUSTRIA, 10Department of Surgery, Klinikum Donaustadt, Vienna, AUSTRIA, 11Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, AUSTRIA, 12Division of Oncology, University of Pittsburgh Medical School, Hillmans Cancer Center, Pittburgh, PA, 13Management, Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, AUSTRIA.
The improved long-term outcome in denosumab-treated postmenopausal women with early luminal breast cancer in ABCSG 18 is driven by progesterone receptor (PR)-positive tumors. Background: The biological effects of progesterone are mediated through the RANK/RANK-ligand system, which promotes tumor growth and malignant behavior in breast cancer models. In the large prospective, double-blind, placebo-controlled, phase 3 ABCSG 18 trial we have demonstrated that the addition of the RANK ligand denosumab dramatically reduces fractures compared to placebo, but also improves disease-free survival (DFS), and overall survival (OS) in aromatase inhibitor-treated postmenopausal patients with early luminal breast cancer. Patients and Methods: Tumor histological grade, as well as immunohistochemically quantified Ki-67, estrogen receptor (ER), and progesterone receptor (PR) data were collected from 2,026 patients. ER and PR were assessed according to the Allred score and considered positive if ≥3. Disease-free survival (DFS), distant recurrence-free (DRFS), and overall survival (OS) data were prospectively collected during a median follow-up (FU) of 8.1 years as part of the ABCSG 18 study. Cox Models were used to evaluate the association with outcome. Results: Overall, luminal A-like tumors had a better DFS, DRFS, and OS than luminal B-like tumors, but PR expression per se was not prognostic in this large prospective clinical trial. Patients with PR positive tumors, however, who were treated with denosumab, enjoyed a considerably better long-term outcome than patients who had been randomized to the placebo arm (HR for DFS: 0.80; 95% CI 0.65-0.98; HR for DRFS: 0.68; 95% CI 0.51-0.90; HR for OS 0.63; 95% CI 0.46-0.88). In contrast, in patients with PR-negative tumors, no differences in long-term outcomes between the denosumab or placebo arm of the trial were observed (HR for DFS: 0.87; 95% CI 0.46-1.64; HR for DRFS: 1.19; 95% CI 0.53-2.66; HR for OS 0.99; 95% CI 0.37-2.65). While the HR for DFS in denosumab vs placebo patients remained stable with increasing Allred scores, HRs for DRFS and OS improved with increasing Allred scores, thereby suggesting that the efficacy of denosumab depends on the degree of intra-tumoral PR expression. Conclusion: Our results indicate that the long-term outcome benefit for adjuvant denosumab in this large prospective randomized placebo-controlled trial is driven by patients with PR-positive tumors. The disruption of PR signaling, either via PR antagonization or by RANKL inhibition, could therefore be a promising therapeutic strategy in luminal breast cancer.
Presentation numberPS2-07-12
Gut microbiome composition predicts pathologic complete response in patients with early-stage HER2-positive breast cancer receiving neoadjuvant HER2-targeted therapy +/- immunotherapy with pembrolizumab
Alexis LeVee, UCLA David Geffen School of Medicine, Los Angeles, CA
A. LeVee1, K. Lee2, S. Rice3, I. Chan3, S. Sridharan3, S. Shiao4, S. Pal5, H. McArthur3; 1Department of Medicine, Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 2TGen Integrated Microbiomics Center (TIMC), Translational Genomics Research Institute (TGen), Flagstaff, AZ, 3Department of Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, 4Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, 5Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA.
Background: The gut microbiome has been shown to influence response to immune checkpoint inhibitors (ICI). This study aimed to investigate the association between gut microbiome profiles and clinical outcomes in patients with early-stage HER2-positive (HER2+) breast cancer receiving neoadjuvant HER2-targeted therapy +/- immunotherapy with pembrolizumab (K) as part of the randomized phase II neoHIP trial. Methods: Patients with stage II-III HER2+ breast cancer were randomized to 3 treatment arms: Arm A consisted of paclitaxel (T), trastuzumab (H), and pertuzumab (P), THP; Arm B of THP-K; and Arm C of TH-K. Fecal samples were collected at baseline, cycle 3 day 1, surgery, and at 30 and 60 days post-surgery and were analyzed using deep shotgun metagenomics sequencing. Pre-operative fecal samples were analyzed according to pathologic complete response (pCR) and onset of diarrhea during the neoadjuvant period. Comparisons were adjusted to control for false discovery rates (q-value). Results: 99 fecal samples (Arm A: n=32; Arm B: n=48; Arm C: n=19) from 27 patients were included in the analysis. Bacterial diversity by Jaccard analysis was significantly different across all time points for all arms (all p<0.05; q<0.05). In the THP and THP-K arms, Jaccard analysis demonstrated differences in gut microbiome diversity in patients who achieved pCR versus non-pCR (p=0.009; q=0.009). In patients treated with THP who achieved pCR, multiple species were in greater abundance including GGB9237 SGB14179 (log fold change [LFC] 6.87), GGB34797 SGB14322 (LFC 6.25), and Faecalicatena fissicatena (LFC 4.61) compared to those with non-pCR (all p<1E-5; q<0.01). In the THP-K arm, patients who achieved pCR had different species in greater abundance, including GGB9365 SGB14341 (LFC 3.15), Hydrogenoanaerobacterium saccharovorans (LFC 2.92), and GGB9502 SGB14899 (LFC 2.56) compared to those with non-pCR (all p<0.0001; q<0.01). Comparing those who achieved pCR in THP vs. THP-K, GGB9715 SGB15260 (LFC 4.89), Brotolimicola acetigignens (LFC 4.88), and Clostridiaceae bacterium Marseille Q4149 (LFC 4.54) were higher in abundance in the THP-K arm, and GGB34797 SGB14322 (LFC 6.27) and GGB3175 SGB4191 (LFC 5.73) were depleted in the THP-K arm (all p<0.0001; q<0.05). In patients with diarrhea vs. without diarrhea, beta diversity was significantly different in the THP arm by Bray Curtis analysis (p<0.05; q<0.05) and in the THP-K arm by Jaccard analysis (p<0.05; q<0.05), with multiple species in different abundances in those with and without diarrhea in both arms (p<0.05). Conclusion: This is the first study to demonstrate that the pre-operative gut microbiome influences response to ICI in patients with early-stage HER2+ breast cancer. Patients who achieved pCR had a significantly different microbiome profile compared to those with residual disease. This study highlights the role of the gut microbiome in influencing response to ICI in patients with breast cancer treated in the curative intent setting.
Presentation numberPS2-07-13
Prognostic and predictive role of RBsig and CCNE1/RB1 gene-expression signatures for patients with early breast cancer treated with endocrine therapy with or without palbociclib in the PALLAS trial (ABCSG-42, AFT-05, BIG 14-03)
Luca Malorni, Hospital of Prato, Azienda USL Toscana Centro, Prato, Italy
L. Malorni1, D. Hlauschek2, E. Mayer3, M. Benelli4, L. Biganzoli5, I. Migliaccio1, A. Kermanidis2, G. Pfeiler6, E. P. Mamounas7, A. M. Brufsky8, M. Bellet-Ezquerra9, K. Clifton10, G. Rubovszky11, T. Foukakis12, M. Goetz13, S. Lee14, M. Ruiz-Borrego15, W. Symmans16, K. V. Ballman13, F. Liu17, J. Machacek-Link2, C. Denkert18, A. DeMichele19, M. Gnant20; 1Oncology Department and Transaltional Research Unit, Hospital of Prato, Azienda USL Toscana Centro, Prato, ITALY, 2Austrian Breast and Colorectal Cancer Study Group, ABCSG, Vienna, AUSTRIA, 3Dana-Farber Cancer Institute, DFCI, Boston, MA, 4Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, ITALY, 5Oncology Department, Hospital of Prato, Azienda USL Toscana Centro, Prato, ITALY, 6Department of Gynecology and Gynecological Oncology, Medical University of Vienna and ABCSG, Vienna, AUSTRIA, 7Orlando Health Cancer Institute, Orlando Health Cancer Institute, Orlando, FL, 8University of Pittsburgh Cancer Institute, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 9Hospital Universitari Vall d’Hebron and Vall d’Hebron institute of Oncology, VHIO, Barcelona, SPAIN, 10Department of Medicine, Division of Oncology, Washington University in Saint Louis, St. Louis, MO, 11National Institute of Oncology Hungary, National Institute of Oncology Hungary, Budapest, HUNGARY, 12Department of Oncology/Pathology, Karolinska Institute, Stockholm, SWEDEN, 13Mayo Clinic, Mayo Clinic, Rochester, MN, 14Department of Haematology-Oncology, National University Cancer Institute (NCIS), Singapore, SINGAPORE, 15Hospital Universitario Virgen del Rocio and GEICAM, Hospital Universitario Virgen del Rocio and GEICAM, Sevilla, SPAIN, 16MD Anderson Cancer Center, University of Texas, Houston, TX, 17Clinical Pharmacology and Translational Sciences, Oncology, Pfizer Inc., San Diego, CA, 18Institute of Pathology, Philipps University Marburg and University Hospital Marburg (UKGM), Marburg, GERMANY, 19University of Pennsylvania, University of Pennsylvania, Philadelphia, PA, 20Austrian Breast and Colorectal Cancer Study Group, Comprehensive Cancer Center, Medical University of Vienna, Vienna, AUSTRIA.
Background:We have identified two potentially predictive signatures of palbociclib resistance: the RBsig, composed of E2F1/E2F2 dependent genes, which is associated with genetic loss of RB1, and the ratio between the gene expression levels of CCNE1 to RB1 (CCNE1/RB1). Both signatures have been previously analyzed in vitro and in neoadjuvant and metastatic studies with palbociclib. The present analysis aims to explore the role of RBsig and CCNE1/RB1 in the phase III adjuvant PALLAS trial, which randomized patients to palbociclib plus endocrine therapy, versus endocrine therapy alone. Materials and methods:Gene expression data from PALLAS were generated using the HTG EdgeSeq Oncology BM Panel. Of the 87 genes composing RBsig, 46 were available within the EdgeSeq dataset and were used for the analyses; CCNE1 and RB1 were both available. RBsig was calculated as the mean of the Z-score scaled gene expression (log) of the 46 genes; CCNE1/RB1 was computed as the log ratio between the mRNA expression of CCNE1 and RB1. The prognostic/predictive effect of the continuous signatures in terms of invasive disease-free survival (IDFS), distant recurrence-free survival (DRFS), and overall survival (OS) was evaluated using Cox proportional hazard models Results:HTG data were available from 2,587 of 5,748 patients from the entire PALLAS intention-to-treat (ITT) cohort. As RBsig and CCNE1/RB1 levels are affected by pre-surgical treatments, we limited the analyses to 1612 pre-treatment samples (530 core biopsies and 1082 surgical specimens) (analysis population). Compared with the entire PALLAS ITT cohort, the analysis population was significantly enriched for patients with low clinical risk (41.3% vs 59.4%) who did not receive prior chemotherapy (17.5% vs 55.9%). In keeping with the PALLAS ITT cohort, no significant benefit from the addition of palbociclib was observed in the analysis population. Higher baseline RBsig or CCNE/RB1 values were significantly and independently associated with worse IDFS (hazard ratio [HR] 1.63, 95% confidence interval [CI] 1.3-2.0; HR 1.22, 95%CI 1.1-1.3, respectively), DRFS (HR 1.94, 95%CI 1.5-2.5; HR 1.26, 95%CI 1.1-1.4, respectively) and OS (HR 1.79, 95% CI 1.3-2.4; HR 1.26, 95%CI 1.1-1.4, respectively) in the analysis population. No predictive effect of palbociclib benefit was observed for either signature in the analysis population, nor according to clinical risk or chemotherapy receipt. Interestingly, higher RBsig was strongly prognostic in patients who did not receive adjuvant chemotherapy (HR 1.83, 95%CI 1.4-2.3) but not in those who received it (HR 1.01, 95%CI 0.6-1.6) (interaction p=0.02) suggesting a potential predictive role of RBsig for chemotherapy benefit. Conclusions:In the PALLAS trial, higher baseline RBsig or CCNE1/RB1 values are associated with worse prognosis, but are not predictive of benefit from palbociclib in the adjuvant setting. RBsig might be associated with differential benefit from chemotherapy. Further study of these biomarkers may strengthen their role in risk stratification.https://acknowledgments.alliancefound.org
Presentation numberPS2-07-14
Characterization of long-term responders to first-line CDK4/6 inhibitors in HR+/HER2- advanced breast cancer
Fara Brasó-Maristany, IDIBAPS, Barcelona, Spain
I. Garcia-Fructuoso1, O. Martínez-Sáez1, M. Romero Vaquerano2, R. Gómez-Bravo1, F. Schettini1, S. Cobo3, E. Sanfeliu4, B. Adamo1, È. Seguí3, B. Walbaum3, M. González-Rodríguez1, M. Bergamino1, O. Castillo3, P. Blasco3, P. Galván3, M. Muñoz1, C. Núria1, T. Pascual1, M. Vidal Losada1, A. Prat1, F. Brasó-Maristany3; 1Medical Oncology Department, Hospital Clinic de Barcelona, Barcelona, SPAIN, 2Medical Oncology Department, Salvadoran Social Security Institute, San Salvador, EL SALVADOR, 3Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, SPAIN, 4Department of PAthology, Hospital Clinic de Barcelona, Barcelona, SPAIN.
Background: CDK4/6 inhibitors (CDK4/6i) plus endocrine therapy (ET) have reshaped first-line treatment of HR+/HER2- metastatic breast cancer (MBC), achieving median progression-free survival (PFS) of 24-33 months in pivotal trials. However, real-world outcomes vary: while some progress early, others achieve long-term control. As triplet therapies and escalation strategies emerge, identifying predictors of prolonged benefit is key to refine patient selection. We aimed to characterize clinical, molecular and pathological factors associated with sustained benefit in this setting. Methods: We analyzed 166 patients (pts) with HR+/HER2- MBC treated with first-line CDK4/6i between March 2014 and April 2022 at Clinic Barcelona Comprehensive Cancer Center, with a minimum follow-up (FU) of 36 months. Clinical and molecular characteristics were compared between long-term responders (LR, defined as PFS ≥36 months) and non-long responders (nLR). Comparisons were performed using chi-square or Fisher’s exact tests. Survival was analyzed using the Kaplan-Meier method. Odds ratios (ORs) for factors associated with LR were estimated using univariate and multivariate logistic regression. Intrinsic subtype (IS) was determined at baseline using a research-based PAM50 assay in 123 pts. Gene expression data from the Breast Cancer 360™ nCounter panel were available for 119 pts. Statistical significance was set at p≤0.05. Results: Median FU was 60.5 months (95% CI: 54.0-67.43). Median PFS and overall survival (OS) for the cohort were 21.9 months (95% CI: 18.3-26.5) and 57.9 months (95% CI: 44.8-74.6), respectively. By data cutoff, 133 pts (80.1%) had progression and 87 (52.4%) had died. Of the total, 53 (31.9%) classified as LR. Most pts received ribociclib (104, 62.7%), followed by palbociclib (58, 34.9%) and abemaciclib (4, 2.4%), with no significant differences in CDK4/6i type between LR and nLR groups (p=0.626). IS distribution differed significantly (p=0.038): 32 (82.1%) of LR were luminal vs. 53 (63.1%) in nLR, and 7 (17.9%) vs. 31 (36.9%) were non-luminal. Specifically, Luminal A (LUMA) was more frequent in LR (51.3% vs. 31.0%, p=0.045), while HER2-enriched tumors were less frequent (5.1% vs. 19.0%, p=0.054). Endocrine-resistant tumors (defined as relapse during adjuvant ET or within 12 months after its completion) were less common in LR (p=0.009), and liver metastases were significantly less frequent in LR (7.6% vs. 38.9%, p20%) was more common in LR (43.4% vs. 21.2%, p=0.005). In univariate analysis, endocrine resistance (OR 0.30, p=0.003), liver metastases (OR 0.13, p20% (OR 2.77, p=0.011), Luminal IS (OR 2.67, p=0.038), and LUMA (OR 2.35, p=0.031) were significantly associated with LR. In multivariate analysis including endocrine resistance, liver metastases, PR >20% and luminal IS, only liver metastases remained independently associated with a lower probability of being LR (OR 0.16, p=0.023). The remaining variables were not independently significant, likely reflecting overlapping biological information related to hormone sensitivity and luminal phenotype. Multivariate logistic regression models adjusted for endocrine resistance, liver metastases, PAM50 subtype, and PR >20%, identified multiple immune-related genes (including HLA-A, HLA-B, HLA-C, HLA-E, CD274, STAT1, CCL5, CXCL10, TAP1, PSMB10, and GZMA) independently associated with LR, highlighting the potential contribution of immune-related pathways to durable benefit from CDK4/6i. Conclusions: In HR+/HER2- MBC, long-term benefit from first-line CDK4/6i was associated with absence of liver metastasis, hormone-sensitivity, luminal features, and upregulation of immune-related genes, highlighting a potential role of the tumor immune microenvironment in durable response.
Presentation numberPS2-07-15
Immune biomarkers of standard-of-care chemoimmunotherapy response and resistance in real-world patient population: TBCRC061
Justin M. Balko, Vanderbilt University Medical Center, Nashville, TN
J. M. Balko1, P. I. Gonzalez-Ericsson1, B. Taylor1, A. Ocampo1, J. Serrenho1, S. Opalenik1, V. Sanchez1, M. Sanders2, M. Rimawi3, Y. Abdou4, S. Nunnery5, R. Nanda6, F. Howard6, J. Anampa-Mesias7, L. Kennedy1; 1Medicine, Vanderbilt University Medical Center, Nashville, TN, 2Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 3Medicine, Baylor College of Medicine, Houston, TX, 4Medicine, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 5Medicine, Tennessee Oncology, Nashville, TN, 6Medicine, University of Chicago, Chicago, IL, 7Medicine, Montefiore Medical Center, Bronx, NY.
Background: Currently, only a small fraction (<10%) of patients with breast cancer receiving immune checkpoint inhibitors (ICI) as standard of care (SOC) achieve clinical benefit. Additionally, immunotherapy carries serious and long-lasting immune-related adverse events, highlighting the need for effective biomarkers for patient selection. Most biomarkers evaluated thus far, including PD-L1, stromal tumor-infiltrating lymphocytes (sTILs), TMB and most gene expression profiles, are generally prognostic or predictive of chemotherapy response, and lack immunotherapy benefit specificity. Some biomarkers, like DetermaIO, tumor-specific (ts)MHC-II have been identified as potential predictive biomarkers for immunotherapy-specific response across multiple tumor types and trials while many others are generally prognostic or predictive of response in both chemotherapy- and chemo-immunotherapy-treated patients. We present the results from the early-stage cohort of TBCRC061, a non-interventional correlative trial leveraging archival tissues from patients treated with SOC ICI (neoadjuvant target accrual n=80, and metastatic n=100) to validate these biomarkers. Methods: Patients with breast cancer receiving SOC chemoimmunotherapy were identified and consented. Pre-treatment formalin-fixed and paraffin embedded archival samples were collected. Relevant patient demographics and response to treatment were extracted from the medical record. The neoadjuvant cohort completed accrual in 2025 and will be presented here. Results: Of 85 consented patients in the neoadjuvant cohort, 67 completed neoadjuvant treatment and had sufficient and suitable tissue for correlative analysis. Forty-seven patients (69%) achieved a complete or near-complete response (defined as RCB 0/I). tsMHC-II demonstrated a trend for enhanced response (ts-HLA-DR≥5%, n=40, 78% vs 59%, p=0.09, Fisher’s exact test). sTILs ≥30% defined numerically higher rates of response (83% vs 66%, p=0.14, Fisher’s exact test). Patients with tumors with immune infiltration in the tumor core (inflamed, n=29) or brisk accumulation of TILs in the tumor margin (margin predominant, n=30) achieved similar response rate (79% vs 73%), however patients with ≤5% sTILs and no accumulation on the margins (immune desert; ID) had drastically lower response rates (17%, n=6, p=0.01, Fisher’s exact test). The combination of tsMHC-I & II and tumor immune spatial organization differentiated response to chemoimmunotherapy (AUC: 0.75 p=0.0012, multiple logistic regression). Discussion: The combination of tsMHC-I & II and tumor immune spatial organization more accurately differentiated response to chemoimmunotherapy than each individual biomarker. Given the lack of a directly comparable chemotherapy-alone cohort, we cannot differentiate immunotherapy-specific benefit from general chemotherapy benefit in this trial, though MHC-II was not prognostic or predictive of chemotherapy response in prior studies. Patients with ID tumors likely require a different therapeutic strategy than the current SOC, as they consistently demonstrate exceptionally poor rates of response across trials. RNA sequencing data from baseline tumor biopsies is pending and will be presented as well in an exploratory analysis, including discovery of potential targetable pathways overrepresented in ID tumors.
Presentation numberPS2-07-16
Clinicogenomic characterization and ctDNA detection of ESR1 fusion positive metastatic breast cancer
Katheryn Santos, Dana-Farber Cancer Institute/Broad Institute, Boston, MA
K. Santos1, A. Lebrón-Torres2, C. Weipert3, S. Morganti4, K. Smith4, A. Patel4, C. Snow4, M. Hughes5, S. Ravikumar4, G. Suggs4, J. Bsat4, S. Oesterreich6, A. V. Lee7, E. P. Winer5, H. J. Burstein4, D. L. Abravanel4, H. A. Parsons4, S. M. Tolaney4, R. M. Jeselsohn4, N. U. Lin4, N. Priedigkeit1; 1Medical Oncology, Dana-Farber Cancer Institute/Broad Institute, Boston, MA, 2Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, 3Clinical Oncology, Guardant Health, Palo Alto, CA, 4Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 5Medical Oncology, Yale Cancer Center, New Haven, CT, 6Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 7Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA.
Background: ESR1 fusions are an emerging mechanism of acquired resistance to endocrine therapy (ET) in hormone-receptor positive (HR+) metastatic breast cancer (MBC). These fusions drive estrogen-independence and eliminate binding sites for all approved ER-targeting agents, posing a significant clinical challenge. Non-invasive detection of ESR1 fusions is limited by diverse gene partners and variable DNA breakpoints, which complicates assembling meaningfully large cohorts. By integrating high quality RNA-sequencing (RNA-seq), a sensitive, methylation-informed circulating tumor DNA (ctDNA), and robust clinical data, we set out to comprehensively characterize ESR1 fusion-positive disease. Methods: Patients with HR+ MBC harboring ESR1 fusions were retrospectively identified in a single academic center using three approaches: (1) whole transcriptome RNA-seq on fresh-frozen biopsies, (2) transcriptome profiling on archival tissue as part of standard clinical testing, and (3) a next-generation methylation-informed ctDNA assay. For fusion-positive cases, comprehensive clinical and outcome data were collected. For cases with RNA-seq, PAM50 assignments and Gene Set Enrichment Analyses (GSEA) were performed. Results: We identified 63 patients with an ESR1 fusion, with a median age at metastatic diagnosis of 56 years (range 30 – 83). Fusions were detected in 4.5% (9 of 202) of patients with luminal metastatic disease via tumor RNA-seq, in 9.9% (45 of 454) of patients with HR+ MBC via ctDNA testing, and in 11 additional cases as part of routine clinical testing; with 2 cases confirmed by both RNA-seq and ctDNA. In patients with longitudinal sampling, 76% (32 of 42) had a prior fusion-negative test, suggesting fusions are predominately acquired events. Fusions were detected a median of 2.6 years (range 0 – 10.9) from metastatic diagnosis and emerged after a median of 4 prior lines of therapy (range 1 – 14). All patients received ET prior to fusion detection (58% SERM, 83% AI, 60% SERD). Post-fusion detection, the longest interval of disease control with combination ET was 10.1 months whereas single agent ET was only 2.8 months. Four patients received an oral selective estrogen receptor degrader (SERD) after fusion detection-with the longest time to next treatment being 3.9 months. Genomically, the most recurrent 3’ partners were: AKAP12 (in 7 cases), CCDC170, PLEKHG1, VTA1 (each in 5 cases), and IMPG1 (in 4 cases), with the majority of partners being promiscuous or n-of-1 events. All ESR1 fusion positive tumors with RNA-seq were PAM50 classified as Luminal B, and GSEA of ESR1-fusion positive cases versus other Luminal B tumors revealed highly significant enrichment in proliferation pathways-including G2M checkpoint, E2F and MYC targets (FDR q-val < 0.25). The median overall survival after fusion detection was 5.4 months and 44 of 63 (70%) patients had passed at the time of this analysis; with prospective analyses ongoing. Additional genomic correlates, detailed clinical outcomes, and clinical case vignettes will be presented. Conclusions: This comprehensive clinicogenomic study of ESR1 fusion-positive MBC further establishes ER fusions as relatively common drivers of ET resistance. Critically, these fusions require more sensitive detection assays and serial testing-making prior estimates of prevalence likely conservative. ESR1 fusions appear acquired in ET resistant disease, are a marker of poor prognosis with a hyperproliferative biology, and confer broad resistance to subsequent ET-including oral SERDs. As next-generation SERDs more effectively target ESR1 point mutations, we hypothesize ESR1 fusions will become an increasingly significant clinical problem. Collectively, these findings highlight an urgent need to characterize ER fusion-positive MBC to establish more effective therapeutic options.
Presentation numberPS2-07-17
Characterizing circulating tumor cell (CTC) enumeration and phenotypic profiling in blood with synchronously obtained tissue in metastatic breast cancer (MBC) in TBCRC /AURORA US
Karthik V Giridhar, Mayo Clinic Comprehensive Cancer Center, Rochester, MN
K. V. Giridhar1, W. C. Nenad2, H. Ye3, G. L. Wheeler4, T. Hinoue5, A. R. Michmerhuizen2, B. M. Felsheim2, U. R. Chandran6, B. J. Kelly7, E. R. Mardis7, P. W. Laird5, J. M. Balko8, A. C. Garrido-Castro9, S. Coppens4, J. Bowen4, R. Nanda10, N. U. Lin9, C. K. Anders11, C. Isaacs12, J. J. Tao13, J. D. Anampa14, N. Jahan15, L. Huppert16, A. DeMichele17, B. H. Park8, S. Vinayak18, A. L. Delson19, M. M. Magbanua20, M. Balic6, H. A. Parsons9, A. V. Lee6, L. A. Carey2, L. Norton21, A. C. Wolff13, N. E. Davidson18, I. Krop22, T. A. King9, C. M. Perou2, K. A. Hoadley2, Aurora US Metastases Network; 1Department of Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 2Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 3Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 4Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, 5Department of Epigenetics, Van Andel Research Institute, Grand Rapids, MI, 6UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 7Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, 8Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 9Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 10Department of Medicine, The University of Chicago, Chicago, IL, 11Duke Cancer Center, Duke Health, Durham, NC, 12Department of Medicine, Georgetown University Lombardi Cancer Center, Washington, DC, 13Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, 14Department of Medical Oncology, Albert Einstein College of Medicine, Bronx, NY, 15O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 16Department of Medicine, University of California San Francisco, San Francisco, CA, 17Rena Rowan Breast Center, Penn Medicine, Philadelphia, PA, 18Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, 19Breast Science Advocacy Core, University of California San Francisco, San Francisco, CA, 20Laboratory Medicine, University of California San Francisco, San Francisco, CA, 21Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY, 22Yale Cancer Center, Yale School of Medicine, New Haven, CT.
Background: In MBC, CTC enumeration is highly prognostic for both progression free survival (PFS) and overall survival (OS). Multiparametric technologies enable for expanded phenotypic profiling of CTCs, including ER and HER2 assessment. Limited information is available on the concordance of CTC ER and HER2 levels compared to synchronously obtained tissue. We assessed the prognostic performance of CTC enumeration and phenotypic profiling of ER and HER2 on CTCs. Methods: The prospective, multi-institutional Aurora study enrolled pts with MBC of any subtype either at diagnosis of metastatic disease or at time of progression on any line of systemic therapy. Pts underwent a metastatic site biopsy, consented to archival primary tissue collection and donated blood samples serially every 3-4 months until progression. CTCs, from blood collected at study entry, were identified as nucleated, EpCAM+/ cytokeratin+/ CD45- cells using an automated imaging system (RareCyte, Seattle). ER and HER2 expression levels were measured by mean fluorescence intensity (MFI), with thresholds above 80 (ER) and 160 (HER2) considered positive. Comparisons were made to total RNAseq from paired metastatic tissue biopsies. PFS and OS were calculated from the time of study entry to last follow up or death. Results: Baseline CTC enumeration was performed in 154 pts. The most common clinical subtype was HR+/HER2- (59.7%), followed by HER2-positive (16.8%) and TNBC (15.5%). The median lines of prior therapy were 1 (range 0-10). At least 1 CTC was identified in 68/154 pts (44%), 30 (19%) had ≥ 5 CTCs, and 15 (9.7%) had CTC clusters. The median CTC value was 0 (0-877). The presence of CTCs (≥1 or ≥5) and CTC clusters was adversely associated with PFS and OS (Table 1). Across all subtypes, the median CTC ER-MFI across was 36.2 (range 4.2-10,016.6) and the median HER2-MFI was 139.6 (range 4.2-2480.5). We observed high intra-patient heterogeneity of ER-MFI (CV: 0.65 [0.34-1.25]) and HER2-MFI (CV: 0.79 [0.42-1.54]). Clinical metastatic subtype from metastasis at study entry was available in 54/68 pts with CTCs. Pts with clinical ER+ MBC had a higher median ER-MFI compared to clinical TNBC (111.9 vs 68.6, p = 0.02). However, 11 pts (26.8%) with clinical ER+ MBC had all CTCs with an ER-MFI <80. Lower ER expression was observed in CTCs collected after prior exposure to a SERD (median ER-MFI: post-SERD [n=8] 27 vs post-aromatase inhibitor [n=4] 252, p<2.22e-16). No correlations were observed between the median CTC ER-MFI or HER2-MFI and paired tissue ESR1 or ERBB2 RNA expression (n=66). Conclusions: CTCs are strongly predictive of PFS and OS. CTC ER- and HER2-MFI were highly heterogeneous without clear correlation with bulk tissue RNA expression. The clinical impact of discordant clinical receptor status and CTC receptor status warrants further exploration.
| Number | Median PFS | PFS HR (95% CI) | Median OS | OS HR (95% CI) | |
| Presence of CTC | |||||
| 0 CTC | 86 | 7.37 (0.03 – 43.1) | reference | 20.72 (0.03 – 50.6) | reference |
| ≥ 1 CTC | 68 | 4.7 (0.43 – 19.5) | 1.65 (1.16-2.36) | 12.04. (0.43-44.1) | 2.13 (1.44-3.16) |
| Presence of ≥ 5 CTC | |||||
| <5 CTC | 124 | 7.7 (0.03 – 43.1) | reference | 19.31 (0.03 – 50.6) | reference |
| ≥ 5 CTC | 30 | 2.88 (0.43 – 18.7) | 1.77 (1.15 – 2.71) | 5.74 (0.43 – 29.3) | 3.59 (2.26-5.70) |
| Presence of clusters | |||||
| 0 clusters | 139 | 7.37 (0.03 – 43.1) | reference | 18.06 (0.03 – 50.6) | reference |
| ≥ 1 cluster | 15 | 1.91 (0.43 – 18.7) | 2.06 (1.16 – 3.68) | 3.42 (0.43 – 18.7) | 5.44 (2.94-10.09) |
Presentation numberPS2-07-18
Prognostic effects of methylation-based HR and HER2 subtyping by liquid biopsy in ESR1 mutation-negative metastatic breast cancer
Pedram Razavi, Memorial Sloan Kettering Cancer Center, New York, NY
P. Razavi1, N. Zhang2, A. Hardin2, A. Das2, M. J. Ellis3; 1Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 2Real World Evidence, Guardant Health, Inc., Redwood City, CA, 3Clinical Development, Guardant Health, Inc., Redwood City, CA.
Background: Analysis of cell-free DNA (cfDNA) in blood has established clinical utility for the detection of somatic genomic alterations in cancer, but epigenetic signatures based on tumor-derived circulating methylated DNA is a less established approach. Here, we describe the application of a cfDNA methylation-based hormone receptor (HR) and HER2 breast cancer molecular subtype (BCMS) feature in clinically assigned HR+ HER2- MBC. Real word evidence (RWE) outcomes by epigenetic HR and HER2 status in ESR1-mutant cases will be reported at ESMO 2025. The aim of this study is to evaluate the prognostic properties of BCMS in patients with endocrine therapy (ET) resistance not associated with an ESR1 mutation. Methods: The GuardantINFORM™ RWE database includes de-identified administrative claims data from Guardant360® testing. Guardant360® integrates genomic and epigenomic components, including a methylation-based cfDNA BCMS feature to identify HR, HER2, and/or triple negative breast cancer (TNBC) status for patients with tumor fraction >0.5% with overall accuracy ranging from 85.73% to 91.88% (Tolkunov et al., AACR 2025). Included here were adult patients with aromatase inhibitor (AI)-treated MBC who received treatment after an ESR1 mutation (ESR1mut) was not detected. Clinical outcomes via “real-world time to treatment discontinuation” (rwTTD) and “real-world overall survival” (rwOS) were compared between patients assigned HR+/HER2- versus other categories (TNBC, HR-/HER2+, or HR+/HER2+). Propensity score matching was applied to balance baseline characteristics between study groups. Results: A total of 769 AI-exposed ESR1mut-negative clinically HR+/HER2- MBC cases had BCMS results available and no prior anti-HER2 therapy. Of these, 620 (80%) were HR+/HER2- by BCMS, while the remaining 20% comprised other subtypes (HR+/HER2+: n = 67, HR-/HER2+: n = 24, and HR-/HER2-: n = 63). Non-HR+/HER2- cases were grouped as there was a different subtype to that assigned clinically (HR+/HER2-). For patients who underwent ET only, rwTTD and rwOS were worse for cases not assigned HR+/HER2- (adjusted HR = 0.37, 95% CI 0.17-0.78, P = 0.009; adjusted HR = 0.18, 95% CI 0.42-0.77, P = 0.02). For patients who received ET plus CDK4/6 inhibition, rwTTD was worse (adjusted HR = 0.44, 95% CI 0.22-0.9, P = 0.024), and for those who received PIK3CA/AKT1 inhibition, there was no difference. Conclusions: cfDNA-based BCMS predicted RWE outcomes in previously AI treated patients receiving second-line therapy without ESR1 mutations. The subset categorized as not HR+/HER2- had significantly worse outcomes if assigned ET monotherapy. Additional RWE will be presented as well as a liquid biopsy-based genomic analysis of BCMS HER2+ cases that were classified as HER2-negative clinically. Epigenetic-based molecular breast subtyping may be a feasible, non-invasive clinical tool to reassess ER and HER2 status in MBC without requiring serial tissue biopsies.
Presentation numberPS2-07-20
Ki-67-guided stratification and resistance mechanism analysis of CDK4/6 inhibitor therapy following adaptiveneoadjuvant endocrine treatment in HR+/HER2− breast cancer
Yong-Sheng Wang, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
Y. Wang1, Z. Bi1, P. Qiu1, J. Zhang2, C. Zheng3, Y. Liu1, B. Cong1, X. Sun1, P. Chen1, C. Wang1; 1Department of Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, CHINA, 2Department of Breast Cancer Center, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA, 3Department of Breast Cancer Center, Weifang People’s Hospital, Weifang, CHINA.
Background: The absolute improvement of invasive disease-free survival with intensive adjuvant CDK4/6 inhibitor (CDK4/6i) therapy ranges from 4.9% to 7.6%. Given the associated adverse events (AEs) and economic implications, identifying patients who are most likely to benefit from CDK4/6i can help avoid under- or overtreatment.Short-term adaptive neoadjuvant endocrine therapy (NET) may facilitate the selection of patients who could benefit from intensified therapy. This study aims to utilize adaptive NET to guide patient selection for combination endocrine therapy with CDK4/6i and to further explore molecular mechanisms underlying resistance to CDK4/6i. Methods: Postmenopausal patients with stage II-III HR+/HER2- breast cancer and a baseline Ki-67 index >10% were prospectively enrolled in a multicenter clinical trial (Clinical trial.gov Identifier: NCT05809024). All patients received 2 weeks of NET with aromatase inhibitors (AIs), followed by a second tumor biopsy. Based on post-treatment Ki-67 levels (cut-off: 10%), patients were stratified into two groups: (A) AI-responsive (Ki-67 ≤10%), who proceeded directly to surgery; and (B) AI-nonresponsive (Ki-67 >10%), who received an additional cycle of AI combined with the CDK4/6i dalpiciclib prior to surgery. Tumor samples were collected at three time points—prior to NET, after AI therapy, and after AI plus CDK4/6i—for single-cell transcriptome sequencing and next-generation sequencing (NGS). Results: Between February 2023 and 2025, 120 patients were enrolled. After 2 weeks of AI therapy, 68.3% (82/120) achieved Ki-67 ≤10% (AI-responsive), while 31.7% (38/120) remained with Ki-67 >10% (AI-nonresponsive). Among the AI-nonresponsive patients, 50.0% (19/38) remained Ki-67>10% following one cycle of AI plus CDK4/6i (CDK4/6i-nonresponsive), while the remaining 50.0% achieved Ki-67 ≤10% (CDK4/6i-responsive). No new drug-related AEs were observed. Single-cell sequencing was conducted in 23 patients: AI-responsive (n=7), AI-nonresponsive (n=12), CDK4/6i-responsive (n=2), and CDK4/6i-nonresponsive (n=2). Seven distinct cell clusters were identified: B cells, CD8+T cells, endothelial cells, epithelial cells, fibroblasts, macrophages, and mast cells. Mast cells was significantly more abundant in the AI-nonresponsive group, and the malignant epithelial C2 cell cluster was notably enriched in the CDK4/6i-nonresponsive group. Bulk transcriptomic sequencing of 52 tumor samples revealed upregulation of FUT6, APOF, and SERPINA6 in the CDK4/6i-nonresponsive group, with FUT6 showing the most significant differential expression. NGS was performed on 61 patients. PIK3CA mutations were detected in 54.5% of patients before AI therapy, rising to 60.6% after AI therapy (72.7% in the AI-nonresponsive group; 53.8% in the AI-responsive group), and declining to 42.8% following AI plus CDK4/6i treatment (62.5% in the CDK4/6i-nonresponsive group; 16.6% in the CDK4/6i-responsive group). TP53 mutations were observed in 36.3% of patients before AI therapy, 42.6% after AI therapy, and 57.1% following AI plus CDK4/6i treatment (75.0% in the CDK4/6i-nonresponsive group; 33.3% in the CDK4/6i-responsive group). Conclusions: In patients initially nonresponsive to AI therapy, the addition of CDK4/6i significantly reduced Ki-67 levels, indicating that these individuals may benefit from intensified CDK4/6i-based therapy. The malignant epithelial C2 cell cluster and FUT6 were identified as potential therapeutic targets associated with CDK4/6i resistance. Moreover, persistent or emerging PIK3CA and TP53 mutations during adaptive therapy may serve as molecular indicators of resistance to sequential AI and CDK4/6i treatments.
Presentation numberPS2-07-21
Tumor Necrosis Factor-related Weak Inducer of Apoptosis (TWEAK) – A Potential Biomarker for Predicting Response and Long-Term Outcomes in Early Breast Cancer Patients
Kerstin Wimmer, Medical University Vienna, Vienna, Austria
K. Wimmer1, D. Hlauschek2, A. Rauch1, M. Sachet1, C. Ramos1, V. Gerakopulos1, G. Pfeiler3, C. Brunner4, G. Pristauz-Telsnigg5, G. Rinnerthaler6, A. Pichler7, F. Fitzal8, S. P. Gampenrieder9, G. Huber10, M. Seifert3, D. Egle11, M. Filipits12, R. Oehler1, C. Singer3, M. Gnant13, Austrian Breast & Colorectal Cancer Study Group; 1Department of General Surgery, Division of Visceral Surgery, Medical University Vienna, Vienna, AUSTRIA, 2ABCSG, Austrian Breast & Colorectal Cancer Study Group, Vienna, AUSTRIA, 3Department of Gynecology, Medical University Vienna, Vienna, AUSTRIA, 4Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, AUSTRIA, 5Department of Gynecology and Obstetrics, Medical University Graz, Graz, AUSTRIA, 6Division of Oncology, Department of Internal Medicine, Medical University Graz, Graz, AUSTRIA, 7Department of Hemato-Oncology, LKH Hochsteiermark-Leoben, Leoben, AUSTRIA, 8Department of General Surgery, Hanusch Hospital, Vienna, AUSTRIA, 9Department of Internal Medicine III with Hematology, Medical Oncology, Hemostaseology, Infectiology, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Salzburg, AUSTRIA, 10Brustzentrum Kärnten, Barmherzige Brüder Hospital, St.Veit/Glan, AUSTRIA, 11Department of Gynecology and Obstetrics, Medical University Innsbruck, Innsbruck, AUSTRIA, 12Center for Cancer Research, Medical University Vienna, Vienna, AUSTRIA, 13Comprehensive Cancer Center, Medical University Vienna, Vienna, AUSTRIA.
Background: Baseline plasma levels of Tumor Necrosis Factor-related Weak Inducer of Apoptosis (TWEAK), Vascular endothelial growth factor A (VEGF-A), Programmed cell death 1 ligand 2 (PD-L2), and Osteoprotegerin (OPG) were analyzed for their predictive value in women with early-stage HER2-negative breast cancer who received neoadjuvant chemotherapy or endocrine therapy, and were randomized 1:1 to additional MUC1-based immunotherapy (tecemotide, L-BLP25). Methods: Plasma levels of the biomarkers were assessed in 314 patients from the prospective, randomized, open-label, 2-arm phase-II ABCSG-34 trial before neoadjuvant treatment. Of these, 240 received chemotherapy and 74 endocrine treatment, with (n=160) or without (n=154) tecemotide, a mucin 1 (MUC1) antigen-specific peptide vaccine. Treatment response was evaluated based on residual cancer burden (RCB) classes (0 to 3) and pathologic complete response (pCR). Long-term outcome data on invasive disease-free survival (iDFS), overall survival (OS) and distant recurrence-free survival (DRFS) were available in 227 patients. Associations of log2-transformed biomarkers with response and with survival were evaluated using ordinal logistic regression and Cox models. A potential predictive role for immunotherapy benefit was tested with interaction terms. For biomarkers demonstrating predictive value, an optimal cut-off was determined using ordinal logistic regression. Results: In vaccinated patients, higher TWEAK levels at baseline correlated with worse RCB class (OR 0.66; 95% CI, 0.48-0.90). This association differed significantly from that observed in non-vaccinated patients (interaction p = 0.01), where baseline TWEAK levels were not associated with RCB classes (OR 1.13; 95% CI, 0.86-1.50).Similar, though non-significant, findings were observed for pCR. Among vaccinated patients, higher baseline TWEAK levels were associated with lower pCR-rates (OR 0.66; 95% CI, 0.41-1.06), while in non-vaccinated patients no such association was observed (OR 1.13; 95% CI, 0.78-1.63; interaction p = 0.08). Apart from TWEAK, no associations were found between treatment response and baseline plasma levels of VEGF-A, PD-L2, or OPG. Consequently, an optimal predictive cut-off for TWEAK (450 pg/ml) for RCB classes was determined based on the highest discrimination (=c-index). After a median follow-up of 7.2 years, 81 iDFS, 53 OS, and 62 DRFS events were documented. When considering TWEAK levels as a continuous variable in the COX model, results for long-term outcomes showed a similar pattern to what was observed for short-term response; however, the number of events was low, and interaction p-values did not reach statistical significance (iDFS: p = 0.63; OS: p = 0.26; DRFS: p = 0.14). Patients with low TWEAK (≤450 pg/ml) seemed to derive greater long-term benefit from immunotherapy: compared to non-vaccinated patients, those treated with tecemotide showed improved 7-year iDFS (72% vs. 58%, HR 0.61), OS (86% vs. 66%, HR 0.43), and DRFS (84% vs. 61%, HR 0.36). In contrast, among patients with high baseline TWEAK, survival differences between groups were minimal, not exceeding 5%. Conclusion: Low baseline TWEAK levels predicted significantly better pathological response measured by RCB to MUC1-based vaccination. Although long-term outcomes did not reach statistical significance, clinically meaningful benefits in iDFS, OS, and DRFS were seen in patients with low TWEAK baseline levels. These findings support TWEAK as a potential predictive biomarker for identifying patients likely to benefit from neoadjuvant immunotherapy. Further validation in larger cohorts and in different neoadjuvant settings including currently used combinations of chemo- and immune checkpoint inhibitor therapy is warranted.
Presentation numberPS2-07-22
Serial, multi-omic, multi-analyte, liquid biopsy monitoring of metastatic lobular breast cancer to detect clinically-relevant heterogeneity and evolution
Andi K. Cani, University of Michigan, Ann Arbor, MI
A. K. Cani1, E. M. Dolce1, E. P. Darga1, K. Hu2, C. Liu3, D. Robinson3, Y. Wu3, C. Paoletti1, G. D. Luker4, S. A. Tomlins3, A. M. Udager3, N. C. Henderson5, D. G. Thomas3, J. M. Rae1, C. M. Arul3, E. F. Cobain1, D. F. Hayes1; 1Internal Medicine, University of Michigan, Ann Arbor, MI, 2Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 3Pathology, University of Michigan, Ann Arbor, MI, 4Radiology, University of Michigan, Ann Arbor, MI, 5School of Public Health, University of Michigan, Ann Arbor, MI.
Precision and immuno-oncology therapies commonly rely on molecular biomarker information obtained from single tissue biopsies, largely missing tumor heterogeneity and evolution. CtDNA offers a minimally invasive, yet limited approach to mitigate tissue shortcomings in monitoring the evolving tumor clonal architecture since it is characterized by low tumor fraction and provides only an aggregate picture of the genomic landscape. We hypothesized that serial, single-cell genomic profiling of circulating tumor cells (CTCs) complements the molecular information obtained from tissue and ctDNA. Invasive lobular carcinoma (ILC) of the breast, a high CTC producing malignancy offers an ideal setting to study the contribution of CTCs. We analyzed 119 individual CTCs and 15 leukocyte controls, 26 ctDNAs, and 24 tissue samples over time from 15 CTC-positive patients with metastatic ILC undergoing various therapies. CTCs were enriched/isolated with the tandem CellSearch®/DEPArray™ system and genomically profiled to detect mutations, copy number alterations, and in a novel application, tumor mutation burden (TMB) and microsatellite instability (MSI) at the single cell level. Molecular findings were combined with clinical information. We confirmed the presence of high CTC levels in metastatic ILC, where almost 80% of patients had >5 CTC/7.5mL blood. Single-cell profiling had a 2.7% rate of artifactual mutation detection and a 16.5% combined rate of miscalling the zygosity of a heterozygous mutation compared to bulk sequencing as truth. CTC copy number alterations were concordant with tissue (R = 0.77) and even allowed detection of single-cell intragenic copy number differences at the single amplicon level. Only 20/36 (56%) targetable genomic alterations were shared in all three specimen types (tissue, ctDNA and CTC). The rest were shared in either 2 or 1 specimen types, except ctDNA, which had no exclusive alterations and had particularly low detection rate of copy number alterations. Adding CTC analysis to tissue, ctDNA or both, contributed 0.49 – 0.69 additional targetable alterations per patient. Interestingly, 1 patient with plentiful CTCs had no detectable ctDNA. Our novel CTC TMB analysis revealed that in only 2/5 patients that were TMB-high by any specimen type, there was agreement between all three specimen types. Notably, one of the other 3 patients was TMB-high in CTCs only, while another in CTC and ctDNA only, and the last in tissue and ctDNA, but not CTCs. Remarkably, TMB heterogeneity among individual CTCs was common, with 3/5 TMB-high patients having CTCs with greater than, and fewer than 10 mut/Mb. Data from one patient’s 6 tissue, 5 ctDNA, and 28 individual CTC samples over 7 timepoints from primary cancer to hospice revealed in unprecedented detail the heterogeneity and evolution in response to endocrine, chemo and immunotherapy pressures not appreciable in ctDNA and tissue. Another patient harbored CTCs of two different clones matching tissue from two different lesions, only one of whom was TMB-high and expressed higher cytokeratin levels. Another patient, displaying ERBB2 copy amplification and HER2 overexpression by IHC, who was treated with anti-HER2 therapy, later showed disappearance of the ERBB2 amplification in CTCs but not in ctDNA. Taken together, these data support the non-invasive monitoring of advanced lobular breast cancer patients by liquid biopsy that incorporates CTCs to complement tissue and ctDNA in informing resistance mechanisms and subsequent treatment approaches.
Presentation numberPS2-07-23
Predictive markers of distant recurrence from randomized chemotherapy regimens for hormone receptor-positive breast cancer patient in the PACS-01 trial.
FREDERIQUE PENAULT-LLORCA, Centre Jean Perrin, CLERMONT FERRAND, France
F. PENAULT-LLORCA1, A. LUSQUE2, T. FILLERON2, K. TRAN3, L. DU3, R. BAEHNER4, F. DALENC5, M. LACROIX-TRIKI6, T. BACHELOT7, F. ANDRE8, P. BOUCHER9, J. LEMONNIER9, F. SYMMANS3; 1Department of Biopathology, Centre Jean Perrin, CLERMONT FERRAND, FRANCE, 2Department of Biostatistics, Oncopole Claudius Regaud, TOULOUSE, FRANCE, 3Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, HOUSTON, TX, 4Department of Pathology, University of California, San Francisco,, CA, 5Department of Medical Oncology, Oncopole Claudius Regaud, TOULOUSE, FRANCE, 6Department of Pathology, Institut Gustave Roussy, VILLEJUIF, FRANCE, 7Department of Medical Oncology, Centre Leon Berard, Lyon, FRANCE, 8Department of Medical Oncology, Institut Gustave Roussy, VILLEJUIF, FRANCE, 9R&D, UNICANCER, PARIS, FRANCE.
Background. SETER/PR index measures transcription that is related to estrogen and progesterone receptors (ER/PR) and not proliferation. Low SETER/PR index (below 0.75) predicted benefit from: 1) dose-dense, versus conventionally dosed, anthracycline-paclitaxel chemotherapy in the CALGB-9741 trial, and 2) the addition of 8 weekly doses of paclitaxel to anthracycline-based chemotherapy in the GEICAM/9906 trial. Therefore, we independently tested whether the SETER/PR index could predict benefit from the addition of docetaxel to anthracycline-based chemotherapy in patients from the PACS-01 trial (Roche et al, JCO 2006, PMID: 17116941) who had node-positive, hormone receptor-positive (HR+) cancer and subsequently received adjuvant endocrine therapy. In PACS-01, patients were randomized to receive weekly cycles of fluorouracil, epirubicin and cyclophosphamide (FEC) for six 3-weekly cycles (6FEC), versus three cycles of FEC followed by three 3-weekly cycles of docetaxel (3FEC + 3D). Methods. SETER/PR index was measured in HR+ tumor samples with the QuantiGene Plex bead-based hybridization assay (ThermoFisher) using an aliquot of RNA that was residual from prior testing of Recurrence Score (RS) at Genomic Health. Pre-defined analyses included the continuous SETER/PR index and the cut point SETER/PR index 25, in patients who received adjuvant endocrine therapy after their chemotherapy. The primary endpoint was distant recurrence-free interval (DRFI). Interaction between chemotherapy regimens and each biomarker on DRFI was tested using a multivariable Cox proportional hazards model including the biomarker, the treatment arm and an interaction term between the treatment arm and biomarker. Results. There were 490 samples with evaluable SETER/PR index and RS from patients who received adjuvant endocrine therapy after their randomized chemotherapy treatment. There was a significant interaction between SETER/PR index as continuous score and treatment arm on DRFI (Pinteraction = 0.028), but not SETER/PR index dichotomized using cut point at 0.75 (Pinteraction = 0.668). Instead, the interaction was described by a cut point of SETER/PR index at 1.50 (Pinteraction = 0.013) and favored 6FEC when SETER/PR index ≥ 1.50 (HR 3.16, 95%CI 1.28-7.85), without significant difference between arms when SETER/PR index was below 1.50 (HR 0.83, 95%CI 0.51-1.35). Recurrence score did not predict outcomes between chemotherapy regimens using cut point RS at 25 (Pinteraction = 0.534) or as a continuous score (Pinteraction = 0.670). Exploratory analyses also illustrated a predictive value of SETER/PR index as continuous score and using cut-point at 1.50 in the subset of patients with 4 or more positive lymph nodes. Conclusions. SETER/PR index predicted between DRFI outcomes from an anthracycline-docetaxel regimen versus an anthracycline regimen without docetaxel, and the predictive cut point was different from previous studies that had assessed paclitaxel. Addition of docetaxel was less effective than continuing FEC treatments when breast cancer had high endocrine-related transcriptional activity (i.e., SETER/PR index ≥ 1.50). Overall, this provides additional new evidence that SETER/PR index values can predict which chemotherapy regimen might be more effective for a patient with HR+ breast cancer.
Presentation numberPS2-07-24
Circulating tumor DNA (ctDNA) biomarker analyses of a phase 1/2 study evaluating vepdegestrant, a PROteolysis TArgeting Chimera (PROTAC) estrogen receptor (ER) degrader, in ER-positive/human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer (aBC)
Seth A Wander, Harvard Medical School, Boston, MA
S. A. Wander1, H. S. Han2, E. P. Hamilton3, S. A. Hurvitz4, M. Lachowicz5, W. Wu6, J. Corradi6, A. Van Acker7, M. A. Dorso7, E. Duperret7, S. Lonning8, S. Dychter9, C. Ma10; 1Massachusetts General Hospital, Harvard Medical School, Boston, MA, 2Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, 3Breast Cancer Research Program, Sarah Cannon Research Institute, Nashville, TN, 4Department of Medicine, Fred Hutch Cancer Center, Seattle, WA, 5Clinical Research, Arvinas Operations, Inc., New Haven, CT, 6Platform – Biology, Arvinas Operations, Inc., New Haven, CT, 7Translational Sciences, Arvinas Operations, Inc., New Haven, CT, 8Biomarkers, Pfizer, Inc., San Diego, CA, 9Clinical Research, Pfizer, Inc., San Diego, CA, 10Section of Medical Oncology, Division of Oncology, Washington University School of Medicine, St. Louis, MO.
Background: Vepdegestrant, an oral PROTAC ER degrader, showed encouraging clinical activity and was well tolerated in a phase 1/2 study (NCT04072952) including patients (pts) with heavily pretreated ER+/HER2- aBC. In a subsequent phase 3 trial (VERITAC-2, NCT05654623), vepdegestrant demonstrated statistically significant and clinically meaningful improvement in progression-free survival vs fulvestrant among pts with previously treated ER+/HER2- aBC and ESR1 mutations (ESR1m). Here, we report exploratory ctDNA biomarker analyses from the phase 1/2 study evaluating vepdegestrant monotherapy.Methods: This multicenter, open-label study included a 3+3 dose escalation (phase 1; vepdegestrant doses: 30−700 mg daily) and dose expansion (phase 2; vepdegestrant 200 mg or 500 mg once daily). Eligible pts had ER+/HER2- metastatic, recurrent, or locally advanced unresectable breast cancer previously treated with ≥1 cyclin-dependent kinase 4/6 inhibitor and ≥2 (phase 1) or ≥1 (phase 2) endocrine regimens. Baseline (cycle [C] 1 day [D] 1) and on-treatment (C1D28) circulating-free DNA (cfDNA) samples from the majority of pts treated with vepdegestrant (all at doses ≥100 mg/day) were analyzed using F1LCDx from Foundation Medicine. For these exploratory analyses of the pooled phase 1/2 dataset, baseline and on-treatment changes in variant allele fraction (VAF) of ESR1-mutated ctDNA and tumor fraction (TF; percentage of cfDNA that originates from the tumor) were assessed in association with the clinical benefit rate (CBR; complete response, partial response, or stable disease for ≥24 weeks) and analyzed by Firth’s penalized logistic regression. Results: Among 154 pts treated with vepdegestrant, 138 had ctDNA samples analyzed with the F1LCDx platform (all had received vepdegestrant ≥100 mg/day). Of these, 81 pts (59%) had ESR1m (D538G 41%; Y537S 39%; Y537N 22%; E380Q 8%; L536P 6%; L536R 5%). When analyzed as single variables, ESR1m VAF and TF at baseline were not positively or negatively associated with CBR; among pts with ESR1m, mean (95% CI) baseline ESR1m VAF was 9.1% (5.7-12.4) in pts with clinical benefit (n=34) and 7.4% (2.6-12.1) in pts without clinical benefit (n=32) and mean baseline TF (95% CI) was 18.4% (12.0-24.8) and 22.2% (15.4-29.0), respectively. However, when baseline ESR1m VAF was normalized to baseline TF, a higher ESR1m VAF/TF ratio was significantly associated with better CBR; mean ESR1m VAF/TF (95% CI) of 0.59 (0.47-0.72) vs 0.35 (0.25-0.45) in pts with vs without clinical benefit (p=0.005). Robust reductions in ESR1m VAF were observed across Y537X, D538X and L536X variants of ESR1 after 1 cycle of treatment, with 87% of ESR1m alleles showing ≥50% reduction from baseline at C1D28. Robust reductions in TF were also observed, with 81.5% of pts with ESR1m demonstrating ≥50% reduction in TF from baseline at C1D28. Among pts with ESR1m, change in TF was significantly associated with clinical benefit; the median percentage change in TF from C1D1 to C1D28 was −99.2% in pts with clinical benefit (n=33) vs −50.4% in pts without clinical benefit (n=32; p<0.001). Conclusions: Patients with higher baseline ESR1m VAF/TF, a likely indicator of ESR1m clonality, had better clinical outcomes with vepdegestrant than pts with lower ESR1m VAF/TF. Greater decreases in ctDNA TF after 1 cycle of treatment were associated with increased likelihood of achieving clinical benefit. Overall, these exploratory biomarker analyses provide potentially clinically useful insights on pt responses to vepdegestrant and add to a growing body of evidence on the use of ctDNA in decisions around treatment continuation for pts with ER+/HER2- aBC.
Presentation numberPS2-07-25
Genomic biomarkers of response to ipatasertib in hormone receptor-positive, HER2-negative metastatic breast cancer
Zahra Bagheri, Harvard Medical School, Boston, MA
Z. Bagheri1, M. Lloyd2, G. Fell2, E. Scott2, J. Keenan2, L. Spring2, J. Shin2, S. Isakoff2, L. Ryan2, S. Padden2, E. Fisher2, A. Newton2, B. Moy2, A. Varkaris2, L. Ellisen2, D. Micalizzi2, D. Haber2, D. Juric2, A. Bardia2, S. Wander2; 1Department of Graduate Education, Harvard Medical School, Boston, MA, 2Cancer Center, Massachusetts General Hospital, Boston, MA.
Background: AKT inhibitor therapy is approved in combination with fulvestrant for patients with AKT1/PIK3CA/PTEN-mutant, HR+/HER2- metastatic breast cancer (MBC). Molecular factors which impact sensitivity to AKT inhibition are poorly defined. The TAKTIC trial tested the combination of ipatasertib with endocrine therapy, with or without palbociclib, for endocrine-resistant MBC (Wander et al., SABCS 2023). This study aimed to identify genomic biomarkers in circulating tumor DNA (ctDNA) that are associated with differences in response to ipatasertib combination therapy. Methods: TAKTIC was a phase Ib trial evaluating ipatasertib with fulvestrant or an aromatase inhibitor (Arms A/B), or fulvestrant plus palbociclib (Arm C) in patients with HR+/HER2– MBC with disease progression after ≥1 prior line of metastatic therapy. An exploratory end point was to identify genomic predictors of response/resistance to AKT inhibitor therapy via ctDNA sequencing. Progression free survival (PFS) on ipatasertib was compared in patients with versus without alterations detected in baseline liquid biopsy samples. Matched baseline and post-progression ctDNA sequencing was also examined for acquired mutations. Eligibility required baseline sequencing within 61 days prior to treatment initiation and post-progression sampling within 30 days pre- or any time post-progression. ctDNA was profiled via Guardant360; only oncogenic/likely oncogenic alterations were analyzed. PFS was estimated via the Kaplan–Meier method, and hazard ratios (HR) with 95% confidence intervals (CI) were estimated using Firth’s Cox models, adjusting for relevant covariates as permitted by sample size. McNemar’s test assessed mutation changes in paired baseline and post-progression ctDNA. Results: Of 77 TAKTIC patients, 39 were included for baseline biomarker analysis. This subgroup included 19 patients treated with doublet therapy (Arms A/B) and 20 with triplet therapy (Arm C). Patients were heavily pretreated (median of 3 prior therapies and 85% with prior CDK4/6i) consistent with the overall population. Median PFS was 5.8 months, comparable to 5.5 months in the overall population. Baseline amplifications in FGFR1 were detected in 13% of ctDNA (n=5/39) and associated with shorter PFS on ipatasertib (HR 5.21, 95% CI 1.68–14.37). Mutations in ERBB2 (n=4/39, 10%) and amplifications in EGFR (n=3/39, 8%) also correlated with worse PFS (HR 3.44, 95% CI 1.05–9.10; and HR 4.75, 95% CI 1.17–15.20, respectively). Based upon multivariate gene analysis combining PI3K pathway alterations, patients with PIK3CA, PTEN, and/or AKT1 mutations had significantly improved PFS on triplet therapy compared to those without (n=9/20; HR 0.21; 95% CI 0.05–0.68); however, no association was seen with doublet therapy or in the overall cohort. ESR1 mutations were detected in 31% of patients and associated with shorter PFS approaching statistical significance (n=12/39, HR 2.09, 95% CI 1.00–4.21). Paired baseline and post-progression ctDNA analysis (n=27) was constrained by limited sample size. FGFR1 amplifications emerged in 4 cases (15%) at progression but were absent at baseline. Further analyses are ongoing and will be presented at the meeting. Conclusions: Genomic biomarkers detected in ctDNA correlated with PFS on ipatasertib therapy in this heavily pretreated population with HR+/HER2- MBC. Activating alterations in FGFR1, ERBB2, EGFR, and ESR1 were linked with inferior PFS, and triplet therapy prolonged treatment benefit in patients with PI3K pathway mutant disease. These exploratory findings are hypothesis generating and require clinical and preclinical validation to inform personalized AKT inhibitor treatment.
Presentation numberPS2-07-26
Clinical performance of Signatera Genome assay for predicting recurrence in patients with breast cancer
Wassim McHayleh, AdventHealth Cancer Institute, Orlando, FL
W. McHayleh1, C. Scalise2, E. Kalashnikova3, B. Sridhar2, J. Feeney2, W. Tan2, A. Hsieh2, A. Butskova2, J. Ortega2, S. Velichko2, C. Palsuledesai2, H. Sethi2, G. Heilek2, J. McKenzie2, A. Rodriguez2, M. Liu2, M. George4; 1Hematology and Medical Oncology, AdventHealth Cancer Institute, Orlando, FL, 2Oncology, Natera, Inc., Austin, TX, 3Oncology, Natera, Inc., Orlando, TX, 4Breast Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ.
Background: Circulating tumor DNA (ctDNA) is a clinically validated biomarker in breast cancer for predicting the risk of recurrence, monitoring response to neoadjuvant treatment, and detecting molecular relapse after adjuvant therapy. Signatera™ is a tumor-informed, multiplex PCR-NGS ctDNA assay that may be developed from a backbone of whole-exome sequencing (WES) or whole-genome sequencing (WGS). Early clinical validation of Signatera Genome in a pan-cancer cohort demonstrated improved performance in certain clinical scenarios. In this study, we specifically assessed the performance of the Signatera Genome assay in the surveillance setting for breast cancer. Methods: A retrospective analysis was performed using clinically annotated residual patient samples from commercial testing with WES-based Signatera. Treatment decisions and cadence of ctDNA testing were at the provider’s discretion. Signatera Genome assays were designed, consisting of 64 high-quality tumor-specific variants, from matched tumor and normal whole genome sequencing data. These assays were then used to detect ctDNA in the corresponding patients’ plasma. ctDNA levels were quantified as mean tumor molecules per mL of plasma (MTM/mL). Longitudinal blood samples represented time points after surgery/definitive treatment until recurrence or the end of follow-up. The correlation between ctDNA status and distant recurrence-free survival (DRFS) was assessed using Cox regression analysis. Results: Clinical performance of the Signatera Genome assay was assessed in a real-world cohort of 228 patients with breast cancer [triple-negative breast cancer (TNBC), n=130; HR+/HER2-, n=85; and HER2+, n=13]. In a landmark analysis, patients with ctDNA-positivity within 3 months of surgery were significantly at a higher risk of distant disease and/or molecular recurrence (HR: 8.1, 95% CI: 3.5-8.6, P <0.001). With longitudinal testing, ctDNA-positivity at any time post-definitive treatment was associated with significantly worse DRFS (HR: 22.2, 95% CI: 11.0-44.7, P <0.0001). ctDNA detection in the surveillance setting preceded clinical recurrence as confirmed by imaging with a sensitivity and specificity of 100%. Upon analyzing ctDNA dynamics, patients with unfavorable ctDNA dynamics (persistently/converted to positive) were observed to have significantly shorter DRFS than those with favorable ctDNA dynamics (serially/converted to negative) (HR with Firth’s penalized likelihood hazard model: 97.8, 95% CI: 11.5-12768.9, P <0.001). Conclusions: These data indicate the robust clinical performance of the Signatera Genome in breast cancer for detecting recurrence after surgery or definitive treatment. Prospective clinical trials are warranted to establish the clinical utility of the assay for guiding treatment decisions for patients with molecular non-radiographic recurrence.
Presentation numberPS2-07-27
Qutils: open-source image-based infiltrating immune cell detection and outcomes in calgb 40502, calgb 40603, and calgb 40601 phase iii breast cancer clinical trials
Mark Vater, The Ohio State University, Columbus, OH
M. Vater1, R. Salgado2, E. Blige1, H. Lefebvre3, A. Strahan3, M. Gatti-Mays4, H. Rugo5, K. Ballman6, M. Watson7, Y. Wen8, R. Leon-Ferre9, K. Niazi10, W. Symmans11, L. Carey12, K. AbdulJabbar13, Z. Li3, D. Stover4; 1Biomedical Informatics, The Ohio State University, Columbus, OH, 2Pathology, Gasthuiszusters van Antwerpen Hospital and Ziekenhuis Netwerk Antwerpen Hospital, Antwerp, BELGIUM, 3Stefanie Spielman Comprehensive Breast Center, The Ohio State University, Columbus, OH, 4Internal Medicine, The Ohio State University, Columbus, OH, 5Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 6Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, 7Pathology and Immunology, Washington University, St. Louis, MO, 8Department of Medicine, University of Chicago, Chicago, IL, 9Medical Oncology, Mayo Clinic, Rochester, MN, 10Pathology, The Ohio State University, Columbus, OH, 11Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 12Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 13Case45, Case45, London, UNITED KINGDOM.
QuTILs: Open-Source Image-Based Infiltrating Immune Cell Detection and Outcomes in CALGB 40502, CALGB 40603, and CALGB 40601 Phase III Breast Cancer Clinical Trials Background: Stromal tumor infiltrating lymphocytes (sTILs) are associated with improved response to chemotherapy and better overall survival (OS) in triple-negative breast cancer (TNBC). Quantification of sTILs via hematoxylin and eosin (H&E) tumor slides offers a widely available method to define a simple, effective surrogate for host anti-tumor immunity. Digitization of H&E slides has rapidly gained acceptance for use in diagnostic pathology and research, opening the door for image-based computational approaches to enumerate sTILs, among other features. Methods: We describe QuTILs – a multilayer perceptron-based framework to detect/enumerate TILs trained on >700,000 cells via The Cancer Genome Atlas Program (TCGA) slide images, implemented in the widely used open-source QuPath digital pathology suite. QuTILs was applied to digital H&E images using standard computing in primary breast tumors from phase 3 breast cancer clinical trials – CALGB 40502 (testing), CALGB 40601 and 40603 (validation) – in total 1,729 total images from 962 unique patients. The aims of this investigation were: 1) to assess the relationship between TIL patterns with overall or event-free survival outcomes; 2) to benchmark diverse image-based characteristics of TIL abundance and spatial architecture; and 3) to evaluate the association between image-based TIL levels and RNA-seq-based immune deconvolution metrics (CIBERSORT, EPIC, quanTIseq). Results: QuTILs average processing time was 7.9 minutes for whole-slide total TIL count and area density on a standard laptop workstation. In CALGB 40502, there was positive but modest correlation between pathologist TILs and QuTILs total TILs %, higher in the TNBC subgroup of CALGB 40502 (Pearson r = 0.55). In Cox proportional hazards models, the CALGB 40502 TNBC cohort showed significant univariate association for total TILs % with increasing decile (higher total TIL%) associated with reduced hazard (hazard ratio [HR]: 0.712, 95% CI: 0.559-0.906; P = 0.006), which remained significant in multivariable model incorporating age, race, and treatment arm (HR: 0.713, 95% CI: 0.556-0.915; P = 0.008). In validation TNBC clinical trial CALGB 40603, marginal TILs % was significant in the univariable Cox model (HR: 0.786, 95% CI: 0.639-0.966). Evaluation of TILs % by QuTILs with RNAseq-based deconvolution metrics in cohort-stratified and deconvolution method-stratified multivariable models showed that decile total TILs % by QuTILs was shown to significantly predict outcome in the EPIC (HR: 0.668, 95% CI: 0.466-0.957) and quanTIseq (HR: 0.646, 95% CI: 0.446-0.936) sets, with a similar magnitude as in clinical Cox, suggesting that RNA-based deconvolution estimates may not supplement the prognostic power of image-based TIL estimates. Exploratory analyses of pre- versus post-neoadjuvant therapy H&E images in CALGB 40603 and CALGB 40601 revealed a significant decrease in QuTILs total TIL% in CALGB 40603 for all pairs (n = 118 pairs, pre-median=9.52%, post-median=6.33%; Wilcoxon signed rank P < 0.001) but no significant change in QuTILs total TIL% among all pairs in CALGB 40601 (n = 82 pairs, pre-median=15.92%, post-median=15.10%; P = 0.886). Conclusions: QuTILs provides a computationally efficient open-source workflow for sTIL identification from digital H&E images that can be deployed on a standard laptop. We present the first evaluation of digital sTIL enumeration in CALGB 40502, CALGB 40603, and CALGB 40601 phase III breast cancer clinical trials and demonstrate significant prognostic association with QuTIL total TIL % among TNBCs across studies.
Presentation numberPS2-07-28
Immune microenvironment features of germline mutation-associated breast tumors revealed by single-molecule imaging in the Breast Cancer Family Registry
Marni B. McClure, National Cancer Institute, Bethesda, MD
M. B. McClure1, L. Mangiante2, C. Weiss2, Z. Ma2, J. Koo2, E. M. John3, C. Curtis3, J. Caswell-Jin3, A. W. Kurian3; 1Medical Oncology, National Cancer Institute, Bethesda, MD, 2Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, 3Department of Medicine, Stanford School of Medicine, Stanford, CA.
Background: Single-cell spatial profiling enables detailed insights into the tumor microenvironment while maintaining the native tissue context. Germline pathogenic variants (PVs) are associated with decreased lymphocyte and increased tumor-associated macrophages from bulk transcriptomic data; however, this relationship has yet to be deconvoluted. To examine how spatial features vary by germline cancer predisposition, we performed single-molecule RNA imaging in a racially and ethnically diverse cohort with detailed genetic and clinical annotation. Methods: We performed single molecule RNA imaging on the Bruker CosMx platform of a cohort in the population-based Northern California Breast Cancer Family Registry (NC-BCFR) consisting of 222 samples from 144 women diagnosed 1995-2005 with >20 years of follow-up. We deployed an in-house machine learning pipeline to perform imaging-based cell segmentation and quantification of cell-level transcriptomic data for 6,519 genes across 1,083,345 cells. Normalization for each of 9 tissue microarrays and batch integration was followed by cell typing using well-established canonical markers and the reference-based SingleR algorithm. Spatial transcriptomics data were used to predict tumor HER2 and hormone receptor status (HR). Cell neighborhood was determined by scikit NearestNeighbors package with neighborhood number determined by finding the inflection point in an ElbowPlot analysis. Co-localization was defined with the colocation quotient analysis. We performed univariate and multivariate analyses to examine the correlation of germline PV status, race/ethnicity, age, tumor stage, and tumor subtype with proportion of each identified cell neighborhood. P-values were corrected for multiple hypothesis testing using Benjamini-Hochberg correction. Results: Of 144 NC-BCFR participants, 28 (19.4%) had at least one germline PV, with 11 BRCA1, 8 BRCA2, 3 PALB2, 2 ATM, and 1 FANCM PV represented. Tumor samples with germline PVs globally contained increased regulatory T cells and CD8+ T cell (Wilcox FDR=0.0059, 0.0070) and decreased fibroblast (Wilcoxon FDR=0.0085) populations compared to non-PV tumors. Cell neighborhood analysis defined 12 distinct microenvironmental compartments including including cancer core (4 distinct clusters), epithelial/fibrotic, epithelial-innate immune, stromal, adaptive immune, innate immune, fibrotic-immune depleted, and mixed (2 clusters). Tumor samples with germline PVs were enriched for neighborhood 0 (Wilcoxon FDR<.001), defined by adaptive B (19%) and T lymphocytes (36%), of which 30% were CD4+, 26% are CD8+, 34% regulatory T cells, and 9.6% NK cells. In a multivariate model that included clinical subtype, age, race/ethnicity, and stage as covariates, the association between germline pathogenic PV and proportion neighborhood 0 remained significant (multivariate p=0.0035). We observed an increase in co-localization of cancer epithelial cells to CD8+ T cells (Wilcoxon FDR= 0.0306) and macrophages (Wilcoxon FDR=0.0349) within tumors from germline PV carriers vs non-carriers. Conclusions: Dynamic interplay among tumor cells and the host immune system is captured by single molecule spatial transcriptomic profiling. In the diverse, population-based, genetically characterized NC-BCFR, we observed substantial differences in the adaptive tumor immune environment of germline PV carriers vs. non-carriers. Our results suggest that inherited genetic risk shapes the tumor-immune landscape, which may inform strategies to optimize germline-targeted therapies.
Presentation numberPS2-07-29
Digital Histology Models Predict Chemotherapy Benefit in Randomized Adjuvant Trials and Real-World Cohorts
Frederick Matthew Howard, University of Chicago, Chicago, IL
F. M. Howard1, S. Kochanny1, A. Li1, S. Hassan1, J. Dolezal2, E. M. Flores3, R. Medenwald4, C. Fan5, M. Watson6, L. McCart7, L. R. Teras8, C. Bodelon8, A. V. Patel8, W. F. Symmans9, D. Stover7, C. Perou5, M. Sullivan10, K. Yao4, A. T. Pearson1, D. Huo11; 1Medicine, University of Chicago, Chicago, IL, 2Medicine, Geisinger Cancer Institute, Danville, PA, 3Pathology, Ingalls Memorial Hospital, Harvey, IL, 4Surgery, NorthShore University HealthSystem, Evanston, IL, 5Genetics, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 6Medicine, University of Chicago, St Louis, MO, 7Medicine, Ohio State University, Columbus, OH, 8Population Science, American Cancer Society, Atlanta, GA, 9Pathology, MD Anderson Cancer Center, Houston, TX, 10Pathology, NorthShore University HealthSystem, Evanston, IL, 11Public Health, University of Chicago, Chicago, IL.
Background: Adjuvant trials have established the benefit of taxane and dose dense chemotherapy (CT) in high risk breast cancer. Digital pathology can provide biologic insights into CT sensitivity and better personalize treatment using data from completed trials. Methods: Digital H&E images were obtained from resection specimens from CALGB 9344 (anthracycline [AC] ± taxane [T]), CALGB 9741 (standard or dose dense, and sequential or concurrent AC-T), cases from American Cancer Society (ACS) Cancer Prevention Studies, and three Chicago area hospitals (University of Chicago, NorthShore, and Ingalls Memorial Hospital). Computational models previously trained to predict 61 distinct gene signatures directly from histology (all with correlation coefficient > 0.5 vs true signature) were applied to predict distant recurrence in CALGB 9344. The most accurate signature as measured by concordance index (C-index) was subsequently tested in other cohorts. The primary outcome was distant recurrence free interval (DRFI); breast cancer specific survival (BCSS) was reported in ACS as DRFI was not recorded. Cox models controlling for age, nodal status, tumor size, and estrogen / progesterone receptor status were used to estimate interaction of signature and CT benefit. Cox models incorporating these same clinical factors and signature predictions were fit in CALGB 9344 and subsequently applied to all cohorts to better identify groups with low / high recurrence risk. Results: This study includes 2,355 cases from CALGB 9344, 1,460 cases from CALGB 9741, 1,122 cases from ACS, and 2,292 cases from Chicago hospitals. CALGB 9344 and 9741 included more hormone receptor negative (~32%) and higher nodal burden (mean 5 involved) cases than the Chicago / ACS cohorts. A histology model for a “bronchioid” gene signature (originally designed to identify a favorable lung adenocarcinoma subtype but also prognostic in breast cancer), best predicted DRFI in CALGB 9344 (C-index 0.60) and performed well in CALGB 9741 (C-index 0.63), Chicago (C-index 0.69), and ACS cohorts (BCSS C-index 0.63). This histology signature outperformed OncotypeDX when available in the Chicago cohort (n = 983, DRFI C-index 0.69 vs 0.65). An explainability analysis demonstrated that tubule formation and low nuclear pleomorphism were associated with bronchioid signature prediction. Patients in the lowest risk tertile for the bronchioid signature did not benefit from the addition of taxane to adjuvant AC in CALGB 9344 (taxane HR in low risk tertile 1.13, 95% CI 0.81 – 1.59, HR in higher tertiles 0.74, 95% CI 0.62 – 0.88, interaction p = 0.04). Significant interaction also seen for distant recurrence free survival (p = 0.002) and overall survival (p = 0.01) in CALGB 9344. Similarly, higher signature tertiles predicted benefit from adjuvant CT (vs no CT) in the Chicago cohort (interaction p = 0.01 for DRFI). Dose dense CT (vs standard schedule) only improved DRFI in the higher signature risk tertiles (HR 0.73, 95% CI 0.53 – 1.00) in CALGB 9741, but interaction testing was not significant (p = 0.46). Cox models incorporating both clinical factors and bronchioid signature improved recurrence prediction (C-index = 0.65, 0.73, and 0.78 for DRFI in CALGB 9344, CALGB 9741, and Chicago, and 0.71 for BCSS in ACS). Low / high risk tertiles from this Cox model (with cutoffs defined from CALGB 9344) consistently identify patients with low (12.8%, 6.2%, 2.9%, 2.4% in CALGB 9344, 9741, Chicago, and ACS) and high (43.4%, 33.7%, 23.9%, 6.7%) 5-year distant recurrence rates (breast cancer survival events for ACS). Conclusion: Digital histology signatures predict adjuvant taxane / CT benefit and recurrence risk in trial and real-world cohorts and could be applied to personalize treatment. Digital pathology can provide biologic insights in datasets with limited tissue availability for genomic testing.
Presentation numberPS2-07-30
Structural Variants in Homologous Recombination Repair/DNA Damage Response Pathways as an Alternative Mechanism of Genomic Instability in Breast Cancer
Arnaud Da Cruz Paula, i3S Instituto de Investigação e Inovação em Saúde, Porto, Portugal
A. Da Cruz Paula1, D. Muldoon2, A. Gazzo2, M. Repetto3, C. Schwartz2, H. Dopeso2, D. Ross2, H. Wen2, H. Zhang2, L. Norton4, E. Brogi2, C. Bandlamudi2, B. Weigelt2, F. Pareja2; 1Cancer Signaling and Metabolism, i3S Instituto de Investigação e Inovação em Saúde, Porto, PORTUGAL, 2Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 3Department of Medicine, Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, NY, 4Department of Medicine, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: Alterations in homologous recombination repair (HRR) and DNA damage response (DDR) pathways drive genomic instability in a subset of breast cancers (BC), with associated therapeutic implications. While inactivating mutations in HRR/DDR genes are well established, the role of structural variants (SVs) disrupting these pathways remains to be determined. Here, through the analysis of a large cohort of BC, we sought to investigate SVs as a mechanism of HRR/DDR gene inactivation and to assess their contribution to genomic instability through homologous recombination deficiency (HRD) genomic scars and mutational signatures. Methods: We interrogated 9,481 BCs (n=4,376 primary; n=5,105 metastasis), previously subjected to clinical tumor/normal targeted sequencing (MSK-IMPACT), to identify SVs affecting the HRR/DDR core genes BRCA1, BRCA2, PALB2, ATM and CHEK2. Loss of heterozygosity of the wild-type allele (LOH) was assessed using FACETS, and the driver probability of fusion genes was evaluated using Oncofuse. Mutational signatures were inferred using SigMA and HRD status was assessed though evaluation of genomic scars by applying an algorithm tailored for MSK-IMPACT data. Results: We identified 58 BCs (n=21 primary; n=37 metastasis) harboring SVs affecting the HRD/DDR genes interrogated, including ATM (n=14), BRCA1 (n=14), BRCA2 (n=18), CHEK2 (n=6) and PALB2 (n=6). The SVs included fusion genes (n=34), deletions (n=16), duplications (n=14) and inversions (n=4). Most SVs (67%; 39/58) were out-of-frame, while 33% (19/58) were in-frame. Bi-allelic inactivation was identified in 55% (26/47) of evaluable cases, in the form of LOH (81%; 21/26) or as a second somatic mutation (19%; 5/26). Among fusion genes, half (17/34) had an Oncofuse driver probability >50%, suggesting that they were likely drivers. BCs with SVs in the HRR genes BRCA1, BRCA2 and PALB2 were classified as HRD in 55%, 82% and 83% of cases, respectively, and showed a dominant HRD-associated mutational signature 3 in 64%, 65% and 50% of cases, respectively. Conversely, BCs with DDR genes CHEK2 and ATM SVs were classified as non-HRD in 60% and 57% of cases, respectively, exhibiting predominantly clock (42% and 60%) and APOBEC (40% and 25%) mutational signatures. Conclusions: We demonstrate that although rare, SVs in core HRR/DDR genes represent an underrecognized mechanism of gene inactivation in a subset of BCs. Notably, their associated HRD genomic scars and mutational signatures mirror those observed in cases harboring inactivating mutations in those genes, supporting the functional relevance of these SVs in shaping HRR/DDR related processes. Our findings highlight alternative mechanisms driving genomic instability, deepen our understanding of the HRR/DDR pathways in BC, and broaden the potential eligibility for targeted therapies exploiting DNA repair deficiencies.
Presentation numberPS2-08-01
A predictive gene signature for benefit from dose-dense adjuvant chemotherapy in patients with high-risk early breast cancer: results from the PANTHER phase III trial
Dimitrios Salgkamis, Karolinska Institutet, Stockholm, Sweden
D. Salgkamis, M. Sarafidis, E. Sifakis, I. Zerdes, S. Agartz, J. Hartman, M. Hellström, T. Foukakis, J. Bergh, A. Matikas; Oncology-Pathology, Karolinska Institutet, Stockholm, SWEDEN.
Introduction: Dose dense adjuvant chemotherapy (DD-ACT) for high-risk breast cancer (BC) improves outcomes compared to standard interval treatment (SD-ACT) regardless of any clinicopathologic factor. SET2,3 is the only gene signature that predicted benefit from DD-ACT in the CALGB C9741 trial for patients with ER-positive BC with low endocrine activity, compared though with a suboptimal control of paclitaxel every three weeks. Based on our previous results, we hypothesized that proliferation and immune gene expression may identify patients benefiting from treatment escalation. Material and Methods: PANTHER (NCT00798070) is an international phase III trial which compared adjuvant sequential epirubicin/cyclophosphamide and docetaxel administered either every 2 (DD-ACT) or every 3 weeks (SD-ACT), with tailored dosing at the dose-dense schedule according to hematologic toxicity for patients with resected node positive or high risk node negative BC. Bulk RNA was extracted from tumor-rich FFPE surgical biospecimens from patients enrolled at Swedish sites and sequenced in the NovaSeq X Plus system. Mean expression was quantified for three predefined gene modules: a mitosis kinase score (MKS), an immune kinase score (IKS), and an immune activation score (IAS). MKS was independently combined with each of the two immune-related scores (IKS and IAS) to stratify patients into low and high activity groups. Primary endpoint of the trial was BC recurrence-free survival (BCRFS) and secondary endpoints were event-free (EFS), distant disease-free (DDFS) and overall survival (OS). The median follow-up time was 10.2 years. Results: RNA sequencing was conducted on 506 FFPE tumor samples, of which 481 were retained for downstream analysis following quality control. Multivariable analysis, adjusted for tumor size, lymph node status, grade, and BC subtype, identified a subgroup of patients (n=157, 32.6% of the cohort) who derived greater benefit from DD-ACT compared to SD-ACT (HR=0.29, 95% CI 0.14-0.59, p=0.001), while the remaining patients (n=322) did not benefit from DD-ACT (HR=1.22, 95% CI 0.74-2.01, p=0.443; pinteraction = 0.02). Results were consistent for all secondary endpoints. This DD-ACT sensitive group was characterized by tumors with low expression of both mitotic and immune kinase signaling genes, or by tumors with high mitotic activity and a cold immune microenvironment. Conclusions: We describe a predictive gene signature that identifies a subgroup of patients who derive greater benefit from DD-ACT compared to SD-ACT across all survival endpoints and BC subtypes, independent of clinicopathologic factors, when compared with an optimal control of docetaxel every three weeks. Finally, we aim to validate our findings in independent patient cohorts.
Presentation numberPS2-08-02
Comprehensive Characterization of PTEN loss by IHC and PTEN alteration by NGS in Metastatic HR-Positive, HER2-Negative Breast Cancer– An Exploratory Analysis of Biomarker Concordance and Co-Occurrence
Jennifer R Klemp, Caris Life Sciences, Irving, TX
R. L. Mahtani1, S. C. Smith2, S. M. Swain3, S. Wu4, J. R. Klemp5, M. Benrashid6, S. Puri7, T. Nicolaides8, J. Xiu4, M. Oberley9, M. Chaki7, G. Sledge10; 1Miami Caner Institute, Baptist Health South Florida, Plantation, FL, 2Pathology, VCU Massey Comprehensive Cancer Center, Richmond, VA, 3MedStar Health, Georetown University Medical Center, Washington DC, DC, 4Clinical and Translational Research, Caris Life Sciences, Irving, TX, 5Biopharma, Caris Life Sciences, Irving, TX, 6R & D and US Medical Affairs, AstraZeneca Pharnaceuticals LP, Gaithersburg, MD, 7US Medical Affairs, AstraZeneca Pharmaceuticals LP, Gaithersburg, MD, 8Precision Oncology Alliance, Caris Life Sciences, Irving, TX, 9Pathology & Genetics, Caris Life Sciences, Irving, TX, 10Medical Affairs, Caris Life Sciences, Irving, TX.
Introduction: PTEN, a key negative regulator of the PI3K/AKT signaling pathway, can get inactivated in hormone receptor positive (HR+), HER2-negative (HER2−) metastatic breast cancer (mBC). While genomic alterations can inactivate PTEN, a subset of tumors lose PTEN protein expression without any PTEN genomic alterations, resulting in a discordance between genomic and protein-based test results. The CAPItello-291 trial (NCT04305496) led to the approval of capivasertib (AKT inhibitor) in combination with fulvestrant in patients with HR+/HER2- mBC with alterations in PIK3CA, AKT1, and/or PTEN. Here, we report a prespecified exploratory analysis evaluating the concordance between PTEN loss of expression by IHC and its genomic alteration by NGS, as well as the respective co-occurrences with PIK3CA and AKT1 mutations. Methods: 2,716 breast tumors underwent comprehensive tumor profiling at Caris Life Sciences (Phoenix, AZ) between August 2024 to June 25, 2025. All tumors were identified as HR positive and HER2 negative by a combination of IHC and CISH based on ASCO/CAP guidelines. Gene mutations were determined by Whole Exome Sequencing. PTEN IHC was performed using 6H2.1 antibody and scored by board-certified pathologists; PTEN loss was defined as complete absence of staining (0+, 0%). Results: Among the 2,716 tumors, 196 (7.21%) demonstrated PTEN loss by IHC. The overall concordance between PTEN IHC loss and PTEN NGS genomic alterations was 92.9%. In the absence of a definitive reference standard, the positive percent agreement was 52%; negative percent agreement was 96%. Among the discordant cases, two distinct patterns were observed. In tumors with PTEN loss by IHC but wild-type (wt) by NGS (N=112), PIK3CA and AKT1 mutation rates were 36.6% (41/112) and 0.89% (1/112), respectively. Conversely, in tumors with intact PTEN by IHC but PTEN alterations (alt) by NGS (N=77), PIK3CA and AKT1 mutation rates were 49.4% (38/77) and 1.3% (1/77), respectively. Notably, 5.41% (70/1293) of tumors that were wild-type for PTEN, PIK3CA and AKT1 by NGS demonstrated PTEN loss by IHC suggesting PI3K pathway activation that would have been missed without IHC testing.Conclusion: To our knowledge, this is the largest study highlighting a substantial discordance between PTEN expression by IHC and genomic alterations by NGS in HR+/HER2− mBC. Our results underscore the necessity for a multimodal approach to patient selection for markers of treatment resistance and suggest that PTEN IHC may identify additional patients with PI3K/AKT pathway activation.
| Group NGS | PTEN-loss IHC | PTEN-intact IHC | Total | ||||||||||||||||||||||||
| PTEN wt | 112 (4.12%) | 2443 (89.9%) | 2555 | ||||||||||||||||||||||||
| PTEN alt | 84 (3.09%) | 77 (2.84%) | 161 | ||||||||||||||||||||||||
| PIK3CA and AKT1 and PTEN wt | 70 (5.41%) | 1223 (94.6%) | 1293 | ||||||||||||||||||||||||
| PIK3CAor AKT1 or PTEN alt | 126 (8.87%) | 1294 (91.1%) | 1420 | ||||||||||||||||||||||||
Presentation numberPS2-08-03
A multi-site evaluation of the performance of the MasSpec Pen for surgical margin analysis during breast cancer surgeries
Ruth Costa, Baylor College of Medicine, Houston, TX
K. Liebenberg, R. Costa, S. Bench, E. Ma, M. Hutchinson, F. Jackobs, J. Mardick, E. Silberfein, C. Hsu, A. Thompson, S. Carter, E. Bonefas, N. Chandandeep, J. Wang, J. Suliburk, L. Eberlin; Surgery, Baylor College of Medicine, Houston, TX.
A multi-site evaluation of the performance of the MasSpec Pen for surgical margin analysis during breast cancer surgeries Keziah E. Liebenberg, Rute A.P. Costa, Sarah Bench, Emily X. Ma, Mary L. Hutchinson, Faith Jackobs, Jacob I. Mardick, Eric J. Silberfein, Cary Hsu, Alastair Thompson, Stacey A. Carter, Elizabeth Bonefas, Chandandeep Nagi, Jing Wang, James W. Suliburk, Livia S. Eberlin Introduction Breast cancer is the second leading cause of cancer deaths among women worldwide. Breast-conserving surgery (BCS), the most common treatment option. Achieving negative margins is critical for prolonged disease-free survival, making accurate intraoperative margin assessment essential. Molecular technologies offer the opportunity to incorporate cancer-specific markers into clinical decision-making to improve cancer detection. Our team has developed the MasSpec Pen (MSPen), a handheld device connected to a mass spectrometer for rapid, gentle molecular analysis of human tissues. This technology, combined with statistical learning, enables tissue disease state classification. Here, we evaluate the performance of the MSPen for intraoperative breast cancer detection.Methods Banked normal breast, ductal carcinoma in situ (DCIS) and invasive ductal and lobular carcinomas, IDC and ILC respectively, were analyzed with the MSPen coupled to Mass Spectrometer. Following analysis, the tissue region analyzed was demarcated, and the samples snap-frozen, sectioned, H&E stained and evaluated by a pathologist. Classification models were built with logistic regression and the least absolute shrinkage and selector operator (LASSO) method to distinguish normal breast from breast cancer tissue. The MSPen system was adapted as a mobile cart for implementation and testing within the surgical workflows of two different hospitals, intraoperatively and in pathology. The classifiers were used to predict on data obtained from the intraoperative analyses and validated against final histopathological reports. Preliminary dataTo date, we have analyzed 478 banked human breast tissue samples using the MSPen, including normal breast (n=205), healthy lymph nodes (n=26), IDC (n=101), metastatic IDC in lymph nodes (n=44), DCIS (n=61), and ILC (n=41). The rich mass spectral data obtained revealed a high relative abundance of metabolites and lipid species previously identified as potentially diagnostic of breast cancer. Using this data, we built three statistical classifiers to differentiate: 1) normal breast from IDC, 2) normal breast from ILC, and 3) normal breast from cancer (IDC, ILC, and DCIS). The classifier distinguishing IDC from normal breast achieved an accuracy of 95.6% on a test set and 95.5% on a validation set. The ILC classifier reached 82.1% accuracy on a test set. The model distinguishing normal from cancer (IDC, ILC, and DCIS) achieved 87.5% accuracy on a test set and 84% on a validation set. Classifier refinements are ongoing to enhance performance. In a pilot clinical study, we demonstrated feasibility for ex vivo and in vivo tissue analysis using the MSPen in 29 breast surgeries. At Ben Taub site, we deployed a compact, mobile, battery-operated MSPen cart, which is easily transported to any operating room without requiring line power.We have successfully performed intraoperative in vivo and ex vivo analyses of breast tissues during BCS and mastectomies. The mobile cart enables real-time analysis of resected specimens alongside pathologists conducting margin assessments, enhancing integration between the MSPen system and clinical surgical workflows. Novel aspect he MSPen is being experimentally used for real-time surgical margin evaluation intraoperatively and in the pathology room at two hospitals.
Presentation numberPS2-08-04
Site specific Genomic and Immune Landscapes Underpinning in metastatic Male Breast Cancer
Dario Trapani, University of Milan, milan, Italy
D. Trapani1, S. Deshmukh2, S. Wu2, J. Xiu2, N. Lin3, G. Curigliano1, P. Spanheimer4, P. Advani5, P. Barreto Coelho6, M. Lustberg7, G. Sledge Jr2, S. Tolaney3, J. Leone3; 1department of oncology and hematology, University of Milan, milan, ITALY, 2Caris Life Sciences, Caris Life Sciences, Phoenix, AZ, 3breast oncology, Dana farber cancer institute, Boston, MA, 4medical oncology, University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, 5medical oncology, Mayo Clinic, Jacksonville, FL, 6medical oncology, University of Miami, Coral Gables, FL, 7breast oncology, Yale School of Medicine, Yale University, New Haven, CT.
Background: Male breast cancer (MaBC) is a rare malignancy, accounting for approximately 1% of all breast cancer cases. Our previous studies suggest that MaBC is biologically distinct from female breast cancer (FeBC), exhibiting a unique mutational profile and immune-genomic microenvironment. While the impact of these molecular alterations on site-specific metastases has been explored in FeBC, it remains poorly understood in MaBC. This gap limits accurate risk stratification and prediction of disease trajectory in male patients. In this study, we investigated site-specific genomic and immune alterations across metastatic sites in MaBC. Methods: 238 MaBC samples were tested by NGS (592, NextSeq; WES, NovaSeq) and WTS (NovaSeq; Caris Life Sciences, Phoenix, AZ). We performed analysis in hormone receptor-positive (HR+) and HER2-negative (HER2-) subtypes which are most common in male. HR+HER2- was defined as estrogen receptor (ER) or progesterone receptor (PR) positive and HER2 IHC 1+ or 2+ with negative chromogenic in situ hybridization (CISH) assay. Immune cells were estimated using WTS deconvolution (Quantiseq). Gene expression profiles were analyzed for T-cell inflamed score, interferon-gamma (IFN-gamma) score and MAPK Activation Score. We classified patients by site of biopsy/specimen site. Statistical significance was assessed with chi-square and Mann-Whitney U tests, p<0.05 is considered statistically significant. Results: Our cohort included 86 HR+HER2- MaBC samples biopsied from primary breast cancer (pBC) (n=39), bone (n=15), liver (n=8), lung (n=8), lymph nodes (n=7), and skin (n=9) mets. We report here only the data on statistically significant differences, but the overall landscape has been depicted across all defined-sites investigated. pBC showed a significantly lower prevalence of PIK3CA mutations (23.6% vs. 53.3%, p<0.05) compared to bone mets. ESR1 mutations were absent in pBC but present in 25% of liver and 25% of lung mets (all comparisons p<0.05). Immune profiling revealed reduced infiltration of M2 macrophages (4.3% vs. 6%, p<0.05) and neutrophil (2.2% vs. 6.9%, p<0.05) in pBC relative to bone Mets. Further, CD4+ T cells were lower in pBC compared to lymph nodes (0% vs. 2.4%, p<0.05). pBC had lower expression of immune checkpoint genes (PDCD1LG2, HAVCR2 and CD47) compared to other mets. pBC exhibited lower MAPK pathway activation score (-1.6 vs. 0.17, p=0.03) and a trend toward lower T cell-inflamed gene expression scores (-65 vs. 8, p=0.06) compared to bone mets. Bone mets had higher infiltration of M2 macrophage compared to lung (6% vs. 4.4%, p<0.05) and lymph node (6% vs. 4.5%, p<0.05). Neutrophil infiltration was elevated in bone versus liver (6.9% vs. 2%, p<0.05) but T regulatory cells were lower in bone compared to lymph nodes (1% vs. 2.4%, p<0.05). Bone showed reduced expression of CD47 (median TPM 50.6 vs. 109.9, p<0.05) compared to skin. Further bone had lower IFN-gamma scores (-0.41 vs. -0.17, p<0.05) compared to lymph node. Data on prior number and types of cancer treatment to give a context to the findings will be presented at the meeting. Conclusions: Our study highlights that each MaBC metastatic site harbors a unique genomic and immune landscape that can influence tumor behavior and potentially impact patient prognosis. Better understanding of metastatic site-specific molecular features in larger cohorts can help in the future research efforts and guiding treatment decisions.
Presentation numberPS2-08-05
Multimodal integration of real world genomic and clinical data for the prediction of brain metastasis development in breast cancer
Anton Safonov, Memorial Sloan Kettering Cancer Center, New York, NY
A. Safonov1, D. Smith2, S. Nandakumar3, L. Boe4, E. Ferraro5, J. Shen6, K. Tsai7, I. Khatri8, R. Kumar9, J. An10, J. Jee1, M. Robson1, N. Schultz11, N. Moss12, W. Chatila11, L. R. Pike13, P. Razavi1; 1Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, 3Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 4Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 5Early Drug Development, European Institute of Onoclogy, Milan, ITALY, 6Graduate Medical Education, Drexel University College of Medicine, Philadelphia, PA, 7Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 8Graduate Medical Education, Warren Alpert Medical School at Brown University, Providence, RI, 9Biochemistry and Molecular Biology, University of Miami, Coral Gables, FL, 10Global Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, NY, 11Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 12Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, 13Department of Radiation Oncologyology, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: Despite advances in metastatic breast cancer (MBC) treatment, brain metastases (BM) continue to pose a major challenge, driving significant morbidity and poor prognosis. In the absence of consensus screening strategies, BMs are typically diagnosed only after neurological sequelae have already developed. This underscores the urgent need for predictive tools to identify patients at risk for BM before clinical symptoms arise. Methods: This study included 3901 patients who underwent sequencing of primary tumor (n = 1874) or non-brain metastasis of samples (n = 2027) with MSK-IMPACT, a custom tumor-normal next generation sequencing assay. We developed lasso machine-learning models to predict onset and timing of BM in two different scenarios. First, we predicted time from to BM diagnosis from the initial biopsy confirming metastatic breast cancer. Second, we developed an “early-stage” model capturing time from curative breast surgery to BM onset. Both models exclusively incorporated clinicopathologic and genomic data available at time of initial metastatic disease and initial surgery, respectively. We developed validative gradient boosting machine (GBM) machine learning and Fine-Gray models, allowing for consideration of variable interactions and competing risks, respectively. Results: Our cohort included 529 BM events over a median follow-up of 50 mos. In metastatic samples, the lasso-based ML model effectively stratified the cohort into high, intermediate and low-risk groups: the high-risk group (25% of the cohort) exhibited a hazard ratio (HR) of 10.1 (95% C.I 6.83 – 17.23) while the intermediate group (50% of the cohort) exhibited an HR of 3.65 (95% C.I 2.30 – 5.78) relative to the low-risk group. The model was driven by several genomic features (functional variants in TP53, RB1, PTEN, MDM2) as well as several clinical features (low ER and PR percentage, number of metastatic sites, metastasis to lung, adrenal or distant lymph node, and pre-menopausal status). In early-stage samples, the ML model stratified the cohort into two groups, with the high-risk group exhibiting an HR of 5.7 (95% C.I 3.83 – 8.53) relative to the low-risk group. Feature importance in the early-stage setting was driven by stage, ER/PR status as well as TP53 mutation status. The predictive performance of these models, along with the relevance of these key variables, was preserved in the GBM and Fine-Gray models. Conclusions: Leveraging a large cohort of genomically profiled breast cancers, we developed a machine learning model that effectively stratifies patients by risk of developing brain metastasis. The most influential variables informing risk stratification were biologically and clinically plausible. For example, recurrent alterations in cell cycle regulation (RB1, TP53) have been linked to brain metastasis tropism in other cancer types and emerged as key features in our analysis. Our clinically actionable multimodal model allows for early identification of patients at risk for brain metastasis, enabling the study of personalized screening and intervention approaches to identify and intercept this debilitating complication.
Presentation numberPS2-08-06
Biomarker analysis of patients treated with abemaciclib rechallenge after progression on abemaciclib plus endocrine therapy from the Phase II AGAIN Study (WJOG14220B)<sup></sup>
Meiko Nishimura, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
M. Nishimura1, T. Kogawa2, K. Sugiyama3, Y. Ozaki1, Y. Tanabe4, C. Funasaka5, S. Kurozumi6, A. Yoshimura7, T. Iwasa8, H. Sakai9, M. Morita10, S. Watanabe11, T. Fujii12, M. Yamamoto13, Y. Tsuji14, M. Terada15, T. Ota16, S. Tokunaga17, M. Shimokawa18, T. Takano1; 1Department of Breast Medical Oncology, Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 2Division of Early Clinical Development for Cancer, Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 3Department of Medical Oncology, NHO Nagoya Medical Center, Nagoya, JAPAN, 4Department of Clinical Oncology, Toranomon Hospital, Tokyo, JAPAN, 5Department of Experimental therapeutics, National Cancer Center Hospital East, Kashiwa, JAPAN, 6Department of Breast Surgery, IUHW Narita Hospital, Narita, JAPAN, 7Department of Breast Oncology, Aichi Cancer Center, Nagoya, JAPAN, 8Department of Medical Oncology, Kindai University Faculty of Medicine, osakasayama, JAPAN, 9Advanced Cancer Translational Research Institute, Showa Medical University, Tokyo, JAPAN, 10Department of Medical Oncology, Hyogo Cancer Center, Akashi, JAPAN, 11Department of Medical Oncology, Kishiwada City Hospital, Kishiwada, JAPAN, 12Department of General Surgical Science, Graduate School of Medicine, Gunma University, Maebashi, JAPAN, 13Department of Breast Oncology, National Hospital Organization, Hokkaido Cancer Center, Sapporo, JAPAN, 14Department of Medical Oncology, Tonan Hospital, Sapporo, JAPAN, 15Department of Breast Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, JAPAN, 16Department of Breast Medical Oncology, Izumi City General Hospital, Izumi, JAPAN, 17Department of Medical Oncology, Osaka City General Hospital, Osaka, JAPAN, 18Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Ube, JAPAN.
Background: We previously conducted a phase II study evaluating abemaciclib (ABE) rechallenge after progression on ABE plus endocrine therapy in patients with hormone receptor (HR)-positive, HER2-negative metastatic breast cancer. This study demonstrated a progression-free survival (PFS) benefit for patients treated with ABE plus fulvestrant (FUL) following progression on ABE plus aromatase inhibitor (AI)/tamoxifen (TAM). However, there are currently no robust biomarkers to identify patients likely to benefit from ABE rechallenge. This study presents the results of a biomarker analysis of liquid biopsies, including those collected at the time of disease progression after ABE rechallenge.Methods: We obtained liquid biopsies before ABE rechallenge, at 2 months (m) after rechallenge, and at disease progression to identify biomarkers associated with response or resistance to ABE. We analyzed circulating tumor DNA (ctDNA) using a targeted next-generation sequencing panel.Results: By the end of the study period, 54 of 64 patients had discontinued treatment due to disease progression, and liquid biopsies were obtained at progression. The most frequently acquired gene alterations at progression were TP53 mutations (n = 7, 13.0%) and CCND1 amplification (n = 5, 9.3%). Median overall survival (OS) in patients with TP53 mutations at progression was 18.8 m (95% confidence interval [CI], 10.8–not applicable [NA]) compared to 28.6 m (95% CI, 25.3–NA) in those with wild-type TP53. Moreover, TP53 mutations at progression were associated with shorter OS (hazard ratio [HR], 0.24; 95% CI, 0.07–0.8; p = 0.02). Among patients who switched from ABE plus AI/TAM to ABE plus FUL (n = 27), the most frequently acquired alternation was CCND1 amplification (n = 4, 14.8%), which was associated with shorter median PFS (HR, 0.19; 95% CI, 0.05–0.7; p = 0.01) and OS (HR, 0.16; 95% CI, 0.03–0.8; p = 0.02). In patients who switched from ABE plus FUL to ABE plus AI (n = 37), TP53 mutation was the most common newly acquired alteration at progression (n = 6, 16.2%).Conclusions: In this study of ABE rechallenge, TP53 mutation at disease progression may be associated with poorer survival outcomes. CCND1 amplification may be associated with resistance to ABE in patients who switched from ABE plus AI/TAM to ABE plus FUL.
Presentation numberPS2-08-07
Differences in Genomic Landscape Between Younger and Older Women with Advanced Breast Cancer
Nicole Casasanta, Yale Cancer Center, Yale School of Medicine, New Haven, CT
N. Casasanta1, E. S. Sokol2, S. Rengarajan3, L. Pusztai1, J. S. Ross4, M. B. Lustberg1; 1Department of Medicine, Division of Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Computational Discovery, Foundation Medicine, Boton, MA, 3Computational Discovery, Foundation Medicine, Boston, MA, 4Computational Discovery, Foundation Medicine, New Haven, CT.
Background: Despite underrepresentation in clinical trials, the treatment of premenopausal breast cancer is extrapolated from trials focusing on postmenopausal women. The same benefit may not translate given differences in tumor biology, grade, and need for ovarian function suppression. In both younger and older breast cancer patients, there is a need to enhance our understanding of predictive biomarkers for response to targeted therapies. The goal of this study was to evaluate genomic differences between women 60 years of age with advanced breast cancer.Methods: FFPE blocks of 1,144 advanced breast cancer cases among women 60 were analyzed by hybrid capture-based comprehensive genomic profiling that evaluated broad types of genomic alterations (GA) including mutations, insertions/deletions, amplifications, homozygous deletions, rearrangements, and fusions. MSI-high (MSI-H) status, tumor mutational burden (TMB), and homologous recombination deficiency signature (HRDsig) were determined from sequencing data. PD-L1 CPS was determined by IHC (Ventana SP142 assay, % of immunocyte/inflammatory cell staining). GA were compared using Fisher’s exact test with the Benjamini-Hochberg multiplicity adjustment.Results: In hormone receptor positive (HR+) and triple negative breast cancer (TNBC) cohorts, non-European genomic ancestry was more prevalent in women 60 (HR+ 38.87% vs 15.41% p=<0.001; TNBC 42.14% vs 22.73%, p=0.004). In HR+ and TNBC cohorts, CDH1 GA were more frequent in those >60 than <50 (HR+ 22.41% vs 8.2% p=0.002; TNBC 10.4% vs. 2.41% p=0.09). In the HR+ group, ESR1 GA were more common in the >60 group vs <50 (18.64% vs 8.98% p=0.06). In the TNBC group, PIK3CA GA were significantly more common in >60 than <50 (31.68% vs 10.84% p=0.007). In HR+ cancer, PIK3CA GA were also more frequent in older women (>60 49.34% vs <50 36.33%, p=0.1). BRCA1 and BRCA2 GA were more common in 60 in the HR+ cohort (BRCA1 3.52% vs 0.56%, p=0.07; BRCA2 9.38% vs 3.01% p=0.03). In the TNBC group, BRCA1 GA were more common in 60 (15.66% vs 4.95% p=0.06). HRDsig positivity was significantly more common 60 (TNBC 45.68% vs 23.74% p=0.002; HR+ 15.54% vs 6.24% p=0.00036). Younger patients were more frequently PD-L1 positive compared to older patients (TNBC 73.39% vs 57.04%, p=0.2; HR+ 40.14% vs 30.18% p=0.1). TMB ≥ 10 mut/Mb was more common in women >60 vs < 50 (TNBC 11.68% vs 2.47% p=0.002; HR+ 8.22% vs 4.15% p=0.06).Conclusions: Older women were more likely to have targetable GA including ESR1, PIK3CA, and TMB ≥ 10 mut/Mb. Higher prevalence of ESR1 GA in older women may reflect longer history of endocrine therapy use. The prevalence of PIK3CA alterations in older women with TNBC demonstrates a potential area for therapeutic investigation. Younger women had a higher prevalence of BRCA1 and BRCA2 alterations, likely reflecting germline alterations and benefit of PARP inhibitors.
| Alteration | HR+ <50 (n=256) | HR+ >60 (n=531) | P-value | Odds Ratio | Alteration | TNBC <50 (n=166) | TNBC >60 (n=202) | P-value | Odds Ratio |
| CDH1 | 8.2% | 22.41% | 0.002 | 0.37 | CDH1 | 2.41% | 10.4% | 0.09 | 0.23 |
| IGF1R | 4.69% | 0.56% | 0.01 | 8.3 | PIK3CA | 10.84% | 31.68% | 0.007 | 0.34 |
| BRCA2 | 9.38% | 3.01% | 0.03 | 3.11 | TERT | 0% | 3.96% | 0.1 | 0 |
| ESR1 | 8.98% | 18.64% | 0.06 | 0.48 | NTRK1 | 7.23% | 1.49% | 0.1 | 4.87 |
| BRCA1 | 3.52% | 0.56% | 0.07 | 6.22 | BRCA1 | 15.66% | 4.95% | 0.06 | 3.16 |
| TP53 | 39.84% | 27.5% | 0.1 | 1.45 | TP53 | 93.98% | 85.64% | 0.7 | 1.1 |
| PIK3CA | 36.33% | 49.34% | 0.1 | 0.74 | ERBB3 | 0% | 3.47% | 0.1 | 0 |
| Multiple PIK3CA | 15.05% (14/93) | 19.47% (51/262) | – | – | Multiple PIK3CA | 5.55% (1/18) | 10.94% (7/64) | – | – |
Presentation numberPS2-08-08
Comparison of anthropometric, systemic and mammary adiposity measures in patients with breast cancer.
Josephine Van Cauwenberge, Catholic University of Leuven, Leuven, Belgium
J. Van Cauwenberge1, I. Bachir2, F. Richard1, M. Maetens1, T. Geukens3, K. Van Baelen1, M. De Schepper4, G. Zels1, H. Nguyen5, D. Lambrechts6, E. Vanderheyden6, T. Van Brussel6, I. Nevelsteen7, A. Smeets7, H. Wildiers3, P. Neven8, G. Floris4, C. Desmedt1; 1Laboratory of Translational Breast Cancer Research, Department of Oncology, Catholic University of Leuven, Leuven, BELGIUM, 2Department of Anesthesiology, Université Libre de Bruxelles, Brussels, BELGIUM, 3Department of General Medical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 4Pathology, University Hospitals Leuven, Leuven, BELGIUM, 5Laboratory of Translational Breast Cancer Research, Catholic University of Leuven, Leuven, BELGIUM, 6Laboratory of Translational Genetics, Department of Human Genetics, VIB-KU Leuven, Leuven, BELGIUM, 7Department of Surgical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 8Department of Gynecological Oncology, University Hospitals Leuven, Leuven, BELGIUM.
Introduction: The obesity-breast cancer (BC) connection is increasingly recognized for its clinical relevance, especially as the proportion of women with BC and overweight/obesity continues to rise. While use of body mass index (BMI) as an adiposity measure is convenient, it is unclear how it relates to other anthropometric, systemic and local adiposity measures as well as with various endocrine, metabolic and inflammatory measures in patients with BC. To explore these complex associations, we launched the prospective FATLAS study (NCT04200768). Methods: FATLAS is a monocentric study set-up at University Hospitals Leuven enrolling cancer treatment naïve patients with BC scheduled for upfront surgery for whom tissue and blood samples are collected. Global adiposity is assessed via body fat percentage, visceral fat (TANITA RD-545), waist-to-hip ratio (WHR) and BMI. Local adiposity is evaluated by the median size of cancer-associated adipocytes (CAA) and distant mammary adipocytes (DA) in the adjacent normal breast tissue (HALO v3.6.4134). Inflammation markers include global (CRP, β-globulin fraction and WBC, eosinophil and lymphocyte count) and local markers (Crown-like Structures (CLS) density, stromal tumor infiltrating lymphocytes (sTIL), IL6, IL18 mRNA expression). Metabolic dysregulation is assessed using global (HbA1c, leptin, HDL, LDL, IGF1 levels, fasting C-peptide and insulin, metabolic syndrome score) and local markers (IGF1, IGF1R and leptin mRNA expression). Correlations were assessed using Spearman coefficients. Results: We included 96 women with ER+/HER2- BC (62% with no special type BC (NST) and 38% with lobular breast cancer (ILC)) of which 41% with normal-weight, 39% with overweight and 20% with obesity. 33% were diagnosed at stage I, 61% at stage II and 5% at stage III. 60% of patients were postmenopausal. Mean age at diagnosis was 57 years. Both body fat percentage and visceral fat showed a strong correlation with BMI (ρ= 0.82, ρ=0.82), but not with WHR (ρ=0.32 and ρ=0.27). Results were consistent across pre/peri- and postmenopausal patients. Adipocyte size increased with BMI in both DA and CAA (ρ=0.37, ρ=0.52) with a more pronounced trend in CAA (Kruskal-Wallis 0.001 and p=0.002 respectively). CAA were consistently smaller than DA across all BMI categories (p<0.05) . The association between adiposity and inflammation was limited when considering all patients, but we did observe significant correlations between global adiposity and CRP (e.g. BMI ρ=0.53). In pre/perimenopausal patients, global adiposity (BMI) correlated stronger with several systemic (e.g. neutrophil count ρ=0.56) and local inflammation markers (IL6: ρ=0.30 and IL18: ρ=0.40). Regarding local adiposity, DA correlated stronger with several systemic inflammation markers (e.g. CRP ρ=0.43, CAA ρ=0.34), while CAA correlated stronger with local inflammation markers (e.g. IL6 CAA: ρ=0.51; DA: ρ=0.25). In postmenopausal patients, all global adiposity markers – except WHR – correlated only with CRP (e.g. BMI: ρ=0.53), while significant correlations were observed between local marker CAA and local inflammation (CLS: ρ=0.48; sTIL: ρ=-0.30) and systemic CRP (ρ=0.38). Regarding metabolic dysregulation, BMI correlated with several systemic (e.g. circulating leptin ρ=0.80) and local markers (e.g. mammary leptin levels ρ=0.52), with this observation being more pronounced in premenopausal patients (ρ=0.84 and ρ=0.69). Conclusion: BMI correlates with body and visceral fat but less with mammary adipocyte size. It remains associated with inflammation and metabolic dysregulation, particularly in pre/perimenopausal patients.
Presentation numberPS2-08-09
Baseline HER2 expression on circulating tumor cells in metastatic breast cancer
Brenno Pastò, University of Udine, Udine, Italy
B. Pastò1, G. Guimarães2, E. Molteni1, N. Bayou2, M. Serafini2, E. Nicolò2, L. Pontolillo2, K. L. Eng2, E. Andreopoulou2, O. Elemento3, L. Gerratana1, F. Puglisi1, C. Reduzzi2, M. Cristofanilli2; 1Department of Medicine (DMED), University of Udine, Udine, ITALY, 2Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY, 3Institute of Computational Biomedicine, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY.
Background: The predictive role of human epidermal growth factor receptor 2 (HER2) in metastatic breast cancer (mBC) is well established. Recently, trastuzumab deruxtecan (T-DXd) has shown efficacy even in tumors with low HER2 expression, prompting a reassessment of how HER2 levels are evaluated. Circulating tumor cells (CTCs) can provide a more granular, non-invasive and real-time assessment of HER2 status. In this study, we aim to investigate baseline (BL) HER2 expression on CTCs and test its potential impact on treatment response and survival in a real-world cohort of patients (pts) treated with trastuzumab-based therapy (antiHER2) or T-DXd. Methods: Pts with mBC who received either antiHER2 or T-DXd and underwent CTCs enumeration and HER2 staining with CellSearch® before treatment started were eligible for analysis. BL counts of total and HER2-positive (HER2+) CTCs were recorded and CTC status was stratified accordingly into indolent (ind; < 5 total CTCs), aggressive-HER2-negative (agg-HER2–; ≥ 5 CTCs, no HER2+) and aggressive-HER2+ (agg-HER2+; ≥ 5 CTCs, ≥ 1 HER2+). The HER2+/total CTC ratio was also calculated and dichotomized based on the overall median (high vs low ratio). Differences in median values by treatment type were assessed using the Mann-Whitney U test. Associations with clinical benefit rate (CBR, dichotomized for this analysis) and progression-free survival (PFS) were evaluated using logistic regression and Kaplan-Meier curves with log-rank test, respectively. Results: A total of 23 pts were included: 16 (69.6%) had de novo mBC and 14 (60.9%) were HER2+ on tissue biopsy. Median follow-up from diagnosis of stage IV disease was 17.1 months. From September 2023 to February 2025, 19 anti-HER2 and 11 T-DXd treatments were initiated (6 pts received > 1 treatment). Of these, 10 were administered in the first-line setting (all antiHER2), 8 in the second-line (2 antiHER2, 6 T-DXd) and 12 in third or later lines (7 antiHER2, 5 T-DXd). AntiHER2 was administered in combination in 18/19 cases (94.7%), mostly with chemotherapy (12/19, 63.2%). BL CTC status was classified as ind in 12 occasions (40.0%), while agg-HER2– and agg-HER2+ phenotypes were observed in 9 cases (30.0%) each. No significant differences were documented in BL median counts of total or HER2+ CTCs between antiHER2 and T-DXd. The median HER2+/total CTC ratio was 0.13% (interquartile range, 0-42.9%) overall, with no significant differences between treatment groups. The overall CBR was 50.0%, with rates of 57.9% for antiHER2 and 36.4% for T-DXd. Compared to ind CTC status, the agg-HER2– phenotype was associated with a significantly lower likelihood of clinical benefit (OR 0.09, p = 0.046), while a higher HER2+/total CTC ratio was strongly linked to increased odds of clinical benefit (OR 28, p = 0.012), regardless of treatment received. Interestingly, although CTC count was not associated with PFS, agg-HER2+ and ind status showed similar and significantly better outcomes than agg-HER2–, both in the overall cohort (log-rank p = 0.027) and in the antiHER2 subgroup (log-rank p = 0.002), but not for T-DXd. Similarly, a higher HER2+/total CTC ratio was significantly associated with improved PFS, both overall and in the antiHER2 group (log-rank p = 0.010 and 0.001, respectively). Conclusions: Agg-HER2+ CTC phenotype and a higher HER2+/total CTC ratio were both associated with greater benefit from antiHER2, remarking the predictive role of HER2 and suggesting validity of CTC-based HER2 assessment. Further research is warranted to identify reliable predictors of response to T-DXd, which may require alternative cutoffs or distinct metrics to more accurately reflect HER2 expression levels. Despite limitations due to the sample size, these preliminary findings provide a rationale for developing CTC-based scoring systems to guide treatment selection with HER2-targeted agents.
Presentation numberPS2-08-10
Peripheral immune profiling during neoadjuvant HER2-targeted therapy in HER2-positive (HER2+) early breast cancer (EBC): Mass cytometry (CyTOF) analysis from the MARGOT trial
Elia Segui, Dana-Farber Cancer Institute, Boston, MA
E. Segui1, H. Heiling2, J. Baginska1, A. N. Nau2, I. Gomez Diaz3, J. Huezo3, P. R. Pohlmann4, M. F. Rimawi5, N. F. Sinclair6, R. L. Yung7, N. U. Lin1, A. C. Wolff8, M. K. DeMeo1, A. Root9, T. Rahman1, A. Mohammed-Abreu1, M. DiLullo1, A. Patel1, E. R. Ogayo10, T. A. King10, E. P. Winer11, E. A. Mittendorf10, N. Tayob2, S. Tolaney1, I. E. Krop11, J. Agudo1, A. G. Waks1; 1Department of Medical Oncology, Breast Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Data Science, Dana-Farber Cancer Institute, Boston, MA, 3Department of Medical Oncology, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, 4Breast Medical Oncology, UT MDACC, Houston, TX, 5Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, 6Department of Medical Oncology, Breast Oncology, Dana-Farber Brigham Cancer Center – Foxborough and Milford, Foxborough, MA, 7Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 8Johns Hopkins Kimmel Comprehensive Cancer Center, Breast & Gyn Malignancies Group, Johns Hopkins School of Medicine, Baltimore, MD, 9., Drexel University College of Medicine, Philadelphia, PA, 10Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, 11Medical Oncology, Yale Cancer Center, New Haven, CT.
Background: Anti-tumor immune activity impacts outcomes in HER2+ EBC, and anti-HER2 antibodies act in part through immune mechanisms such as antibody-dependent cell-mediated cytotoxicity (ADCC). Margetuximab is an anti-HER2 antibody engineered to enhance ADCC through increased affinity for FcγRIIIa. Early changes in circulating immune populations during anti-HER2 therapy remain poorly characterized. We used CyTOF to profile peripheral blood mononuclear cells (PBMCs) at baseline and early on-treatment in patients receiving neoadjuvant chemotherapy plus HER2-directed therapy. Methods: The investigator-initiated MARGOT trial (NCT04425018/TBCRC052) randomized 171 patients with stage II-III HER2+ EBC 1:2 to receive neoadjuvant THP (trastuzumab/pertuzumab/paclitaxel) or TMP (margetuximab/pertuzumab/paclitaxel). The primary endpoint, pathologic complete response (pCR), was previously reported; pCR was numerically higher with TMP but not statistically significant. This PBMC sub-study included all patients with paired PBMCs at baseline and cycle 2 day 1 (C2D1; 3 weeks on-treatment) from sites meeting quality criteria before the data cut-off. A total of 91 patients (THP n=25, TMP n=66) underwent CyTOF profiling at both timepoints using 22 surface and 13 functional markers, identifying 26 immune cell populations. Descriptive statistics were used to summarize baseline and on-treatment cell type changes (Δ). Associations with pCR were evaluated using logistic regression; on-treatment changes by treatment arm were analyzed using linear regression. Multiple testing was addressed using false discovery rate (q-values); raw p-values <0.05 were considered of interest. We hypothesized that ADCC effector populations (e.g., NK cells, monocytes) would change more with margetuximab, and that evidence of ADCC would correlate with response. Results: For the 91 patients (56 HR+, 35 HR-), pCR rates were similar to the overall trial: 52% TMP, 48% THP. Baseline cell types associated with pCR overall and by treatment arm are shown in Table 1. After 3 weeks of therapy, classical monocytes showed the largest change across all cell populations in both treatment arms (mean Δ -5.1% (p<0.001, q<0.001) in TMP and -4.6% (p=0.013, q=0.110) in THP). A decrease in one activated NK cell subset (NK CD56dimCD57+CD8-) was observed only with TMP (TMP: mean Δ -1.83% vs THP: mean Δ -0.47%; mean difference TMP vs THP=-1.36; p=0.002, q=0.052). Functional marker analyses will be presented at the meeting. Conclusions: Neoadjuvant chemotherapy plus anti-HER2 therapy induces early changes in circulating immune populations in HER2+ EBC. PBMC profiling reveals a rapid decline in circulating monocytes and NK cell subsets after 3 weeks of therapy, potentially reflecting tumor migration and contribution to ADCC.
| Overall population (n=91) | TMP (n=66) | THP (n=25) | |
| Classical monocytes | OR 1.07 (95% CI 1.01-1.13), p=0.030, q=0.330 | OR 1.09 (95% CI 1.01-1.18), p=0.027, q=0.279 | – |
| Nonclassical monocytes | OR 1.45 (95% CI 1.04-2.15), p=0.043, q=0.333 | – | – |
| CD8 central memory T cells | – | OR 0.75 (95% CI 0.55-0.97), p=0.043, q=0.279 | – |
| CD8 terminal effector memory RA+ T cells | – | OR 0.88 (95% CI 0.77-0.98), p=0.041, q=0.279 | – |
Presentation numberPS2-08-11
Artificial Intelligence (AI)-based spatial assessment of tumor-infiltrating lymphocytes (TIL) and outcome in early HER2+ breast cancer (BC): ancillary study of the phase III adjuvant ShortHER trial.
Maria Vittoria Dieci, University of Padova, Padova, Italy
M. Dieci1, K. AbdulJabbar2, G. Bisagni3, S. Bartolini4, L. Cavanna5, A. Musolino6, F. Giotta7, A. Rimanti8, O. Garrone9, E. Bertone10, K. Cagossi11, A. Ferro12, F. Piacentini13, E. Orvieto14, M. Sanders15, F. Miglietta1, H. Yan16, S. Balduzzi17, R. D’Amico13, P. Conte1, R. Salgado18, V. Guarneri1; 1Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, ITALY, 2NA, Case45, London, UNITED KINGDOM, 3Department of Oncology and Advanced Technologies, Azienda USL-IRCCS, Reggio Emilia, ITALY, 4Department of Nervous System Medical Oncology, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, ITALY, 5Internal Medicine and Oncology, Clinica Piacenza, Piacenza, ITALY, 6Medical Oncology, Breast and GYN Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, ITALY, 7Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, ITALY, 8Carlo Poma Hospital, ASST Mantova, Mantova, ITALY, 9Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, ITALY, 10Medical Oncology, S. Anna Hospital, Torino, ITALY, 11Breast Unit Ausl Modena, Ramazzini Hospital, Carpi, ITALY, 12Rete Clinica Senologica-Oncologia Medica S. Chiara, APSS, Trento, ITALY, 13Department of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena, Modena, ITALY, 14Pathology unit, ULSS 8 Berica, Vicenza, ITALY, 15Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 16case45, case45, London, UNITED KINGDOM, 17NA, University of Modena and Reggio Emilia, Modena, ITALY, 18Department of Pathology, ZAS Hospitals, Antwerp, BELGIUM.
Background: We previously demonstrated the prognostic role of TIL (manual score) in patients with early HER2+ BC enrolled in the ShortHER trial at long follow up (Dieci, JAMA Oncol 2025). Here, we analyze the added prognostic value of AI-based spatial TIL assessment. Methods: The ShortHER study randomized 1254 pts with HER2+ early BC to 9 weeks vs 1 year of adjuvant trastuzumab combined with chemotherapy. Manual TILs were scored according to international accepted standard developed by the TILs-WG. A zero-shot, explainable AI pipeline (Case45), was used to analyze H&E stained slides, focusing on TIL and their spatial interplay with cancer cells. The following AI-metrics were considered: AI-TIL (ratio of TILs adjacent to cancer nests relative to stromal cells); immune hotspots (normalized fraction of immune cell aggregates in relation to cancer and tissue). Immune metrics were considered as continuous and as categorical (cut-offs: <20%/>20% for manual TIL; <75%/>75% percentile for AI-TIL and immune hotspots). Survival endpoint was DDFS (distant relapse, death). Cox regression models were adjusted for clinicopathological factors (age, stage, hormone receptor, treatment arm, menopause, histologic grade). Likelihood ratio test evaluated the added prognostic value of TIL metrics to prognostic models. Results: 858 were cases evaluable by AI, median FU was 9.0 years. AI-TIL showed a moderate positive correlation with manual TIL (p<0.001, Spearman’s rho 0.500). Patients with high AI-TIL (n=211) experienced a better DDFS vs low AI-TIL: 8-yr rate 93.2% vs 86.7%, log-rank p=0.003, adj HR 0.47 (95%CI 0.26-0.82). Added prognostic value of AI-TIL was observed for patients with intermediate TIL scores between 10% and 29% (n=205): 8-yr DDFS rate was 94.8% for high AI-TIL vs 87.8% for low AI-TIL, p=0.062, mostly driven by the subgroup of patients with manual TIL 20-29% (8-yr DDFS 100% for high AI TIL vs 82.9% for low AI-TIL, p=0.017). Patients with high immune hotspots had a significantly better DDFS vs others (8-yr rates 92.9% vs 86.8%, p=0.028, adj HR 0.66, 95% CI 0.39-1.10). The prognostic role of immune hotspots was confined to patients with low TIL, either manual or AI. In low manual TIL, 8-yr DDFS was 92.9% vs 85.2% for high vs low immune hotspots (p=0.029; adj HR 0.67, 95% CI 0.36-1.25). In low AI-based TIL, 8-yr DDFS was 92.6% vs 84.7% for high vs low immune hotspots (p=0.024; adj HR 0.67, 95%CI 0.38-1.17). Table shows the added prognostic information provided by TIL metrics to multivariable models (Model 1: clinico-pathological factors + manual TIL; Model 2: clinico-pathological factors + AI-TIL).Conclusions: TIL AI-based spatial analysis complements manual TIL scoring in refining prognostic stratification early HER2+ BC. This added value is more pronounced in cases with low to intermediate manual TIL scores.
| Model | Likelihood ratio chi-square | P value | |||
| AI-TIL added to Model 1 | 5.61 | 0.018 | |||
| Manual TIL added to Model 2 | 4.17 | 0.041 | |||
| Immune hotspots added to Model 1 | 1.80 | 0.180 | |||
| Immune hotspots added to Model 2 | 3.26 | 0.071 |
Presentation numberPS2-08-12
Age-associated divergence in breast cancer: clinical, molecular, and genomic insights from a large real-world cohort
Julia Foldi, UPMC, Pittsburgh, PA
J. Foldi1, S. Satta2, B. Taylor2, P. Dutta2, S. Oesterreich1, A. Lee1, E. Kalashnikova2, A. Rodriguez2, M. Liu2, M. Balic1, A. Partridge3; 1University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, UPMC, Pittsburgh, PA, 2Oncology, Natera, Inc., Austin, TX, 3Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Introduction: Breast cancer (BC) in younger individuals is often clinically aggressive and biologically distinct. However, comprehensive real-world evidence comparing molecular underpinnings in very early-onset BC (age <35 years) vs. BC in older age groups (ages 36-50 years and age >50 years) remains limited. Here, we evaluate clinicopathologic features and molecular characteristics between BC diagnosed in younger (<35 years) and older (>35 years) age groups, with a focus on subtype distribution, ctDNA dynamics, relapse patterns, and genomic alterations.Methods: This retrospective analysis included patients diagnosed with early-stage BC who underwent tumor-informed circulating tumor DNA (ctDNA) testing (Signatera™, Natera, Inc.) in a real-world clinical setting (2019-2024). Genomic alterations were derived from whole-exome sequencing of tumor tissue as part of their ctDNA testing. Histology, molecular subtype, lymph node (LN) status, recurrence events, ctDNA detection over time, and relapse patterns were compared. Results: The cohort included 10,122 patients stratified by age <35 years (N=334), ages 36-50 years (N=2,501), and age >50 years (N=7,287). Distribution across age groups based on histology, subtype, and nodal status revealed that the age <35 years group exhibited a higher prevalence of aggressive subtypes (triple negative breast cancer [TNBC]: 22.4%, and hormone receptor [HR]−/human epidermal growth factor receptor 2 [HER2]+: 6.7%), predominantly ductal histology (77.6%), and the highest rate of lymph node involvement (44.1%); ages 36-50 years showed intermediate distributions across all features; while age >50 years were enriched for HR+/HER2− tumors (35.7%), had a higher frequency of lobular histology (12.4%), and demonstrated the lowest rate of nodal involvement (38%, p=0.016). Genomic profiling revealed greater prevalence of PIK3CA mutations in the age >50 years group (31%, p <0.0001) and enrichment in TP53 mutations in the age <35 years group (notably in HR−/HER2+: 51%, p=0.12 and TNBC: 53%, p50 years group demonstrated the highest prevalence of elevated TMB/MSI-High (53.3%), while the age <35 and age 36-50 years groups exhibited higher rates of TMB/MSS-low tumors (60.6% and 56.3%, respectively) compared to patients with age >50 years (46.6%, p<0.001). With respect to ctDNA detection, we observed the highest cumulative ctDNA positivity (29%) in the age <35 age group, particularly in those with HR-/HER2+ disease (46.2%, p=0.09). Consistent with this finding, patients with HR-/HER2+ BC in the age <35 group exhibited the highest relapse rates (46.7%, p<0.0001) with a high risk of multi-site relapse (13.3%). Notably, patients in the HR+/HER2- age <35 group exhibited a shorter median time to relapse (48 days, p=0.02) compared to other subtypes.Conclusions: Our data confirm that very early-onset breast cancer represents a biologically and clinically distinct entity characterized by more aggressive subtypes, shorter time to ctDNA positivity, higher metastatic burden at time of relapse, and differential patterns of genomic alterations. These findings highlight the need for intensified surveillance and tailored therapeutic strategies in younger BC patients, with a potential role for ctDNA monitoring.
Presentation numberPS2-08-13
Spatially resolved tumor subclones in HER2-positive breast cancer
Lucie Cervenkova, Université Libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, Belgium
L. Cervenkova1, M. Rediti2, M. Serra1, F. Lifrange3, C. Tommasi4, N. Occelli1, X. Wang1, D. Vincent1, G. Rouas1, L. Buisseret1, D. Larsimont5, F. Libert6, A. Lefort1, N. Van Renne1, D. Venet1, F. Rothé1, C. Sotirou1; 1Breast Cancer Translational Research Laboratory, Université Libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, BELGIUM, 2IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, ITALY, 3Department of Pathology, University Hospital Center of Liege, Liege, BELGIUM, 4Department of Medicine and Surgery, University of Parma, Parma, ITALY, 5Department of Pathology, Université Libre de Bruxelles (ULB), Institut Jules Bordet, Brussels, BELGIUM, 6Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), Université Libre de Bruxelles (ULB), Brussels, BELGIUM.
Background: HER2-positive breast cancer displays significant tumor and tumor microenvironment (TME) heterogeneity, which directly impacts treatment response and survival outcomes. Treatment resistance can arise due to the presence of somatic tumor subclones and is further driven by interactions with TME. Spatial transcriptomics (ST) enables mapping of these subclones in the tissue context. Methods: We performed spatial transcriptomics (10X Genomics Visium) on frozen HER2-positive breast cancer surgical samples from untreated primary tumors (n = 25). We then utilized H&E images of the ST slides to annotate morphological structures at the single-cell level (QuPath). Next, we developed a pipeline to characterize spatial ‘tumor subclones’. We first defined ‘tumor microregions’ as spatially distinct clusters of tumor-enriched spots separated by stromal components. We then inferred their copy number profiles and computed a similarity score for each tumor microregion pair. These scores served as an input distance matrix for hierarchical clustering to identify subclones. To characterize the subclones at the gene expression level, we computed their marker genes (p-value 1) and subjected them to gene set over-representation analysis. Subsequently, we queried subclone markers against LINCS L1000 drug perturbation profiles (Enrichr) to nominate compounds predicted to modulate subclone-specific transcriptional programs. Finally, we reconstructed evolutionary relationships among subclones with MEDICC2. Results: We identified a total of 71 spatial tumor subclones in our cohort. Our results demonstrate substantial multi-clonality of HER2-positive breast cancer (median of 3 subclones per sample), with only 5 samples comprising a single subclone. Over-representation analysis results suggest that multi-clonal samples contain subclones with diverse gene expression profiles, exhibiting varying associations (FDR < 0.05) to pathways related to immune-rich (e.g., interferon alpha/gamma response, IL2 STAT5 signaling), stromal (e.g., epithelial mesenchymal transition), or luminal phenotypes (e.g., estrogen response early/late). We further demonstrate that subclones with similar phenotypes within the same sample often share a recent common ancestor, indicating that they diverged after acquiring shared copy number alterations that likely enable specific, conserved transcriptional programs. Conversely, subclones with different phenotypes emerge on distinct evolutionary branches, likely due to the absence of these key copy number changes. Additionally, we observe statistically significant differences (Kruskal-Wallis, p-value < 0.05) in ERBB2 expression between subclones within the sample in the majority of our cohort (18 out of 25 samples). We hypothesized that the presence of low-ERBB2 subclones may hinder the response to targeted anti-HER2 treatment. Additionally, enrichment tests on drug perturbations gene sets reveal that subclones within the same sample show varying treatment responses to the commonly used chemotherapy regimen. We present a case of a patient‐derived sample that recurred after adjuvant carboplatin-docetaxel therapy. Enrichment analysis of spatial subclones revealed striking heterogeneity in carboplatin sensitivity, with only one of three subclones showing a significant response (FDR = 0.006). This intra-sample variation in treatment response underscores the importance of profiling spatial subclones. Conclusions: Our findings underscore the intra-tumor somatic heterogeneity of HER2-positive breast cancer depicted by ST and its potential clinical impact. Further analyses focused on the interactions of existing subclones with the TME could refine our understanding of the mechanisms driving treatment resistance in this disease.
Presentation numberPS2-08-14
Chemokine signatures improve PD-L1-based prediction of pembrolizumab response in triple-negative breast cancer
Shipra Gandhi, Winship Cancer Institute of Emory Univeristy, Atlanta, GA
S. Gandhi1, S. Deshmukh2, S. Wu2, J. Xiu2, G. Sledge Jr.2, D. Trapani3, J. Leone4, S. Chumsri5, M. Lustberg6, C. Hong7, S. Yao8, K. Takabe9, M. Ernstoff10, M. Opyrchal11, P. Chalasani12, C. Paulos13, G. Lesinski13, A. Madabhushi14, S. Badve15, K. Kalinsky1, P. Kalinski16; 1Medicine, Winship Cancer Institute of Emory Univeristy, Atlanta, GA, 2Oncology, Caris Life Sciences, Phoenix, AZ, 3Oncology, Europena Institute of Oncology, Milan, ITALY, 4Oncology, Dana Farber Cancer Institute, Boston, MA, 5Oncology, Mayo Clinic, Jacksonville, FL, 6Oncology, Yale University, New Haven, CT, 7Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 8Cancer prevention and control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 9Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 10Developmental Therapeutics Program, National Institute of Health, Bethesda, MD, 11Medicine, Indiana University, Bloomington, IN, 12Medicine, The George Washington University, Washington, DC, 13Immunology, Winship Cancer Institute of Emory Univeristy, Atlanta, GA, 14Biomedical Engineering, Emory University, Atlanta, GA, 15Pathology, Winship Cancer Institute of Emory Univeristy, Atlanta, GA, 16Oncology, Magee-Women’s Research Institute, Pittsburgh, PA.
Background: Triple-negative breast cancer (TNBC) is an aggressive yet immunogenic subtype of breast cancer (BC) and the only one with FDA-approved immune checkpoint inhibitor (ICI) – pembrolizumab. Currently, PD-L1 (22C3) is the companion biomarker for predicting pembrolizumab benefit, but its utility is limited by spatial/temporal heterogeneity, assay variability, and modest correlation with treatment response. Chemokines such as CXCL9, CXCL10, and CCL5 recruit CD8⁺ T cells into the tumor microenvironment and have been associated with improved response to ICI, but their predictive value alongside PD-L1 remains unclear. Here, we examined the relationship between these chemokines, PD-L1, and pembrolizumab efficacy in TNBC. Methods: We analyzed 3662 TNBC tumors profiled by NGS (592, NextSeq; WES/WTS, NovaSeq; Caris Life Sciences). Immune cells were estimated using WTS deconvolution (Quantiseq). PD-L1 positivity was defined as >10% CPS (PharmDx 22C3). CXCL9/CXCL10/CCL5-high (H) and -low (L) TNBC were classified by RNA expression above or below 50th percentile. Real-world median overall survival (mOS) was derived from insurance claims and calculated from start of pembrolizumab to last contact using Kaplan-Meier. Statistical significance was assessed by chi-square and Mann-Whitney U with multiple comparison adjustments (q<0.05). Results: TNBC expressed higher levels of CXCL9, CXCL10, and CCL5 (N = 3662, median TPM: 5.3, 14.8, and 15.1) compared to HER2+ (N = 1344; 4.8, 10.1, 10.6) and HR+HER2- BC (N = 6097; 2.7, 6.6, 9.4), all q<0.05. Moderate correlations were observed between PD-L1 CPS and each chemokine (r = 0.41, p<.01). PD-L1-positive tumors had better mOS (24.3 months (m) vs 18.6 m, HR 0.75, 95% CI 0.6-0.92, p=0.007) compared to PD-L1-negative TNBC post-pembrolizumab. Among PD-L1-negative tumors, those with CXCL9-H had better mOS (26.9 m vs 15.8 m for CXCL9-L, HR 0.61, 95% CI 0.45-0.82, p=0.001) post-pembrolizumab. Among PD-L1-positive tumors, CXCL10-H and CCL5-H was associated with improved mOS (CXCL10-H: 25.7 m vs. 20.7 m for CXC10-L, p = 0.03; CCL5-H: 26.7 vs. 18.5 m for CCL5-L, p = 0.03) post-pembrolizumab. PD-L1 negative CXCL9-H, PD-L1 positive CXCL10-H and PD-L1 positive CCL5-H groups showed higher infiltration of CD8+ T cell, B cells, M1/M2 macrophages, Tregs. They also exhibited higher T cell inflamed score, IFNy score and MAPK activation score vs. CXCL9-L, CXCL10-L and CCL5-L groups (Table). Conclusions: In TNBC, a chemokine signature comprising CXCL9, CXCL10 and CCL5 refines PD-L1 based prediction of pembrolizumab benefit. Specifically, high CXCL10 or CCL5 expression in PD-L1-positive tumors, and high CXCL9 expression in PD-L1-negative tumors, were associated with improved survival post pembrolizumab and immune-rich microenvironment. These findings support the use of chemokine profiling to complement PD-L1 and enhance patient selection for ICI therapy in TNBC.
| PD-L1 positive CCL5-High | PD-L1 positive CCL5-Low | q-value | PD-L1 positive CXCL10-High | PD-L1 positive CXCL10-Low | q-value | PD-L1 negative CXCL9-High | PD-L1 negative CXCL9-Low | q-value | |||||||||||||||||||||
| B cell (median %) | 4.36 | 3.63 | <0.001 | 4.5 | 3.37 | <0.001 | 4.21 | 3.51 | <0.001 | ||||||||||||||||||||
| M1 Mɸ (median %) | 4.52 | 3.26 | <0.001 | 4.45 | 3.53 | <0.001 | 3.01 | 2.31 | <0.001 | ||||||||||||||||||||
| M2 Mɸ (median %) | 3.51 | 2.83 | 0.001 | 3.52 | 2.8 | <0.001 | 3.37 | 2.83 | <0.001 | ||||||||||||||||||||
| CD8+ T cell (median %) | 1.64 | 0.37 | <0.001 | 1.77 | 0.15 | <0.001 | 0.81 | 0 | <0.001 | ||||||||||||||||||||
| Tregs (median %) | 3 | 1.7 | <0.001 | 3 | 1.75 | <0.001 | 2.18 | 1.06 | <0.001 | ||||||||||||||||||||
| Neutrophil (median %) | 8.08 | 4.83 | <0.001 | 4.09 | 4.71 | 0.01 | 4.23 | 4.59 | 0.02 | ||||||||||||||||||||
| T-Cell Inflamed Score | 110 | -44 | <0.001 | 111 | -75 | <0.001 | 78 | -77 | <0.001 | ||||||||||||||||||||
| IFNy score | 0.01 | -0.31 | <0.001 | 0.01 | -0.28 | <0.001 | -0.24 | -0.34 | <0.001 | ||||||||||||||||||||
| MAPK Activation Score | -0.97 | -1.39 | 0.08 | -0.87 | -2.03 | <0.001 | -0.6 | -1.07 | <0.001 |
Presentation numberPS2-08-16
Validation of a multi-omic biosignature to predict locoregional recurrence risk and the benefit of adjuvant radiation therapy in women with early-stage invasive breast cancer treated with breast-conserving surgery and endocrine therapy.
G Bruce Mann, Royal Melbourne and Royal Women’s Hospital, Parkville, Australia
G. Mann1, K. Mittal2, R. de Boer3, C. MacCallum1, M. Alvarado4, M. Christie5, F. Vicini6, C. Shah7, Y. V. Zou8, M. Mentrikoski9, J. Savala9, D. Dabbs9, S. C. Shivers2, P. Whitworth10, T. Bremer2; 1The Breast Service, Royal Melbourne and Royal Women’s Hospital, Parkville, AUSTRALIA, 2R&D, PreludeDx, Laguna Hills, CA, 3Medical Oncology, Royal Melbourne Hospital, Parkville, AUSTRALIA, 4Breast Surgical Oncology, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco,, CA, 5Department of Pathology, Royal Melbourne Hospital, Parkville, AUSTRALIA, 6Department of Radiation Oncology, Michigan Healthcare Professionals, Farmington Hills, MI, 7Department of Radiation Oncology, Allegheny Health Network, Pittsburgh, PA, 8Department of Radiation Oncology, Genesis Care, Parkville, AUSTRALIA, 9Department of Pathology, PreludeDx, Laguna Hills, CA, 10Medical Affairs, PreludeDx, Laguna Hills, CA.
Background: Randomized clinical trials demonstrate that radiotherapy (RT) after breast conserving surgery (BCS) significantly reduces risk of invasive locoregional recurrences (LRR) in women with early-stage hormone receptor positive (HR+) HER2 negative invasive breast cancer (IBC). However, RT de-escalation trials also demonstrate that some patients may omit RT based on clinicopathological factors. To better inform shared decision making, a multi-omic test was developed (n=939) to predict women’s invasive LRR risk and RT benefit. Herein, we evaluate this test in a retrospective observational cohort treated at Royal Melbourne Hospital (RMH), Australia. Methods: Women with HR+HER2- T1T2N0M0 IBC were treated at RMH with definitive BCS between 2006-2017 (n=526) and with adjuvant ± RT ± endocrine therapy (ET) ± chemotherapy (CT). Treatment and outcome were collected by RMH and transferred to a third-party biostatistics group (MCG, AUS). After RMH central pathology review, FFPE tumor samples were sent to a CLIA lab (Laguna Hills, CA) to assay protein expression by multiplex-immuno-fluorescence and RNA expression by targeted Next Generation Sequencing (blinded to treatment and outcome). The test combines multi-omic data using biosignatures to calculate a Decision Score (DS) to determine LRR prognosis and a Radio Resistance Index (RRI) to determine individualized RT benefit. Test results were transferred to and analyzed by MCG to validate the test using a pre-specified analysis plan. The association of continuous DS and RRI with LRR risk and RT benefit were analyzed in multivariable analysis. The absolute risk and RT benefit within Low and Elevated categorical risk groups were assessed using pre-specified thresholds (DS=5, RRI=5). Hazard ratios (HR) were determined using Cox proportional hazards analysis and the interaction of the RRI and RT benefit was assessed. Kaplan-Meier survival analysis was used to calculate absolute LRR risk. Results: Complete biomarker and clinical information were available in 426 women treated with ET of whom 319 patients received RT. There were 43 invasive LRR and 22 contralateral breast events within 10-years. The study demonstrated that the test was prognostic for LRR risk, where increasing continuous DS was associated with higher invasive LRR risk (LRR HR=2.4 per 5 units, p<.001). The test was also predictive for RT benefit. Overall, patients substantially benefited from RT (HR=0.1, p<.001) but RT treated patients with increasing RRI had less RT benefit (LRR HR=5.9 per 5 units, RRI:RT interaction p<.001). For example, in a categorical analysis, in a Low Risk group (DS≤5, n=188), RT had no significant association with LRR (HR=0.6, p=.58), where mean 10-yr LRR was 8% without RT and 6.5% with RT. In contrast, the mean 10-yr LRR was 26% without RT in the Elevated Risk group (DS>5, n=238). In this group (DS>5), patients with RRI ≤5 substantially benefited from RT (HR=0.2, p=0.002), with a corresponding 18% reduction in 10-yr LRR risk; however, for patients with RRI>5, RT was not associated with a statistically significant decrease in LRR risk (HR=0.9, p=.8), where the10-yr LRR risk remained at 22% after RT. Conclusion: Women diagnosed with early-stage HR+ HER2- IBC and treated with BCS and ET ±RT were stratified into those with lower and higher LRR risk and RT benefit using a multi-omic test. In this external blinded validation in a retrospective observational cohort, the test stratified women into those with low LRR risk who had no significant benefit from RT, those with elevated LRR risk who had a significant benefit from RT, and those with elevated LRR risk who had minimal therapeutic benefit from RT.
Presentation numberPS2-08-17
Prevalence and clinical significance of exon 6 ESR1 gene fusions in advanced breast cancer after disease progression on aromatase inhibitors
Seth A. Wander, Massachusetts General Hospital Cancer Center, Boston, MA
S. A. Wander1, D. G. Stover2, J. T. Lei3, N. Zhang4, A. Hardin4, M. J. Ellis3; 1Department of Medical Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, 2Internal Medicine, Ohio State University Comprehensive Cancer Center, Columbus, OH, 3Clinical Development, Guardant Health, Inc., Redwood City, CA, 4Real World Evidence, Guardant Health, Inc., Redwood City, CA.
Background: Endocrine therapy resistance (ETR) is a frequent challenge in metastatic ER+ HER2- breast cancer (MBC) and is driven by different molecular mechanisms. Since the first observation of a transcriptionally active ESR1 fusion involving exons 1-6 (e6) of ESR1 fused to YAP1 (Li et al., Cell Reports, 2013), e6 ESR1 gene fusions are increasingly recognized as a cause of ETR. However, subsequent studies have found that 3’ fusion partners are highly heterogeneous, leading to questions regarding clinical actionability. Here, we report an ESR1 fusion classification system with real world evidence (RWE) support. Methods: ESR1 fusions were detected in circulating tumor DNA (ctDNA) by sequencing the intron between exons 6 and 7 to identify fusion gene breakpoints. Based on the study by Gou et al. (Cancer Research, 2021), a 4-group classification system was devised: Group (G)1: known active (lab tested); G2: likely active (not lab tested but transcription factor [TF] or transcriptional co-activator [CoA] partner and at least 2 instances in GuardantINFORM™ RWE database of 1,276 cases); G3: possibly active (not TF or CoA partner but at least 3 recurrent cases); and G4: inactive fusion (all other fusions [i.e., ESR1 fusions not involving exon 6, out of frame, chromosome 6 partner in frame, exon to exon, or ESR1 to ESR1]). Propensity matching was used to account for different levels of ctDNA, age, and lines of therapy. Results: The most prevalent partners by fusion group were YAP1 (n = 21), ARNT2 (n = 9), LPP (n = 2) for G1; SS18 (n = 4, previously reported sarcoma fusion partner), TAZ (n = 4), ESRRG (n = 4), NFAT5 (n = 4) for G2; PXN (n = 9), TFG (n = 5, previously reported breast cancer fusion partner), PROSER1 (n = 4) for G3; and in frame examples of SYNE1 (n = 62), PLEKHG1 (n = 57), SNAP91 (n = 29), CCDC170 (n = 44, latter case previously reported to be functionally inactive [Lei et al., Cell Reports, 2018]) for G4. Since G1, 2 and 3 had similar adverse prognostic effects, outcome analysis involved comparisons to G5, defined as “no fusion detected” and fusion groups 1, 2 and 3 in combination. In a multivariate analysis, patients with G1-3 ESR1 fusions receiving ET monotherapy as second-line treatment had a rwOS HR of 3.0 (95% CI 1.7-5.5, P = 0.0002) when compared to G5 (no fusion). Supporting the inactive classification, G4 had an indistinguishable clinical course from G5. Patients with G1-3 ESR1 fusions receiving heterogeneous second-line ET combinations (including chemotherapy and antibody drug conjugates) also had an increased risk of death compared to G5 based on a multivariate analysis (HR 2.0, 95% CI 1.2-3.3, P = 0.01). Conclusions: Despite low prevalence, estimated at 2.1%, in ER+ HER2- MBC cases post AI therapy, G1-3 ESR1 fusions were associated with an increased risk of early death when managed with second-line ET monotherapy or heterogeneous second-line ET combinations. These results highlight the need for novel therapeutic options for patients with G1-3 ESR1 fusions.
Presentation numberPS2-08-18
Cathepsin Protease Expression and Therapeutic Outcomes to Trastuzumab Deruxtecan (T-DXd) in Metastatic Breast Cancer
Samer Alkassis, UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA
S. Alkassis1, A. Dugan2, U. Jariwala2, B. Yilma2, S. Fragkogianni2, M. Lipsyc-Sharf1, A. LeVee1, Y. Yuan3, F. Meric-Bernstam4, H. Rugo5, J. O’Shaughnessy6, M. Rimawi7, R. Schiff7, D. Elashoff1, J. Glaspy1, M. McDermott1, A. Cummings1, J. Mercer2, A. Bardia1; 1Hematology/ Oncology, UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA, 2Medical affairs, Tempus AI, Inc., Chicago, IL, 3Hematology/ Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, 4Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX, 5Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, 6Medical Oncology, Texas Oncology, Sara Cannon Research Institute, Dallas, TX, 7Hematology/ Oncology, Baylor College of Medicine, Houston, TX.
Background: T-DXd is FDA approved for patients with metastatic breast cancer (MBC) and HER2+ or HER2 low/ultra-low status. However, response and resistance mechanisms associated with T-Dxd are not well understood and how they differ across HER2 subtypes requires further interrogation. Cathepsins are a family of proteolytic enzymes that play crucial roles in cellular processes including protein degradation and turnover in endosomes/lysosomes and when secreted into the extracellular space can contribute to matrix remodeling. Proteases are also important for ADC linker cleavage and payload release. Recent preclinical data revealed that specific extracellular and intracellular proteases, including cathepsins, may alter the efficacy of ADCs with cleavable linkers, such as T-DXd, as opposed to ADCs with non-cleavable linkers like T-DM1 that depend on complete lysosomal degradation. Here, from a clinicogenomic database, we characterized the transcriptomic expression of proteases prior to T-DXd vs T-DM1 treatment (tx) to understand their impact on clinical outcomes in MBC. Methods: Using the Tempus database, the transcriptomic expression of two protease genes (cathepsin L “CTSL”, cathepsin B “CTSB”) was generated from pre-tx biopsies collected from patients with MBC (N = 453; 36% TNBC, 26% HR+/HER2-, 19.2% HER2+, 19% NOS) 1 year prior to or up to 15 days post-tx with SG (n = 204), T-DXd (n = 178), or T-DM1 (n = 71). RNA-sequencing data were generated and processed with the Tempus xR assay. Cox proportional hazards models with risk set adjustment were used to test for associations between pre-tx gene expression and overall survival (OS), where gene expression was modeled as a continuous linear predictor. The proportional hazards assumption for OS was tested. Given the exploratory nature of the analyses, all p-values are uncorrected and nominal statistical significance was set at p < 0.05. Results: This cohort had a median age of 52 and was diverse (55% White, 14% Black, 7.1% Other, 24% Unknown). Among patients treated with T-DXd (n = 170), high expression of CTSB and CTSL was associated with significantly worse OS in the HER2+ subgroup (n = 35; HR 4.35; p = 0.013 and HR 6.04; p = 0.002, respectively) but correlated with improved outcomes in patients with HR+/HER2− disease (n = 75; HR 0.51; p = 0.024 and HR 0.56; p = 0.013, respectively). In the TNBC cohort (n = 28), we observed the following trends: CTSB: HR 0.63, p = 0.4 and CTSL: HR 0.75, p = 0.5. In contrast, in the T-DM1 cohort, no significant associations were found between protease expression and OS (entire cohort: n = 59, HR 1.03; p = 0.9 and HR 0.96; p = 0.9 for CTSB and CTSL, respectively; HER2+cohort: n=39, HR 0.55; p = 0.10 and HR 0.69; p = 0.2 for CTSB and CTSL, respectively). Conclusions: Using a real-world multimodal dataset, here we build upon recent in-vitro findings and demonstrate the potential role of proteases as biomarkers of response to T-DXd in HER2 low/ultralow, but not HER2 positive/amplified breast cancer. These findings highlight the potential differential dependence on cathepsins based on HER2 expression and impact on payload release modulating efficacy in MBC. These findings need to be validated in additional datasets and can guide biomarker-driven clinical application of ADCs.
Presentation numberPS2-08-19
Ki67 Spatial Heterogeneity as a Predictive and Prognostic Marker in Breast Cancer: A Spatial Image Analysis Approach
Christophe Van Berckelaer, Antwerp University Hospital (UZA), Edegem, Belgium
C. Van Berckelaer1, K. Zwaenepoel2, L. Cox1, D. Charlotte1, D. Julie1, E. Louise1, H. Fleur1, L. Evy1, G. R. Devi3, A. Ramadhan2, S. Koljenovic2, P. Van Dam1; 1MOCA, Antwerp University Hospital (UZA), Edegem, BELGIUM, 2Department of Pathology, Antwerp University Hospital (UZA), Edegem, BELGIUM, 3Department of Surgery, Division of Surgical Sciences, Duke University School of Medicine, Durham, NC.
Background: Ki67 is a well-established proliferation marker in breast cancer. Current clinical use focuses on the proportion of Ki67-positive cells, ignoring spatial heterogeneity in expression. However intra-tumoral heterogeneity has demonstrated to be associated with worse outcome. We hypothesized that spatial Ki67 heterogeneity carries clinical information beyond conventional scoring and aimed to evaluate its added value for predicting pathological complete response (pCR) after neoadjuvant chemotherapy (NACT) and for stratifying recurrence risk using genomic expression profiling (GEP). Using digital image analysis (DIA), precise and spatial quantification of biomarker distribution is possible. Methods: We analyzed two retrospective breast cancer cohorts using an AI-assisted DIA pipeline. Tumor sections stained for ER, PR, Ki67, and HER2 were digitized and analyzed in QuPath. Using AI, individual tumor cells were recognized and four tumor regions (0.5mm x 0.5mm) with the highest tumor/stroma ratio were selected for analysis. Spatial Ki67 heterogeneity was quantified using the Morisita-Horn Index (MHI) after the Ki67-positive and Ki67-negative tumor cells were mapped using XY-coordinates and square tessellation (100×100 µm tiles) was applied. The MHI was used to compare the similarity in cell composition between all pairs of tiles within a region. MHI values range from 0 to 1, with higher values indicating a more uneven distribution of Ki67+ cells. We used logistic regression and model comparison with Akaike Information Criterion (AIC), likelihood ratio test (LRT) or Vuong test, to evaluate the predictive value of Ki67 heterogeneity. In the first cohort (n=45), spatial heterogeneity was assessed on pretreatment biopsies from patients treated with NACT. In the second cohort (n=79), heterogeneity was evaluated in HR+/HER2- breast cancer patients stratified as high or low risk of recurrence based on GEP. Results: In the GEP cohort, both a higher proportion of Ki67- positive cells and greater Ki67 heterogeneity were significantly associated with high genomic risk. The median MHI was 0.24 (0.03–0.35) in the high-risk group compared to 0.14 (0.01–0.43) in the low-risk group (P = 0.008). This higher MHI indicates more heterogeneous regionally clustered Ki67 expression, suggesting biologically distinct proliferative zones. In multivariate models, Ki67 heterogeneity remained a significant predictor of high-risk classification (OR 0.22, P = 0.036). Furthermore, in nested model comparison using LRT, addition of Ki67 heterogeneity significantly improved the model for predicting genomic risk (P = 0.034). These findings were consistent across biopsy and resection specimens, highlighting the robustness of heterogeneity measures. In the NACT cohort, Ki67 heterogeneity was higher in patients who achieved pCR (median MHI 0.26 [0.17–0.35]) compared to those who did not (median MHI 0.22 [0.12–0.40], P = 0.023). In multivariate modeling, Ki67 heterogeneity emerged as an independent predictor of pCR (OR 23.5, P = 0.038), outperforming Ki67 density and improving model fit (AIC 31.6 vs. 36.1; P = 0.038). Finally, in both cohorts, DIA-derived Ki67 models slightly outperformed traditional pathologist scoring, although Vuong tests did not show a statistically significant difference. Conclusion: Spatial Ki67 heterogeneity provides additional prognostic and predictive value beyond conventional Ki67 scoring. This heterogeneity indicates distinct areas of higher proliferation, clinically relevant biological variation, not captured by simple percentage positivity. Although validation in larger, prospective cohorts is necessary before clinical implementation, DIA provides a more objective and reproducible alternative to manual scoring, particularly when incorporating spatial heterogeneity.
Presentation numberPS2-08-20
Translational analysis of cerebrospinal fluid (CSF) and plasma circulating tumor DNA (ctDNA) from breast cancer patients (pts) with leptomeningeal disease (LMD) treated with trastuzumab deruxtecan (T-DXd) in the DEBBRAH trial
Amanda Fitzpatrick, King’s College London, London, United Kingdom
A. Fitzpatrick1, T. Shanmugalingam1, S. Haider1, M. Iravani1, L. Garrigós2, J. García-Sáenz3, P. Cortez4, F. Racca5, S. Blanch6, M. Ruiz-Borrego7, G. Martrat8, M. Mancino9, J. Guerrero10, J. Pérez-García11, M. Vaz Batista12, S. Braga13, A. Llombart-Cussac14, J. Cortés15; 1Comprehensive Cancer Centre, King’s College London, London, UNITED KINGDOM, 2Oncology, International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group; Hospital Universitari Dexeus, Barcelona, SPAIN, 3Oncology, Hospital Clínico San Carlos, Madrid, SPAIN, 4Oncology, IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, SPAIN, 5Oncology, IOB Institute of Oncology, Quiron Group, Barcelona, SPAIN, 6Oncology, Instituto Valenciano de Oncologia, València, SPAIN, 7Oncology, Hospital Universitario Virgen del Rocío, Sevilla, SPAIN, 8Trial, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 9Translational, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 10Data Analytics, Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 11Oncology, International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group; Medica Scientia Innovation Research (MEDSIR), Barcelona, SPAIN, 12Oncology, Hospital Professor Doutor Fernando Fonseca EPE; Medica Scientia Innovation Research (MEDSIR), Lisbon, PORTUGAL, 13Oncology, Hospital Professor Doutor Fernando Fonseca EPE, Lisbon, PORTUGAL, 14Medicine, Hospital Arnau de Vilanova; Translational Oncology Group, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU; Medica Scientia Innovation Research (MedSIR), Valencia, SPAIN, 15Oncology, 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), Madrid, SPAIN.
Rationale:LMD represents a severe and life-threatening complication of metastatic cancer, historically associated with extremely poor outcomes. Therapeutic options are limited, and these pts are routinely excluded from clinical trials investigating novel systemic therapies. Radiological response assessment in pts with LMD is challenging and CSF ctDNA has emerged as a potential minimally invasive biomarker for the diagnosis and disease monitoring of LMD. The DEBBRAH trial was a multi-cohort phase II study assessing T-DXd in HER2-positive and HER2-low breast cancer pts with brain metastases and/or LMD, with Cohort 5 dedicated to LMD. The primary endpoint of overall survival indicated antitumour response (median OS 13.3 months) in this cohort. This translational analysis aims to evaluate intra- and extracranial T-DXd activity through serial ctDNA measurement in CSF and plasma collected from pts in DEBBRAH cohort 5, and to determine molecular features associated with response and treatment resistance. Methods:Seven pts with cytology positive LMD were enrolled in DEBBRAH cohort 5 and received T-DXd 5.4 mg/kg intravenously every 21 days. Radiological response was measured by RECIST v.1.1 for extracranial disease and RANO-BM for intracranial lesions. CSF was collected at baseline from six pts (in one case, collection occurred after cycle 1), and serially prior to each T-DXd dose up to cycle 7 in four pts (three HER2-positive and one HER2-low). Plasma was obtained at baseline and prior to cycle 2 in all pts, and at end of treatment in four pts. Both sample types were collected in Streck Cell-Free DNA BCT tubes and centrifuged to remove cellular content, prior to cell-free DNA (cfDNA) extraction. cfDNA underwent shallow WGS (median 0.5X coverage), for large scale copy number calling and tumor fraction estimation by ichorCNA. Results:CSF ctDNA was detected in all baseline samples (tumor fraction range 0.11 – 0.83), whereas plasma ctDNA was only detected in three of seven pts (tumor fraction range 0.06 – 0.82). All four pts with serial CSF samples showed a reduction in ctDNA tumor fraction during T-DXd treatment, and two of them achieved complete CSF ctDNA clearance within 1 to 2 treatment cycles. The two pts with CSF ctDNA complete clearance were HER2-positive and received 7 and 12 cycles of T-DXd, respectively. In both cases, CSF remained ctDNA-negative at the end of CSF sampling (cycle 7) and treatment was discontinued due to extracranial progression according to RECIST v.1.1. Of the remaining two pts with serial CSF samples, one HER2-positive pt achieved a 15% reduction in ctDNA from baseline, completed 28 cycles of T-DXd, and remains alive at 18.5 months. The other, a HER2-low pt, had a 59% reduction in ctDNA from baseline and completed 15 cycles of T-DXd before developing extracranial progression according to RECIST v.1.1. An increase in plasma ctDNA tumor fraction was observed in three of the four patients with samples taken at the end of treatment, suggesting progression of extracranial disease.Mutational analysis to determine molecular variants associated with treatment response and resistance is being performed, and results will be displayed at the conference.Conclusion:These findings suggest that CSF ctDNA offers superior sensitivity over plasma ctDNA for detecting LMD. Although CSF ctDNA clearance was observed, which could be associated with LMD response, these data could not be correlated clinically since LMD-specific radiological and clinical assessment tools were not used in this trial. CSF ctDNA appears to be a promising response assessment biomarker for LMD in clinical trials, and merits further study in larger cohorts, incorporating LMD-specific assessment tools.
Presentation numberPS2-08-21
Circulating tumor DNA (ctDNA) Dynamics in Early-stage Breast Cancer Patients (pts) with Brain Metastases
Amy J. Xu, Memorial Sloan Kettering Cancer Center, New York, NY
A. J. Xu1, B. Dwivedi2, E. Kalashnikova2, J. Ortiz2, J. McKenzie2, A. Rodriguez2, M. C. Liu2, L. A. Huppert3, C. K. Anders4, N. U. Lin5, S. L. Sammons6; 1Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 2Oncology, Natera, Inc., Austin, TX, 3Radiation Oncology, University of California San Francisco, San Francisco, CA, 4Medical Oncology, Duke University, Durham, NC, 5Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 6Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.
Introduction: Brain metastases in breast cancer (BC) present significant clinical challenges. The utility of circulating tumor DNA (ctDNA) in detecting intracranial progression is not well established. This study investigated ctDNA kinetics before and after brain relapse from early-stage BC in pts undergoing routine surveillance with a tumor-informed ctDNA assay. Methods: To identify pts with brain metastases from early-stage breast cancer and relevant ctDNA timepoints, we utilized Natera’s proprietary real-world database, which is linked to Komodo’s Healthcare Map® claims database and available results of tumor-informed ctDNA testing (SignateraTM, Natera, Inc.) from 2019 to 2024. Hormone receptor and HER2 status and date of relapse were inferred from ICD-10 claims codes associated with interventions and secondary neoplasms. We evaluated ctDNA kinetics before and after brain relapse, including relative timing of ctDNA positivity, ctDNA levels (mean tumor molecules (MTM)/mL), and ctDNA clearance after initiation of therapy. Results: We identified 77 pts with early-stage breast cancer who developed subsequent brain relapse, including 28 pts with brain-only metastasis (BOM) and 49 pts with brain and extracranial metastases (BM+ECM) at the time of initial metastatic diagnosis. Among 28 pts with BOM, TNBC was the most represented subtype (40%; 11/28), followed by HER2+ (32%; 9/28) and ER+/HER2- (28%; 8/28). BOM were diagnosed at a median of 21.2 months (range: 1.8 – 87.0) and BM+ECM at a median of 19.2 months (range: 1.21 – 90.4) after surgery. The overall ctDNA detection rate prior to relapse was 54% (15/28) for pts with BOM and 92% (45/49) for pts with BM+ECM. Median time from ctDNA detection to BOM was 3.21 months (range: 0.2 to 14.5 months) and to BM+ECM was 2.66 (range: 0.03 to 39.2 months), acknowledging that the timing of ctDNA assessments was not controlled in this real-world dataset. Median ctDNA level prior to BOM was 1.27 MTM/mL (range: 0.04 – 227) compared to a median of 47.7 MTM/mL (range: 0.09 – 3854) in pts with BM+ECM. Among pts with BOM with matching pre- and post-relapse timepoints (n=17), 9 pts were ctDNA positive prior to BOM; of these, ctDNA clearance was observed in 44.4% (4/9) following post-relapse treatment, while the remaining pts were persistently ctDNA-positive [55.5% (5/9)]. Eight pts were ctDNA negative at BOM, and 25% (2/8) converted to positive after the diagnosis of isolated brain relapse. Among 34 pts with BM+ECM and matching pre- and post-relapse ctDNA timepoints, 30 pts were ctDNA positive prior to BM+ECM; of these, 13.3% (4/30) achieved clearance following post-relapse treatment, and 86.6% (26/30) remained positive. Four pts were ctDNA negative at BM+ECM, and 25% (1/4) converted to ctDNA-positive after the diagnosis of BM+ECM. Conclusions: Our RWD study provides evidence that tumor-informed ctDNA test results were positive in >50% of early-stage BC pts with BOM and >90% of early-stage BC pts with BM+ECM prior to the date of metastatic relapse. As expected, median ctDNA levels were generally lower in those with BOM compared to BM+ECM. Our data confirm the utility of ctDNA to identify pts who are at high risk of relapse, and provide evidence that ctDNA can detect CNS-limited recurrences. Given the potential for CNS involvement in pts with BC, pts with ctDNA-positivity and negative extracranial scans should be considered for CNS imaging to exclude BOM.
Presentation numberPS2-08-22
Clinical Relevance of ctDNA-Guided Molecular Profiling After Progression on CDK4/6 Inhibitor plus Endocrine Therapy in HR+/HER2- Metastatic Breast Cancer
Antonio Marra, European Institute of Oncology IRCCS, Milano, Italy
A. Marra1, D. Presti2, K. Venetis3, G. Castellano1, V. Peruzzo3, R. Adorisio3, A. Ranghiero3, A. Borghi3, D. Vacirca3, E. Giordano1, S. Perazzo1, K. Qeraj1, M. Lombardi3, P. Zagami1, D. Trapani1, E. Munzone2, E. Guerini Rocco3, N. Fusco3, G. Curigliano1; 1Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milano, ITALY, 2Medical Senology, European Institute of Oncology IRCCS, Milano, ITALY, 3Pathology, European Institute of Oncology IRCCS, Milano, ITALY.
Background: Circulating tumor DNA (ctDNA)-based molecular profiling is used to guide therapeutic decision-making in hormone receptor-positive/HER2-negative (HR+/HER2-) metastatic breast cancer (mBC) following progression on cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i) and endocrine therapy (ET). The predictive value of post-progression genomic alterations for subsequent lines of treatment remains unclear. Methods: We analyzed all consecutive patients with HR+/HER2- mBC treated at our Institution with ctDNA testing after progression on CDK4/6i-based therapy between November 2023 and June 2025. ctDNA sequencing was performed using a 77-gene liquid biopsy next-generation sequencing assay. Clinical, pathological, treatment, and outcome data were extracted from medical records. Oncogenic alterations were assessed both individually and by oncogenic pathways, as described by Sanchez-Vega et al. (Cell, 2018). Real-world progression-free survival (PFS) was defined as the time from each treatment initiation to disease progression or death, whichever occurred first. The log-rank test was used to compare PFS among groups, and Cox proportional-hazards models were applied to compute hazard ratios (HRs) and 95% confidence intervals (CIs). Results: A total of 256 patients were included (median age: 49 years, interquartile range [IQR] 43-57; 51% premenopausal). ctDNA was collected following progression on CDK4/6i in combination with either aromatase inhibitors (51%) or fulvestrant (49%). The median time from metastatic diagnosis to ctDNA testing was 36 months (IQR 18-65), with most patients tested after first- (n=135, 53%) and second-line treatment (n=62, 24%). Based on CDK4/6i treatment duration, 11% showed primary resistance (PFS 24 months). Oncogenic alterations associated with reduced benefit from CDK4/6i plus ET were BRCA2 (HR 2.54, 95% CI 1.44-4.49, p<0.001) and of the receptor tyrosine kinase (RTK) pathway (HR 2.96, 95% CI 1.66-5.26, p<0.001). ctDNA analysis revealed PIK3CA (33%) and ESR1 (32%) as the most common actionable alterations, followed by PTEN (6%) and AKT1 (4%), in line with prior evidence. Following ctDNA profiling, patients received oral selective estrogen receptor degraders (SERDs; 10.5%), Pi3k/Akt pathway inhibitors with ET (7%), everolimus with exemestane (17.6%), oral chemotherapy (51.2%), or intravenous chemotherapy (19.1%). Patients with 24 months of exposure to CDK4/6i in 6.1% and 24.1% of cases, respectively. Genomic alterations of the Pi3k/Akt pathway were associated with a trend toward reduced PFS on oral SERDs (HR 4.95, 95% CI 0.99-24.75, p=0.052), while MAPK pathway alterations to reduced benefit from everolimus with exemestane (HR 2.98, 95% CI 1.00-8.88, p=0.050). Oncogenic alterations of ESR1 were correlated with shorter PFS on single-agent chemotherapy (HR 1.99, 95% CI 1.19-3.32, p=0.008). Conclusions: ctDNA-guided molecular profiling after progression on CDK4/6i and ET reveals clinically-relevant genomic alterations that may influence subsequent treatment outcomes in HR+/HER2- mBC. These findings further support integrating ctDNA analysis into treatment planning beyond first-line progression in HR+/HER2- mBC.
Presentation numberPS2-08-23
Association Between Peripheral Eosinophils and Survival Outcomes in HR+HER2- Breast Cancer: An Analysis of 15,262 Patients
Ze-Qing Li, Fudan University Shanghai Cancer Center, Shanghai, China
Z. Li, S. Wu, X. Wu, L. Ge, C. Liu, Y. Jiang, Z. Shao; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Background: Breast cancer exhibits complex immune regulation involving both the tumor microenvironment and systemic circulation. While lymphocyte-dominated immune responses in HR+/HER2- breast cancer have been extensively investigated, the functional roles of granulocytic populations—particularly eosinophils—remain poorly understood, including their clinical relevance, tumor-immune interactions, and spatial organization. Methods: We analyzed a multi-omics discovery cohort (n=472) and a large clinical validation cohort (n=14,790) with matched preoperative peripheral eosinophil counts to assess their prognostic value in HR+/HER2- breast cancer. Integrated analyses of whole-exome sequencing (WES), copy number alterations (CNAs) and RNA sequencing (RNA-seq) revealed distinct genomic and transcriptomic profiles between eosinophil-high and -low groups. The functional roles of eosinophils in tumor immunity were investigated through immunohistochemistry (IHC), adoptive transfer experiments, and flow cytometry analyses. Furthermore, we integrated IHC with digital pathology to map their spatial distribution in tumors. Results: In both the discovery and validation cohorts, higher circulating eosinophil levels were associated with improved survival in HR+/HER2- breast cancer. Genomic analysis revealed that higher circulating eosinophil levels were associated with lower HLA homozygosity. Circulating eosinophils were associated with increased intratumoral immune activation and more eosinophil infiltration within tumors. Tumors with high eosinophil infiltration exhibited significantly stronger anti-tumor immune responses. Mechanistically, eosinophils exerted anti-tumor effects through direct cytotoxicity and recruitment of CD8⁺ T cells, as evidenced by reduced tumor burden in eosinophil-adoptive transfer models. Spatial analysis using digital pathological profiling further suggested that tumor-infiltrating eosinophils were located closest to lymphocytes, followed by stromal cells and tumor cells. Conclusions: Through integrative multi-omics analysis and spatial immune profiling, our study provides evidence for the anti-tumor role of eosinophils in HR+/HER2- breast cancer. These findings establish eosinophils as a potential prognostic biomarker in this subtype, highlighting their relevance in the tumor immune microenvironment and potential utility in risk stratification.
Presentation numberPS2-08-24
Detection of predictive mutations and HER2/neu expression from circulating tumor cells of metastatic breast cancer patients: new opportunities by analyzing high blood volumes obtained by diagnostic leukapheresis
André Franken, University Hospital Duesseldorf, Duesseldorf, Germany
A. Franken1, D. Karayel1, M. Rivandi1, J. Oles2, M. Sudarsanam2, C. Driemel2, R. P. Neves2, N. Krawczyk1, B. Jäger1, F. Meier-Stiegen1, N. Stamm1, B. Alberter3, B. Polzer3, T. W. Friedl4, S. Riethdorf5, B. Volz6, A. Koch6, K. Schäfer7, T. Rau7, J. C. Fischer8, E. Ruckhäberle1, K. Pantel5, W. Janni4, N. H. Stoecklein2, T. Fehm1, H. Neubauer1; 1Department of Obstetrics and Gynecology, University Hospital Duesseldorf, Duesseldorf, GERMANY, 2General, Visceral and Pediatric Surgery, University Hospital Duesseldorf, Duesseldorf, GERMANY, 3Division “Personalized Tumor Therapy”, Fraunhofer Institute for Toxicology and Experimental Medicine, Regensburg, GERMANY, 4Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, GERMANY, 5Department of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, GERMANY, 6Department of Obstetrics and Gynecology, University of Tübingen, Tübingen, GERMANY, 7Department of Pathology, University Hospital Duesseldorf, Duesseldorf, GERMANY, 8Institute for Transplantation Diagnostics and Cell Therapeutics, University Hospital Duesseldorf, Duesseldorf, GERMANY.
Background: Tumor-derived material obtained through liquid biopsy—most commonly circulating tumor DNA (ctDNA)—has become a key biomarker for guiding targeted therapy in metastatic breast cancer. However, ctDNA is not released by all tumors and does not provide information on protein expression. In contrast, circulating tumor cells (CTCs) are intact cells that allow for multiparametric analyses, including both genomic profiling and protein-level assessments such as HER2 expression.Despite these advantages, the clinical utility of CTCs is often limited by their extremely low abundance. To enhance CTC yield, the diagnostic leukapheresis (DLA) offers a powerful alternative. By processing several liters of blood, DLA enables the density-based pre-enrichment of mononuclear cells (MNCs), including rare CTCs.In this study, we compared the utility of CTCs with ctDNA and tissue biopsies to assess their respective and complementary roles as biomarkers in metastatic breast cancer. Material & Methods: To detect CTCs, 7.5 mL blood samples from 139 patients with metastatic breast cancer were analyzed using the CellSearch system, supplemented by immunofluorescence-based assessment of HER2 expression. Of these, 41 samples were retrospectively selected from a cohort of the DETECT-III clinical trial; 98 samples were collected prospectively.Single CTCs were isolated via micromanipulation, followed by whole genome amplification of genomic DNA. Targeted sequencing was performed using a next generation sequencing (NGS) panel covering predictive hotspot mutations in PIK3CA, ESR1, AKT1, and ERBB2, and results were compared with matched tissue biopsies. In parallel, ctDNA isolated from matched plasma samples was analyzed by digital droplet PCR for hotspot mutations in PIK3CA and AKT1.Additionally, from DLA products of eight patients, CTCs were enriched from samples adjusted to 2 × 10⁹ MNCs using the flow enrichment target capture Halbach (FETCH) technology. Single CTCs were then isolated by fluorescence-activated cell sorting and analyzed by NGS. Results: Sequencing of single CTCs was successful in 100% of samples containing ≥18 CTCs. In contrast, samples with lower CTC counts (1-8 CTCs) showed a reduced success rate of 38.5%. In case CTC sequencing was successful, mutations identified in matched tissue biopsies were also detected in CTCs. Notably, in a substantial fraction of patients, additional mutations were identified in CTCs that were not detectable in tissue. In CTC-positive cases (≥1 CTC), a high concordance (82.6%) between detected mutations in CTCs and ctDNA was observed. However, ctDNA failed to detect 50% of the predictive PIK3CA and AKT1 mutations identified in tissue, resulting in a superior sensitivity of CTCs over ctDNA when ≥19 CTCs were available for analysis. As expected, application of DLA significantly increased CTC yield. While standard 7.5 mL blood samples yielded ≥19 CTCs in only 12.5% of patients (≥1 CTCs in 62.5%), DLA yielded ≥19 CTCs and resulted in successful sequencing in all 8 cases. In 62.5% of the DLA samples, mutations were identified that were often not detectable from standard CTC analysis due to low counts or absent CTCs in blood. HER2-positive CTCs were found in 37.5% of the DLA samples. Overall, actionable alterations were identified in CTCs from 87.5% of the DLA patients. Conclusion: If present in sufficient numbers, CTCs enable more sensitive detection of predictive mutations than ctDNA and additionally provide HER2 protein expression data. DLA is critical to achieve the required CTC yield. CTC analysis from DLA products offers a valuable complement to tissue biopsies, particularly when metastases are inaccessible, biopsies require considerable effort, or are declined by patients.
Presentation numberPS2-08-25
Endocrine Sensitivity and Predicted Benefit of Extended Endocrine Therapy based on Breast Cancer Index (BCI) in BRCA1, BRCA2 and CHEK2 Pathogenic Variant Carriers with ER+/HER2- Breast Cancer
Siddhartha Yadav, Mayo Clinic, Rochester, MN
S. Yadav1, B. O’Neal2, S. Yasir3, N. Siuliukina2, A. Karki1, Y. Zhang2, A. K. Anderson2, K. Treuner2, M. P. Goetz1, F. J. Couch3; 1Department of Oncology, Mayo Clinic, Rochester, MN, 2NA, Biotheranostics, Inc., San Diego, CA, 3Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
Introduction: Patients with ER+/HER2- breast cancer associated with germline pathogenic variants (PVs) in BRCA1, BRCA2 and CHEK2 exhibit distinct clinicopathological features compared to sporadic cases. However, the optimal endocrine therapy strategies for these patients remain undefined. The Breast Cancer Index (BCI) is a validated gene expression-based signature that stratifies patients based on the risk of overall (0-10 years) and late (post-5 years) distant recurrence and predicts the likelihood of benefit from extended endocrine therapy (EET) in early-stage, HR+ breast cancer. Here we utilize BCI to investigate endocrine sensitivity and the predicted EET benefit in a cohort of ER+/HER2- breast cancer patients with BRCA1, BRCA2 and CHEK2 PVs. Methods: BRCA1, BRCA2 and CHEK2 PV carriers with ER+/HER2- breast cancer diagnosed at Mayo Clinic between 1996 and 2024 at Mayo Clinic were identified from a registry tracking germline test result. For comparison, we utilized the BCI Registry, a prospective, multi-institutional tumor registry of patients with early-stage ER+/HER2- breast cancer undergoing BCI testing, regardless of germline PV status. The analysis was restricted to women with invasive, early-stage locoregional breast cancer and available pre-treatment tumor specimens. BCI testing was performed by RT-PCR blinded to clinical outcome. BCI prognostic and BCI predictive categories were compared between the Mayo Clinic germline PV cohort and the BCI Registry using descriptive statistics and Fisher’s exact test. Results: The final analysis included 123 PVcarriers (BRCA1: 36; BRCA2: 40; CHEK2: 47) from the MayoClinic cohort and 2,563 women from the BCI Registry. The proportion of patientsdiagnosed before age 50 was higher among PV carriers—52.8% in BRCA1,50.0% in BRCA2, and 21.3% in CHEK2—compared to 7.0% in the BCIRegistry. PV carriers were also more likely to be premenopausal andnode-positive at diagnosis. Based on BCI score, a significant higher proportionof BRCA1 (75.0%, p=0.002) andBRCA2 (70.0%, p=0.01) PV carriers wereclassified as high risk, compared to 48.5% of patients in the BCI Registry, butno significant difference was seen for CHEK2 PV carriers (46.8%,p=0.88). A significantly higher proportion of BRCA1 PV carriers werepredicted to benefit from EET compared to the BCI Registry cohort (77.8% vs.33.9%, p<0.001), whereas no significant differences in BCI Predictivecategories were observed for BRCA2 or CHEK2 PV carriers. Overall,compared to breast cancer cases in the BCI registry, a significant differencein the distribution of the combined BCI prognostic and predictive categorieswas noted for BRCA1 (p<0.001)and BRCA2 (p=0.02) PV carriers,but not for CHEK2 PV carriers (p=0.66). In particular, BRCA1 andBRCA2 PV carriers were enriched for high-risk tumors with a high likelihoodof EET benefit (66.7% and 37.5%, respectively vs 25.1%, p<0.001 and p=0.02),whereas only a small non-significant increase was observed for CHEK2 PVcarriers (29.8% vs 25.1%, p=0.66) compared to breast cancer cases in the BCIRegistry. Conclusions: Genomicclassification by BCI reveals distinct patterns of endocrine sensitivity andpredicted EET benefit among germline PV carriers, particularly in BRCA1and BRCA2 PV carriers. These findings warrant further investigation andmay inform personalized endocrine therapy strategies for women with germlinePV-associated ER+/HER2- breast cancer.
Presentation numberPS2-08-26
Pathologist- and artificial intelligence-based TILs assessment in patients with early triple-negative breast cancer treated with neoadjuvant chemo-immunotherapy: real-world evidence from a nationwide cohort
Ioannis Zerdes, Karolinska Institutet & Karolinska University Hospital, Stockholm, Sweden
I. Zerdes1, A. Papakonstantinou2, B. Acs3, N. Tsiknakis4, S. Steen3, E. Karlsson5, X. Liu4, K. Wang4, S. Agartz4, G. Manikis4, J. Bergh2, J. Hartman3, T. Foukakis2; 1Oncology-Pathology & Theme Cancer, Karolinska Institutet & Karolinska University Hospital, Stockholm, SWEDEN, 2Oncology-Pathology & Breast Center, Karolinska Institutet & Karolinska University Hospital, Stockholm, SWEDEN, 3Oncology-Pathology & Clinical Pathology/Cancer Diagnostics, Karolinska Institutet & Karolinska University Hospital, Stockholm, SWEDEN, 4Oncology-Pathology, Karolinska Institutet, Stockholm, SWEDEN, 5Oncology-Pathology & Nuclear Medicine/Hospital Physics, Karolinska Institutet & Karolinska University Hospital, Stockholm, SWEDEN.
Background: Neoadjuvant chemo-immunotherapy is the standard of care for patients with stage II-III triple negative breast cancer (TNBC). Real-world data on tumor-infiltrating lymphocytes (TILs) indicate a correlation of higher pre-treatment TILs levels with increased pCR rates. Digital pathology and artificial intelligence (AI) tools could provide value towards a more standardized and objective TILs assessment and additional spatial insights. In this study we aimed to assess the performance and prognostic value of both pathologist-based and digital pathology-based TILs scoring methods.Methods: The study population included a nationwide cohort of patients with early TNBC treated with neoadjuvant chemo-immunotherapy according to the KEYNOTE-522 regimen, in 20 Swedish hospitals between 2022-2024. TILs assessment on H&E-stained diagnostic biopsy tissue sections was performed i) manually (sTILs), based on the International Immuno-Oncology Working Group guidelines and ii) digitally (AI-TILs = TILs/Stromal cells). using the previously validated deep learning-based scoring algorithm HoverNet. Correlation with pCR was performed using univariate logistic regression model.Results: 337 patients were included in this nationwide cohort. 51.5% of the patients achieved pCR and 35% had RCB 2/3. Patients with baseline pathologist-based sTILs ≥30% were significantly associated with higher pCR rates and lower RCB score (chi-square test, p<0.0001). Both sTILs and AI-TILs were available for 84 patients and demonstrated a moderate correlation (Spearman’s rho= 0.59, p<0.0001). Both variables (continuous) were significantly associated with increased pCR rates (OR=1.068, 95% CI: 1.04-1.11, p<0.001 for sTLs; OR=1.072, 95% CI: 1.03-1.12, p<0.0001 for AI-based TILs). In non-lymphocyte predominant breast cancer (non-LPBC; sTILs <50%), patients with high AI-TILs (cut-off: median) were associated with higher likelihood of achieving pCR compared to those with low AI-TILs abundance (OR= 4.1; 95% CI: 1.3-14.1, p=0.018) Conclusions: In this nationwide study, both pathologist- and AI-based TILs were predictive for pCR. AI-TILs assessment could potentially contribute to a better response discrimination in patients with non-LPBC treated with neoadjuvant chemo-immunotherapy. Validation in additional clinical cohorts is warranted.
Presentation numberPS2-08-28
Artificial Intelligence-Based Histopathology Model Predicts Recurrence Risk in Older Patients with Early HR+/HER2− Breast Cancer: Results from the Basel University Hospital Cohort
Elena Diana Chiru, Cancer Center Baselland, Liestal, Switzerland
E. D. Chiru1, L. Sojak1, J. Witowski2, K. Zeng2, C. Kurzeder3, S. Muenst4, M. Vetter1; 1Medical Oncology, Cancer Center Baselland, Liestal, SWITZERLAND, 2Medical Oncology, ATARAXIS AI, New York, NY, 3Breast and Gynecological Oncology, Breast Center, Basel, SWITZERLAND, 4Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, SWITZERLAND.
Background: Older patients with hormone receptor-positive (HR+)/HER2-negative (HER2−) early-stage breast cancer (BC) face frequent uncertainty in treatment decisions, as tools like Oncotype DX (ODX) are less validated in this age group. Ataraxis Breast RISK (ATX) is a novel artificial intelligence (AI) model that integrates digitized H&E Histology slides and clinical data to predict recurrence. We assessed the prognostic performance of ATX in patients ≥70 years of age treated at Basel University Hospital (USB) with a focus on reclassification of intermediate-risk ODX cases using ATX. Methods: ATX scores were computed using a locked AI model based on diagnostic histopathology and clinical features. The primary endpoint was disease-free interval (DFI). Harrell’s concordance index (C-index) and hazard ratios (HR) per 0.1 score increase were used to assess prognostic accuracy. We analyzed 267 HR+/HER2− early BC patients from USB, all of whom had prior ODX testing. Subgroup analyses focused on patients ≥70 years (n=40, 5 events) and <50 years (n=41). Reclassification was assessed within the ODX-intermediate-risk group (n=167). Pooled results from five external cohorts (totaling 611 patients ≥70 years) were also referenced for external context. Results: Among USB patients ≥70 years, ATX achieved a C-index of 0.79 (95% CI: 0.51-1.00) and an HR of 2.12 (95% CI: 1.18-3.78), indicating strong predictive accuracy. In contrast, ODX performance was limited (C-index: 0.55; HR: 1.76, p=0.13). In patients <50 years, ATX also demonstrated robust prognostic value (HR: 4.36, p=0.03).Stratifying patients ≥70 by a 10% 5-year recurrence threshold, ATX separated a high-risk group (n=31) with substantially worse DFI from a low-risk group (n=9), supporting its potential clinical utility.Within the ODX-intermediate-risk group (n=167), ATX reclassified 128 patients (77%) to low risk and 39 (23%) to high risk. Although the separation in DFI curves was less pronounced (HR: 1.61, p=0.393; C-index: 0.69), ATX offered definitive risk categorization, addressing the ambiguity inherent to ODX intermediate scores. Across 5 external validation cohorts including 611 patients ≥70 years, ATX maintained strong performance with a pooled C-index of 0.74 (95% CI: 0.70-0.81) and HR of 1.82 (95% CI: 1.51-2.20), reinforcing its generalizability in older BC populations. Conclusions: ATX provides high prognostic accuracy in older HR+/HER2− BC patients and outperforms ODX in the USB cohort. It consistently stratifies patients across age groups and resolves the uncertainty associated with intermediate genomic risk scores. These findings highlight ATX’s potential to support more individualized, evidence-based CHT decisions in elderly patients—overlooked population in BC risk modeling.
Presentation numberPS2-08-29
Plasma hepcidin as a prognostic biomarker in obese and non-obese patients with early breast cancer (BC): a pooled analysis of two prospective cohort studies
Evan A Cescon, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
E. A. Cescon1, P. J. Goodwin2, W. Jacot3, A. Bobrie3, S. Thezenas3, T. Ganz4, E. Nemeth4, A. E. Lohmann5, M. Ennis6, K. J. Jerzak7; 1Sunnybrook Research Institute, Evaluative Clinical Sciences, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA, 2Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, CANADA, 3Department of Medical Oncology, Institut du Cancer de Montpellier, Universite de Montpellier, Montpellier, FRANCE, 4Department of Medicine, University of California Los Angeles, Los Angeles, CA, 5Department of Oncology, London Health Sciences Centre, University of Western Ontario, London, ON, CANADA, 6Department of Biostatistics, Sunnybrook Health Sciences Centre, Toronto, ON, CANADA, 7Department of Medicine, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, CANADA.
Background: BC is one of the leading causes of cancer death among women globally, with obesity recognized as a risk factor for adverse clinical outcomes. Chronic inflammation and metabolic dysregulation, often observed in obese individuals, are hypothesized to drive this risk. The potential prognostic significance of the peptide hormone hepcidin, which is a well-known marker of inflammation and iron dysregulation, is therefore of interest in patients with BC. Methods: We performed a pooled analysis of individual patient data within two prospective cohort studies, the “Canadian” (n=518; Jerzak et al, 2020) and “French” (n=144; Durigova et al., 2013) cohorts. Plasma hepcidin levels were measured postoperatively and prior to systemic therapy initiation using the same gold-standard and commercially available ELISA assay (La Jolla, California). Distant disease-free survival (DDFS) was defined as the time between BC diagnosis and first distant recurrence. Overall survival (OS) time was defined as time between BC diagnosis and death from any cause.Cox regression models were fitted using both data sets, treating hepcidin as a continuous variable and with a hepcidin-by-BMI interaction (BMI dichotomized at 30). Cohort-specific ‘beta’ coefficients and standard errors (SE) from each survival model were combined meta-analytically using a random effect model (using the R package ‘metafor’), with results presented as hazard ratios (HRs) and 95% confidence intervals (CIs). HRs are expressed as the hazard at the 75th vs the 25th percentile of the hepcidin distribution in the combined datasets: P25=11.6, P75=31.3 ng/ml. Results: In total, 622 patients were included. At BC diagnosis, 46% were age ≥50, 41% were post-menopausal and 16% were obese (BMI ≥30). Most patients had T1 (57%) or T2 (32%) tumors and 66% had N0/N1mi disease; 75% of BCs were hormone receptor positive in the pooled population and 22% were HER2+ in the French cohort. HER2 status in the Canadian cohort is unknown. Over a median follow-up of 11.6 (95% CI 11.5-11.8) years, 145 patients (22%) experienced a distant relapse and 139 (21%) died.In our pooled analysis of obese BC patients, a significant prognostic interaction between hepcidin levels and obesity was observed. Specifically, among obese women (BMI ≥ 30), elevated plasma hepcidin was significantly associated with shorter DDFS (adjusted HR 2.23; 95% CI 1.23-4.07) and shorter OS (adjusted HR 1.87; 95% CI 1.03-3.39). Conversely, no statistically significant associations were identified among non-obese women (BMI <30) for either DDFS (HR 0.84; 95% CI 0.58-1.22) or OS (HR 1.00; 95% CI 0.72-1.38). Conclusion: Higher plasma hepcidin is independently linked to shorter DDFS and OS in obese, but not non-obese, women with early stage BC. Underlying mechanisms warrant further study.
| Outcome | Coefficient | SE of coefficient | HR (95% CI) | Interaction P-value | |
| Distant Disease-Free Survival for Hepcidin 31.3 vs 11.6 | Canadian cohort | 0.024 | |||
| BMI<30 | -0.062 | 0.1751 | 0.94 (0.67-1.33) | ||
| BMI≥30 | 0.735 | 0.3170 | 2.09 (1.12-3.88) | ||
| French cohort | 0.068 | ||||
| BMI<30 | -0.494 | 0.3514 | 0.61 (0.31-1.22) | ||
| BMI≥30 | 1.713 | 1.1554 | 5.55 (0.58-53.38) | ||
| Meta-analysis | |||||
| BMI<30 | -0.170 | 0.1873 | 0.84 (0.58-1.22) | ||
| BMI≥30 | 0.803 | 0.3057 | 2.23 (1.23-4.07) | ||
| Overall Survival for Hepcidin 31.3 vs 11.6 | Canadian cohort | 0.113 | |||
| BMI<30 | 0.037 | 0.1798 | 1.04 (0.73-1.48) | ||
| BMI≥30 | 0.587 | 0.3090 | 1.80 (0.98-3.29) | ||
| French cohort | 0.249 | ||||
| BMI<30 | -0.225 | 0.4135 | 0.80 (0.37-1.8) | ||
| BMI≥30 | 1.866 | 1.7603 | 6.46 (0.21-203) | ||
| Meta-analysis | |||||
| BMI<30 | -0.005 | 0.1649 | 1.00 (0.72-1.38) | ||
| BMI≥30 | 0.625 | 0.3043 | 1.87 (1.03-3.39) |
Presentation numberPS2-09-01
Optimizing Transfer Learning for Early Prediction of Pathologic Complete Response in Breast Cancer: A Multicenter Study Using Pretreatment DCE-MRI
Kanika Bhalla, Washington university School of Medicine in st louis, St. Louis, MO
K. Bhalla1, A. Sanchez1, J. Luna1, E. Podany2, T. Ahmad1, D. Bennett1, A. Davis2, A. Gastounioti1; 1Radiology, Washington university School of Medicine in st louis, St. Louis, MO, 2Medicine, Washington university School of Medicine in st louis, St. Louis, MO.
Background: Pathologic complete response (pCR) after neoadjuvant therapy (NAT) in breast cancer is associated with improved outcomes; however, pCR rate varies dramatically (e.g., from 8-65%) depending on molecular subtype and choice of therapy. While transfer learning from pretrained deep learning models has shown promise in advancing early prediction of pCR with DCE-MRI, the impact of the pretraining data domain remains underexplored, particularly across molecular subtypes. Objective: To evaluate the impact of general-domain versus domain-specific pretraining in transfer learning for pCR prediction from pretreatment DCE-MRI. Materials and Methods: We leveraged the multicenter MAMA-MIA dataset of pretreatment DCE-MRI from patients undergoing NAT (1357 patients; pCR: 424, non-pCR: 933) to evaluate ResNet-50 models pretrained on (1) ImageNet: a multi-source dataset of 1.28M natural images and thousands of labels, and (2) RadImageNet: a single-site dataset of 1.35M radiologic images and 185 labels. Both models were fine-tuned (top 10% layers) and evaluated for pCR prediction with pretreatment DCE-MRI, using identical hyperparameters and five-fold stratified cross-validation preserving center ratios. Predictive performance was assessed overall and across tumor subtypes: luminal A, luminal B, triple-negative, and HER2-enriched. Areas under the curve (AUCs) were compared using DeLong’s test. The combined performance of the best fine-tuned model and clinical features (HER2, HR status) was also assessed. Results: Transfer learning from ImageNet significantly outperformed RadImageNet in pCR prediction with pretreatment DCE-MRI overall (AUC: 0.63 vs. 0.59; p = 0.02). The performance of both fine-tuned models varied across different tumor subtypes, with statistically significant AUC gains observed for ImageNet relative to RadImageNet in luminal A (0.66 vs. 0.61; p = 0.01) and HER2-enriched tumors (0.68 vs. 0.53; p = 0.009). No significant differences between the two models were observed for luminal B or triple-negative tumors. Combination of the fine-tuned ImageNet model with clinical features (HER2, HR status) further enhanced pCR prediction, with the combined model achieving an AUC of 0.68. Conclusion: Based on our preliminary findings, ImageNet outperformed RadImageNet in transfer learning for predicting pCR from pretreatment breast DCE-MRI. This superior performance is likely due to the richer and more diverse features learned from the multisource general-domain images in ImageNet. Furthermore, the tumor subtype significantly influenced the performance of both models, and performance improvement with ImageNet. Our results provide valuable insights towards optimizing the use of transfer learning in predictive modeling for early prediction of pCR with DCE-MRI in breast cancer patients undergoing NAT.
| Group | Model | AUC [95% CIs] | Balanced Accuracy [95% CIs] | PPV [95% CIs] | NPV [95% CIs] | p-value for AUC diff. | ||||||||||||||
| Overall (N = 1357) | ImageNet | 0.63 [0.59, 0.66] | 0.59 [0.58, 0.61] | 0.41 [0.40, 0.43] | 0.75 [0.74, 0.77] | p = 0.02 | ||||||||||||||
| RadImageNet | 0.59 [0.57, 0.61] | 0.56 [0.54, 0.58] | 0.39 [0.37, 0.40] | 0.73 [0.71, 0.75] | ||||||||||||||||
| Luminal A (N = 359) | ImageNet | 0.66 [0.60, 0.73] | 0.58 [0.53, 0.64] | 0.23 [0.13, 0.33] | 0.88 [0.85, 0.91] | p = 0.01 | ||||||||||||||
| RadImageNet | 0.61 [0.58, 0.65] | 0.60 [0.55, 0.64] | 0.24 [0.16, 0.32] | 0.88 [0.81, 0.94] | ||||||||||||||||
| Luminal B (N = 137) | ImageNet | 0.63 [0.54, 0.72] | 0.58 [0.47, 0.70] | 0.49 [0.30, 0.68] | 0.68 [0.62, 0.74] | p = 0.50 | ||||||||||||||
| RadImageNet | 0.59 [0.52, 0.67] | 0.54 [0.49, 0.59] | 0.48 [0.42, 0.55] | 0.62 [0.54, 0.71] | ||||||||||||||||
| Triple-negative (N = 466) | ImageNet | 0.59 [0.50, 0.68] | 0.58 [0.54, 0.61] | 0.45 [0.41, 0.49] | 0.70 [0.64, 0.75] | p = 0.79 | ||||||||||||||
| RadImageNet | 0.60 [0.56, 0.65] | 0.58 [0.55, 0.62] | 0.50 [0.46, 0.53] | 0.68 [0.63, 0.73] | ||||||||||||||||
| HER2-enriched (N = 143) | ImageNet | 0.68 [0.64, 0.73] | 0.65 [0.58, 0.72] | 0.68 [0.56, 0.80] | 0.63 [0.52, 0.74] | p = 0.009 | ||||||||||||||
| RadImageNet | 0.53 [0.45, 0.61] | 0.51 [0.47, 0.54] | 0.49 [0.35, 0.63] | 0.50 [0.44, 0.56] | ||||||||||||||||
| Overall (N = 1357) | ImageNet + clinical | 0.68 [0.62-0.75] | 0.64 [0.61-0.67] | 0.41 [0.40, 0.43] | 0.84 [0.80, 0.89] | N/A |
Presentation numberPS2-09-02
Neutrophil extracellular traps signature to predict pathological complete response in patients with triple negative breast cancer after neoadjuvant systemic therapy
Takeo Fujii, National Cancer Institute, Bethesda, MD
T. Fujii1, S. You2, H. Furuya3, M. Nomura4, M. Muto4; 1Women’s Mlignancies Branch, National Cancer Institute, Bethesda, MD, 2Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, 3Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, 4Department of Clinical Oncology, Kyoto University Hospital, Koyo-city, JAPAN.
BACKGROUND: While neoadjuvant systemic therapy has improved outcomes in triple-negative breast cancer (TNBC), patients who do not achieve a pathological complete response (pCR) remain at high risk of recurrence and poor survival. As pCR is a surrogate marker for long-term outcomes and a key endpoint in early-stage TNBC trials, identifying patients unlikely to achieve pCR is critical for optimizing therapy. There is an unmet need for predictive biomarkers to guide treatment decisions. Neutrophils, abundant in the tumor microenvironment, form neutrophil extracellular traps (NETs), which are reported to have a context-dependent role in promoting or suppressing tumor progression and metastasis. We hypothesize that a NET-related gene signature predicts pCR following neoadjuvant systemic therapy in TNBC. Our primary objective is to develop and externally validate a NET-based predictive score using publicly available transcriptomic datasets in TNBC. MATERIALS AND METHODS: We used a transcriptomic dataset (GSE1238545) of baseline pretreatment tumor biopsies from patients with TNBC who underwent neoadjuvant systemic therapy to develop a transcriptome-based NET-related gene score, termed “TNBC-NET score” (training cohort). Differentially expressed genes (DEGs) between pCR and non-pCR groups were identified using a false discovery rate (FDR) <0.1 and log2 fold change≥0.3 as cut-off. The TNBC-NET score was externally validated in an independent TNBC transcriptomic dataset (GSE164458; validation cohort). Logistic regression model was used to assess the association between the TNBC-NET score and pCR. Fisher’s exact test was used to compare categorical variables. Gene set enrichment analysis (GSEA) was performed to identify pathways significantly enriched between high and low TNBC-NET score groups. RESULTS: Among 131 previously reported NET-related genes, 13 were significantly upregulated in patients who achieved pCR (n=16) compared to those who did not (n=23) in the training cohort. A TNBC-NET score was calculated for each patient based on the expression of these 13 genes. In logistic regression models treating the TNBC-NET score as a continuous variable, a higher score was significantly associated with increased likelihood of pCR in both the training (n=39; odds ratio [OR]: 4.13, 95% confidence interval [CI]: 1.06-16.1, p=0.04) and validation (n=160; OR: 3.22, 95% CI: 1.52-6.80, p=0.002) cohorts. When the validation cohort was stratified into high and low TNBC-NET score groups using the median as the cutoff, the pCR rate was significantly higher in the high-score group compared to the low-score group (71.7% vs. 43.3%, p=0.003). Gene set enrichment analysis (GSEA) revealed that the epithelial-mesenchymal transition (EMT) pathway was enriched in tumors from the low TNBC-NET score group, which was associated with a lower likelihood of achieving pCR. CONCLUSIONS: We developed and validated a 13-gene TNBC-NET score predictive of pCR following neoadjuvant systemic therapy in TNBC. The EMT pathway was enriched in patients with low TNBC-NET scores, suggesting a potential role in treatment resistance. The TNBC-NET score may serve as a clinically actionable biomarker to guide personalized treatment and risk-adapted clinical trial enrollment aimed at improving pCR rates. Prospective studies are warranted to validate its utility and investigate resistance mechanisms in TNBC-NET score low group.
Presentation numberPS2-09-03
Th1-biased cytokine signatures as biomarkers of clinical benefit following SV-BR-1-GM cancer vaccination in breast cancer.
Pravin Kesarwani, BriaCell Therapeutics, Philadelphia, PA
P. Kesarwani1, S. Modi2, G. Woodfield1, B. Bayer3, V. Bhardwaj1, S. Pachhal1, G. D. Priore3, C. L. Wiseman3, W. V. Williams3, M. Lopez-Lago1; 1R&D, BriaCell Therapeutics, Philadelphia, PA, 2Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 3Clinical Trials Research, BriaCell Therapeutics, Philadelphia, PA.
Background: Although cancer immunotherapy has shown great promises, many cancers remain resistant to treatment due to tumor heterogeneity, immune tolerance, and insufficient immune activation. Bria-IMT is a therapeutic cancer vaccine composed of SV-BR-1-GM, a breast cancer cell line engineered to release GM-CSF. This therapy has completed a phase I/II clinical trial (NCT03066947) and is under evaluation in a phase III study (NCT06072612) for advance metastatic breast cancer. Mechanistically, SV-BR-1-GM cells function as antigen-presenting cells, directly activating CD4+ T cells in an antigen-specific, HLA-restricted manner (as shown in vitro). They also deliver a broad repertoire of cancer antigens to stimulate endogenous immune responses. Building on evidence that suggests changes in circulating cytokines and chemokines reflect treatment-induced immune activation, this study aims to characterize serum cytokine and chemokine profiles as potential biomarkers of immunological response and clinical outcome following cancer immunotherapy. Methods: This retrospective analysis evaluated 35 different cytokines/chemokines in serum samples collected from 30 patients enrolled in the Phase I/II trial (NCT03066947) of Bria-IMT alone or in combination with an anti-PD-1 checkpoint inhibitor (CPI). Serum was isolated from blood pre- and post-therapy. We used multiplex ELISA using meso-scale-discovery (MSD) technology to measure 35 different cytokines/chemokines known to be associated with immune regulation. Data was analyzed using MSD discovery workbench, and plotted using Origin 2025. Results: Patients were categorized by RECIST into stable disease (SD, n=13), progressive disease (PD, n=10), partial responders (PR, n=5), and non-evaluable (NE, n=2). Following three or more treatment cycles, patients with SD and PR demonstrated increased levels of proinflammatory cytokines IL-2 and IL-15 compared to baseline; by contrast, an increase was not observed in patients with PD. IL-2 is produced by activated T cells and supports their proliferation, while IL-15, typically presented by dendritic cells and monocytes, promotes NK cell and memory CD8⁺ T cell activation. This dual increase suggests coordinated activation of both the adaptive and innate immune compartments. MCP-1 (CCL2) and IP-10 (CXCL10), chemokines involved in the recruitment of monocytes, T cells, and dendritic cells, were also significantly elevated in patients with SD and PR. Both chemokines are known to be secreted by activated dendritic cells, further suggesting dendritic cell engagement in response to therapy. In contrast, these cytokines and chemokines did not increase in patients with PD. Importantly, immunosuppressive cytokines such as IL-10, IL-13, and IL-4 were not upregulated following treatment. This cytokine profile indicates a Th1-skewed, proinflammatory immune response associated with disease control. The absence of such changes in PD patients suggests a lack of effective immune activation in those with progressive disease. Conclusion: SV-BR1-GM induces a cytokine profile consistent with a Th1-polarized immune response, marked by elevated IL-2 and IL-15 in patients with clinical benefit (SD and PR). These cytokines, along with increased levels of MCP-1 and IP-10, reflect enhanced activation and recruitment of cytotoxic T cells, NK cells, and antigen-presenting cells. In contrast, there was no induction of Th2- or regulatory-associated cytokines such as IL-4, IL-10, or IL-13, suggesting that the vaccine does not promote an immunosuppressive or tolerogenic environment. This Th1-biased response aligns with effective anti-tumor immunity and suggests that serum cytokine profiles may serve as informative biomarkers of immunologic activity and clinical benefit following SV-BR-1-GM vaccination.
Presentation numberPS2-09-04
Predicting outcomes for patients with mixed ductal/lobular carcinoma of the breast based on circulating tumor DNA positivity patterns
Julia Foldi, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA
J. Foldi1, S. Satta2, C. Palsuledesai2, S. Oesterreich1, A. Lee1, J. McKenzie2, A. Rodriguez2, E. Kalashnikova2, M. Liu2, M. Balic1; 1Department of Medicine, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 2Oncology, Natera, Inc., Austin, TX.
Background: Invasive lobular carcinoma (ILC) exhibits distinct histopathologic characteristics and prognoses when compared to breast carcinomas of no special type (NST). However, mixed NST/ILC, an uncommon and less studied subtype, poses unique clinical challenges due to its mixed histology and heterogeneous expression of hormone receptors and HER2. In this study, we developed a quantitative model to classify these tumors based on their “NST-like” or “ILC-like” molecular relapse patterns as defined by circulating tumor DNA (ctDNA) results. Methods: In this study, patients with early-stage breast cancer (eBC) were identified using Natera’s proprietary real-world database, which is linked to Komodo’s Healthcare Map® claims database and available results from tumor-informed ctDNA testing (SignateraTM, Natera, Inc.). Receptor status and the date of relapse were inferred from the claims codes associated with interventions and secondary neoplasms. The quantitative model developed to classify tumors involved comparing mixed NST/ILC ctDNA profiles against cumulative ctDNA positivity curves of canonical NST and ILC using squared error minimization technique. To validate the model, we performed a separate analysis based on clinical recurrences. We assigned each patient with mixed NST/ILC who recurred to the “NST-like” or “ILC-like” category by comparing their clinical recurrence behavior against the subtype-specific cumulative molecular relapse distributions derived from known NST and ILC cases. Thus, the concordance between ctDNA-based and recurrence-based classifications was assessed. Results: A total of 10,122 patients with eBC (NST, N=7,073; ILC, N=1,147; mixed NST/ILC, N=271) were identified with inferred clinical outcomes and longitudinal ctDNA results for analysis. ctDNA-positivity at any time point after surgery was observed in 47 patients with mixed NST/ILC, of whom 16 (34%) were classified as “NST-like” and 31 (66%) as “ILC-like” by the ctDNA-based classification model. A secondary neoplasm was reported in 21/47 (44.6%) of patients. A 100% (21/21) concordance was observed between the ctDNA-based and recurrence-based classifications. We next analyzed the mutational landscape of newly classified tumors using whole-exome sequencing data. Interestingly, for “NST-like” tumors, TP53 and CDH1 were mutated in 38% (6/16) of cases, followed by PIK3CA and USP40 (31%, 5/16). For “ILC-like tumors”, the most frequently altered genes were: TP53 (45%, 13/29), PIK3CA (34%, 10/29), UNC13C (28%, 8/29), and CDH1 (21%, 6/29). Conclusion: In phenotypically diverse disease types, such as mixed NST/ILC, ctDNA-based relapse patterns can help predict the clinical trajectory of the disease and discriminate between “NST-like” vs “ILC-like” phenotype. Upon further refinement and validation, the model can potentially inform prognosis among patients with mixed NST/ILC.
Presentation numberPS2-09-05
Feasibility of Virtual RNA Inference from Histopathology to Identify Gene Signatures of Neoadjuvant Chemotherapy and Immune Checkpoint Inhibitor (ICI) Response in Triple-Negative Breast Cancer (TNBC)
Yuan Yuan, Cedars Sinai Medical Center, Los Angeles, CA
Y. Yuan1, V. Pujara2, J. Lownik2, G. Murray3, J. Bitar1, L. Chen1, P. Najafzadeh3, K. Dabirian3, D. Lin1, F. Kolling IV4, J. Marotti5, L. Vaickus5, K. Yao2, L. Vahdat6, K. Le2, Z. Azher2, J. Levy2; 1Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA, 2Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, 3Department of Medicine, Huntington Health, Pasadena, CA, 4Center for Quantitative Biology, Dartmouth College Geisel School of Medicine, Lebanon, NH, 5Department of Pathology and Laboratory Medicine, Dartmouth Health, Lebanon, NH, 6Department of Medicine, Dartmouth Health, Lebanon, NH.
Background: TNBC remains a biologically aggressive subtype with limited biomarkers to predict pathologic response in the neoadjuvant setting. While spatial transcriptomics (ST) can reveal features of the tumor microenvironment (TME) associated with response and resistance, its clinical application is limited by cost and scalability. As part of ongoing efforts to investigate mechanisms of resistance to neoadjuvant chemotherapy and ICI in breast cancer, we evaluated the feasibility of virtual RNA inference (VRI)—a deep learning approach for prediction of spatial gene expression from routine H&E-stained slides. We hypothesized that inferred ST patterns localized to the tumor could predict pathologic response. Methods: To develop VRI, development cohort of 25 TNBC cases from Dartmouth Health was assembled, comprising primarily of surgical specimens with matched Visium ST data. All slides were sectioned at 5 μm and scanned at 40x (Leica Aperio GT450). A Vision Transformer model (UNI), pretrained on large-scale histopathology datasets, was fine-tuned to predict expression values for a 1,000-gene panel at the level of individual Visium spots (~55 μm resolution), resulting in over 125,000 paired image-expression samples. Spearman correlations were computed per gene using cross-validation.For the validation cohort, H&E slides from retrospective TNBC resections (Cedars-Sinai, 20x, Leica Aperio GT450, Hamamatsu Nanozoomer S360, 01/2018-07/2024) were collected from patients who received neoadjuvant chemotherapy±ICI (N=58 total, 28 pCR, 20 ICI). VRI was applied to predict spatial gene expression. A deep-learning-based semantic segmentation model trained on over 2,000 annotated cases was used to segment tumor and various TME compartments (e.g., tumor stroma, tumor infiltrating lymphocytes). Inferred gene expression was averaged over intratumoral regions to generate patient-level profiles. Differential gene expression, adjusting for ICI, cross-validation, and pathway analysis were used to identify spatially localized gene signatures associated with complete vs. partial pathologic response to ICI. Results: In the development cohort, VRI achieved a median Spearman correlation of 0.55 across 1,000 genes, demonstrating reliable inference of ST from histology. Application to the Cedars-Sinai validation cohort identified a tumor-specific gene expression signature associated with complete pathologic response. A few of many notable predictive genes included LAD1, MDK, Ki67, and CDKN2A, enriched in hallmark cancer pathways such as E2F targets (p < 0.001), G2M checkpoint (p < 0.001), and epithelial-mesenchymal transition (p = 0.03), among others. A composite score derived from these pathways effectively stratified responders from non-responders, achieving an AUC of 0.82 (sensitivity=0.77, specificity=0.73). Ongoing works include expansion of include cohort size, additional subgroup analysis to factor in treatment regimens (e.g., ICI), and adjust for clinical covariates such as stage, grade, residual tumor, and tumor cellularity. Conclusion: This pilot study demonstrates the feasibility of using VRI to infer tumor-specific gene expression patterns predictive of neoadjuvant chemotherapy response from standard H&E slides in TNBC. Inferred spatial transcriptomic features capture biologically relevant response signatures without the need for molecular assays. Future work will formulate inferred ST into spatially resolved TME features—such as immune cell distribution and stromal context—that may inform drug response and guide future biomarker discovery.
Presentation numberPS2-09-08
Tertiary lymphoid structure (TLS)-related gene expression and outcomes in HER2-Positive (HER2+) breast cancer (BC) in the Real-World Database
Saranya Chumsri, Mayo Clinic, Jacksonville, FL
S. Chumsri1, S. Deshmukh2, Y. Liu3, S. Wu4, J. Xiu4, N. Norton5, S. Shipra Gandhi6, E. Torres7, M. Lustberg8, E. Perez1, A. Nassar9, G. Sledge4, E. Thompson5, K. Knutson10; 1Hematology Oncology, Mayo Clinic, Jacksonville, FL, 2N/A, Caris Life Sciences, Phoenix, AZ, 3Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 4Caris Life Sciences, Caris Life Sciences, Phoenix, AZ, 5Cancer Biology, Mayo Clinic, Jacksonville, FL, 6Hematology Oncology, Emory, Atlanta, GA, 7Hematology Oncology, University of Southern California, Los Angeles, FL, 8Hematology Oncology, Yale Cancer Center, New Haven, CT, 9Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 10Cancer Immunology, Mayo Clinic, Jacksonville, FL.
Background: Tertiary lymphoid structures (TLS), which are organized ectopic lymphoid aggregates resembling secondary lymphoid organs, have been associated with improved outcomes across various cancer types. However, limited data exist on the prognostic relevance of TLS in HER2+ BC pts treated with trastuzumab. In our prior work, we demonstrated that the presence of functional TLS, defined by lymphoid aggregates with elevated expression of BCL6 or IL21R, correlated with improved outcomes in the NCCTG N9831 trial. In this study, we evaluated TLS-related gene and outcomes in the real-world database. Methods: 1344 HER2+ (HR-HER2+: 560; HR+HER2+: 784) BC samples were analyzed via NGS (592-gene panel, NextSeq; WES/WTS, NovaSeq; Caris Life Sciences). Above (high) or below (low) the 50th percentile expression levels of TLS-related genes were used to calculate overall survival (OS). IL21R-high (H) and -low (L) tumors were classified by RNA expression above or below the 50th percentile. Real-world OS was derived from insurance claims and calculated from date of biopsy to last contact using Kaplan-Meier. Statistical significance was assessed using chi-square and Mann-Whitney U with multiple comparison adjustments (q < 0.05).Results: Among pts with HR-HER2+ BC, high expression of IL21R, CXCL9, CD38, and CXCL10 were associated with improved OS (Table). In contrast, among pts with HR+HER2+ BC, high expression of CXCR3, PDCD1, TBX21, CD200 and IL21R, were associated with improved OS. In both HR-HER2+ and HR+HER2+, IL21R-H groups had 1) higher infiltration of B cells (HR-HER2+: 4.4% vs 2.9%; HR+HER2+: 5.6% vs 4.2%), M1 MΦ (HR-HER2+: 3.9% vs 2.6%; HR+HER2+: 3% vs 2.1%), M2 MΦ (HR-HER2+: 4% vs 3.2%; HR+HER2+: 5.7% vs 4.4%), NK cells (HR-HER2+: 2.8% vs 2.5%; HR+HER2+: 2.7% vs 2.4%), CD8+ T cell (HR-HER2+: 0.9% vs 0%; HR+HER2+: 0.7% vs 0%), Tregs (HR-HER2+: 2.9% vs 1.4%; HR+HER2+: 2.8% vs 1.5%), all q<.05; 2) higher T cell inflamed score (HR-HER2+: 87.5 vs -95; HR+HER2+: 71 vs -86), IFNy score (HR-HER2+: -0.19 vs -0.48; HR+HER2+: -0.27 vs -0.49), all q<.05; 3) higher expression levels of MHC class I genes (HLA-A, HLA-B, HLA-C, HLA-E, FC: 1.6-2.3; all q<.05), MHC class II genes (HLA-DQA1, HLA-DOA, HLA-DPA1, HLA-DMA, FC: 2.2-3.2; all q<.05) compared to IL21R-L group.Conclusion: Consistent with our prior findings, overexpression of TLS-related genes was associated with improved outcomes in HER2+ BC in real-world data. Notably, the prognostic impact of TLS-related genes differed by hormone receptor (HR) status. Among these, IL21R emerged as the only gene significantly associated with clinical outcomes across both datasets and HR subgroups. Higher IL21R expression is associated with more B cell, M1 MΦ, CD8 T cell and NK cells. These findings underscore the potential importance of the IL21/IL21R axis in HER2+ BC and warrant further investigation to validate its role and therapeutic relevance.
| HR-HER2+ | HR-HER2+ | HR-HER2+ | HR-HER2+ | HR-HER2+ | HR+HER2+ | HR+HER2+ | HR+HER2+ | HR+HER2+ | HR+HER2+ | |
| HR | 95% CI | High | Low | p-value | HR | 95% CI | High | Low | p-value | |
| IL21R | 0.77 | 0.61-0.97 | 41.7 m (N=280) | 35.2 m (N=280) | 0.02 | 0.77 | 0.62-0.93 | 50.5 m (N=390) | 43.6 m (N = 390) | 0.009 |
| CXCL9 | 0.73 | 0.58-0.92 | 41.9 m (N=280) | 35.2 m (N=280) | 0.007 | 0.83 | 0.68 – 1 | 50 m (N=390) | 46.3 m (N = 390) | 0.08 |
| CD38 | 0.79 | 0.63-0.99 | 41.71 m (N=280) | 35.4 m (N=280) | 0.04 | 0.94 | 0.77 – 1.1 | 47.6 m (N=390) | 47.8 m (N = 390) | 0.5 |
| CXCL10 | 0.69 | 0.55-0.87 | 46.5 m (N=280) | 33 m (N=280) | 0.002 | 0.95 | 0.77 – 1.2 | 43.8 m (N=390) | 49 m (N = 390) | 0.62 |
| CXCR3 | 0.88 | 0.7-1.1 | 39.7 m (N=280) | 36.1 m (N=280) | 0.28 | 0.74 | 0.6-0.9 | 53 m (N=390) | 42.3 m (N = 390) | 0.003 |
| PDCD1 | 0.84 | 0.66-1 | 39.7 m (N=280) | 36.6 m (N=280) | 0.14 | 0.77 | 0.63-0.94 | 53.3 m (N=390) | 43.5 m (N = 390) | 0.01 |
| TBX21 | 0.84 | 0.66-1 | 38 m (N=280) | 36.6 m (N=280) | 0.14 | 0.81 | 0.66-0.99 | 49.5 m (N=390) | 45.2 m (N = 390) | 0.03 |
| CD200 | 0.85 | 0.68-1 | 41.7 m (N=280) | 35.5 m (N=280) | 0.18 | 0.73 | 0.66-0.1 | 49.6 m (N=390) | 45.4 m (N=390) | 0.04 |
Presentation numberPS2-09-09
Early on-treatment PD-L1 expression and stromal tumor infiltrating lymphocytes (sTILs) refine the prediction of EFS in the NeoTRIP trial
Matteo Dugo, IRCCS Ospedale San Raffaele, Milan, Italy
M. Dugo1, G. Viale1, H. Chiun-Sheng2, D. Egle3, M. Callari4, R. H. Ali5, B. Bermejo6, C. Zamagni7, X. Wang5, M. Thill8, A. Antón-Torres9, S. Russo10, E. Ciruelos11, R. Greil12, V. Semiglazov13, M. Colleoni14, L. Del Mastro15, G. Viale16, L. Gianni4, G. Bianchini1; 1Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, ITALY, 2Department of Medical Oncology, National Taiwan University Hospital, College of Medicine, National Taiwan University and Taiwan Breast Cancer Consortium, Taipei, TAIWAN, 3Department of Obstetrics and Gynecology, Medical University Innsbruck, Innsbruck, AUSTRIA, 4Fondazione Michelangelo, Fondazione Michelangelo, Milan, ITALY, 5Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UNITED KINGDOM, 6Department of Medical Oncology, Hospital Clínico Universitario de Valencia, Biomedical Research Institute INCLIVA, Valencia, SPAIN, 7Dipartimento Malattie Oncologiche ed Ematologiche, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, ITALY, 8Klinik für Gynäkologie und Gynäkologische Onkologie, Agaplesion Markus Krankenhaus, Frankfurt Am Main, GERMANY, 9Departamento de Medicina, Psiquiatría y Dermatología, Universidad de Zaragoza, Instituto Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, Zaragoza, SPAIN, 10Dipartimento di Area Medica, Azienda Sanitaria Universitaria Friuli Centrale, Udine, ITALY, 11Servicio Oncología Médica, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 12Department of Internal Medicine III, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute-CCCIT, Cancer Cluster Salzburg, Salzburg, AUSTRIA, 13Breast Cancer Department, N.N.Petrov Research Institute of Oncology, Saint Petersburg, RUSSIAN FEDERATION, 14Senologia Medica, Istituto Europeo di Oncologia IRCCS, Milan, ITALY, 15Clinica di Oncologia Medica, University of Genova; IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 16Dipartimento di Patologia e Medicina di Laboratorio, Istituto Europeo di Oncologia IRCCS, Milan, ITALY.
Background We previously showed that on-treatment high sTILs were significantly associated with improved EFS and were independent of pathological complete response (pCR) in absence or upon addition of atezolizumab to neoadjuvant chemotherapy. The same is true for PD-L1 positivity but only for patients receiving immunotherapy (Bianchini G ASCO 2025). We hypothesized that on-treatment up-regulation of PD-L1 may be a new marker of benefit from neoadjuvant atezolizumab. Here we analyzed pre-treatment RNA-Seq and imaging mass cytometry (IMC) data to identify baseline features that could predict PD-L1 up-regulation during treatment. We next assessed whether combining on-treatment sTILs and PD-L1 improves prediction of EFS. Methods NeoTRIP randomized 280 patients to nab-paclitaxel/carbo for 8 cycles, alone (CT) or with atezolizumab (CT/A). As Per-Protocol Population, 258 patients were evaluable for EFS, the primary endpoint of the study. We collected samples at baseline (n=258/258; 100%) and on Day 1 Cycle 2 (D1C2) (n=230/258; 89.2%). We centrally assessed sTILs (≥30% to be considered high) and PD-L1 expression (SP142) on immune cells (IC) (IC≥1% considered positive). Association with EFS was evaluated by Kaplan-Meier curves and log-rank test. Pre-treatment IMC and RNA-seq data of tumours PDL1- at baseline and converting to PDL1+ at D1C2 were compared with those remaining PDL1- at D1C2. P-values were corrected by false discovery rate (FDR). Results No differentially expressed genes were observed after FDR correction between tumors that up-regulate versus those that do not up-regulate PD-L1 during CT/A treatment. In the CT arm, up-regulation of PD-L1 was associated with significantly higher baseline expression (FDR < 0.05) of two immunoglobulin genes (IGHV3-64D and IGKVD2-28). Gene set enrichment analysis showed that baseline tumors up-regulating PD-L1 at D1C2 were enriched for interferon gene sets in both arms. Samples from CT arm also showed an enrichment for macrophage-related gene sets, while samples from CT/A arm were enriched for hypoxia and cell cycle gene sets. Immunoglobulin and B cell gene sets were positively and negatively enriched in tumors up-regulating PD-L1 in CT and CT/A arms, respectively. IMC showed no significant differences in baseline cell phenotype densities or cell-cell interactions between the two groups.In the CT arm, combined D1C2 PD-L1 and sTILs did not significantly improve EFS prediction in comparison to sTILs alone (likelihood ratio test p-value = 0.89). In CT/A arm instead, the combination of D1C2 PD-L1 and sTILs was more informative of EFS prediction than sTILs alone (likelihood ratio test p-value = 0.01). PD-L1-/sTILs-Low patients (N=19) had the worst outcome (5-years EFS = 0.37%, 95% C.I. = 0.20%-0.66%]). Conversely, patients with PD-L1+/sTILs-High (N=25) had the best EFS (5-years EFS = 0.86%; 95% C.I. = 0.72%-1.00%). Compared to the latter group, patients with PD-L1+/sTILs-Low tumors (N=30) had less favorable outcome (5-years EFS = 0.71%; 95% C.I. = 0.56%-0.90%). Only one patient was classified as PD-L1-/sTILs-High.The association of PD-L1/sTILs groups with EFS was maintained in the subset of patients with residual disease after surgery (log-rank p-value = 0.001). Conclusions Specific biology might be underlying the up-regulation of PD-L1 during neoadjuvant atezolizumab treatment, although we have not identified statistically robust baseline biomarkers that can predict this behavior. By combining the two clinically established biomarkers PD-L1 and sTILs evaluated on-treatment, we improved outcome prediction indicating an independent predictive-prognostic value of these markers. This association was independent of pCR status. Whether these findings may be translated to neoadjuvant pembrolizumab treatment remains to be defined.
Presentation numberPS2-09-10
Aneuploid-associated molecular features and therapeutic strategies in hormone receptor-positive HER2-negative breast cancer
Eo-Ryeong Lee, Fudan University Shanghai Cancer Center, Shanghai, China
E. Lee, L. Ge, S. Wu, Y. Jiang, Z. Shao; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Background: Hormone receptor-positive, HER2-negative (HR⁺/HER2⁻) breast cancer represents the most prevalent subtype of breast cancer and generally confers a favorable prognosis. However, its substantial biological heterogeneity poses challenges for risk stratification, as a subset of patients experience aggressive disease and treatment resistance. Aneuploidy is widely recognized as a hallmark of cancer associated with poor prognosis, yet its clinical significance in hormone receptor-positive, HER2-negative (HR⁺/HER2⁻) breast cancer remains underexplored. Identifying aneuploid-associated molecular features and therapeutic vulnerabilities within this group is crucial for precision oncology. Methods: We conducted a multi-omics analysis integrating genomic sequencing, transcriptomics, metabolomics, and digital pathology data from a well-annotated cohort of 1,082 breast cancer patients (519 HR⁺/HER2⁻). Tumor ploidy was determined using the ASCAT algorithm. Subsequently, we performed comprehensive bioinformatics analyses to characterize genomic instability, transcriptomic and metabolic alterations, immune microenvironment features, and developed a digital pathology-based classifier to predict tumor ploidy. Therapeutic implications were validated using patient-derived organoid (PDO) drug sensitivity assays. Results: Aneuploid tumors constituted a high-risk subgroup within HR⁺/HER2⁻ breast cancer, associated independently with significantly worse recurrence-free and distant metastasis-free survival. Aneuploid tumors demonstrated extensive genomic instability, elevated homologous recombination deficiency scores, and higher mutation burdens. Transcriptomic profiling revealed pronounced activation of cell cycle pathways, suggesting increased sensitivity to CDK4/6 inhibitors. Metabolic analyses identified heightened nucleotide metabolism, particularly in purine and pyrimidine pathways, indicating potential vulnerability to antimetabolites. Immune analyses showed subtle immunosuppressive features despite comparable overall immune infiltration. Our digital pathology model accurately predicted tumor ploidy, demonstrating high clinical applicability. Functional PDO assays confirmed the enhanced sensitivity of aneuploid tumors to PARP inhibitors, CDK4/6 inhibitors, and antimetabolites. Conclusions: Our study elucidates a distinctive multi-omics landscape of aneuploid HR⁺/HER2⁻ breast cancer, highlighting critical molecular vulnerabilities suitable for targeted therapies. The development of a robust, scalable digital pathology classifier offers a practical tool for clinical risk stratification and therapeutic decision-making, supporting the integration of ploidy-based personalized treatment strategies into clinical practice.
Presentation numberPS2-09-11
Beyond the tumor: a CAF-Driven Gene Signature for Early Clinical Stratification of Metastasis Risk in Breast Cancer
Daniela P. Barrera, Environ, Santiago, Chile
D. P. Barrera1, J. Contreras-Riquelme1, V. Toledo2, J. Sapunar2, B. Chahuán2, L. Moyano3, B. Prieto1, J. Cerda-Infante1; 1Medical Devices, Environ, Santiago, CHILE, 2Instituto Oncológico, Fundación Arturo López Pérez, Santiago, CHILE, 3Anatomía Patológica, Instituto Nacional del Cáncer, Santiago, CHILE.
Introduction. Despite advances in breast cancer (BC) diagnosis and treatment, there is still a lack of robust early prognostic tools to accurately identify patients at high risk of metastatic progression. This gap limits timely access to personalized therapies, leading to overtreatment in low-risk cases and delayed intervention in those who develop metastases. Current prognostic models rely mainly on tumor-intrinsic features, such as size, histological grade, or hormone receptor status, without integrating key components of the tumor microenvironment (TME). Among these, cancer-associated fibroblasts (CAF) are non-neoplastic cells that, in response to signals like TGF-β, PDGF, and IL-6, acquire a myofibroblast phenotype with high extracellular matrix (ECM) remodeling capacity, promoting stromal desmoplasia and facilitating tumor invasion. CAF also drive EMT, angiogenesis, and local immunosuppression, contributing to tumor aggressiveness and therapy resistance. Given their role in tumor progression, CAF are a promising source of prognostic biomarkers. We hypothesized that CAF from primary tumors harbor a transcriptomic signature with independent prognostic value for distant metastasis.Methods. A total of 182 FFPE breast tumor biopsies from women with invasive carcinoma were analyzed. Patients had ≥5 years of clinical follow-up, represented all five molecular subtypes, and had not received neoadjuvant chemotherapy, radiotherapy, or systemic treatment. Reactive stroma was quantified using H&E and Masson’s trichrome staining analyzed in QuPath. Associations with metastasis-free survival (MFS) and overall survival (OS) were assessed using Kaplan-Meier and Cox models. RNA was extracted from FFPE and CAF-enriched explants. Differential expression analysis between patients with and without metastases identified 233 CAF-associated genes. A 10-gene prognostic signature was derived using LASSO regression and validated in an independent retrospective cohort and external datasets. Predictive performance was evaluated via AUC, specificity, and Cox models. Results. A reactive stroma content greater than 53.2% was significantly associated with poor outcomes, including a 3.75-fold higher risk of distant metastasis (HR=3.75; 95% CI: 1.98-7.09; p<0.01). The 10-gene CAF signature identified in metastatic tumors was involved in biological processes such as ECM organization, cell adhesion, and migration. This signature demonstrated strong predictive performance in the training cohort (AUC=0.938), and in independent datasets (AUC=0.956; specificity 100%), outperforming models based solely on conventional clinicopathological parameters. Patients expressing the signature showed a 19.95-fold increased risk of metastasis (p<0.001), independently of hormone receptor status, histological grade, or tumor size. Notably, the signature retained prognostic value across molecular subtypes and sequencing platforms, supporting its clinical robustness. Conclusion. This study highlights the prognostic relevance of the stromal compartment, particularly CAF, in BC metastasis. The validated 10-gene signature provides a novel, TME-based biomarker that enables early risk stratification at the time of diagnosis. By incorporating this signature into clinical workflows, oncologists may identify high-risk patients earlier and guide personalized therapeutic decisions, ultimately improving outcomes and reducing mortality in BC. Acknowledgments. CORFO 22CVID-206707; ANID VIU24P0138.
Presentation numberPS2-09-12
Splenomegaly and response to chemo-immunotherapy in Triple-Negative Breast Cancer: Findings from MMTV-R26<sup>Met</sup> mouse model and patients
Jean MONATTE, INSERM, CNRS, Aix-Marseille Université, Marseille, France
J. MONATTE1, A. TASSIN DE NONNEVILLE2, N. CORVAISIER1, O. CASTELLANET1, R. FERRARA3, P. MICHEA1, J. P. BORG1, F. MAINA1, A. GONCALVES2, F. LAMBALLE1; 1Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM, CNRS, Aix-Marseille Université, Marseille, FRANCE, 2Medical Oncology, Institut Paoli-Calmettes, Marseille, FRANCE, 3Nuclear Medecine, Institut Paoli-Calmettes, Marseille, FRANCE.
Background: Triple-negative breast cancer (TNBC) is marked by pronounced heterogeneity in immune microenvironments and variable response to immunotherapies. Preclinical models often fail to capture this diversity, limiting translational progress and biomarker discovery for patient stratification. Method: We developed the immunocompetent MMTV-R26Met mouse model, which mimics key TNBC features. Multi-omics characterization (histology, proteomics, genomics, immunophenotyping, scRNA-seq) enabled direct comparison with human TNBC. We generated syngeneic orthotopic transplants that preserved tumor-immune heterogeneity across passages. Treatment studies evaluated the effects of epirubicin, anti-PD-1 and combination therapy. Clinical validation involved 125 stage II-III TNBC patients receiving neoadjuvant pembrolizumab plus chemotherapy, with comprehensive clinical, pathological, and imaging data collection. Predictive models used machine learning approaches (logistic regression). Results: Our multi-omics analysis revealed strong similarities between the MMTV-R26Met mouse model and TNBC patients, such as high-grade tumors, inter-tumoral diversity, and distinct molecular subtypes. Additionally, immune profiling revealed four distinct immune subtypes (depicting enrichment in macrophages, neutrophils, dendritic cells or T lymphocytes). Interestingly, the neutrophil-enriched subtype exhibited a marked splenomegaly which remained through successive transplants and correlated with systemic immune activation. Moreover, our in vivo experiments revealed that treatment responses varied according to the immune profile of the tumor. Neutrophil-enriched tumors exhibited dramatic tumor regression upon epirubicin+anti-PD-1 treatment, accompanied by a reduction in spleen size and enhanced cytotoxic T cell infiltration (n=17). In contrast, macrophage-enriched tumors displayed limited immune remodeling and responded preferentially to anti-PD-1 monotherapy with persistent immunosuppressive features (n=11). In univariate analysis using a ROC-derived cutoff, patients with a larger spleen size had nearly fourfold higher odds of achieving pathological complete response (pCR) to chemo-immunotherapy (OR = 3.99, 95% CI: 1.76-9.80, p = 0.0015) mirroring the murine outcomes. Ki-67 (OR=1.03, 95% CI :1.01-1.04, p=0.004), Breast SUV max (OR=1.13, 95% CI :1.01-1.27, p=0.045) and family history (OR=0.43, 95%CI : 0.20-0.91, p=0.028) emerged as top associated markers with pCR in univariate analysis. Importantly, in multivariate logistic regression adjusting for key clinical and biological covariates, spleen vertical size remained an independent predictor of pCR (OR = 1.06, 95% CI: 1.01-1.13, p = 0.033). The final multivariate model demonstrated excellent discriminative power, achieving an area under the ROC curve (AUC) of 0.91 These findings highlight spleen size as a clinically relevant and independent predictor of response to chemo-immunotherapy in triple-negative breast cancer. Conclusion: The MMTV-R26Met mouse model recapitulates the immune heterogeneity of human TNBC, identifying splenomegaly as a hallmark of neutrophil-enriched tumors with heightened sensitivity to chemo-immunotherapy. This phenotype translates to improved response in patients with larger spleens, establishing spleen size as a novel, actionable biomarker for immunotherapy benefit. The MMTV-R26Met mouse model provides a robust platform for personalizing TNBC treatment and advancing biomarker-driven clinical strategies.
Presentation numberPS2-09-14
When interception needs a second chance: ctDNA dynamics in ER+/HER2- advanced breast cancer treated with 1st line CDK4/6 inhibitors – Insights from the CICLADES study
Vincent Massard, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, France
V. Massard1, M. Betz2, A. Diallo3, J. Salleron2, A. M. Savoye4, D. Spaeth5, F. Clatot6, P. Barthelemy7, R. Longo8, Y. Tazi9, M. Gardner10, A. Witz2, J. Dardare2, D. Bechet11, J. L. Merlin2, A. Harlé2; 1Département d’oncologie Médicale, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, FRANCE, 2Service de Médecine de Précision, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, FRANCE, 3Département de biostatistiques, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, FRANCE, 4Département d’oncologie Médicale, Institut Jean Godinot, Reims, FRANCE, 5Département d’oncologie Médicale, Hôpital privé Nancy-Lorraine – ELSAN, Nancy, FRANCE, 6Département d’oncologie Médicale, Centre Henri Becquerel, Rouen, FRANCE, 7Département d’oncologie Médicale, ICANS, Strasbourg, FRANCE, 8Département d’oncologie Médicale, CHR Metz-Thionville, Ars-Laquenexy, FRANCE, 9Département d’oncologie Médicale, Clinique Sainte Anne, Strasbourg, FRANCE, 10Département d’oncologie Médicale, Hôpital Claude Bernard, Metz, FRANCE, 11Département de Recherche clinique, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, FRANCE.
Background: Circulating tumor DNA (ctDNA) is a valuable biomarker to monitor therapeutic resistance and disease progression in advanced breast cancer (ABC). In ER+/HER2- ABC, mutations in ESR1, PIK3CA, and TP53 are frequent and may impact response to endocrine therapy (ET) combined with CDK4/6 inhibitors. The CICLADES study aimed to evaluate the prognostic and temporal dynamics of these mutations and ctDNA-based progression signals.Methods: A prospective cohort of 117 patients with ER+/HER2- ABC treated with first line ET and CDK4/6 inhibitors was analyzed. Plasma ctDNA was profiled using a 35-gene NGS panel at baseline, at follow-up visits, and at radiologic progression. Associations between mutation status and progression-free survival (PFS) were examined for ESR1, PIK3CA, and TP53. Additional analyses explored mutation emergence, polyclonality, and molecular progression, defined as a ≥50% increase in variant allele frequency (VAF). Transient variants not detected at baseline or progression were evaluated as potential markers of subclonal dynamics.Results: Main results are presented in Table1. Baseline ESR1 mutations (n=6) and those acquired during follow-up (n=26) were associated with significantly shorter PFS. Most ESR1 mutations at progression were newly acquired, particularly in ligand-binding domains. PIK3CA and TP53 mutations also predicted shorter PFS at baseline, but not significantly during follow-up. While PIK3CA and TP53 mutations were largely present from baseline, most ESR1 mutations emerged under treatment. Molecular progression, defined by ≥50% increase in VAF, was detected in 16 patients and preceded radiologic progression in 11, with a median lead time of 2.7 months. Polyclonality was not associated with prognosis. Fourteen patients exhibited transient low-VAF variants during follow-up, suggestive of subclonal shedding rather than resistance; these did not correlate with clinical progression.Conclusion: In ER+/HER2- ABC, pre-existing ESR1, PIK3CA and TP53 mutations, and acquired ESR1 mutations, are significantly associated with shorter PFS under CDK4/6i-based therapy.Molecular progression detected via ctDNA may precede radiologic progression by several months, supporting the extension of the concept of molecular progression beyond ESR1 mutations and into real-world settings, and highlighting a window of opportunity for earlier therapeutic intervention. However, episodes of subclonal shedding, reflected by transient low-VAF variants, underscore the importance of confirmatory sampling before clinical decision-making. These results support longitudinal ctDNA monitoring as a refined tool for real-time disease interception.
| Mutation | Timepoint | n Mutated | n WT | Median PFS (Mut) | Median PFS (WT) | p-value | HR (95% CI) | ||||||||
| ESR1 | Baseline | 6 | 111 | 12.9 mo | 25.7 mo | 0.011 | 0.35 (0.15-0.82) | ||||||||
| Follow-up | 26 | 91 | 16.0 mo | 28.8 mo | 0.00638 | 0.51 (0.31-0.83) | |||||||||
| PIK3CA | Baseline | 31 | 86 | 13.5 mo | 30.9 mo | 0.0284 | 0.57 (0.34-0.95) | ||||||||
| Follow-up | 47 | 70 | 18.0 mo | 26.2 mo | 0.239 | 0.76 (0.48-1.2) | |||||||||
| TP53 | Baseline | 16 | 101 | 10.7 mo | 28.7 mo | 0.0032 | 0.41 (0.22-0.76) | ||||||||
| Follow-up | 42 | 75 | 22.8 mo | 25.5 mo | 0.989 | 1 (0.62-1.62) |
Presentation numberPS2-09-15
Application of CNSigs, An R Package for the Identification of Copy Number Mutational Signatures, in Early and Advanced Breast Cancer
Shawn Striker, Ohio State University, Columbus, OH
S. Striker1, D. Tallman1, K. Collier2, E. Blige1, M. Vater1, D. G. Stover2; 1CCC – Comprehensive Cancer Center, Ohio State University, Columbus, OH, 2Medicine – IM Medical Oncology, Ohio State University, Columbus, OH.
Application of CNSigs, An R Package for the Identification of Copy Number Mutational Signatures, in Early and Advanced Breast Cancer Shawn Striker, David Tallman, Katharine Collier, Elijah Blige, Mark Vater, Daniel G. Stover Background: Copy number aberrations (CNAs) are gains and losses of large genomic segments present across most cancer types and are a hallmark of cancer genomic alterations. However, the processes underlying CNAs and characteristic patterns of CNAs are poorly understood. Using single nucleotide variant (SNV) data, bioinformatic advances have identified underlying mutational signatures resulting from distinct mutational processes. Mutational signatures have led to a variety of discoveries, several of which are being investigated in clinical management of cancer. The development of algorithms able to uncover similar signatures for CNAs, rather than SNVs, is still in its infancy. Here we present an analysis package for the R programming language called CNSigs that allows for the robust and reproducible derivation of copy number signatures and apply CNSigs in early and advanced breast cancer. Methods: Using segmented data files from DNA sequencing, six copy number features are extracted by CNSigs for signature determination: segment size, the number of breakpoints per 10 megabase bin, number of copy number oscillation events, average size of changepoints, average copy number, and number of breakpoints per chromsome arm, along with ploidy. Based on extracted copy number features, mixed model approaches and non-negative matrix factorization are utilized to derive CNA signatures across cancer types. CNSigs was applied to two large publicly-available cohorts of primary breast cancers (TCGA and METABRIC), a validation cohort of paired tissue and circulating tumor DNA/ctDNA (n=24 pairs), and ctDNA from a large cohort of patients with metastatic triple-negative breast cancer (mTNBC; n=171 patients). Results: To verify the reproducibility of the signatures, we derived five signatures from two independent breast cancer datasets that use distinct copy number segmentation approaches (TCGA – ABSOLUTE; METABRIC – ASCAT). From these two independent datasets, we identified five copy number signatures with high accuracy (average cosine similarity = 0.89). These five signatures are distinct from known breast cancer receptor-based or expression-based subtypes, yet reveal unique associations with underlying mutations, mutational processes, and transcriptional programs. We then took a pan-cancer approach to assess the robustness of the algorithm, and applied the pipeline to 11 different cancer types from TCGA dataset, deriving 13 pan-cancer signatures. To investigate potential application to ctDNA, we evaluated patients with paired tumor and ctDNA sequencing acquired at the same time (n=24 pairs), demonstrating that CNSigs are detectable via ctDNA. Exploratory application of CNSigs in a large cohort of mTNBC patients revealed association of presence of one specific signature, CNSig 11, with extended response to taxane chemotherapy. Conclusions: The CNSigs R package allows researchers to easily analyze their own samples to derive copy number signatures and evaluate clinical associations. We demonstrate potential application in ctDNA and association with treatment response. The development of this package allows further investigation of underlying processes that may be responsible for these CNA fingerprints.
Presentation numberPS2-09-16
Optical genome mapping and multi-modal characterization of breast tumor and organoid samples reveal structural variation and gene fusion signatures
Rahul Kumar, UPMC Hillman Cancer Center and Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA
R. Kumar1, O. Shah1, B. Schlegel1, J. Hooda1, D. Brown2, H. Chang3, A. Chang1, J. Chen1, J. Xavier1, P. Mattila1, L. Fairfull1, G. Tseng4, R. Bhargava5, S. Oesterreich1, A. Lee1; 1Women’s Cancer Research Center, UPMC Hillman Cancer Center and Magee-Women’s Research Institute, University of Pittsburgh, Pittsburgh, PA, 2Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, 3Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, 4School of Public Health, University of Pittsburgh, Pittsburgh, PA, 5Department of Pathology, University of Pittsburgh, Pittsburgh, PA.
Background: Despite progress in diagnosis and treatment, the heterogeneity of breast cancer (BC) continues to challenge therapeutic strategies. To better understand the genomic and transcriptomic alterations driving BC progression, we applied an integrative approach combining optical genome mapping (OGM) and RNA-seq. OGM is a powerful tool for detecting large structural variants (SVs), genomic instability, and fusion. We independently analyzed breast tumor tissues and patient-derived organoids (PDOs) models using both platforms. By comparing genomic and transcriptomic landscapes across these models, we aim to identify key alterations and evaluate the ability of organoids to reflect critical features of BC biology. Methods: The study cohort consists of 62 samples, including 27 breast tumor tissues and 35 PDOs, categorized based upon origin (tumor vs. PDO), tissue source (primary vs. metastatic), and histological subtype (invasive lobular carcinoma, ILC, and no special type, NST). OGM was performed on all 62 samples using the Bionano Saphyr platform. Bulk RNA sequencing was conducted on a subset of 55 samples, comprising 22 tumor tissues and 33 PDOs. RNA-seq libraries were prepared and sequenced in paired-end mode using the Illumina platform. Fusion analysis was performed by the nf-core/rnafusion module, and Oncofuse tool was used to predict the oncogenic potential of fusion genes. Results: SVs analysis revealed a complex landscape of genomic alterations across PDOs and tumor samples. A total of 3,007 SVs were identified in PDOs and 2,241 SVs in tumors. In both cohorts, deletions represented the most shared type of SVs, followed by inter/intra-chromosomal translocations. Intrachromosomal SV size distributions were comparable, with tumors showing a slightly larger median size. Cytoband analysis revealed 42 regions enriched for SVs, with hotspots on chromosomes 8 and 1. However, over 95% of SVs were non-recurrent, highlighting predominant patient-specific structural alterations. To assess functional impact, SVs were mapped to BC-related genes from the OncoVar database. Recurrent top alterations were identified in ESR1, PPM1D, CCND1, and ERBB3 across both PDOs and tumors, spanning diverse SV types and gene roles. Genome instability profiling showed a wide range of homologous recombination deficiency (HRD) scores in both groups, calculated from large-scale state transitions (LST), loss of heterozygosity (LOH), and telomeric allelic imbalance (TAI) components. HRD-high samples (score >42) were present in both PDOs and tumors, independent of histology. Circos plots revealed structural complexity ranging from stable to highly rearranged genomes. A total of 216 amplicons were detected in PDOs, including 5 break fusion bridge (BFB) positive amplicons, while 131 amplicons were identified in tumor samples, with 2 BFB-positive amplicons. Chromothripsis events were observed in both PDO and tumor samples, with varying levels of confidence. The most frequently affected chromosomes included chr8, chr12, and chr17, suggesting recurrent loci of catastrophic genomic rearrangement. Fusion detection using RNA-seq and OGM identified 411 and 1,178 events, respectively, with 37 overlapping fusions. Shared fusions showed both up- and downregulated expression patterns. High-confidence driver fusions included ESR1–ARNT2, DENND5A–SORL1, TAB3–DMD, and ARHGEF7–ATP11A, implicating them as potentially functional alterations. Conclusions: This study provides a comprehensive multi-platform characterization of SVs using both tumor tissues and PDOs. Collectively, these findings highlight the genomic fidelity of PDOs in modeling primary breast tumors and underscore the value of integrating OGM and transcriptomic profiling to uncover novel structural drivers of BC.
Presentation numberPS2-09-17
Comprehensive peripheral immune profiling in Triple Negative Breast Cancer (TNBC) links immune cell dynamics to therapeutic response and irAEs: insight from the IRIS Study
Simone Nardin, AOU Maggiore della Carità, Novara, Italy
L. Negrini1, V. Martini2, B. Conte1, I. Taglialatela1, T. L. Landolfo1, S. Gobbato1, B. Ruffilli1, S. Nardin2, F. D’Avanzo2, V. Rossi2, C. Branni1, J. Gennari1, A. Turki1, G. Di Foggia1, M. Bitrus1, A. Giacobino3, E. Rota Caremoli4, I. Romaniello5, A. M. Vandone6, V. Prati7, V. Guarneri8, A. Gennari1; 1Department of Translational Medicine (DIMET), University of Eastern Piedmont, Novara, ITALY, 2Medical Oncology dept., AOU Maggiore della Carità, Novara, ITALY, 3Medical Oncology dept., ASL Biella, Biella, ITALY, 4Cancer Center, ASST Papa Giovanni XXIII, Bergamo, ITALY, 5Medical Oncology dept., ASL13 Novara, Borgomanero, ITALY, 6Medical Oncology, AO S. Croce e Carle, Cuneo, ITALY, 7Oncology Unit, Michele Pietro Ferrero Hospital, ASL CN 2, Verduno, ITALY, 8Surgery, Oncology and Gastroenterology dept., University of Padova, Padova, ITALY.
Background: Triple-negative breast cancer (TNBC) is the most aggressive subtype with limited targeted options. Immune checkpoint inhibitors (ICIs) have reshaped treatment approaches in both metastatic and early-stage TNBC. In the neoadjuvant setting, ICIs combined with chemotherapy improve pathological complete response (pCR) rates over chemotherapy alone. Despite their benefits, ICIs pose risks of immune-related adverse events (irAEs), requiring careful patient selection and monitoring. Comprehending the immune context may be fundamental for tailoring therapy and predicting outcomes in TNBC. Methods: Between March 2021 and April 2025, peripheral blood samples were collected at baseline from patients with TNBC treated with ICIs and enrolled in the translational, prospective and multicentre (12 Italian sites) IRIS study investigating circulating immune cells as predictive or prognostic biomarkers. In 60 patients (49 in the neoadjuvant and 11 in the metastatic setting), immunophenotype was performed by flow cytometry (FACS) to characterize circulating B cells, T cells, dendritic cells (DC), and Natural Kiler cells (NK) by a manual gating strategy. To capture immune cell heterogeneity, high-dimensional analysis was applied, including t-distributed stochastic neighbour embedding (t-SNE) for dimensionality reduction and FlowSOM for unsupervised clustering. For consistency, datasets were down sampled to 6000 T cells and 1600 B cells per sample, excluding sample below this cut-off (FlowJoTMv10). Statistical analysis included Mann-Whitney U test, univariate and multivariate Cox models for time to first irAE (TTirAE), defined as the interval between the first ICI cycle and onset of irAE (any grade), and Logistic regression (R software v.4.4.3). Results: In patients with TNBC, those in the metastatic setting exhibited significantly lower percentages of circulating unswitched and switched memory B cells (p = 0.0004 and p = 0.035) and higher frequencies of naive B cells (p = 0.0046). Additionally, dysfunctional central memory (CM) Th1-like cells and dysfunctional transitional CD4+ T cells were enriched in the metastatic group. Focusing on TTirAE (No irAEs: n = 34; irAEs: n = 13), a higher baseline percentage of peripheral plasmacytoid dendritic cells (pDCs) was associated with a lower risk of irAEs (HR = 0.89; 95% CI: 0.80 – 0.99; p = 0.040), whereas increased frequencies of CM Th1 cells were linked to a higher risk (HR = 2.10; 95% CI: 1.05 – 4.20; p = 0.036). These associations remained statistically significant after adjusting for age at diagnosis and treatment setting. Considering only patients with early-stage TNBC, baseline circulating immune profiling revealed distinct differences between patients achieving pCR (n = 29) and those with residual disease (RD, n = 11). Patients with pCR showed higher percentages of dysfunctional CD4+CD28-Tregs (p = 0.014) and NKT cells (p = 0.038), whereas those with RD had increased frequencies of mature plasma cells (p = 0.015), transitional Th1/Th17 cells (p = 0.031), and CM Tc1 cells (p = 0.043). Although not statistically significant, higher baseline frequency of CD8+ naive T cells showed a trend toward a positive association with pCR (OR = 1.05; 95% CI: 1.00-1.11; p = 0.068). In contrast, CM Tc1 cells, plasma cells, and transitional marginal zone-like B cells were negatively associated with pCR (OR < 1.0; 0.070 < p < 0.080). Conclusion: Our preliminary data highlight the potential utility of peripheral immunophenotyping in TNBC as a non-invasive tool to identify predictive biomarkers and guide patient stratification in immunotherapy-based approaches.
Presentation numberPS2-09-18
Real-time liquid biopsy assessment in early-stage breast cancer: methylation based-ctDNA analysis and CTCs detection as potential predictors of response to neoadjuvant therapy
Caterina Gianni, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
C. Gianni1, I. Azzali2, L. Pontolillo3, B. Pastò4, E. Nicolo’5, M. Serafini5, N. Bayou5, M. Velimirovic6, A. J. Medford7, M. D. Lipsyc-Sharf8, A. Davis9, C. Reduzzi5, A. Musolino1, A. Bardia8, M. Cristofanilli5; 1Medical Oncology and Breast Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, ITALY, 2Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, ITALY, 3Policlinico Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma, ITALY, 4Department of Medicine (DMED), University of Udine, Udine, ITALY, 5Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, 6Department of Medicine, Cleveland Clinic, Cleveland, OH, 7Medicine, Mass General Cancer Center, Boston, MA, 8Division of Hematology/Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 9Division of Oncology, WashU Medicine, St Louis, MO.
Background: In early-stage breast cancer (EBC), liquid biopsy may help refine risk stratification and therapeutic decision-making. Historically, the detection of circulating tumor cells (CTCs)>1/7.5 ml has a strong prognostic value for high risk of relapse, but rarely are detected. We explored ctDNA and CTC detection at diagnosis and their association with tumor characteristics and response to neoadjuvant therapy (NAT). Methods: We retrospectively analyzed 40 patients with stage I-III breast cancer who underwent GR and Cellsearch testing at diagnosis and/or during NAT. Guardant Reveal™ (GR) is a tissue-naïve, methylation-based cell-free DNA assay capable of detecting circulating tumor DNA (ctDNA) with high sensitivity, even in low-input samples. CellSearch® detects CTCs via immunomagnetic enrichment of EpCAM-positive cells and cytokeratin-based immunostaining, followed by automated enumeration. Associations between baseline ctDNA and CTC detection and clinical features—including tumor grade, stage, molecular subtype—and response to NAT were assessed. Results: In total, 71 Guardant Reveal™ tests were performed across 40 patients, with a median of 1 test per patient (range 1-7). Thirty patients had a baseline GR before starting NAT. Seventeen patients (43%) underwent more than one GR test during or after NAT, and 15/40 patients (38%) had at least one GR test after surgery. The median time between GR tests was 3.2 months (IQR 2.7-5.0; range 0.3-15.1), while the median interval between surgery and the post-surgery GR test was 3.5 months (IQR 1.9-4.7; range 0.5-8.3). The median age of the cohort was 45 (range 28-77); most patients had invasive ductal carcinoma (88%) and received NAT (85%). Among the 33 patients who underwent testing pre-NAT, ctDNA was detected in 17 (52%). ctDNA detection was significantly associated with tumor grade and molecular subtype, being more frequent in G3 than G1-G2 tumors (72% vs. 27%, p=0.02), andin triple-negative and HR-positive subtypes compared to HER2-enriched (79% vs. 35% vs. 0%, p=0.04). Inflammatory breast cancer and stage III showed a trend toward higher ctDNA tumor fraction (median methylation tumor fraction 0.43% and 0.02%, p=0.04 and 0.05, respectively). Lower ctDNA levels at diagnosis were observed in patients achieving pCR (pCR vs. no pCR, median methylation tumor fraction 0.03% vs. 0.09%), although the association is not statistically significant (p=0.26). CTC testing with CellSearch® was performed in 29 patients, for a total of 56 tests. 15 patients (52%) underwent more than one CTC test, enabling longitudinal assessment. Among CTC-tested patients, only one patient was positive (11 CTC/7.5mL), converting to negative after 2 months of NAT. No additional CTC positivity was observed during follow-up. At the time of analysis, no recurrences had been observed (median follow-up: 13.6 months; range 6.6-18.2 from diagnosis to last contact) and four patients are still receiving NAT. Conclusion: In EBC, ctDNA detection using a methylation-based assay identified patients with aggressive features and potential lower likelihood of achieving pCR. The use of methylation-based ctDNA testing may hold promise as a tool to guide escalation or de-escalation strategies, supporting a more personalized approach to treatment when combined with other biomarkers such as CTCs. Further validation in larger cohorts is needed. Updated data will be presented at the conference.
Presentation numberPS2-09-19
Tonsl, an immortalizing oncogene on chr.8q24.3 amplicon, confers intrinsic resistance to cdk4/6 inhibitors in breast cancer.
Harikrishna Nakshatri, Indiana University School of Medicine, Indianapolis, IN
H. Nakshatri1, A. S. Khatpe1, U. R. Kamdar1, G. Sandusky2, S. K. Althouse3, S. K. Deshmukh4, S. Wu4, N. Kulkarni4, L. A. Poole4, P. Coelho5, M. B. Lustberg6, G. W. Sledge4, K. D. Miller7, P. Bhat-Nakshatri1; 1Surgery, Indiana University School of Medicine, Indianapolis, IN, 2Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 3Biostatistics, Indiana University School of Medicine, Indianapolis, IN, 4Medical Affairs, Caris Life Sciences, Tempe, AZ, 5Medicine, University of Miami, Miami, FL, 6Center for Breast Cancer, Yale School of Medicine, New Haven, CT, 7Medicine, Indiana University School of Medicine, Indianapolis, IN.
Background: Advanced Estrogen receptor-positive (ER+) breast cancers benefit from the inhibitors of cell-cycle kinases CDK4 and CDK6. However, 20% of breast cancers show intrinsic resistance to these inhibitors. Although the mechanisms of acquired resistance to CDK4/6 inhibitors have been studied extensively, little is known about mechanisms of intrinsic resistance. We had recently reported a gene called TONSL, a DNA repair and chromatin organization-associated gene located on chromosome 8q24.3 amplicon, as an immortalizing oncogene and its overexpression in ER+ breast cancer is associated with poor outcome. In preclinical models, TONSL-immortalized breast epithelial cell lines generated ER+ adenocarcinomas upon transformation. Here we investigated whether TONSL amplification confers intrinsic resistance to CDK4/6 inhibitors. Here we compare the survival of TONSL amplified vs not amplified ER+ breast cancer patients treated with CDK4/6 inhibitors.Methods: 10,980 ER+ breast cancer samples (TONSL amplified, n=313; Not amplified, n=10,667) were analyzed by next-generation sequencing (WES, NovaSeq) and Whole Transcriptome Sequencing (WTS; NovaSeq) (Caris Life Sciences, Phoenix, AZ). Copy number variations were determined using CNVKit. A tissue microarray comprising ~200 ER+ breast cancers was examined for TONSL and phospho-RB expression levels by immunohistochemistry. The role of TONSL in sensitivity to abemaciclib was examined in vitro in a TONSL amplified ER+ breast cancer cell line (generated from MCF-7 derived tumor in nude mice) with and without shRNA-mediated TONSL knockdown using bromodeoxyuridine (BrdU) incorporation ELISA. Real-world median overall survival (mOS) was derived from insurance claims and calculated from the biopsy to last contact or start of CDK4/6 inhibitor (abemiciclib/palbociclib/ribociclib) to last contact or first to last dose of CDK4/6 inhibitor treatment (time on treatment) using Kaplan-Meier. Statistical significance was assessed using chi-square and Mann-Whitney U and p<0.05 was considered significant.Results: MCF-7 derivative, which is intrinsically resistant to CDK4/6 inhibitor abemaciclib, regained drug sensitivity as assessed using cell proliferation assay upon TONSL knockdown (p<0.05). TONSL amplification/overexpression correlated with elevated RB-loss and P53-loss gene signatures, but not RB phosphorylation. In real-word database, moderate correlations were observed between TONSL copy number and RNA expression (r=0.48, p<0.05). Median age of breast cancer patients with TONSL amplification was lower (62 vs 64, p<0.05) compared to TONSL not amplified. TONSL amplification was more common in Black or African American patients (20% vs 14%, p<0.05), but less common in White patients (68.6% vs 77.7%, p<0.05). TONSL amplified breast cancer patients had worse mOS (28.4 months (m) vs 41.9, HR 1.4, 95% CI 1.2-1.6, p<0.0001) compared to TONSL not amplified. Importantly, TONSL amplified breast cancer patients treated with CDK4/6 inhibitors had worse mOS (41.6 m vs 56.3, HR 1.43, 95% CI 1.1-1.8, p=0.002). Moreover, TONSL amplified breast cancer patients had shorter mOS with palbociclib time on treatment (8.9 m vs 11 m, HR 1.31, 95% CI 1-1.6, p=0.01) compared to TONSL not amplified.Conclusions: TONSL amplification in ER+ breast cancer is associated with poor outcome and is potentially a biomarker of intrinsic resistance to CDK4/6 inhibitors. Since activity of TONSL is governed by multiple protein complexes, disruptors of TONSL-protein complexes could potentially be developed as sensitizers to CDK4/6 inhibitors.
Presentation numberPS2-09-20
Circulating Tumor DNA Dynamics and Anatomical Patterns of Relapse Following Curative Therapy in Early-Stage Breast Cancer
Daniel Stover, Ohio State University, Columbus, OH
D. Stover1, B. Dwivedi2, P. Dutta2, S. Beyer1, H. LeFebvre1, E. Kalashnikova2, J. McKenzie2, A. Rodriguez2, M. Liu2; 1Oncology, Ohio State University, Columbus, OH, 2Oncology, Natera, Inc., Austin, TX.
Background: Predicting the anatomical patterns of relapse after an initial diagnosis of early-stage breast cancer (BC) and understanding their association with circulating tumor DNA (ctDNA)-based molecular residual disease (MRD) detection by tumor subtype and treatment history may inform surveillance strategies and enable earlier therapeutic intervention. Methods: This retrospective study explored the association between sites of relapse and ctDNA detection rates and dynamics in a real-world cohort (2019-2024) of patients (pts) with early-stage BC. We utilized Natera’s proprietary real-world database linked to Komodo’s Healthcare Map® claims database and results of tumor-informed ctDNA testing (SignateraTM, Natera, Inc.). Hormone receptor (HR), HER2 status, and the date of relapse were inferred from the claims codes associated with interventions and secondary neoplasms (i.e., metastases). Pts with a diagnosis code of secondary neoplasms were categorized by the anatomic site(s) of recurrence. ctDNA detection rate (required time points: ctDNA positivity within 6 months prior to relapse; for ctDNA-negative pts, three or more time points were included), and dynamics were analyzed in relation to subtype and relapse site.Results: Evaluation of Natera’s real-world database identified 532 pts with early-stage breast cancer who developed subsequent distant metastatic disease. The distribution of patients by subtype was: HR+/HER2-: 52% (276/532), triple-negative (TNBC): 34% (182/532), and HER2+: 14% (74/532). Among pts with isolated relapses (N=259), the most common sites were bone ([HR+HER2-: 59%; 82/138], [HER2+: 36%; 14/39] [TNBC: 30%; 25/82]) and lung ([HR+HER2-:16%; 22/138, [HER2+: 31%; 12/39], [TNBC: 34%; 28/82]), concordant with common sites of metastasis by subtype. Relapses occurred within a median of 1.84 years (yrs) after surgery, varying by subtype (TNBC: 1.33 yrs, HER2+: 1.92 yrs, HR+HER2-: 2.41 yrs). Among pts with required timepoints (77%, 411/532), ctDNA was detectable prior to/at the time of recurrence in 91% (373/411) of cases; 90% (130/145) for TNBC, 90% (187/208) for HR+HER2- and, 97% (56/58) for HER2+ disease. For the subset of patients with an isolated relapse, the rate of ctDNA positivity at the time of recurrence was the highest for liver (100%; 23/23), followed by bone (93%; 95/102) metastases. In terms of ctDNA quantification, median ctDNA levels differed by subtypes, with TNBC showing the highest levels (16.4 MTM/mL, range: 0.03-17053) compared to HR+/HER2- (5.58 MTM/mL, range: 0.018-8343) and HER2+ (5.44 MTM/mL, range: 0.03-2879). ctDNA levels among secondary neoplasms prior to/at the time of recurrence were highest for multi-site relapses (median = 15 MTM/mL, IQR:1.5-143), followed by lung (median: 19.9, IQR: 1.5 – 82), bone (median: 5.6, IQR: 0.9-29), and liver (3.4, IQR: 0.52-76.8) only metastases. Serial ctDNA measurements preceding the date of secondary neoplasms showed rising ctDNA levels over time in >90% of cases, with the most rapid increase observed in TNBC. The cases that exhibited declining ctDNA levels without achieving clearance were predominantly HR+HER2- pts with bone metastases.Conclusions: Our findings highlight the potential utility of serial ctDNA monitoring in clinical practice to identify pts at high risk of relapse across multiple organ sites, including isolated progression in individual organs. These findings could potentially inform future ctDNA-guided intervention strategies, including increased surveillance by imaging for pts with ctDNA positivity.
Presentation numberPS2-09-21
Real-world clinical outcomes and genomic insights for patients with PIK3CA-mutant metastatic breast cancer following progression on CDK4/6 inhibitor therapy
Maxwell R Lloyd, Beth Israel Deaconess Medical Center, Boston, MA
M. R. Lloyd1, L. Bucheit2, D. Hintz3, J. Liao3, N. Vasan4, K. Clifton5, C. Ma6, S. Wander7; 1Department of Medicine, Division of Hematology / Oncology, Beth Israel Deaconess Medical Center, Boston, MA, 2Medical Affairs, Guardant Health, Palo Alto, CA, 3Outcomes and Evidence, Guardant Health, Palo Alto, CA, 4Department of Medicine, Perlmutter Cancer Center, NYU Langone Health, New York, NY, 5Department of Medicine, Washington University in St Louis, St Louis, MO, 6Department of Medicine, Washington University in St. Louis, St. Louis, MO, 7Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, MA.
Background: PIK3CA mutations (PIK3CAmut) are detected in approximately 40% of hormone receptor-positive metastatic breast cancers (mBC) and are actionable therapeutically. The emergence of targeted therapies for mBC with PIK3CAmut, ESR1 mutations (ESR1mut), and those with dual mutant disease has made therapy decisions following progression on a CDK4/6 inhibitor (CDK4/6i) increasingly complex. Here we assessed treatment selection for patients (pts) with PIK3CAmut mBC post-CDK4/6i, examining clinical outcomes and associated genomic findings via circulating tumor DNA (ctDNA). Methods: Pts tested with Guardant360 (a ctDNA sequencing assay) since November 2023 (capivasertib approval date) were identified in Guardant INFORM, a real-world clinical-genomic dataset which combines de-identified genomic results with associated claims data. Eligible pts had mBC, were treated with a CDK4/6i after metastatic diagnosis, and had ctDNA-detected PIK3CAmut after CDK4/6i but prior to the start of the next line of therapy (LOT). The therapy selected following PIK3CAmut detection was assessed. Exposures of interest included treatment with capivasertib (CAPI), alpelisib (ALP), and elacestrant. Subgroup exposures included baseline and post-treatment genomic alterations detected on ctDNA, and dual PIK3CAmut/ESR1mut disease when alterations were identified on the same test. Outcomes analyzed included real-world time to treatment discontinuation (rwTTD), and overall survival (rwOS) in months using the Kaplan Meier method; log-rank tests compared time-to-event outcomes. Results: 460 pts met inclusion criteria of which 115 (25%) subsequently received CAPI, 22% a CDK4/6i regimen, 15% aromatase inhibitor (AI) monotherapy, 14% elacestrant, 12% chemo and 6% ALP. rwTTD was numerically longer for CAPI compared to ALP [6.0 vs. 5.5 months; HR: 0.82 (95% CI:0.46-1.44); p=0.48]; rwOS was not reached at this time. When assessing genomics, ctDNA testing post-CAPI (n=15), found several alterations with frequency >10% including TP53 single nucleotide variant (SNV) (53%), FGFR1 copy number variant (CNV) (20%), CCND1 CNV (20%), KRAS SNV (13%), and ESR1 SNV (13%). FGFR1 CNV, CCND1 CNV, and KRAS SNV frequencies increased substantially in post-treatment samples compared to baseline (FGFR1: 20% vs. 6%; CCND1: 20% vs. 10%; KRAS: 13% vs. 4%), suggesting post-progression enrichment and potential roles in CAPI resistance. For PIK3CAmut/ESR1mut tumors (n=165, 35%), 66 (40%) received elacestrant as next LOT, 28 (17%) received CAPI, 21 (13%) received AI only, 19 (12%) each received CDK4/6i or chemo, and 10 (6%) received ALP. In PIK3CAmut disease treated with CAPI, pts without ESR1mut had numerically longer rwTTD compared to those with ESR1mut (6.0 vs. 4.9 months; HR: 1.23 (95% CI: 0.63-2.39); p=0.54]. Outcomes need additional follow-up time for maturity and updated results will be presented at the meeting. Conclusions: In this real-world analysis, pts with PIK3CAmut mBC and CDK4/6i progression were most frequently treated with CAPI as subsequent therapy when no ESR1mut was identified; co-mutant disease received elacestrant most often. CAPI responses may differ based upon genomic alterations in baseline ctDNA, including the presence of ESR1mut, though these exploratory findings require additional follow up for data maturity. Higher frequencies of FGFR1, CCND1, and KRAS alterations at CAPI progression warrant further testing as potential acquired resistance mediators.
Presentation numberPS2-09-22
The Impact of Breast Cancer Histology and Tumor Molecular Subtype on Nodal Positivity
Erin Elizabeth Burke, The Ohio State University, Columbus, OH
S. Myers1, Y. Gokun2, B. Slover2, K. Johnson3, A. M. Roy3, D. Stover3, N. Williams3, E. E. Burke1; 1Surgery, The Ohio State University, Columbus, OH, 2School of Biomedical Science – Center for Biostatistics, The Ohio State University, Columbus, OH, 3Medicine, The Ohio State University, Columbus, OH.
Introduction: Previous studies have demonstrated higher rates of nodal metastasis in invasive lobular carcinoma (ILC) compared to invasive ductal carcinoma (IDC) of the breast. Although nodal burden also varies by tumor molecular subtype, how this relationship differs by histology (ILC vs IDC) is incompletely understood. The preponderance of ILC tumors are ER+ HER2 non-amplified with poor response to neoadjuvant chemotherapy. Recent data highlighting efficacy of targeted therapy in HER2 low disease provide the opportunity to identify additional patients for whom a neoadjuvant approach might confer eligibility for de-escalation of axillary management. This National Cancer Database (NCDB) analysis characterizes pathological nodal disease (pN+) among patients with clinically node negative (cN0) IDC and ILC hypothesizing that pN+ disease occurs more frequently in ER+ HER2 low ILC compared to IDC. Methods: The NCDB was queried for patients with cN0 invasive breast cancer from 2018-2021 who underwent surgery with axillary staging. ER and HER2 status was defined using College of American Pathologists guidelines with HER2 IHC 1/2+ with FISH non-amplified considered as HER2 low. Multivariable logistic regression was used to identify factors associated with pN+ in the entire cohort and for the cohort with ER+ disease adjusting for age, race, ethnicity, grade and subtype as well as the interaction between histology type and tumor size. Results: Of the 404,132 patients identified, 86.3% had IDC and 13.7% had ILC. Most patients (73.3%) had a tumor size of 2-20mm, well/moderately differentiated tumor grade (79.5%) and ER/PR+ and HER2- tumors (74.7%). Although the majority had pN0 disease (83.8%), lobular histology was associated with greater pN+ (20.4% ILC vs 15.5% IDC; p<0.001). ER+ HER2 low lobular carcinomas had higher pN+ than IDC with similar molecular subtype (21.4% ILC vs 17% IDC; p<0.001). However, on multivariable analysis histology was no longer significantly associated with nodal positivity for the entire cohort or for the cohort with ER+ tumors with no missing data on tumor molecular subtype (N=118,533) (Table 1). Rather, increasing tumor size, age < 51 years, and African American race were associated with increased likelihood of pN+ disease. On assessing the interaction between tumor size and histology it was noted that ILC tumors sized 21-50mm had 7% lower odds of pN+ disease compared to IDC (aOR 0.93, 95% CI 0.87-1.00, p=0.039). However, ILC tumors sized >51 mm had 53% higher odds of pN+ disease compared to IDC patients (aOR 1.53, 95% CI 1.33-1.77). Conclusion: In this study, exploring the relationship of nodal positivity by tumor molecular subtype and breast cancer histology, tumor size was the dominant contributor to pN+ status. As such further investigation is needed to delineate which tumors, particularly as it relates to ILC, may benefit from upfront systemic therapy.
Presentation numberPS2-09-23
Preliminary Results from the NEOMET Trial: Nutritional and Exercise Interventions During Neoadjuvant Chemotherapy in Early Breast Cancer
Ida Taglialatela, University of Eastern Piedmont, Novara, Italy
I. Taglialatela1, B. Conte1, S. Nardin2, F. D’Avanzo2, V. Rossi2, B. Ruffilli1, T. Landolfo1, G. Isingrini1, G. Griguolo3, D. Soldato4, A. Coassolo1, F. Condorelli1, E. Del Grosso1, C. Bengala5, C. Branni1, J. Gennari1, G. Di Foggia2, M. Rossato6, G. Tarantino6, M. Manfredi1, V. Martini1, S. Gobbato1, L. Boni4, L. Del Mastro4, V. Guarneri3, A. Gennari1; 1Traslational Medicine, University of Eastern Piedmont, Novara, ITALY, 2Oncology Unit, AOU Maggiore della Carità, Novara, ITALY, 3Department of Oncology, University of Padova, Padova, ITALY, 4Unit of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 5Oncology Unit, S. Chiara Hospital, Novara, ITALY, 6Pharmanutra S.P.A, Pharmanutra S.P.A, Pisa, ITALY.
Background: Host metabolic status and body composition are increasingly recognized as modulators of treatment response and prognosis in early breast cancer (eBC). Nutritional and exercise interventions during neoadjuvant chemotherapy (NAT) may influence systemic inflammation, insulin sensitivity, and lipid metabolism, factors implicated in therapy resistance. However, prospective randomized data assessing their impact on metabolic health and treatment efficacy are limited. Based on prior findings from the SeNEOAD study we identified a metabolomic signature associated with a higher probability of achieving pCR after NAT. Among 445 small molecules analyzed, 61 were differentially expressed in patients with pCR versus residual disease (RD) (p<0.05); patients with RD showed lower levels of fatty acids, such as 9-hexadecenoic acid (p <0.001) and doconexent (p<0.001). These results support the rationale for a nutritional supplementation of these molecules in order to improve pCR rates. Methods: NEOMET is an ongoing randomized prospective multicenter study whose primary aim is to evaluate whether plasma metabolomic signatures can be modified by lifestyle interventions including dietary supplements and physical exercise in eBC patients undergoing NAT. Eligible patients are randomized to one of four groups: a) NAT according to molecular subtype; b) NAT plus nutritional supplementation; c) NAT plus supervised physical exercise; d) NAT plus nutritional supplementation plus supervised physical exercise. Nutritional supplementation consists of two main long-chain polyunsaturated fatty acids omega-3 (n-3 Lc-PUFA), EPA and DHA, plus a source of palmitoleic acid (hexadecenoic acid). Body composition is assessed by bioelectrical impedance analysis (BIA) at baseline and at the end of NACT, before surgery. Quality of life (QoL) is evaluated using the EORTC QLQ-C30 and QLQ-BR45 questionnaires. In this preliminary analysis we present data on BIA and QoL assessments. Results: Between October 2024 and June 2025, 27 patients with stage I-III eBC were enrolled. Of these, 6 are allocated to arm A, 8 to arm B, 6 to arm C and 7 to arm D. Median age is 53 years (range 30-69), and median BMI is 25 kg/m². Tumor subtypes included HER2+ (n=12), triple-negative (n=7), and luminal-like (n=8). Fifteen patients completed NACT and had paired data on body composition and quality of life (QoL). QoL assessments using EORTC QLQ-C30 and BR45 revealed a significant deterioration in global health status (mean score from 148.8 to 116.7, p=0.041), fatigue (26.2 to 45.2, p=0.033), and pain (4.8 to 23.8, p=0.017); physical functioning improved significantly (8.1 to 21.0, p=0.009) and emotional functioning remained stable. Paired BIA assessments showed a trend toward improved body composition: fat-free mass increased from a mean of 44.9 kg to 46.4 kg (p=0.096) and skeletal muscle mass increased from 21.2 kg to 22.1 kg; fat mass remained stable (18.9 kg to 19.0 kg, p=0.47). Adverse event rates were assessed after at least 1 cycle in 26 patients. No significant differences in grade ≥2 toxicities (CTCAE v5.0) were observed between patients receiving standard treatment and those in the experimental arms. Conclusion: The preliminary analysis from the ongoing NEOMET trial supports the safety and feasibility of integrating nutritional and exercise interventions during NAT in eBC. Despite the limited sample size, observed trends suggest potential benefits in preserving lean body mass and improving physical functioning. These findings will be further investigated in the NEOMET trial as more patients will be randomized, to clarify the clinical impact on treatment tolerance and patient well-being. Additional clinical and translational results, including omega-3 index and metabolomic analysis, will be presented at the meeting.
Presentation numberPS2-09-24
Aurora Kinase A (AURKA) a new druggable target in ER+ Inflammatory Breast Cancer
Tanu Sharma, The University of Texas MD Anderson Cancer Center, Houston, TX
T. Sharma1, A. Nasrazadani2, W. Ma3, J. Chen4, M. Kai2, B. Lim2, M. Lewis5, L. Dobrolecki5, N. Kotlov6, O. Baranov6, A. Evdokimova6, F. Paradiso6, M. Hensley6, The MDACC Inflammatory Breast Cancer Team, R. Layman2, W. Woodward1; 1Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 5Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 6BostonGene RnD, BostonGene Corporation, Waltham, MA.
Background-ER+ inflammatory breast cancer (IBC) is under-appreciated and under-investigated as an aggressive subset of ER+ breast cancer, despite having rates of metastasis comparable to triple negative (TN) IBC. Recent genome-wide profiling of tumors from patients with metastatic breast cancer has shown that IBC samples have a higher frequency of AURKA amplification than non-IBC samples. Our pilot studies in IBC validate these findings and suggest AURKA is amplified in TNIBC. We hypothesize that Aurora Kinase A (AURKA) is a viable therapeutic target in metastatic IBC and in particular ER+ IBC due synthetic lethality with ARID1A and SMARCA4 in IBC. Methods-We reviewed RNA and DNA data from clinically ordered Boston Gene tumor assays for IBC versus non-IBC breast cancer cases at MDACC. Using ER- IBC cell lines we examined the protein expression of AURKA and tested Alisertib efficacy was tested using the crystal violet staining in IBC cell lines. Senescence was examined using B-gal and ELISA assay. ER+ patient derived organoids were established from ER+PDX as an ER+ IBC model. Results-We previously demonstrated using a large multi-institutional IBC data set including 137 IBC patient tissues analyzed using Affymetrix gene expression arrays, no clear clustering among ER response genes by ER+ subtypes and high AURKA expression in both ER+ and ER- cases. Comparing 21 ER+ IBC to 432 ER+ NON-IBC patient samples, we find AURKA amplified in 14 cases, co-occurrence of AURKA amplification and proposed synthetic lethal targets in 14 cases, and a statistically significant inverse relationship between AURKA and SMARCA4 in IBC. In ER- IBC compared to non-IBC there is no enrichment in AURKA amplification in IBC, but it is highly prevalent occurring in 16 cases out of 28 and with co-occurrence of proposed synthetic lethal gene alterations in 15 cases. Expression of AURKA by immunoblotting in IBC cell lines demonstrates AURKA expression in four IBC cell lines, including two HER2+ (IBC3 and KPL4) and two triple negative (A3250 and SUM149). Higher doses promote reversible senescence assayed by SA-βgal assay and high IL6 by ELISA validated senescence associated secretory phenotype. In collaboration with the PDX program at Baylor College of Medicine led by Dr. Michael Lewis our team has developed a patient derived xenograft (PDX) from an ER+ IBC patient and passaged patient derived organoids for alisertib assessment in a novel ER_ IBC model. Conclusions-AURKA is commonly amplified in IBC and often associated with gene alterations that may predict synthetic lethality to AURKA targeting. In ER+ IBC AURKA amplification is inversely correlated with SMARCA4. We have characterized IBC models with high AURKA expression and demonstrated sensitivity to AURKA inhibition but senescence at higher doses which may provide a direction for targeting resistance or escape. Further studies in ER+ organoids are ongoing.
Presentation numberPS2-09-25
Monitoring Upregulation of PD-L1 in Cancer Associated Macrophage-Like Cells in Blood Predicts Clinical Outcomes in Metastatic Cancer Patients Treated with PD-L1 Immunotherapies
Daniel L Adams, CreatvBio, Monmouth Junction, NJ
D. L. Adams1, M. Cristofanilli2, C. Reduzzi2, W. V. Williams3, G. Del Priore3, S. Chumsri4, T. Iwase5, N. T. Ueno6, A. B. Duffy7, C. Tang8; 1R&D, CreatvBio, Monmouth Junction, NJ, 2Department of Medicine, Weill Cornell Medicine, New York, NY, 3Management, BriaCell Therapeutics Corp, Philadelphia, PA, 4Oncology, Mayo Cancer Clinic, Jacksonville, NJ, 5Clinical Trials Office, University of Hawaiʻi Cancer Center, Honolulu, HI, 6Medicine, University of Hawaiʻi Cancer Center, Honolulu, HI, 7R&D, Creatv Microtech, Inc, Monmouth Junction, NJ, 8Management, Creatv Microtech, Inc., Rockville, MD.
BackgroundNumerous studies have shown the efficacy of PD-L1/PD-1 based immunotherapies (IMT) for treating patients with metastatic Breast Cancer (MBC) when there is high expression of PD-L1 in their tumor cells and/or tumor immune cells. However, many patients with high PD-L1 tumors do not respond to IMT, and some patients with negative/low PD-L1 tumors may respond to IMTs. The poor correlations between IHC scores and IMT responses are often attributed to the dynamic nature of PD-L1 which may upregulate after chemotherapy, radiation, or other systemic therapies, which is not quantified by IHC tumor immunostaining. Recently, a number of clinical trials have described PD-L1 upregulation in giant phagocytic stromal macrophages found in circulation, i.e. Cancer associated macrophage-like cells (CAML), that may correlate with IMT responses in solid tumors. We conducted a prospective pilot study to monitor the peripheral blood of n=67 MBC patients undergoing systemic treatment with IMT in combination with other therapies, to evaluate CAML PD-L1 prior to and post IMT induction (~47 days) with clinical outcome analysis at 2 years. MethodsIn a prospective pilot study of previously treated MBC patients n=67, all starting new lines of systemic therapy in combination with IMT (pembrolizumab [n=57], nivolumab [n=2], or atezolizumab [n=8]) with active progressive metastatic disease. Of the patients, median age was 58.2 (range: 32-73). Median prior lines of therapy in the metastatic setting was 2 (range 0-14). Race demographics (Caucasian=43, Black=7, Hispanic=4, Asian=3 & unknown/other=11). ECOG 0 (n=34) and ECOG 1 (n=33). Visceral metastasis (n=43) and non-visceral (n=24). ER positivity was 37% (n=25), PR positivity was 39% (n=26), HER2 positivity was 13% (n=9) and 52% were triple negative breast cancer (n=35). Histology was IDC (n=52), Other (n=11), ILC (n=2), & IBC (n=2). We isolated CAMLs from 7.5 ml baseline (T0) blood using the LifeTracDx® PD-L1 test and scored PD-L1 as high or low. If possible, a follow-up sample (T1) was taken (~47 days) after IMT induction. Patient’s progressive free survival (PFS) and overall survival (OS) hazard ratios (HRs) were analyzed by censored univariate analysis based on RECIST v1.1 over 2 years. Results Baseline T0 PD-L1 CAML data was available for 94% (n=63/67) of patients, with 44% (n=28/63) having high CAML PD-L1 which was not correlated with improved PFS (HR=1.3, 95%CI=0.7-2.3, p=0.5376) or OS (HR=1.3, 95%CI=0.7-2.5, p=0.5915). T1 PD-L1 CAML data was available for 78% (n=52/67) of patients, with 58% (n=30/52) having high CAML PD-L1 which significantly correlated with improved PFS (HR=3.1, 95%CI=1.3-7.2 p=0.0170,) and improved OS (HR=4.2, 95%CI=1.6-11.4, p=0.0102). In comparing CAML PD-L1 change post IMT, it was found that consistently low PD-L1 at T0 & T1 had the poorest responses, median PFS (mPFS)=2.2 months and median OS (mOS)=7.2 months. In contrast, patients who increased CAML PD-L1 after IMT induction had better responses, mPFS=6.3 months and mOS=13.1 months. Further, patients with consistently high PD-L1 at T0 & T1 had the best response rates, mPFS=9.5 months and mOS=19.7 months. Conclusion Here we describe a blood-based biopsy which can monitor dynamic PD-L1 changes in circulating tumor immune cells and may predict enhanced clinical benefit to PD-L1 IMTs in MBC. While an initial pilot study on a small population of patients, PD-L1 expression on CAMLs appears to identify patients with improved IMT responses and suggests larger validation studies are needed.
Presentation numberPS2-09-26
Circulating microRNA and cytokine signatures associated with interstitial lung disease in patients treated with trastuzumab deruxtecan.
Manuel Alva Bianchi, Hospital 12 de Octubre, Madrid, Spain
M. Alva Bianchi1, M. Cobos2, R. Sánchez-Bayona1, P. Tolosa1, I. Diaz3, L. Lema1, A. Madariaga1, J. Montes1, W. Parra1, C. Martín-Arriscado2, M. Luis1, L. Manso1, M. Dueñas4, E. Ciruelos Gil1; 1Medical Oncology, Hospital 12 de Octubre, Madrid, SPAIN, 2Medical Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, SPAIN, 3Medical Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12)., Madrid, SPAIN, 4Medical Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, Spain., Madrid, SPAIN.
Background: Interstitial lung disease (ILD) is a clinically relevant adverse event associated with trastuzumab deruxtecan (T-DXd) in HER2-positive and HER2-low metastatic breast cancer. Most ILD cases are low grade, but severe forms can occur, particularly if diagnosis is delayed. To better understand the biology of T-DXd-related ILD, we conducted a translational study analyzing circulating microRNAs (miRNAs) and cytokines at the time of ILD diagnosis, comparing patients who developed ILD (cases) with those who did not (controls). Other anticancer agents, including mTOR and CDK4/6 inhibitors, have also been linked to ILD. Since these therapies are often administered sequentially during the disease course, an exploratory cohort of ILD cases from non-T-DXd treatments was also included to characterize potential shared or treatment-specific molecular signatures.Methods: We conducted a prospective, single-center case-control translational study including patients with HER2-positive or HER2-low metastatic breast cancer treated with T-DXd. The primary objective was to characterize circulating miRNA and cytokine profiles in patients who developed ILD (cases) compared with those who did not (controls). Controls were matched 1:1 by treatment duration and tumor subtype (HER2+ or HER2-low). An exploratory cohort included ILD cases attributed to other anticancer agents (everolimus and abemaciclib) administered sequentially during the disease course. Plasma samples were collected at the time of ILD diagnosis or at an equivalent treatment cycle for controls. ILD diagnosis was made at the investigator’s discretion and graded according to CTCAE version 5.0. miRNA expression was analyzed using the NanoString nCounter® Human v3 miRNA panel, and cytokine levels were measured using LEGENDplex™ multiplex immunoassays targeting immune response and checkpoint markers. Differential expression analyses were performed across cohorts.Results: 12 ILD cases and 12 matched controls treated with T-DXd were included. Mean age was 62.7 vs 57.4 years, with balanced distribution of tumor subtype (HER2-positive vs HER2-low), treatment duration, and baseline clinical characteristics. Differential expression analysis identified 11 overexpressed and 5 underexpressed plasma miRNAs in ILD cases compared to controls (p<0.005) (Table 1). Among the underexpressed miRNAs, hsa-miR-n and hsa-miR-o have been associated with inflammatory pathways, as well as hsa-miR-a, which is overexpressed. However, hsa-miR-q and hsa-miR-p, found underexpressed, are linked to fibrotic processes. Cytokine profiling showed increased levels of IL-6, CXCL10 and IFN-γ; and lower levels CCL2 in ILD cases. Inverse expression patterns were observed between selected miRNAs and cytokines: hsa-miR-a and CCL2, hsa-miR-n and IL-2, hsa-miR-q and Tim-3. In the exploratory cohort (n=6; 3 everolimus, 3 abemaciclib), hsa-miR-m, hsa-miR-n and hsa-miR-o were underexpressed; while hsa-miR-a, hsa-miR-b and hsa-miR-c were overexpressed, which correlates with the expression profile observed in T-DXd-related ILD.Conclusions: This is the first study to characterize circulating microRNA and cytokine profiles associated with ILD in patients treated with trastuzumab deruxtecan. Distinct inflammatory and fibrotic signatures differentiated ILD cases from matched controls, with consistent associations observed between selected miRNAs and immunemediators. A subset of these alterations was also found in ILD cases attributed to everolimus or abemaciclib. These findings support the potential utility of circulating biomarkers in drug-induced ILD and may provide a basis for future studies focused on early detection and patient stratification.Elevated expression levelsin ILD cases compared tocontrols (UP)HSA-miR-aHSA-MIR-bHSA-MIR-cHSA-MIR-d+HSA-MIR-eHSA-MIR-fHSA-MIR-gHSA-MIR-hHSA-MIR-iHSA-MIR-jHSA-MIR-kHSA-MIR-lDecreased expressionlevels in ILD casescompared to controls(DOWN)HSA-miR-mHSA-miR-nHSA-miR-oHSA-miR-pHSA-miR-qTable 1 List of candidate mRNAs as biomarkers for ILD.
Presentation numberPS2-09-27
Patterns of central nervous system (CNS) and leptomeningeal (LM) involvement in advanced breast cancer (BC) with invasive lobular histology: insights from a large multicenter study with a focus on estrogen receptor-positive (ER+)/HER2- disease
Federica Miglietta, University of Padova, Padova, Italy
F. Miglietta1, C. Vernieri2, T. Giarratano3, G. Griguolo1, M. Bottosso1, A. Vingiani2, F. Piacentini4, M. Cacciatore5, A. Botticelli6, G. Fotia2, P. Napolitano1, G. Landa1, G. Vernaci3, E. Di Liso3, L. Casadei7, D. Palleschi8, G. Pruneri9, A. Dei Tos10, V. Guarneri1, M. Dieci1; 1Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, ITALY, 2Medical Oncology Department, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milano, ITALY, 3UOC Oncologia 2, Istituto Oncologico Veneto (IOV) – IRCCS, Padova, ITALY, 4Dept. of Medical and Surgical Sciences for Children and Adults, University Hospital, Azienda Ospedaliero – Universitaria Policlinico di Modena, Modena, ITALY, 5natomia Patologica,, Presidio Ospedaliero S. Maria di Ca Foncello Azienda ULSS 2 Marca Trevigiana, Treviso, ITALY, 6Department of Clinical and Molecular Medicine,, Policlinico Umberto I, Roma, ITALY, 7Dept. of Medical and Surgical Sciences for Children and Adults, University Hospital, Azienda Ospedaliero – Universitaria Policlinico di Modena,, Modena, ITALY, 8Oncologia, Presidio Ospedaliero S. Maria di Ca Foncello Azienda ULSS 2 Marca Trevigiana, Treviso, ITALY, 9Department of Advanced Diagnostics, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milano, ITALY, 10Department of Medicine, University of Padova, Padova, ITALY.
Background: Invasive lobular carcinoma (ILC) is the second most common BC histological subtype and the most frequent among special histological types, accounting for 10-15% of all BCs. While ILC is known to exhibit distinct metastatic patterns compared to carcinoma of no special type (NST, formerly invasive ductal carcinoma), little is known about its specific propensity for CNS and LM involvement, particularly in ER+(>=10%)/HER2- BC. Methods: We leveraged a large multicenter study of 1710 pts with advanced BC. Classification of tumor histology: NST, ILC, other special-types. CNS events: brain-only (parenchymal), LM (sub-classified as isolated-LM: no concurrent parenchymal brain mets; all-LM events: isolated + co-occurring cases). Survival endpoints: CNS-free survival (from BC diagnosis to the first CNS event/last follow up), LM-free survival (from BC diagnosis to LM as first CNS event/last follow up), post-LM survival (from LM as first CNS event to death/last follow up). Fine-Gray competing risk model was used to test the association between histology (ILC vs NST) and the risk of LM as first CNS event, accounting for brain-only as a competing event. Results: Among 1587 pts with known tumor histology, 15.5% (n=245) had ILC and 4.5% (n=72) other special-type histology, with an ER+/HER2- phenotype in 64.5% (n=158) and 41.6% (n=30) of cases, respectively. ILC was significantly enriched in ER+/HER2- tumors compared to triple-negative and HER2+ subtypes (p<0.001). A total of 526 pts developed a CNS event (n=219 in ER+/HER2-), including 466 brain-only (n=181 in ER+/HER2) and 60 LM (n=38 in ER+/HER2-). LM was diagnosed by radiological imaging in 90% of cases, while 10% were diagnosed by CSF cytology, in the absence of radiological evidence. At a median follow up of 16 years, among ER+/HER2- BC pts, those with ILC and NST experienced similar CNS-free survival, both numerically longer than that of other special-type histologies (10-yr CNS-free survival: NST 70.0%; ILC 68.1%; other 34.9%, p=0.083). In contrast, LM-free survival was significantly shorter in ILC compared with NST and other special histologies both when considering all LM events and isolated-LM (all-LM events, 10-yr LM-free survival: NST 95.6%; ILC 89.0%; other 92.3%, p=0.005; isolated-LM, 10-yr LM-free survival: NST 96.0%; ILC 89.9%; other 95.2%, p=0.003). In multivariate Cox models (adjusted for age, histologic grade, % estrogen/progesterone receptor expression, % ki67 and visceral involvement as first site of relapse), ILC was independently associated with worse LM-free survival than NST both when considering all-LM events and isolated-LM (all-LM events: adjusted HR for ILC vs NST [ref] = 5.21 [95%CI 1.54-17.60] p=0.008; isolated-LM; adjusted HR for ILC vs NST [ref.] = 5.36 [95%CI 1.59-18.14] p=0.007). Competing risk analysis confirmed a significantly increased risk of LM involvement as the first CNS event in ER+/HER2- BC pts with ILC compared to NST (HR for ILC vs NST [ref] 2.94 [95%CI 1.56-5.56], p<0.001). Among pts with LM disease, ILC was associated with significantly worse post-LM survival compared to NST (1-yr post-LM survival: NST 30.0% vs ILC 5.9%, p=0.036; HR of ILC vs NST [ref] 2.10 [95%CI 1.04-4.23] p=0.036). Conclusions: In ER+/HER2- advanced BC, compared with NST, ILC was significantly and independently associated with a marked tropism for LM involvement as first CNS event. Moreover, among pts with LM disease, those with ILC experienced particularly poor outcome, with a post-LM survival significantly worse than that of pts with NST-associated LM. These findings underscore the need for closer clinical vigilance and possibly tailored CNS surveillance strategies in pts with ER+/HER2- advanced BC of ILC histology.
Presentation numberPS2-09-28
Use of DiviTum<sup>®</sup>TKa assay to assess CDK4/6 inhibitor medication compliance and drug interactions.
Mariya Rozenblit, Yale University School of Medicine, Yale Cancer Center, New Haven, CT
M. Rozenblit1, A. Kahn1, N. Wiesendanger1, S. Schellhorn1, N. Fischbach1, D. Boyd1, M. DiGiovanna1, K. Fenn1, W. Kidwai1, K. Sabbath1, I. Krop1, T. Sanft1, A. Silber1, J. Du1, W. Amy2, M. Lustberg1, L. Pusztai1; 1Medical Oncology, Yale University School of Medicine, Yale Cancer Center, New Haven, CT, 2Clinical Development and Medical Affairs, Biovica International, San Diego, CA.
Background: CDK4/6 inhibitors (CDK4/6i) + endocrine therapy (ET) are 1st line treatment for metastatic hormone positive breast cancer (ER+mBC). CDK4/6is have several known drug interactions (i.e. with CYP3A inducers) leading to decreased serum concentrations and potentially shorter progression free survival (PFS). The DiviTum®TKa assay by Biovica was designed to measure thymidine kinase, a marker of cell proliferation, and FDA approved in 2022 as a prognostic marker in ER+mBC. TKa suppression by CDK4/6is is associated with prolonged PFS. We assessed if TKa values at C1D15 could identify low compliance or drug interactions leading to sub-optimal TKa suppression and if counseling and adjusting concurrent medications could restore TKa suppression. Methods: This is a single institution pilot study of patients with ER+mBC treated with 1st line CDK4/6i+ET. Serum samples from routine blood draws were sent to CLIA-certified Biovica lab. Participants with sustained TKa levels (>100 DuA) at C1D15 were assessed for medication compliance and drug interactions using Morisky Adherence Scale and chart review of all medications. TKa levels were then repeated at subsequent standard blood draws. Results: 30 patients enrolled 1/2024 – 5/2025, median age 64, all female, 77% white, 57% on ribociclib, 77% without visceral metastases. All participants had at least 3 consecutive TKa levels. Prior to treatment TKa results were available for 11 participants, among these median DuA was 228, 5 had <100 DuA. 15/16 participants had suppressed (2000 on therapy and had progression of disease (POD) in 2 mo. 3 participants had initial decrease in TKa levels followed by subsequent rise. One participant was mistakenly taking abemaciclib qdaily and after correcting to bid her TKa levels decreased. Another participant was briefly on amoxicillin-clavulanate for sore throat and TKa levels decreased after antibiotic completion. The third participant could not tolerate ribociclib due to fatigue and TKa levels were elevated during treatment hold, then decreased when she was switched to palbociclib. One participant continued to have sustained TKa levels on CDK4/6i after an initial decrease, no medication adherence or drug interactions were identified, and she had POD within 3 months. All participants with suppressed TKa levels (25/30) remain on treatment with stable disease at median follow up of 8 mo. Conclusions: The Divitum ® TKa assay can be a useful tool in monitoring adherence and drug interactions in the clinic that could be addressed with counseling and concomitant medication review. Persistently elevated TKa levels early on-treatment predict short PFS, whereas suppression of TKa levels is associated with prolonged PFS.
| N | % or range | |
| Participants | 30 | |
| Age | Median 64 | 35-88 |
| White | 23 | 77% |
| Black | 4 | 13% |
| Asian | 1 | 3% |
| Unknown | 2 | 7% |
| Visceral Disease | 7 | 23% |
| Non-visceral disease | 23 | 77% |
| Ribociclib | 17 | 57% |
| Abemaciclib | 11 | 37% |
| Palbociclib | 2 | 6% |
| Follow Up | Median 8 mo | 3-18mo |
| Pre-treatment TKa | Median 228 DuA | 27-2600 DuA |
| C2D28 TKa | <100 DuA | 83% |
| N | mPFS | |
| TKa consistently suppressed (<100DuA) | 25 | not reached |
| TKa elevated then suppressed | 3 | not reached |
| TKa consistently elevated (DuA>100) | 2 | 2.5mo |
Presentation numberPS2-09-29
Validity of the p53 antibody DO-7 as a surrogate for predicting TP53 mutation status in breast cancer
Joshua Van Allen, University of Connecticut, Farmington, CT
J. Van Allen1, A. Alvarez Soto1, P. Hegde2, M. Sanders2, K. Claffey3, B. Malowitz4, S. Tannenbaum1; 1Hematology and Oncology, University of Connecticut, Farmington, CT, 2Pathology and Laboratory Medicine, University of Connecticut, Farmington, CT, 3Department of Cell Biology, University of Connecticut, Farmington, CT, 4School of Medicine, University of Connecticut, Farmington, CT.
Background: In breast cancer (BC), the presence of TP53 mutation is a negative prognostic factor, also predicting endocrine resistance. Still, the clinical usefulness of determining TP53 mutation status in BC is arguable. In uterine cancer, where pathologists utilize immunohistochemistry (IHC) identifying p53 protein as a surrogate for TP53 mutation status, treatment is impacted; mutated tumors are treated more aggressively. Utilizing the well-established DO-7 p53 antibody and interpretative guidelines from uterine cancers, we sought to prove the validity of IHC testing for p53 in BC. Methods: DO-7 antibody validity in BC was performed in both laboratory and clinical samples. In the laboratory, 4 BC cell lines were utilized; 2 TP53 mutated (TP53m) (MDA-231, T47D) and 2 wild-type (MCF7, ZR75). DO-7 antibody was utilized in immunofluorescence and also western blots separated into total lysate, nuclear, and cytoplasmic compartments utilizing standard techniques. In clinical studies, 70 samples, paraffin-embedded, from 57 patients, all of whom had tissue Next Generation Sequencing (NGS), were reviewed for H&E, ER, and DO-7 staining. Standard scoring utilizing uterine cancer criteria was performed by two independent pathologists, and results were reported as wild-type or mutated and compared to NGS as the gold standard in this learning cohort. Diagnostic testing accuracy was performed for sensitivity, specificity, positive predictive value (PPV) and negative predictive value(NPV). Results: Cell line data: expected DO-7 staining was seen in TP53m lines; minimal nuclear staining in wild-type. Expectation for p53 staining in wild-type would be weak or no staining in the nucleus. Western blots showed protein expression in areas expected with increased intensity both in the nucleus and cytoplasm in TP53m. Clinical data: 57 high-risk patients, 16 neoadjuvant, 41 metastatic with 24 TP53m. Of the 49 ER+ specimens, 37% were TP53m. NGS, 31/70 tissue samples were TP53m. Concordance between IHC and NGS was 94% (66/70) with 100% specificity, 87% sensitivity for TP53m detection (see Table). In the 31 samples with IHC positive TP53m, the most observed pattern was diffuse nuclear (14/31), null/complete absence (8/31), cytoplasmic (5/31). There were 4 false-negative results. Conclusion: Our study suggests that IHC utilizing DO-7 antibody in BC specimens reflects mutation status by NGS with high sensitivity and 100% specificity. A larger testing series is planned for validation of these preliminary findings. The utilization of IHC with high specificity may impact treatment choices for BC patients, particularly for those with ER+ breast cancer in the early stage setting where NGS is not routinely performed and knowledge of endocrine sensitivity is critical.
| NGS TP53 Mutated | NGS TP53 Wild-type | ||
| IHC p53 Mutated | 27 | 0 | PPV: 100% |
| IHC p53 Wild-type | 4 | 39 | NPV: 90.7% |
| Sensitivity: 87.1% | Specificity: 100% |
Presentation numberPS2-09-30
Combination of a novel MELK inhibitor with standard of care treatment results in tumor regression and improved survival in a triple-negative inflammatory breast cancer xenograft model.
Mohd Mughees, UT MD Anderson Cancer Center, Houston, TX
M. Mughees1, R. Pathania2, N. Fowlkes3, J. Wang4, S. Krishnamurthy5, S. Damodaran1, W. Woodward6, K. Hunt7, A. Sahin5, K. Dalby8, B. Chandra1; 1Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, 2Veterinary Medicine & Surgery, UT MD Anderson Cancer Center, Houston, TX, 3Stem Cell Transplantation, UT MD Anderson Cancer Center, Houston, TX, 4Biostatistics, UT MD Anderson Cancer Center, Houston, TX, 5Pathology, UT MD Anderson Cancer Center, Houston, TX, 6Breast Radiation Oncology, UT MD Anderson Cancer Center, Houston, TX, 7Breast Surgical Oncology, UT MD Anderson Cancer Center, Houston, TX, 8Chemical Biology & Medicinal Chemistry, The University of Texas at Austin, Austin, TX.
Background Patients with highly aggressive breast cancers, such as triple-negative breast cancer (TNBC) and inflammatory breast cancer (IBC)—face clinical challenges, due to advanced stage at diagnosis, high risk for development of metastasis, and limited treatment options. Therefore, the development of novel treatment strategies is an urgent unmet need. We previously demonstrated that MELK is highly expressed in TNBC and IBC, and plays a key role in promoting stemness, epithelial-mesenchymal transition (EMT), and metastasis in a xenograft model. The inhibition of MELK reduced fibronectin (FN1) expression in vitro and in a xenograft mouse model, impairing the formation of extracellular fibronectin fibrils and ultimately decreasing cancer cell motility in TNBC/IBC cells. In the current study, we extended our investigation to patient samples to assess MELK expression, EMT markers, and macrophage infiltration in the tumor microenvironment. Additionally, to support clinical translation, we evaluated the efficacy of MELK inhibitors in combination with standard-of-care agents—paclitaxel (PT) and eribulin (ERB) in TNBC/IBC xenograft models. Materials and Methods We evaluated the efficacy and toxicity of MELK-inhibitors (MELK-In-17, MELK-In-30e, and OTS167), alone or in combination with paclitaxel or eribulin, using the 4T1(TNBC) and SUM149 (TN-IBC) mouse models. The multiplex immunofluorescence (mIF) staining was performed on tissue microarrays from 179-TNBC and 278-invasive breast cancer samples from the MD Anderson Breast Tumor Tissue Bank. The panel of markers included MELK, N-cadherin, E-cadherin, vimentin, fibronectin, PANCK, arginase1, CD163, IBA-1, CD206, and PD-L1. Results In the 4T1 model, MELK inhibitors alone or in combination with paclitaxel significantly reduced tumor growth compared to vehicle. MELK-In-30e was more effective than OTS167, though no synergistic effect was observed with paclitaxel combinations. All treatment groups showed reduction in FN1 and Ki67expression, with the greatest decrease in 30e and 30e+paclitaxel groups. In the SUM149 tumor model, treatment with 30e plus eribulin resulted in significantly greater tumor suppression compared to 30e alone (p = 0.02), while 30e plus paclitaxel did not demonstrate synergy. OTS167 showed modest tumor growth inhibition as a monotherapy and in combination with paclitaxel or eribulin showed limited effect. Survival was improved with 30e+paclitaxel and 30e+eribulin combinations. Ki-67 expression was reduced across all treatment groups, with the most substantial decrease in the 30e+eribulin group. 30e alone or 30e+eribulin also led to decreased expression of MELK, FN1, and vimentin. No significant toxicity was observed in the monotherapy or combination treatments. mIF staining of patient tissues revealed variable expression of MELK and EMT markers, with consistent moderate to high PDL1 expression and frequent infiltration of macrophages exhibiting immunosuppressive “M2”-like phenotypes frequently noted. Conclusions and Future Directions MELK-In-30e combined with eribulin significantly inhibited tumor growth and suppressed EMT and proliferative markers. mIF analysis of patient samples revealed a tumor microenvironment enriched in PD-L1 expression and M2 macrophages, suggesting immune evasion mechanisms. These findings provide a rationale for further investigation into MELK-targeted combination therapies to enhance the efficacy of existing therapies and immune activation in aggressive cancers. We plan to assess the prognostic value of MELK, EMT, and immune markers based on mIF analysis of patient samples.
Presentation numberPS2-10-01
Mutational profiles from liquid biopsies in early versus late progressors treated with CDK inhibitors in 1st line metastatic breast cancer
Marie Brichard, Cliniques universitaires Saint-Luc (CUSL), Brussels, Belgium
M. Brichard1, C. Roussel-Simonin2, M. Ibanez Alda3, J. Araujo4, J. Doxon-Douglas5, C. Bousrih2, C. Poisson2, A. Sabau2, T. Grinda2, A. Viansone2, L. Antoun2, S. AKLA2, E. Rassy2, J. Zeghondy2, F. Giugliano5, B. Verret2, C. Nicotra6, M. NgoCamus6, A. Bayle7, B. Pistilli2, A. Italiano8, J. M. Ribeiro2, A. Fernandez-Martinez6; 1Medical oncology department, Cliniques universitaires Saint-Luc (CUSL), Brussels, BELGIUM, 2Breast unit, medical oncology department, Gustave Roussy Cancer Campus, Villejuif, FRANCE, 3Medical oncology department, Hospital Universitario Marqués de Valdecilla, Santander, SPAIN, 4Department of Medical Oncology, Unidade Local de Saude Santa Maria, Lisboa, FRANCE, 5Unit 981, Molecular Predictors and New Targets In Oncology, Gustave Roussy Cancer Campus, Villejuif, FRANCE, 6Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, FRANCE, 7Bureau Biostatistique et Epidémiologie, Gustave Roussy, Villejuif, FRANCE, 8Department of Medical Oncology, Institut Bergonié, Bordeaux, FRANCE.
Background: Approximately 10% of patients (pts) with hormone receptor-positive, HER-2 negative (HR+/HER2-) metastatic breast cancer (MBC) experience disease progression within 12 months (m) of initiating first-line treatment with CDK4/6 inhibitors (CDK4/6i). Early progressors (EP) have a particularly poor prognosis, underscoring a need for personalized strategies guided by integrated clinical and molecular profiling. Our objective is to compare clinical and molecular features between EP and late progressors (LP) to uncover mechanisms driving early progression. Methods: We conducted a retrospective analysis of clinical and plasma-derived circulating tumor DNA (ctDNA) genomic data from HR+/HER2- MBC pts treated with CDK4/6i enrolled in the STING molecular screening program (NCT04932525). Eligible pts were identified through the Molecular Tumor Board database at Gustave Roussy. Genomic profiling was carried out by FoundationOne Liquid CDx assay, which analyzes 324 cancer-related genes on ctDNA in peripheral blood. Pts were classified as EP or LP, with EP defined as progression occurring within 12m after initiation of first-line CDK4/6i. Liquid biopsy was performed at disease progression to CDK4/6i. Clinicopathological and molecular characteristics were compared between EP and LP, using a Wilcoxon rank-sum for continuous variables and Chi-squared or Fisher exact tests for categorical variables. Results: From Feb 2021 to Aug 2022, 116 pts were identified. 58% (n=67) were classified as EP and 42% (n=49) as LP. This higher proportion of EP aligns with the fact that these pts may have undergone more liquid biopsies. With a follow-up of 48m, the median real-world progression-free survival on CDK4/6i (PFS) in the overall population was 11m (9-13m); 6m (4-9m) for EP, and 21m (16-28m) for LP. Overall, 23 pts (20%) had de novo MBC, of which 10 pts were EP and 13 pts were LP. No significant differences were found for age, menopausal status, hormone sensitivity (defined as the absence of progressive disease on or within the first year of completion of adjuvant hormone therapy), visceral metastases or tumor histology (ductal, lobular or mucinous). A total of 77% of pts (n = 90) had a liquid biopsy just after disease progression on first-line CDK4/6i, including 58 EP and 32 LP. Notably, EP exhibited a statistically significantly higher frequency of TP53 (EP 52% vs LP 16%; p < 0.001) and a non-significant higher proportion of PIK3CA mutations (EP 41% vs LP 22%; p = 0.079). ERBB2 mutations were identified exclusively in EP (n= 4, 3.4%), with a median real-world PFS of 7.6m. ESR1 mutations were numerically more frequent in LP (EP 21% vs LP 27%, p = 0.18). Median tumor mutational burden (TMB) was significantly higher in EP (EP 2.53 vs LP 1.26 mutations/Mb p=0.026); circulating tumor fraction was higher in EP (mean EP 11.8 vs mean LP 5.21, p=0.047). No significant differences were found in copy number alterations or gene rearrangements. Conclusion: Early progression to first-line CDK4/6i in HR+/HER2- MBC is associated with distinct genomic features, including a significantly increased frequency of TP53 mutations, increased TMB and increased circulating tumor fraction. These characteristics may be reflective of more aggressive disease biology and a non-luminal phenotype. TP53 is emerging as a known resistance mechanism to CDK4/6i and preclinical data suggest this could be overcome by CDK2 inhibition, indicating that ctDNA-based genomic profiling could be used as a tool to identify high-risk patients and support the development of precision treatment approaches for early progressors.
Presentation numberPS2-10-02
Liquid Biopsy Tracking of PI3K-mTOR Residual Disease Signatures in Metastatic Breast Cancer
Sudha Rao, QIMR Berghofer Medical Research Institute, Herston, Australia
M. Melino1, W. Tu1, S. Goh1, A. Bain1, J. Friend2, T. Prasanna3, D. Yip3, A. Ives4, M. Leahy4, D. Lewis4, M. Nottage5, S. Rao1; 1Cancer Research, QIMR Berghofer Medical Research Institute, Herston, AUSTRALIA, 2Kazia Therapeutics Ltd, Kazia Therapeutics Ltd, Sydney, AUSTRALIA, 3Medical Oncology, The Canberra Hospital, Garran, AUSTRALIA, 4Cancer Care Services, The Royal Brisbane and Women’s Hospital, Herston, AUSTRALIA, 5Medical Oncology, The Royal Brisbane and Women’s Hospital, Herston, AUSTRALIA.
Background Breast cancer is the most commonly diagnosed cancer globally and remains the leading cause of cancer-related death in women. HER2-positive breast cancer, comprising 15-20% of cases, is defined by overexpression of the HER2 tyrosine kinase receptor. While HER2-targeted therapies have transformed clinical outcomes, resistance, recurrence, and metastasis continue to pose significant challenges. Immunotherapy has revolutionised the treatment of several solid tumors by enhancing anti-tumor immunity; however its efficacy in HER2-positive breast cancer has been limited. There is a pressing need for biomarkers that can both track metastatic burden and identify patients most likely to benefit from immunotherapeutic strategies. In this study, we present novel preclinical data from liquid biopsy analyses and propose a biomarker-driven approach targeting the PI3K-mTOR pathway to enhance immune reinvigoration in HER2-positive metastatic breast cancer. Methods Twenty five stage IV HER2-positive metastatic breast cancer (mBC) patients underwent liquid biopsy profiling using an epigenetic biomarker targeting the PI3K-mTOR signaling pathway. We applied digital pathology, Duolink® proximity ligation assays, global immune profiling, and flow cytometry to characterize biomarker expression and immune landscape features. Ex vivo drug screening with PI3K-mTOR inhibitors, including the brain-penetrant paxalisib, was performed to evaluate their effects on mesenchymal phenotypes, disruption of circulating tumor cell (CTC) clusters, and immune reinvigoration. Results We identified a novel epigenetic switch that drives the transition to a more aggressive, drug-resistant mesenchymal phenotype in HER2-positive breast cancer, associated with activation of the nuclear chromatinised PI3K signaling pathway. This phenotype, detectable via liquid biopsy, correlates with metastatic burden and immune exhaustion. In our pilot cohort, we evaluated the interplay between PI3K mutational burden and activation of this mesenchymal epigenetic program. Functional drug response profiling of patient-derived samples demonstrated that dual PI3K-mTOR inhibition not only facilitated mesenchymal-to-epithelial transition but also suppressed key metastatic signatures, including those linked to the highly aggressive persister cell phenotype (p65, FOXQ1, NRF2, and NNMT) and cancer drug resistance (ABCB5, SNAIL, and ALDH1). Importantly, PI3K-mTOR inhibition disrupted metastatic progenitors, including circulating tumor cell (CTC) clusters, and enhanced anti-tumor immune responses, as evidenced by modulation of both progenitor-like and terminally exhausted T cell populations. Conclusions Our preclinical findings support a biomarker-driven therapeutic strategy targeting the PI3K-mTOR pathway to induce epigenetic reprogramming and immune modulation. This dual mechanism may enhance anti-tumor efficacy by converting immunologically ‘cold’ tumors into ‘hot’, more immunogenic tumors. By reshaping the tumor microenvironment, this approach has the potential to improve the clinical utility of immunotherapy and expand treatment options for patients with HER2-positive metastatic breast cancer.
Presentation numberPS2-10-03
Impact of Circulating Tumor DNA (ctDNA) monitoring on Patient Anxiety and Clinician Decision-Making in Early-Stage Breast Cancer (PACE-ctDNA)
Devora Isseroff, Yale University, New Haven, CT
D. Isseroff1, N. Wiesendanger1, A. Kahn1, D. O’Neil1, M. Cohenuram1, A. Bulgaru1, K. Fenn1, J. LaSala1, S. Kert1, N. Fischbach1, J. Kanowitz1, R. Legare1, M. DiGiovanna1, A. Silber1, T. Sanft1, S. Schellhorn1, H. Sethi2, E. Kalashnikova2, S. Rivero-Hinojosa2, M. Liu2, I. Krop1, E. Winer1, W. Wei1, M. Lustberg1, L. Pusztai1, M. Rozenblit1; 1Medical Oncology, Yale University, New Haven, CT, 2Natera, Natera Inc, Austin, TX.
Background: Prior studies show ctDNA is a promising biomarker for detecting minimal residual disease (RD), but impact on patient (pt)-reported outcomes and physician decision-making is unknown. With growing adoption of ctDNA assays such as the Signatera™ RD Test, understanding the effect of ctDNA monitoring on pt anxiety and physician treatment choices is increasingly important. We previously reported 6 month (mo) interim results and here present updated findings at 12mo.Methods: Women ≥18y.o. with triple negative (TNBC) RD or hormone receptor positive (HR+) BC were enrolled during adjuvant surveillance. CtDNA testing was q6mo and continued for 12mo or until recurrence. Pt-Reported Outcomes Measurement Information System (PROMIS) and Provider Decision Assessment Tool (PDAT) scores were collected q6mos. Both used 5-point Likert scales: higher PROMIS scores reflect greater anxiety; higher PDAT scores reflect greater physician confidence. Mean scores were compared baseline vs 6mo and vs 12mo.Results: 67 pts enrolled (mean age: 56 y; range: 24-84). Of 45 (67.2%) pts with TNBC, stage I, II, III distribution was 26.7%, 53.3%, 20.0%, respectively. Of 22 (32.8%) pts with HR+ BC, stage I, II, III was 13.6%, 68.2%, 18.2%, respectively. 64 pts completed ctDNA testing; 7/64 (10.9%) were ctDNA+ at baseline. Of these, 3/7 (42.8%) had imaging-confirmed recurrence; 1/7 (14.3%) had negative baseline imaging and progression at 6 mos; 3/7 (42.9%) had negative imaging at baseline with ctDNA clearance on adjuvant therapy at 6mos. 4 pts who were initially ctDNA- became ctDNA+ on subsequent testing. 3/4pts had recurrence on scans at the time of ctDNA+; 1/4pts had ctDNA clearance at 12mos. Recurrent pts came off study and did not continue PROMIS testing. 23 ctDNA- pts completed ctDNA testing and PROMIS surveys at baseline, 6 and 12mos. There was no statistically significant change in scores over time and a trend toward decreasing anxiety scores in ctDNA- pts. 4 ctDNA+ pts remained on study and had PROMIS scores before and after ctDNA result (table 1). 5 physicians managing ctDNA+ pts completed a PDAT after ctDNA results. Overall decision-making scores were similar after ctDNA+ or ctDNA- results (37.8 vs. 37.1, p = 0.76); physicians caring for ctDNA+ pts self-reported lower confidence in treatment decisions (3.6 vs. 4.3). All ctDNA+ pts had imaging.Conclusion: Pt-reported anxiety was stable over time in ctDNA- pts with a trend toward lower anxiety scores. PROMIS scores after ctDNA+ result were available for only 4 pts who remained on study; as pts with recurrence came off study, we cannot fully assess how ctDNA+ results affect anxiety but present raw data. CtDNA+ results were associated with lower self-report of confidence in treatment decisions among 5 responding physicians, suggesting that without clear clinical guidelines, ctDNA surveillance can introduce uncertainty for providers.
| ctDNA- (N=23) | Baseline | 6 months | 12 months | |||
| Mean Score | 49.6 | 46.3 | 45.0 | |||
| Standard Deviation | 17.7 | 18.1 | 19.1 | |||
| ctDNA+ | Before | Before | After | After | Follow Up | Follow Up |
| Score | ctDNA Result | Score | ctDNA Result | Score | ctDNA Result | |
| Pt1 | 60 | Positive | 57 | Positive | — | — |
| Pt2 | 39 | Positive | 41 | Positive | — | — |
| Pt3 | 44 | Positive | 49 | Negative | 45 | Negative |
| Pt4 | 32 | Positive | 28 | Negative | 44 | Negative |
Presentation numberPS2-10-04
Tumor-infiltrating lymphocytes – a promising predictive and prognostic biomarker for optimizing neoadjuvant treatment response: a single-institution retrospective analysis
Bibiana Vertakova Krakovska, St.Elizabeth Cancer Centre, Bratislava, Slovakia
B. Vertakova Krakovska1, B. Mrinakova1, I. Waczulikova2, L. Vanovcanova3; 11st Department of Oncology Comenius University, St.Elizabeth Cancer Centre, Bratislava, SLOVAKIA, 2Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Bratislava, SLOVAKIA, 32nd Department of Radiology Comenius University, St.Elizabeth Cancer Centre, Bratislava, SLOVAKIA.
Introduction: Tumour-infiltrating lymphocytes (TILs) play a crucial role in the immune response against breast cancer and hold varying prognostic and predictive value depending on the molecular subtype. Elevated levels of TILs have been associated with improved response to chemotherapy and enhanced survival outcomes, particularly in triple-negative (TNBC) and HER2-positive (HER2+) breast cancers. Emerging evidence suggests a strong correlation between TIL levels and the likelihood of achieving an objective response to neoadjuvant therapy (NAT), influenced by breast cancer subtype. This study aimed to assess the impact of TILs on the overall outcomes of NAT across the whole spectrum of breast cancer subtypes, with a specific focus on TNBC and HER2+ subtypes. We evaluated the association between TIL levels and the likelihood of achieving lower Residual Cancer Burden (RCB) scores (RCB 0 and I). Methods: We retrospectively analysed 181 early-stage patients with breast carcinomas treated with NAT. RCB (179 patients) was used to assess pathological response, categorised as RCB 0-III. TILs were scored on diagnostic biopsies and grouped into three categories: I. 41%. For HER2+ patients, we compared outcomes based on receipt of anti-HER2 therapy. For TNBC, we assessed response differences between patients with NAT alone versus those treated with NAT and immune checkpoint inhibitors (ICI). Associations between TILs, treatment, and RCB response were explored. Results: Total cohort (n=179): When stratified by molecular subtype, RCB 0-I was achieved as follows: none of Lum A, 22.2% of Lum B, 48.6% of Lum B HER2+, 85.7% of HER2+ and TN in 59.4%. Immunogenic character is strongly associated with HER2+ and TN tumors where TILs II+III levels were described in Lum B HER2+ (86.5%), HER2+ (82.1%) and TN (86.2%). Low TILs levels were associated with a 3.58-fold higher likelihood of residual disease (non-RCB 0) compared to tumors with high TILs levels. HER2-positive cohort (n = 65): Among HER2-positive patients, 60 received dual anti-HER2 targeted therapy, and 5 did not. The overall rate of excellent response (RCB 0-I) was higher in patients who received targeted treatment (40/60; 66.7%) compared to those who did not (2/5; 40%). Within the treated group, higher TILs levels were associated with better outcomes (Goodman-Kruskal’s gamma = -0.361; P = 0.0625). RCB 0-I was achieved in 63.4% of patients with TILs II and 90% of those with TILs III, compared to only 55.6% in TILs I. Triple-negative cohort (n = 64): In the TNBC group, 33 patients received standard NAT alone, and 31 received NAT with ICI. The RCB 0-I rate was 57.6% in the non-immunotherapy group versus 61.3% in the immunotherapy group. The incorporation of ICI to patients with TILs I resulted in RCB 0 in 80%, while none of the patients with TILs I treated without ICI reached RCB 0. Conclusion: Our findings support the use of TILs as a biomarker in identifying patients most likely to benefit from biologic or immunologic therapies. Notably, HER2-positive and TNBC subtypes demonstrated the most favourable pathological responses (RCB 0-I). In the HER2-positive group receiving targeted treatment, higher immune infiltration (TILs II-III) resulted in a significantly improved pathological response. In TNBC, particularly in patients with low TILs, the incorporation of ICI resulted in a better outcome in comparison to patients with the same TILs level without ICI. In patients who received NAT without ICI, the response was strongly influenced by TILs levels only. These findings support the use of TILs as a biomarker to guide treatment intensification strategies in early-stage breast cancer, particularly in identifying patients most likely to benefit from biologic or immunologic therapies.
Presentation numberPS2-10-05
Association of clinicopathologic features with plasma protein expression in primary invasive breast cancer patients
Anupama Praveen Kumar, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA
A. Praveen Kumar1, P. Raj-Kumar1, J. Liu1, B. Deyarmin2, B. Mostoller3, C. Larson2, H. Blackburn2, D. Teeter2, K. Miller2, M. Johnson2, M. Russo1, L. Fantacone-Campbell4, J. Hooke4, C. Shriver5, H. Hu1, A. Kovatich4, X. Lin1; 1Bioinformatics, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA, 2Biobank, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA, 3Informatics Infrastructure, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA, 4Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University, Bethesda, MD, 5Murtha Cancer Center Research Program, Walter Reed National Military Medical Center, Bethesda, MD.
Background: Liquid biopsies using blood provide a less invasive method for cancer detection and treatment assessment compared to conventional tissue biopsies. The plasma proteome involves secreted proteins from various organs and tissues, making it a prime candidate for extensive biomarker discovery. However, the vast dynamic range of protein abundance in plasma presents a challenge. In this study, we employed a highly sensitive aptamer-based assay to explore plasma proteins associated with breast cancer (BC). Methods: The subjects were enrolled and specimens were collected through the Clinical Breast Care Project using IRB-approved protocols. We analyzed 260 heparinized plasma samples from breast cancer patients at primary diagnosis, prior to neoadjuvant chemotherapy and excisional surgery. Patients with stage IV, prior DCIS, synchronous breast cancer, or relapse within 6 months were excluded. SomaScan Assay 7k (Somalogic Inc, USA) using 7289 aptamers for human proteins was performed. The association of each clinicopathologic feature of the cohort with plasma protein expression was evaluated using linear regression. The clinical features with notable effect size were further subjected to differential expression analysis using Limma with matched samples followed by pathway analysis using Ingenuity Pathway Analysis (IPA) tool. Results: The protein expression levels were significantly (FDR < 0.05) associated with age, race and stage groups. Differential expression using matched subsets identified 359 significant (FDR 1.2) differentially expressed proteins (DEPs) between old (>60 years, n = 44) and young (<40 years, n = 44), and 184 between Caucasian (n = 67) and African American (n = 67) sample groups. We utilized data from Peptide Atlas, and existing literature on normal aging and established cancer biomarkers to contextualize the significant proteins. Pathways significantly (p val 2) upregulated in the old group included immune response (cytokine signaling, neutrophil degranulation etc.) and tumor related (PAK, SNARE signaling etc.) pathways. Pathways identified in young were PPAR, DHCR4 signaling, and LXR/RXR activation. For the race group, there were 11 significant pathways upregulated in Caucasians that included FAK and S100 family signaling. For the higher (n = 62) and lower (n = 62) stage comparison, there were 127 significant DEPs (p val 1.2) with several cancer-related signaling pathways upregulated in higher stage group (VEGF, SCF-KIT etc.). To explore potential markers of relapse, we conducted a subgroup analysis within the higher-stage cohort, comparing patients who relapsed (n = 30) to those who did not (n = 30) that identified 24 significant DEPs (p 1.2) LASSO regression with leave-one-out cross-validation (LOOCV) resulted in a predictive model comprising 18 proteins, achieving a classification accuracy of 0.92. Conclusion: This study identified plasma proteins associated with age group, race and tumor stage in breast cancer patients. These findings indicate that BC related plasma protein expression differs across age and racial groups, a factor to consider in drug target development. We also identified 18 proteins that are potential markers for BC relapse. The identified proteins need to be validated in samples of large cohort. Disclaimer: The contents of this publication are the sole responsibility of the author(s) and do not necessarily reflect the views, opinions, or policies of Uniformed Services University of the Health Sciences (USUHS), the Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., the Department of Defense (DoD) or the Departments of the Army, Navy, or Air Force. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. government.
Presentation numberPS2-10-06
Proteogenomic profiling of treatment-naïve breast cancer biopsies from patients receiving neoadjuvant chemotherapy
Eric Jaehnig, Baylor College of Medicine, Houston, TX
E. Jaehnig1, M. Holt1, J. Zecha2, M. S. Glover2, W. Kelly3, B. Nuttall3, B. McCue1, I. Marin4, S. Carter4, E. Bonefas4, L. Healy4, G. Miles1, M. Anurag1, S. Hilsenbeck1, B. Zhang1, S. Hess2, A. Thompson4; 1Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 2Dynamic Omics, Center for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, 3Translational Medicine, Early Oncology, Oncology R&D, AstraZeneca, Waltham, MA, 4Department of Surgery, Baylor College of Medicine, Houston, TX.
Background: Breast cancer is a heterogeneous disease, comprising approximately 70% hormone receptor-positive (HR+), 15% HER2-positive (HER2+), and 15% triple-negative breast cancer (TNBC) cases. As standard of care, HR+ tumors are treated with hormone therapy, often after surgery. However, chemotherapy may also be used before surgery (neoadjuvant setting). HER2+ breast cancer responds well (50-70% pathological Complete Response, pCR) to targeted anti-HER2 therapies. In contrast, TNBC is primarily treated with neoadjuvant chemotherapy with only 30-50% achieving pCR. Identifying biomarkers predictive of pCR or that indicate alternative therapeutic strategies can help tailor therapy. Recent studies utilizing proteogenomics data (DNA, RNA, protein, and phosphorylation site profiles from the same tumor) from clinical trials highlighted candidates including LIG1 loss in TNBC and GPRC5A/TPBG overexpression in HER2+ tumors as potential predictive biomarkers and therapeutic targets. This study applies proteogenomic profiling to pre-treatment biopsies from tumors treated with physician-selected neoadjuvant chemotherapy to further explore putative targets for precision oncology. Methods: Microscaled proteogenomic profiling was conducted on 51 treatment-naive primary breast tumor core needle biopsies. Subtyping was determined via IHC: HER2+ (IHC 3+), HER2 Equivocal (IHC 2+), HR+ (ER or PR positive), or TNBC (negative for all three receptors). All received physician-selected neoadjuvant regimens, and pCR was evaluated after surgical resection. Multi-omics profiling of tumor sample from each biopsy included whole-exome sequencing (WES), RNA-seq, and mass spectrometry-based proteomics and phosphoproteomics. Both data-dependent acquisition (DDA) and more sensitive data-independent acquisition (DIA) proteomics were used. Results: Proteomic profiles were obtained for 16 HER2+, 17 HR+, and 18 TNBC patients. WES data were acquired for 50 patients; RNA-seq for 26. DIA quantified more total proteins/sites than DDA, though measurements for top variable genes were highly correlated. To assess the potential for each proteomics data type to inform precision oncology, both were included in all subsequent analyses. The highest pCR rate (69%) was achieved in HER2+ tumors treated with dual antibody therapy where all 4 non-pCR tumors were HR+, a known resistance marker, and 3 were HER2 equivocal with lower HER2 protein expression. Both the DIA and DDA data for GPRC5A protein also confirmed significantly higher expression in non-pCR vs. pCR HER2+ tumors. HR+, HER2- tumors had the lowest pCR (12.5%), but two pCR cases co-treated with pembrolizumab showed high expression of cell cycle and immune proteins. TNBC showed 35% (6/17) pCR overall, improving to 56% (5/9) when treatment included pembrolizumab. Non-pCR tumors had lower cell cycle/immune protein levels and higher epithelial-mesenchymal transition protein levels versus pCR tumors. Detailed proteogenomic profiles for FDA-approved drug targets, TNBC-specific kinases, and cell membrane proteins enriched in the context of non-pCR TNBC cases will be presented. Conclusion: This study demonstrates the feasibility of proteogenomic profiling from clinical breast tumor biopsies and highlights mechanisms of anti-HER2 resistance, potential factors underlying exceptional chemotherapy response in ER+ cancers, and candidate alternative therapeutic targets for non-pCR TNBC, offering a template for future precision oncology efforts.
Presentation numberPS2-10-08
Quantitative measurement of HER2 and TROP2 predicts outcomes with trastuzumab deruxtecan in metastatic breast cancer
Charles J Robbins, Yale School of Medicine, New Haven, CT
C. J. Robbins1, W. Wei2, N. N. Chan1, M. He1, J. Benanto1, D. B. Doroshow3, I. E. Krop4, D. L. Rimm1; 1Pathology, Yale School of Medicine, New Haven, CT, 2Medical Oncology and Hematology, Yale School of Medicine, New Haven, CT, 3Internal Medicine, Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 4Medical Oncology, Yale School of Medicine, New Haven, CT.
Background: Antibody-drug conjugates (ADCs) with targets of TROP2 (sacituzumab govitecan) and HER2 (trastuzumab deruxtecan, T-DXd) have expanded treatment options for metastatic breast cancer (mBC), including HR-positive/HER2-non-amplified and triple-negative subtypes. Nevertheless, the optimal selection of patients and treatment sequencing for these ADC therapies remains a clinical challenge. While both drugs are thought to be targeted therapies, the clinical value of assessment of HER2 and TROP2 expression has not been established. Here, we evaluated whether high-sensitivity, simultaneous, quantitative immunofluorescence (QIF) measurement of HER2 and TROP2 (TROPLEX™ assay) predicts clinical outcomes in T-DXd treated patients in a Yale retrospective cohort. Methods: We identified 65 patients with mBC who received T-DXd at Yale between 4/2020 to 2/2025 and retrieved available FFPE specimens. Sections were stained on a Leica BOND RX autostainer for HER2, TROP2, and cytokeratin using the TROPLEX™ assay. Samples were imaged with the CyteFinder II HT multiplex fluorescent microscope (Rarecyte). Image analysis was performed in QuPath using an image processing plugin developed for automated QIF/IHC analysis (Qymia). Tumor regions were annotated by a pathologist, and QuPath with the Qymia plugin quantified protein concentration (attomoles/mm²) for HER2 and TROP2 against a mass-spectrometry-calibrated cell line standard curve. Time to treatment failure (TTF) was defined as time from T-DXd initiation to radiographic progression, clinical progression, or treatment discontinuation due to toxicity up to 2 years of follow-up. Biomarker expression was dichotomized at the cohort median for Kaplan-Meier analysis with log-rank testing. Results: Samples from 65 T-DXd treated patients (19 HER2+, 35 HER2-/HR+, 11 TNBC) were analyzed with the TROPLEX™ assay. Specimens included metastatic biopsies in 51 patients (78.5%), local recurrences in 8 (12.3%), and primary tumor in 6 (9.2%). Median interval from tissue sampling to T-DXd was 18.4 months (range 0.7-30.2). For this specimen cohort, HER2 median expression was 3,554 amol/mm2 (range 15 – 8,232) and TROP2 median expression was 2,868 amol/mm2 (range 0 – 23,383). For HER2 expression, patients with above-median HER2 had significantly longer TTF (median 12.8 vs. 7.5 months; log-rank p=0.019) and a hazard ratio (HR) of 0.51 (95% CI 0.29-0.91). Additionally, there was an inverse association between TROP2 expression and TTF, where patients with above-median TROP2 had significantly shorter TTF (median 5.5 vs. 12.8 months; log-rank p=0.050) and HR of 1.74 (95% CI 1.01-3.04). In a continuous analysis of TROP2 expression, each 2,000-unit increase was associated with shorter TTF (HR 1.17, 95% CI 1.07-1.29, p=0.0010). These associations for TROP2 remained consistent in non-triple-negative (n=54; HR 1.18, 95% CI 1.05-1.32; p=0.0039) and non-HER2-amplified (n=46; HR 1.14, 95% CI 1.04-1.28; p=0.0056) subgroups. Conclusions: Quantitative assessment of HER2 in amol/mm2 is correlated with TTF of T-DXd for mBC using the TROPLEX™ assay. Surprisingly, in this cohort, there appears to be an association between TROP2 amol/mm2 and TTF for T-DXd. Due to the small size of the cohort and the lack of known biological relationship between the biomarkers, this finding requires further validation with larger patient cohorts.
Presentation numberPS2-10-09
Evaluation of spatial collagen morphometry in TNBC versus non-TNBC biopsies: a cross-sectional SHG/TPE and AI-based analysis of tumor and non-tumor regions
Kutbuddin Akbary, HistoIndex, Singapore, Singapore
K. Akbary1, L. Jiaojiao1, R. Yayun1, D. Tai1, C. Hsiao2, K. Huang2; 1Research and Development, HistoIndex, Singapore, SINGAPORE, 2Dept of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, TAIWAN.
BACKGROUND: Triple-Negative Breast Cancer (TNBC) is a heterogeneous and aggressive subtype of breast cancer characterized by the absence of estrogen receptor, progesterone receptor, and HER2 amplification. The tumor microenvironment in breast cancer plays a critical role in disease progression [Acerbi et al. Integr Biol (Camb). 2015 Oct;7(10):1120-34], yet the specific spatial characteristics and morphometric patterns of collagen within different tissue regions remain poorly defined, especially when comparing TNBC and non-TNBC pathologies. Quantitative collagen analysis using second harmonic generation/two-photon excitation (SHG/TPE) imaging and AI-based collagen morphometric evaluation provides an opportunity to characterize fibrosis patterns beyond conventional histopathology. This study aims to delineate and compare collagen morphometric features between tumor and non-tumor regions in TNBC biopsies versus those in non-TNBC biopsies, and to describe region-specific patterns of fibrosis and their similarities and differences. METHODS: 266 unstained breast cancer biopsy samples acquired from Dept of Surgery, National Taiwan University Hospital were analysed using SHG/TPE microscopy and AI analysis. Samples were divided into TNBC (n=21) and non-TNBC (n=245) groups. Collagen morphometric features were identified using a proprietary AI-based analysis using the Genesis®200 scanner (HistoIndex Pte Ltd, Singapore). Four anatomically distinct tissue regions were evaluated: tumor, lobule and duct, stroma-fat, and stroma-fibrosis, with the latter three comprising the non-tumor regions. Normalised Median Relative Differences in feature values were computed to compare collagen parameters between TNBC and non-TNBC groups for each region (Parameter-wise normalization scaled median differences within the scale of -1 to +1). The Wilcoxon rank-sum test was used to assess statistical significance. RESULTS: Collagen morphometric analysis revealed region-specific differences between TNBC and non-TNBC biopsies. In tumor regions, collagen parameters were largely comparable between the two groups, with no significant differences detected. In the non-tumor stroma-fibrosis region, some parameters related to collagen fibre distribution showed numerical differences favouring the TNBC group, but these differences did not reach statistical significance. In contrast, the lobule and duct region demonstrated statistically significant differences in multiple collagen morphometric features. Specifically, TNBC biopsies exhibited increased levels of collagen strings (defined as single collagen-connected areas comprising one or more intersecting fibres) and string aggregation features, including those involving various string dimensions such as thick and thin strings. Additionally, several parameters reflecting collagen fibre distribution were significantly elevated in TNBC samples compared to non-TNBC. CONCLUSIONS: SHG/TPE-based AI analysis demonstrates that TNBC is associated with a distinct fibrosis profile, particularly characterized by significant alterations in collagen morphometric features within the lobule and duct regions of the non-tumor compartment compared to non-TNBC cases. These region-specific differences in collagen architecture likely reflect underlying desmoplastic remodelling unique to TNBC and may serve as candidate imaging biomarkers for subtype differentiation. Further exploration of these spatial collagen morphometric patterns may enhance understanding of tumor-stroma dynamics and support the development of targeted therapeutic strategies in breast cancer.
Presentation numberPS2-10-10
Ten-year outcomes of Chinese early breast cancer patients with PIK3CA/AKT1 mutations: A retrospective cohort study
Ting Luo, West China Hospital, Sichuan University, China, Chengdu, China
T. Luo1, W. Zeng1, J. Peng2, J. Yan2, Y. Gui3, Y. Song1, Z. Li1, X. Zhong1; 1Breast Center, Cancer Center, Institute of Breast Health Medicine, West China Hospital, Sichuan University, China, Chengdu, CHINA, 2West China School of Medicine, West China Hospital, Sichuan University, China, Chengdu, CHINA, 3Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, China, Chengdu, CHINA.
Background: Multiple clinical trials have demonstrated that PI3Kα and AKT inhibitors improved survival in advanced hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)- breast cancer patients with PIK3CA/AKT1 mutations. However, their prognostic relevance in early breast cancer (EBC) remains unclear. This study aimed to evaluate the prevalence and long-term prognostic impact of PIK3CA/AKT1 mutations among EBC patients in China.Methods: Based on the Breast Cancer Information Management System (BCIMS) in West China Hospital (WCH), Sichuan University, we retrospectively enrolled patients diagnosed with stage I-III breast cancer between Jan 2008 and Dec 2014 from WCH BC Cohort. Patients with available fresh-frozen tumor tissue were included. Those receiving neoadjuvant therapy were excluded. Clinical and follow-up data were extracted from BCIMS. Whole exons of PIK3CA and AKT1 were detected in fresh-frozen primary tumors by next-generation sequencing. Descriptive statistics were performed for baseline characteristics. Prognostic outcomes were analyzed by Kaplan-Meier survival curves, and hazard ratios (HRs) were calculated via multivariable Cox regression models.Results: A total of 569 EBC patients were included, with a median follow-up duration of 12.4 years. The overall mutation rate of PIK3CA/AKT1 was 50.1% (HR+/HER2-: 59.6%, HER2+: 41.3%, TNBC: 29.6%). Specifically, 47.3% patients harbored PIK3CA mutations, while 3.9% carried AKT1 mutations. Forty-five patients (7.9%) carried ≥2 variants in PIK3CA and/or AKT1. PIK3CA mutations were more common in HR+ (vs. HR-, 50.7% vs. 36.9%), HER2- (vs. HER2+, 50.0% vs. 41.3%), and WHO grade I-II tumors (vs. grade III, 52.8% vs. 44.0%). 522 patients with ≥10 years of follow-up were included in survival analysis. The 10-year invasive disease-free survival (iDFS), distant disease-free survival (DDFS), and overall survival (OS) rates for patients with PIK3CA/AKT1 mutations were 87.1%, 87.5%, and 91.1%, respectively. However, compared to wild-type patients, those with PIK3CA/AKT1 mutations showed no significant outcome differences. Furthermore, after adjusting for age, menopausal status, T stage, N stage, and treatment, multivariable Cox regression indicated that patients with ≥2 PIK3CA/AKT1 mutations conferred a significantly higher risk of progression or death compared to wild-type patients (aHR = 2.13, 95% CI: 1.02-4.45, p = 0.045), whereas single PIK3CA/AKT1 mutation did not show significant impact on prognosis. Similar trends persisted within the HR+ subgroup treated with endocrine therapy (≥2 mutations vs. wild-type, aHR = 2.39, 95% CI: 1.07-5.34, p = 0.034).Conclusion: Half of Chinese EBC patients harbored PIK3CA/AKT1 mutations, with a notably higher prevalence in the HR+/HER2- subgroup. While the long-term prognosis of patients with PIK3CA/AKT1 mutations overall was generally comparable to that of wild-type patients, those harboring multiple mutations may exhibit an increased risk of disease progression or mortality.
Presentation numberPS2-10-11
Evaluating the impact of neighborhood deprivation on CDK4/6 inhibitor use, outcomes, and circulating tumor DNA (ctDNA) alterations in patients with metastatic breast cancer
Emily L. Podany, Washington University in St. Louis, St. Louis, MO
E. L. Podany1, S. Tapiavala1, L. Foffano2, A. Medford3, N. Heater4, C. Reduzzi5, E. Nicolo5, L. Pontolillo5, A. Putur3, D. Jaber4, K. K. Clifton1, M. Lipsyc-Sharf6, F. Ademuyiwa1, F. Puglisi2, W. Gradishar4, C. X. Ma1, L. Gerratana2, A. Bardia6, M. Cristofanilli5, A. A. Davis1; 1Internal Medicine, Washington University in St. Louis, St. Louis, MO, 2Medicine, University of Udine, Udine, ITALY, 3Internal Medicine, Massachusetts General Hospital, Boston, MA, 4Internal Medicine, Northwestern University, Chicago, IL, 5Internal Medicine, Weill Cornell Medicine, New York, NY, 6Internal Medicine, UCLA Health, Los Angeles, CA.
Background: We previously showed lower rates of PI3K-targeted treatment use and shorter overall survival in patients with metastatic breast cancer (MBC) living in disadvantaged neighborhoods (Podany et al., ASCO 2025) using the validated Neighborhood Atlas Area Deprivation Index (ADI, Kind et al., NEJM 2018). ADI includes 17 measures of neighborhood disadvantage, including poverty, employment, and education. We sought to determine the use of CDK4/6 inhibitors (CDK4/6i), prognostic associations in patients on CDK4/6i with ctDNA somatic profiles, and outcomes of patients with or without CDK4/6i by ADI. Methods: This retrospective, multi-institution cohort study included 1127 patients with MBC and ctDNA testing with the Guardant360 assay treated at Washington University in St. Louis (N=634), Massachusetts General Hospital (N=313), Northwestern University (N=71), and Weill Cornell (N=109). Patient-reported 9-digit zip codes were converted into national ADI ranks (0-100) and divided into high deprivation (HDep, rank ≥60) and low deprivation (LDep, rank <60) based on prior studies. Patients were evaluated for use of CDK4/6i in the first and second line setting by ADI. Progression-free survival (PFS) was evaluated in the first line by CDK4/6i use and ADI. Multivariate models were then designed to determine prognostic differences in somatic mutations by ADI. Results: 335 patients (29.7%) lived in HDep neighborhoods, including 59 of the 203 patients (29.0%) with hormone receptor-positive (HR+) HER2 negative (HER2-) MBC and evaluable zip codes and first line treatment information. 144 of these HR+ HER2- patients received CDK4/6i in the first line and 40 received CDK4/6i in the second line. There was no significant difference in CDK4/6i use in the first line by ADI (74.3% in LDep vs 62.7% in HDep, p=0.099) or second line (29.1% vs 28.6%, p=0.96). Patients living in LDep neighborhoods were more likely to experience disease progression on CDK4/6i if they had a baseline ESR1 single nucleotide variation (snv) on ctDNA profiling (Hazard ratio [HR] 2.22, 95% confidence interval [CI] 1.08-4.54, p=0.03) despite equal incidence (7.5% in LDep, 8.1% in HDep). Patients living in HDep neighborhoods were more likely to have progression on CDK4/6i if they had an ERBB2 snv on ctDNA profiling (HR 8.20, 95% CI 1.57 – 42.93, p=0.013). There was no statistically significant difference in PFS between patients in LDep and HDep neighborhoods on CDK4/6i (12.0 vs 15.9 months, p=0.5). Discussion: Patients with MBC living in more impoverished, HDep neighborhoods were equally likely to receive CDK4/6i and had similar PFS to patients on therapy compared to patients living in LDep neighborhoods. These data are reassuring that patients with MBC were equally likely to receive standard-of-care therapy with CDK4/6i therapy regardless of neighborhood deprivation. This finding differs from our previous work, which identified disparities in PI3K inhibitor use, and suggests more equitable use of a therapy that does not require identification of a molecular target. Patients in LDep neighborhoods were more likely to have disease progression on CDK4/6i if they had an ESR1 mutation. This finding is of potential clinical interest and will be the focus of future research, including assessing choice of endocrine therapy and CDK4/6i by ADI.
Presentation numberPS2-10-12
Perioperative minimal residual disease monitering using a tumor-informed ctDNA assay in stage II-III breast cancer
Han-Byoel Lee, Seoul National University College of Medicine, Seoul, Korea, Republic of
J. AHN1, J. Jung2, Y. Cha1, S. Heo1, E. Kang3, W. Chung4, H. Kim1, D. Bang5, S. Song1, H. Lee3, W. Han3, T. Kim1; 1IMBDx, IMBDx, Seoul, KOREA, REPUBLIC OF, 2Department of Surgery, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu, KOREA, REPUBLIC OF, 3Department of Surgery, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 4DCGen, DCGen, Seoul, KOREA, REPUBLIC OF, 5Department of Chemistry, Yonsei University, Seoul, KOREA, REPUBLIC OF.
Background Tracking minimal residual disease (MRD) via circulating tumor DNA (ctDNA) is associated with treatment efficacy and recurrence. This study presents perioperative tumor-informed ctDNA monitoring in stage II-III breast cancer (BC).Patients and Methods Patients with stage II-III BC were prospectively enrolled at Seoul National University Hospital (Feb 2023-Jul 2024). In the neoadjuvant chemotherapy (NAC) arm, whole blood (WB) was collected at seven perioperative points: baseline (N00), post-cycle 1 (N01), post-NAC (N03), 1-2 months (P00), 6 months (P01), 12, 24, and 36 months post-surgery (P02, P04, P06). In the adjuvant chemotherapy (AC) arm, WB was collected at P00, P01 or post-AC (P99), P02, P04, P06, and 48 months (P08). Plasma was analyzed using CancerDetect™ (IMBdx), a personalized tumor-informed MRD assay. Samples with ≥2 mutations were ctDNA+.Results Among 231 patients, 138 provided evaluable samples (130 in NAC). Median age was 51.5 (range 25-83). Subtypes included HER2+ (41.3%), TNBC (34.1%), HR+/HER2- (24.6%), with 59.4% stage II and 40.6% stage III. The median number of targeted variants was 56 (range 3-100), significantly higher in TNBC and HER2+ than HR+/HER2- (p<0.001).In NAC patients, ctDNA positivity was 80.6% (104/129) at N00, 36.5% (46/126) at N01, 20.2% (24/119) at N03, 12.5% (16/128) at P00/P01, and 8.7% (6/69) at P02. In AC-only patients, positivity was 25% (1/4) at P00, 12.5% (1/8) at P01/P99, 25% (2/8) at P02, and 50% (1/2) at P04.Multivariable logistic regression for baseline (N00) ctDNA+ showed associations with subtype (p=0.015), T stage (p=0.025), and histologic grade (p=0.040). ctDNA positivity at N00 was 90.7% in TNBC, 80.7% in HER2+, and 64.3% in HR+/HER2-. Median ctDNA concentration at N00 (PPM) was 5,660.5 in TNBC (range 21.5-270,615.4), 442.9 in HER2+ (3.5-40,818.3), and 184 in HR+/HER2- (8.8-84,892.3), with TNBC associated with significantly higher levels (p=0.015).The pCR rate in NAC arm was 45.0% (58/129). ctDNA positivity after NAC (N01) was 8.0% (4/50) in pCR patients vs 29.4% (20/68) in non-pCR (p=0.003). At the median follow-up of 523 days (range 255-876), five recurrences occurred. All 3 recurrences in the NAC group were ctDNA+ at N03 and non-pCR. Recurrence was significantly associated with ctDNA positivity at N01 (p=0.014) and N03 (p=0.024). ctDNA concentration at P00 (PPM) also significantly predicted recurrence (p=0.001).Conclusions Perioperative tumor-informed ctDNA monitoring reflects treatment response and risk of recurrence in stage II-III breast cancer. Both binary ctDNA status and quantitative concentration (PPM) demonstrate prognostic value at key clinical time points.
Presentation numberPS2-10-13
Impact of infusion Time-of-Day of Administration (ToDA) of neoadjuvant immunochemotherapy on pathological response in patients with early Triple-negative Breast Cancer treated with KEYNOTE-522 regimen (PEMCLOCK study)
Clémentine Bouchez, Hôpital Saint Louis APHP, Faculté de médecine Paris Cité, Paris, France
C. Bouchez1, D. Loirat2, B. Verret3, A. De Nonneville4, P. Heudel5, S. Bécourt6, L. Someil7, M. Benderra8, A. Hardy-Bessard9, J. Wasserman10, P. Charveriat11, F. Giugliano12, L. Cabel2, Q. Aussedat13, F. Levi14, K. Desseaux15, S. Revillon16, S. Brand17, A. Ballesta16, S. Giacchetti18; 1Breast Disease Unit, Hôpital Saint Louis APHP, Faculté de médecine Paris Cité, Paris, FRANCE, 2Department of Medical Oncology, Institut Curie, Paris, FRANCE, 3Breast Unit, Medical Oncology Department, Institut Gustave Roussy, Villejuif, FRANCE, 4Department of Medical Oncology, Institut Paoli-Calmettes, Université Aix-Marseille , CNRS, INSERM, Marseille, FRANCE, 5Department of Medical Oncology, Centre Léon Bérard, Cancer Research Center of Lyon (UMR Inserm 1052 – CNRS 5286), Lyon, FRANCE, 6Department of Medical Oncology, Centre Oscar Lambret, Lille, FRANCE, 7Breast Disease Unit, Hôpital Saint Louis APHP, Paris, FRANCE, 8Department of Medical Oncology and Cellular Therapy, Hôpital Tenon APHP, Faculté de médecine Sorbonne Université, Paris, FRANCE, 9Department of Medical oncology, Centre CARIO – HPCA, Plérin, FRANCE, 10Department of Medical Oncology, Hôpital de la Pitié-Salpêtrière APHP, Faculté de médecine Sorbonne Université, Paris, FRANCE, 11Department of Medical oncology, Centre hospitalier de Chartres, Le Coudray, FRANCE, 12Breast Unit, Medical Oncology Department, Unit 981, Molecular Predictors and New Targets In Oncology, Gustave Roussy Cancer Campus, Paris, FRANCE, 13Department of medical oncology, Institut Paoli-Calmettes, Marseille, FRANCE, 14UPR “Chronotherapy, Cancers and Transplantation”, Faculté de médecine Paris Saclay, Villejuif, FRANCE, 15Department of Biostatistics, Hôpital Saint Louis APHP, Paris, FRANCE, 16INSERM UMR-S 900, Institut Curie, MINES ParisTech CBIO, PSL Research University, Saint Cloud, FRANCE, 17Department of Biostatistics, Hôpital Saint-Louis, Université Paris Cité, Paris, FRANCE, 18Breast Disease Unit, UPR “Chronotherapy, Cancers and Transplantation”, Hôpital Saint Louis APHP, Université Paris Saclay, Paris, FRANCE.
Background: Neoadjuvant chemotherapy (NAC) combined with pembrolizumab according to KEYNOTE-522 study is the standard treatment for patients (pts) with stage II or III triple-negative breast cancer (TNBC). Circadian rhythms regulate both immune cells function and trafficking, and cancer treatment responses over the 24-hours. Evidence from 31 retrospective studies and one randomized trial supports the benefit of early time-of-day administration (ToDA) of immunotherapy or immunochemotherapy across 10 different cancer types. However, no such data exist for breast cancer patients, nor regarding sex-based differences.Objective: PEMCLOCK evaluates the clinical relevance of ToDA of neoadjuvant pembrolizumab combined with paclitaxel carboplatin for 4 cycles, then by anthracycline cyclophosphamide for 4 cycles for histological tumor response, treatment toxicity, and survival outcomes in pts with high-risk early-stage TNBC. Methods: This French multicenter retrospective study enrolled pts with stage II-III TNBC at 9 cancer centers from 5/2021 to 12/2023. Patients received at least one neoadjuvant pembrolizumab infusion with documented start ToDA. Median ToDA of neoadjuvant pembrolizumab was computed for each patient. The primary endpoint was pathological complete response (pCR) according to the Residual Cancer Burden classification (RCB, 0 to III). Secondary endpoints included treatment-related toxicities and event-free survival. Patients were categorized into early or late ToDA groups based on the overall median of individual median ToDAs across all pembrolizumab infusions. Results: The study included 614 pts with a median follow up of 25 months [95 CI, 24 to 26]. Median age was 49.5 years [IQR: 41.1-58]. Among them, 57% were premenopausal, and 16% carried a constitutional BRCA mutation (12% BRCA1, 4% BRCA2). The median body mass index (BMI) was 25.15 [IQR: 21.8-29.3]. Tumor stages included T1c (8 %), T2 (66 %), and T3-T4 (25 %), with 82% of tumors classified as grade III. Clinically positive axillary nodes were observed in 52 % of pts, with pathological confirmation in 218 cases. Patients received a median number of 8 immunochemotherapy cycles [IQR: 6-8]. The overall median ToDA for pembrolizumab was 12:31 [IQR: 11:34-13:40], which served as the cut-off to define early (n=355) and late (n=252) ToDA groups. Interpatient variability in pembrolizumab ToDA was 4:36 hours [IQR: 3:14-5:44]. Baseline characteristics were balanced between the early and late ToDA groups. A total of 410 patients (67%) achieved pCR (RCB = 0), including 63 % of the patients in the early ToDA group and 74 % in the late ToDA group. There are currently 52 and 35 events for Event-Free Survival and Overall Survival, respectively, thus requiring additional follow up for assessing relevance of ToDA for survival outcomes on the whole study sample. Conclusions: PEMCLOCK is the first real-world large multicenter study which explores the concept of immunochemotherapy ToDA in pts with high-risk early-stage TNBC. Ongoing analyses on treatment-related response, toxicity and survival outcomes along with innovative mathematical modeling approaches will provide a comprehensive understanding to the relations between ToDA and immunochemotherapy efficacy in patients with TNBC.
Presentation numberPS2-10-14
Blood-based metabolomic profiling and transcriptomic analysis of circulating cd3⁺ t cells reveal associations with treatment response and immune activation in early breast cancer (ebc)
Benedetta Conte, University of Eastern Piedmont, UPO, Novara, Novara, Italy
V. Martini1, B. Conte2, L. Negrini2, M. Manfredi2, B. Elettra2, S. D’Alfonso3, M. Mellai3, D. Corà2, F. Favero2, T. Landolfo2, I. Taglialatela1, B. Ruffilli1, S. Nardin1, V. Rossi1, F. D’Avanzo1, S. Gobbato2, M. Bitrus2, A. Turky2, C. Branni2, J. Gennari2, A. Gennari1; 1Department of Translational Medicine – DIMET, Azienda Ospedaliero Universitaria, Maggiore della Carità, Novara, Novara, ITALY, 2Department of Translational Medicine – DIMET, University of Eastern Piedmont, UPO, Novara, Novara, ITALY, 3Department of Health Sciences, University of Eastern Piedmont, UPO, Novara, Novara, ITALY.
Title: Blood-Based Metabolomic Profiling and Transcriptomic Analysis of Circulating CD3⁺ T CellsReveal Associations with Treatment Response and Immune Activation in Early Breast Cancer (eBC)Veronica Martini1, Benedetta Conte2, Letizia Negrini2, Marcello Manfredi2, Elettra Barberis2 Sandra D’Alfonso3, MartaMellai3, Davide Cora’2, Francesco Favero2, Simone Gobbato2, Tommaso Lupo Landolfo2, Matthew Bitrus2, IdaTaglialatela1, Beatrice Ruffilli2, Simone Nardin2, Valentina Rossi1, Francesca D’Avanzo1, Carmen Branni2, Jacopo Gennari2,Alessandra Gennari1,21. Division of Oncology, University Maggiore Hospital della Carità, Novara, Italy2. Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy3. Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.Background:Pathological complete response (pCR) is a strong predictor of long-term outcomes in early breastcancer (eBC), establishing neoadjuvant chemotherapy (NACT) as the standard of care for aggressiveBC subtypes. Recently, targeting host and tumor metabolism has emerged as a promising approachto boost antitumor immunity and improve NACT efficacy. However, the specific metabolicbiomarkers involved, and their immunological effects remain poorly defined.Methods:We performed untargeted metabolomics on baseline plasma from 84 eBC patients undergoing NACTat our institution between 2021–2023 (22% luminal, 36% HER2+, 42% triple-negative; 48% achievedpCR). Data were analysed using univariate (t-test), multivariate (PLS-DA), and pathway enrichmentapproaches. In a subset of 49 patients (58%), bulk RNA was extracted from sorted CD3+ circulatingT cells for gene expression analysis. Gene Ontology enrichment analysis was used to characterizeupregulated pathways in T cells.Results:Pathway enrichment analysis of metabolomic data identified alpha-linolenic acid (ALA, omega-3)and linoleic acid (LA, omega-6) metabolism as top pathways associated with pCR and residualdisease, respectively. Specifically, the VIP score analysis showed that docosahexaenoic acid (DHA, along-chain omega-3 derived from ALA) and dihomo-gamma-linolenic acid (DGLA, a downstreamomega-6 derived from LA) has the strongest association with pCR and RD respectively. Additionaltop ranked metabolites at VIP analysis included palmitoleic acid (POA, an omega-7 lipokine withimmunomodulatory role) and conjugated linoleic acid (CLA, an omega-6 known to counteract theinflammatory effects of classical non-conjugated omega-6). We developed an “Omega Signature”tracking the balance between anti-inflammatory omega-3/6/7 and pro-inflammatory omega-6 asPOA + [(DHA + CLA)/DGLA]. This signature was significantly associated with pCR in a multivariatelogistic regression model adjusted for age and BMI (OR 1.53, 95% CI 1.33–1.88, p < 0.0001). To assesswhether the Omega Signature reflects T cells polarization, we performed transcriptomic profiling ofcirculating T lymphocytes in 49/84 patients. Patients with high Omega Signature (above the median)showed upregulation of gene pathways involved in T cell activation and adaptive immune responsescompared to those with low scores.Conclusion:Our findings reveal that a specific circulating fatty acid profile seems to be associated with treatmentresponse and immune activation in eBC. This signature reflects a favorable balance ofimmunomodulatory lipids and may serve as a non-invasive biomarker to identify patients more likely to achieve pCR. Correlation analyses with tissue metabolomics and gene expression data are ongoingwill be presented at the Congress.
Presentation numberPS2-10-15
Prospective evaluation of a mRNA ERBB2 score as a tool to guide HER2 status and accelerate neoadjuvant decision-making in ER-negative breast cancer
Marta Gonzalez-Rodriguez, Hospital Clínic Barcelona; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
M. Gonzalez-Rodriguez1, E. Sanfeliu2, M. Alva3, T. Pascual4, S. Pernas5, A. Waks6, M. Vidal4, P. Tarantino6, E. Ciruelos3, S. Tolaney6, M. Marín-Aguilera7, L. Paré7, W. Buckingham8, F. Brasó-Maristany9, A. Prat4; 1Medical Oncology, Hospital Clínic Barcelona; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN, 2Pathology Department, Hospital Clínic Barcelona, Barcelona, SPAIN, 3Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 4Medical Oncology, Hospital Clínic Barcelona, Barcelona, SPAIN, 5Medical Oncology, Catalan Institute of Oncology (ICO)-L’Hospitalet de Llobregat, Barcelona, SPAIN, 6Medical Oncology, Dana-Farber/Harvard Cancer Center, Boston, MA, 7Scientific department, Reveal Genomics, Barcelona, SPAIN, 8Market access, Reveal Genomics, Barcelona, SPAIN, 9Medical Oncology, Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN.
Background: In early-stage estrogen receptor (ER) negative breast cancer (BC), whether HER2-positive (HER2+) or triple negative (TNBC), neoadjuvant therapy is frequently employed, making diagnostic turnaround time a critical factor in treatment planning. The ERBB2 mRNA score, developed and validated to predict HER2 status according to ASCO/CAP criteria, has demonstrated high accuracy (AUC=0.97; EBioMedicine 2022). HER2DX and TNBCDX assays, both of which incorporate this score, are performed using the same RNA-based platform, enabling rapid and integrated molecular profiling. However, whether this transcriptomic approach can replace conventional HER2 immunohistochemistry/ in situ hybridization (IHC/ISH) testing in ER-negative tumors has not yet been prospectively evaluated. Methods: In this prospective pilot study, consecutive patients with ER-negative early breast cancer (ER<10% by IHC) at the Breast Cancer Unit at the Clínic Barcelona Comprehensive Cancer Center were enrolled. RNA was extracted from formalin-fixed paraffin-embedded (FFPE) tumor tissue and analyzed using Reveal Genomics’ RNA platform. Based on ERBB2 mRNA expression levels, tumors classified as ERBB2-low received the TNBCDX Research Use Only (RUO) report, whereas those with medium or high ERBB2 levels received the HER2DX report. All cases underwent parallel HER2 IHC and ISH testing according to ASCO/CAP guidelines. The primary endpoint was the concordance between the HER2DX ERBB2 mRNA score and final HER2 status by IHC/ISH. Additionally, we explored the ERBB2 score in 257 ER-negative tumors (93 HER2+ and 164 TNBC) from 4 cohorts (i.e, BIONHER, DAPHNe, ATEMPT and a combined TNBC Spanish cohort from Clínic Barcelona Comprehensive Cancer Center, and Hospital Universitario 12 de Octubre). Results: As of July 1st 2025, 30 patients with newly diagnosed ER-negative BC (8 HER2+ and 22 TNBC) have been enrolled. Concordance between the HER2DX ERBB2 mRNA score and HER2 IHC/ISH status was 100%. Eight cases (26.6%) were classified as ERBB2 medium/high and received the HER2DX report, while 22 cases (73.4%) were ERBB2 low and received the TNBCDX report. The proportion of HER2+ tumors (i.e., IHC 3+ or IHC 2+ with ISH amplification) was consistent with the ERBB2 medium/high group (26.6%), whereas the ERBB2-low group was TNBC (i.e., IHC 0, 1+, 2+ without ISH amplification). Four patients had IHC 2+ tumors and underwent reflex ISH testing. Two showed no ISH amplification, consistent with the ERBB2-low classification, while the other two had confirmed amplification, aligning with the ERBB2-medium/high group. Finally, in the retrospective combined ER-negative cohort of 257 samples, the ERBB2 score accurately predicted HER2 IHC/ISH status, with 92.6% concordance and an AUC of 0.98. Among the ERBB2 medium/high group, 17 patients (15.7%) were classified as HER2-negative; whereas only 2 out of 149 (1.3%) in the ERBB2 low group were categorized as positive. Conclusions: This ongoing pilot study demonstrates the feasibility of using an upfront ERBB2 mRNA-based score to guide selection of HER2DX or TNBCDX testing in patients with newly diagnosed ER-negative breast cancer. Preliminary results show perfect concordance with standard HER2 IHC/ISH testing and support the potential of this approach as a practical and timely tool in the neoadjuvant setting. An updated analysis will be presented at the conference.
Presentation numberPS2-10-16
Distinct Immune Landscapes in Inflammatory and Metaplastic Breast Cancer: Insights from Transcriptomic Profiling
Elyse R Lopez, The University of Texas MD Anderson Cancer Center, Houston, TX
E. R. Lopez1, A. Nasrazadani2, O. Baranov3, V. Smirnov3, A. Love3, F. Paradiso3, M. Hensley3, R. Layman2, J. Mouabbi2, C. Yam2, B. Nelson2, S. Sadia4, V. Valero4, M. Stauder4, W. Woodward4, A. Lucci4, S. Sun4, G. Whitman2, M. Patel4, H. Le-Petross4, Y. Lu4, A. Marx4, A. Alexander4, C. Yajima4, M. Kai4, L. Villarreal4, H. Lopez4, S. H. Giordano2, J. K. Litton2, L. Bora2; 1Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 3BostonGene, BostonGene Corporation, Waltham, MA, 4MDACC Inflammatory Breast Cancer (MDACC-IBC) Team, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Inflammatory breast cancer (IBC) and metaplastic breast cancer (MpBC) are rare subtypes associated with poor prognosis and limited therapeutic targets. While both subtypes exhibit resistance to immune checkpoint inhibitors (ICIs), the mechanisms of immune evasion and immune cell exclusion are not fully understood. To address this, we performed a comparative immune transcriptomic analysis of IBC and MpBC tumors using a large institutional dataset, aiming to define distinct features. Methods: A total of 341 breast cancer tumors were evaluated using BostonGene’s comprehensive AI-driven transcriptomic analysis platform. The cohort included: IBC (n=60), MpBC (n=4), invasive lobular carcinoma (ILC) (n=84), and invasive ductal carcinoma (IDC) controls (n=193). Gene expression profiling was conducted using a panel of 504 cancer;immune-related genes, including key immune checkpoint regulators (PD-L1/CD274), epithelial-mesenchymal transition (EMT) markers (VIM, SNAI2), and antibody-drug conjugate (ADC) targets such as TROP2 (TACSTD2). Immune findings are derived from the RNA-seq data provided by BostonGene but were not categorized within a predefined immune-related gene set. Pathway-level difference analysis using Gene Set Expression Analysis and PAM50 intrinsic subtyping were also conducted. Due to the small number of MpBC cases, results were interpreted in the context of biological trends rather than statistical significance. Results: Transcriptomic profiling revealed differences in immune/stromal features between IBC and MpBC. IBC demonstrated upregulation of JAK/STAT and NF-κB signaling pathways, consistent with an inflamed tumor milieu. Our analysis showed no significant difference in expression of PD-L1 (CD274) between IBC and IDC. PD-L1 expression was low among MpBC tumors, although not statistically significant. In contrast, MpBC tumors displayed a enrichment in EMT signatures, including high expression of VIM. This phenotype is consistent with an immune-cold TIME, where T cell infiltration is minimal and suppressed checkpoint molecule expression. Expression of TROP2 (TACSTD2) was elevated in both IBC and MpBC, though MpBC samples exhibited greater heterogeneity. PAM50 subtyping revealed that IBC and MpBC were predominantly basal-like, with ILC cases largely luminal and IDC displaying mixed profiles. Chi-squared analysis showed no significant difference between IBC and IDC subtypes. Conclusions: IBC and MpBC exhibit distinct but clinically significant immune landscapes. IBC shows features of an inflamed but immunosuppressed microenvironment, characterized by active cytokine signaling and checkpoint expression. MpBC expresses higher level of EMT signaling and stroma related genes. These findings suggest IBC may respond to ICIs, particularly when PD-L1 expression or T cell infiltration is evident, while MpBC may require combination strategies that overcome EMT-mediated immune suppression and stromal exclusion. Additionally, the heterogeneous TROP2 expression may predict response to TROP2-targeted ADCs, such as sacituzumab govitecan. While future validation is needed, our data highlight the importance of immune phenotyping in rare breast cancer subtypes to guide therapeutic strategies and trial design. Acknowledgement: The transcriptomic analyses were performed via collaboration with BostonGene and Morgan Welch IBC Research and Clinic Program (MW IBC program). MW IBC program is funded by the State of Texas Rare and Aggressive Breast Cancer Grant.
Presentation numberPS2-10-17
Potential of MEK Inhibition to overcome Trastuzumab deruxtecan Resistance in MAPK-Activated Triple-Negative Breast Cancer PDX Models
Adrian Gonzalez Gonzalez, Washington University in St Louis, St Louis, MO
A. Gonzalez Gonzalez1, Z. Guo1, T. D. Lorentzen2, J. Luo3, L. Zhao2, X. Shan1, M. Highkin1, J. Hoog1, S. Davies4, L. Ding1, S. Li1, A. Vindigni1, R. Bose1, A. Paulovich5, C. X. Ma1; 1Oncology, Washington University in St Louis, St Louis, MO, 2Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, 3Surgery – Public Health Sciences (PHS), Washington University in St Louis, St Louis, MO, 4Surgical Oncology, Washington University in St Louis, St Louis, MO, 5Translational Science and Therapeutics Division, Fred Hutchison Cancer Center, Seattle, WA.
Background:Since its approval in 2020 by the FDA, the antibody-drug conjugate (ADC) Trastuzumab deruxtecan (T-DXd), has demonstrated its efficacy not only targeting HER2+ tumors, but HER2-low breast cancers, including triple negative breast cancer (TNBC) subtype. However, mechanisms of primary and acquired resistance to T-DXd in TNBC remain poorly understood. To identify predictive biomarkers and rational therapeutic strategies to overcome resistance, we performed proteomic profiling and functional validation studies in a panel of TNBC patient-derived xenograft (PDX) models and TNBC cell lines.Methods:We conducted LC-MRM (liquid chromatography–multiple reaction monitoring) mass spectrometry analysis of tumor lysates from 19 TNBC patient-derived xenograft (PDX) models previously evaluated for T-DXd response. The targeted proteomic panel included 91 peptides representing 63 proteins spanning HER family receptors, DNA damage response (DDR) components, and key signaling effectors. Correlative analysis between protein expression and tumor growth inhibition (%TGI) revealed a significant negative association between T-DXd efficacy and MAPK pathway activation, including elevated levels of MAPK3, pMAPK3_pT202, pMAPK3_pY204. Conversely, positive correlations were observed with proteins such as HER2, pATR_pT1989, PMS1, LIG1, and TUBB. These findings implicated hyperactive MAPK signaling as a potential driver of T-DXd resistance in TNBC, independent of HER2 expression level.Results:To verify our hypothesis, we evaluated the therapeutic potential of combining T-DXd with the MEK inhibitor selumetinib in a panel of TNBC cell lines (sensitive and resistant) and PDX derived organoids. Our preliminary data indicates a synergistic anti-tumor effect between T-DXd and the MEK inhibitor. We further performed in vivo studies using two T-DXd-resistant TNBC PDX models, WHIM3 and WHIM4, both exhibiting high MAPK pathway activation. The combination of T-DXd and the MEK inhibitor selumetinib demonstrated significantly greater antitumor activity than either agent alone, effectively overcoming resistance in both models. Additional T-DXd-resistant TNBC PDX models and cell lines are currently being evaluated for MAPK pathway activation and response to MEK inhibition. Short-term biomarker studies are underway to investigate the mechanisms of action of the combination therapy, and these results will be presented.Conclusions:Proteomic profiling implicates MAPK pathway activation as a key mechanism of T-DXd resistance in TNBC. MEK inhibition with selumetinib restored T-DXd sensitivity in resistant PDX models, supporting MAPK blockade as a rational strategy to overcome ADC resistance. These findings provide preclinical justification for evaluating T-DXd and selumetinib in HER2-low TNBC. Given an ongoing clinical protocol (NCI 10733) evaluating this combination in pancreatic cancer, these results may facilitate translational efforts in metastatic TNBC.
Presentation numberPS2-10-18
Epithelial-mesenchymal transition (EMT) Signature Score of a Bulk Tumor May Not Reflect the EMT of Malignant Epithelial Cells
Yoshihisa Tokumaru, Central Japan International Medical Center, Minokamo, Japan
Y. Tokumaru1, R. Wu2, M. Oshi3, S. Ando1, M. Takeuchi1, T. Ishikawa2, M. Nobuhisa4, M. Futamura5, K. Takabe3; 1Breast Surgery, Central Japan International Medical Center, Minokamo, JAPAN, 2Breast Surgery, Tokyo Medical University, Tokyo, JAPAN, 3Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 4Gastroenterological Surgery and Pediatric Surgery, Gifu University, Gifu, JAPAN, 5Breast Surgery, Gifu University Hospital, Gifu, JAPAN.
Background: Epithelial-mesenchymal transition (EMT) is a process in which polarized epithelial cells acquire mesenchymal features, contributing to tumor progression, metastasis, and treatment resistance. Our group has been showing the clinical relevance of pathway activity scores generated by Gene Set Variation Analysis (GSVA) of Hallmark gene sets in breast cancer. Here, we hypothesized that breast cancer with high EMT score is associated with aggressive features and poorer outcomes compared to EMT-low tumors. Material and Methods: We analyzed three independent large breast cancer cohorts: TCGA (n = 1,077), METABRIC (n = 1,904), and SCAN-B (n = 3,069). Survival outcomes (OS, DSS, DFS) were assessed. Tumor immune microenvironment (TIME) was analyzed using xCell and Thorsson et al.’s immune metrics. Chemosensitivity was examined using four independent datasets (GSE21094, GSE22358, GSE25066, GSE32646). Mutation burden and neoantigen load were also evaluated. To explore EMT signatures across cell types, single-sample GSEA was applied to single-cell RNA-seq data. Results: EMT score was generated by Gene Set Variation Analysis of Hallmark EMT gene set and each cohort was divided into high and low score groups by the median as the cutoff. Unexpectedly, high EMT tumors were associated with significantly lower Nottingham histological grade, and 4 cell proliferation-related Hallmark gene sets; E2F Targets, G2M Checkpoint, MYC Targets V1, and MYC Targets V2, were enriched to low EMT tumors consistently in all three cohorts, TCGA, METABRIC, and SCAN-B (all FDR < 0.01). Notably, EMT-high tumors did not exhibit features of a tumor-permissive immune microenvironment, as assessed by xCell. Surprisingly, high EMT scores were not associated with lymph node (N) or distant (M) metastasis in any of the three cohorts. Indeed, high EMT tumors were not associated with any of the survival parameters in TCGA and METABRIC, nor SCAN-B. To investigate this unexpected lack of prognostic significance of EMT score, we assessed the relationship of EMT score and chemosensitivity in four independent cohorts but observed no significant associations. Similarly, no association was found between EMT score and mutation load nor neoantigen burden. xCell analysis revealed that EMT-high tumors exhibited significantly higher infiltration of stromal cells, including adipocytes, endothelial cells, pericytes, and fibroblasts (all p < 0.001), in all three cohorts. With ssGSEA of single-cell RNA sequencing cohort, cancer-associated fibroblasts (CAFs) was found to express the highest EMT scores compared to the other cell populations (e.g., cancer cells, T cells, B cells, and myeloid cells). These findings suggest that EMT signatures derived from bulk tumor transcriptomes likely reflect the stromal compartment, particularly CAFs, rather than intrinsic properties of malignant epithelial cells. Conclusion: Our findings suggest that EMT signal from transcriptome of a bulk tumor largely represent stromal components, particularly CAFs, rather than intrinsic EMT activity in malignant cells. This highlights the need for analyses of single-cell resolution to accurately interpret EMT biology in the tumor microenvironment.
Presentation numberPS2-10-19
Circulating tumor DNA (ctDNA) as a strong prognostic biomarker of minimal residual disease (MRD) using a tissue-free, epigenomic assay in early-stage breast cancer
Wolfgang Janni, University Hospital Ulm, Ulm, Germany
W. Janni1, B. Rack1, P. A. Fasching2, A. Hartkopf3, H. Tesch4, R. Lorenz5, G. Heinrich6, J. Blohmer7, T. Fehm8, V. Müller9, A. Schneeweiss10, M. W. Beckmann2, M. Rübner2, N. Harbeck11, K. Pantel12, D. Dustin13, M. Cai13, T. W. Friedl1; 1Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, GERMANY, 2Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, GERMANY, 3Department of Gynecology and Obstetrics, Tübingen University Hospital, Tübingen, GERMANY, 4Onkologische Gemeinschaftspraxis, Onkologische Gemeinschaftspraxis, Frankfurt, GERMANY, 5Schwerpunktpraxis Onkologie, Frauenärztliche Gemeinschaftspraxis Casparistr., Braunschweig, GERMANY, 6Schwerpunktpraxis für Gynäkologische Onkologie, Department of Gynecologic Oncology, Fürstenwalde, GERMANY, 7Department of Gynecology and Breast Center, Charité University Hospital Campus, Berlin, GERMANY, 8Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, GERMANY, 9Department of Gynecology and Obstetrics, University Hospital Hamburg-Eppendorf, Hamburg, GERMANY, 10National Center for Tumor Diseases, University Hospital and German Cancer Research Center, Heidelberg, GERMANY, 11Breast Center, Department of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, München, GERMANY, 12Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, GERMANY, 13Global Scientific Affairs, Guardant Health, Palo Alto, CA.
Background: ctDNA may serve as an early biomarker of recurrence and could facilitate identification of patients with asymptomatic distant metastasis who might benefit from early treatment intervention. Here, we utilized a tissue-free, epigenomic assay for the detection of ctDNA in patients with early-stage breast cancer. Patients and Methods: Plasma samples were analyzed in 359 patients with stage I-III breast cancer who were enrolled in the SUCCESS-A phase 3 clinical trial (NCT02181101). All plasma samples were collected approximately two years after completion of adjuvant chemotherapy from patients without evidence of prior disease recurrence. In this updated analysis of a previously presented study, samples were re-analyzed using the Guardant Reveal assay featuring an updated bioinformatics algorithm for ctDNA detection. This algorithm was independently trained and validated in breast cancer samples. The primary objective of the ctDNA analysis was to predict distant metastatic recurrence from a single post-treatment time point in patients with early-stage breast cancer. Results: Out of 359 samples tested, 313 (87.2%) were evaluable; 46 samples failed quality control (QC) due to low plasma cfDNA levels and low ctDNA enrichment. The ctDNA positivity rate was 5.8% (18/313), and ctDNA positivity was strongly associated with worse outcomes, including a significantly shorter distant recurrence-free interval (HR 33.3, 95% CI 4.12-268, p<0.0001) and poorer overall survival (HR 27.3, 95% CI 1.15-647, p<0.0001). The sensitivity for distant recurrence in patients who had a sample collected within one year of recurrence was 73% (11/15). Of all ctDNA-positive samples, 94% (17/18) were from patients who subsequently developed a distant recurrence, with ctDNA detected at a median interval of 7.9 months prior to recurrence. The negative predictive value for distant recurrence was 93% (274/295), and the specificity was 99.6% (267/268). Conclusion: Detection of ctDNA approximately two years after adjuvant chemotherapy was highly prognostic in this early-stage breast cancer cohort. These data support the conduct of treatment intervention trials for patients with molecular relapse and negative staging for distant metastases to demonstrate clinical utility.
Presentation numberPS2-10-20
Dual biomarker approach for minimal residual disease detection in early breast cancer using high-volume blood samples
Iñaki Comino-Méndez, Institute of biomedical research of Malaga (IBIMA), Malaga, Spain
I. Comino-Méndez, J. Pascual, J. Velasco-Suelto, M. Quirós-Ortega, A. Godoy-Ortiz, M. Álvarez, E. Alba; Medical Oncology, Institute of biomedical research of Malaga (IBIMA), Malaga, SPAIN.
Background: Early detection of minimal residual disease (MRD) through liquid biopsy may enable timely identification of relapse in early-stage breast cancer (BC), yet most strategies rely on single-analyte approaches. Circulating tumour DNA (ctDNA) and circulating tumour cells (CTCs) reflect distinct aspects of tumour biology, but their combined utility for MRD detection remains poorly characterised. Patients and methods: We conducted a prospective study of 58 early BC patients treated with either neoadjuvant chemotherapy or primary surgery followed by adjuvant therapy. Patient-specific droplet digital PCR assays targeting tumour-derived mutations were used to analyse ctDNA and CTCs in serial high-volume blood samples collected at baseline, after surgery, and throughout follow-up. A composite score (L-MRD) integrating six clinical and molecular variables was developed to stratify relapse risk. Results: At baseline, ctDNA and/or CTCs were detectable in 67.2% of patients, with higher detection rates in the neoadjuvant cohort. Combined biomarker positivity persisted in 53.6% of cases post-surgery. MRD detection during follow-up anticipated all clinical relapses, achieving 100% sensitivity and a median lead time up to 34.60 months. The L-MRD score stratified patients into low- and intermediate/high-risk groups with 5-year recurrence rates of 2.2% versus 41.6%, respectively (HR=6.6; P=0.04). Prognostic discrimination remained significant for distant relapse and demonstrated strong predictive performance (AUC=0.891; NPV=97.8%). Conclusions: This dual-biomarker approach enables ultra-sensitive and early MRD detection. Integration of longitudinal ctDNA and CTCs data into a composite score improves risk stratification and may inform personalised surveillance strategies in early BC.
Presentation numberPS2-10-21
Associations Between Circulating Tumor DNA Genomic Profiles and Serum Thymidine Kinase Patterns in Patients with Advanced, Hormone Receptor-Positive, HER2-Negative (HR+/HER-) Breast Cancer Initiating Treatment with a CDK4/6 Inhibitor (CDK4/6i)
Elena M. Michaels, Massachusetts General Hospital, Boston, MA
E. M. Michaels1, N. A. Bagegni2, B. Moy1, R. O. Abelman1, N. Vidula1, L. W. Ellisen1, K. Clifton2, A. A. Davis2, C. X. Ma2, A. S. Dedeoglu1, A. Williams3, M. R. Lloyd4, S. A. Wander1; 1Medical Oncology, Massachusetts General Hospital, Boston, MA, 2Medical Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO, 3Medical Oncology, Biovica Inc, San Diego, CA, 4Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA.
Background: Identifying reliable biomarkers to assess treatment response and improve disease monitoring remains an unmet need in breast cancer (BC). Thymidine kinase, an enzyme for DNA synthesis upregulated during cell proliferation, has emerged as a promising biomarker in advanced HR+/HER2- BC. Higher baseline serum thymidine kinase activity (TKa) correlates with worse prognosis. Serial TKa assessment during the first cycle of a CDK4/6i with endocrine therapy (ET) revealed three response patterns – suppressed, rebound, and unsuppressed. The latter two are associated with inferior outcomes but the relationship between TKa response and tumor genomics is unexplored. This case series evaluates TKa patterns in relation to baseline circulating tumor DNA (ctDNA) profiles in patients with HR+/HER2- metastatic BC (MBC) treated with a CDK4/6i and ET. Methods: We evaluated a group of patients from two cancer centers with HR+/HER2- MBC initiating treatment with a CDK4/6i (ribociclib, abemaciclib, palbociclib) and ET (an aromatase inhibitor/AI or the selective estrogen receptor degrader/SERD, fulvestrant). Triple therapy with the PI3K inhibitor, inavolisib, was permitted for PIK3CA-mutated tumors. A subset of patients are participants in the TK IMPACT study (NCT04968964, Washington University). Baseline ctDNA was assessed within one month of treatment start using Guardant360 or Tempus xF. TKa was measured via the DiviTum® TKa assay (Biovica, USA) at baseline prior to initiation of CDK4/6i, Cycle 1 Day 15 (C1D15), and C2D1. A TKa level of 50 DuA (DiviTum unit of Activity) is the lower limit of quantification of the assay. Suppressed pattern was defined as TKa levels that declined to <50 DuA at C1D15 of treatment and remained <50 at C2D1; rebound was defined as TKa that declined to <50 DuA at C1D15 but increased to >50 at C2D1; unsuppressed remained >50 DuA at C1D15 and C2D1. Progression-free survival (PFS) was assessed. Results: Twenty-two patients were included; 32% with de novo MBC, 68% with a metastatic recurrence. Most patients were on first-line therapy (95%). Ribociclib was the most common CDK4/6i (86%) while 14% received palbociclib. Most patients were treated with an AI (77%); a smaller subset received fulvestrant (23%). Baseline ctDNA revealed several potential ET and CDK4/6i resistance mediators including AKT/PTEN mutations (18%), TP53 (14%), RAS/RAF pathway mutations (14%), ERBB2 (14%), ESR1 (9%), RB1 loss (5%), FGFR1 (5%) and CCNE1 amplification (5%). One patient had undetectable baseline TKa and no identifiable mutations in ctDNA. The following TKa patterns were observed: 7 suppressed, 11 rebound, and 3 unsuppressed. Notable cases included a patient with an unsuppressed TKa pattern while on ribociclib/AI who experienced progression within 3 months. Baseline ctDNA revealed an ERBB2 mutation and ATM, BRCA2, and CHEK2 copy number loss. Another patient with RB1 loss and multiple co-occurring mutations (ESR1, PIK3CA, KRASG12V, ATM and BRCA2 loss) had a reduction in TKa from 242 at baseline to 68 at C2D1 after starting palbociclib, inavolisib, and fulvestrant. Conclusions: TKa is a novel biomarker with important clinical implications. Understanding the genomic drivers underlying treatment response is an area of active investigation. This hypothesis-generating study compares baseline ctDNA profiles with TKa response patterns in patients with HR+/HER2- MBC starting treatment with CDK4/6i-based therapy. Longitudinal TKa monitoring alongside imaging will continue, including evaluation at progression for acquired genomic changes. Updated findings in these and additional patients will be presented at the meeting.
Presentation numberPS2-10-22
Proteomic analysis of residual disease identifies potential prognostic biomarkers of triple-negative breast cancer after neoadjuvant chemotherapy
Xi Chen, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, beijing, China
X. Chen, L. Ji, H. Lv, G. Song, Q. Li, J. Wang, Y. Fan, Y. Luo, B. Lan, S. Chen, R. Cai, F. Ma, B. Xu, P. Zhang; Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, beijing, CHINA.
Background:Triple negative breast cancer(TNBC) patients with residual disease after neoadjuvant chemotherapy have a poor prognosis. However, not all such patients have a bad outcome. Residual cancer burden (RCB) can provide some prognostic information, but there is still a lack of effective molecular markers. This study is aimed at exploring the prognostic biomarkers in TNBC patients with residual disease after neoadjuvant therapy from the perspective of proteomics. Methods:TNBC patients with residual disease after neoadjuvant chemotherapy were divided into a short-term recurrence group (recurrence free survival≤3 years) and a non-recurrence group (recurrence free survival> 3 years), and baseline characteristics between the two groups were matched. Sixty residual tumor samples from patients were selected for whole proteome analysis using Liquid Chromatography-Mass Spectrometry (LC-MS). Differential protein screening, pathway enrichment analysis, protein – protein interaction network analysis, and immune infiltration analysis were performed between the two groups. Finally, the expression of differential proteins was validated at the immunohistochemical(IHC) level. Results:A total of 60 postoperative residual tumor samples were tested, with 30 in the recurrence group and 30 in the non-recurrence group, and baseline characteristics were balanced between the two groups. A total of 49 up-regulated and 237 down-regulated proteins were identified when comparing the recurrence and non-recurrence groups. GO enrichment analysis indicated that the down-regulated proteins were mainly enriched in pathways related to leukocyte-mediated immune response, T cell activation, and mitochondrial metabolism. KEGG enrichment analysis showed that the down-regulated proteins were enriched in the T cell receptor signaling pathway and osteoclast differentiation pathway. Both pathway enrichment analyses revealed that immune-related proteins, especially those in the T cell pathway, were significantly down-regulated in recurrent patients compared to non-recurrent patients. Protein-protein interaction network analysis identified 25 key proteins, several of which are immune-related, including CD1C, CD247, CD2, CD300A, CD3D, CD40, CD68, CD84, GZMB, CD20, and CD150. Survival analysis based on these 25 key proteins showed that down-regulation of CD68 and CD40 most significantly affected patient survival. Immune infiltration analysis revealed down-regulated immune cell-related expression in recurrent patients compared to non-recurrent patients, with decreased expression of CD4+ effector T cells, dendritic cells, macrophages, M2 macrophages, naive B cells and plasmacytoid dendritic cells in the recurrence group. Based on the analysis of key differential proteins and immune infiltration, IHC was used to verify the expression of CD4, CD40, and CD68 in recurrence and non-recurrence groups. Results showed that the recurrence group had significantly lower expression levels. The tumor-infiltrating lymphocytes in the recurrence group were also significantly lower than in the non-recurrence group (median 10% vs. 20%, P<0.001). Conclusion:The study found that in patients with residual disease after neoadjuvant therapy for TNBC, the downregulation of immune-related proteins, particularly in the T cell pathway, is closely associated with short term recurrence. Immunohistochemistry confirmed that CD4, CD40, and CD68 are downregulated in the recurrent group. Our study preliminarily confirms that the downregulation of immune-related proteins is an important prognostic factor for TNBC patients with residual disease, offering valuable insights for future research and clinical practice.
Presentation numberPS2-10-23
Circulating Tumor DNA Dynamics in Patients with Hormone Receptor-Positive Human Epidermal Growth Factor Receptor 2-Negative Early Breast Cancer Assessed Using an Ultrasensitive Tumor-Informed Assay
Carlos H Barcenas, University of Texas MD Anderson Cancer Center, HOUSTON, TX
C. H. Barcenas1, A. Contreras2, A. Alexander1, H. Lopez1, R. Yu3, S. Shete3, Y. Chen4, C. Abbott4, K. Keough4, S. M. Boyle4, R. O. Chen4, D. Tripathy1, V. Valero1; 1Breast Medical Oncology, University of Texas MD Anderson Cancer Center, HOUSTON, TX, 2Pathology, University of Texas MD Anderson Cancer Center, HOUSTON, TX, 3Biostatistics, University of Texas MD Anderson Cancer Center, HOUSTON, TX, 4NA, Personalis, Fremont, CA.
PURPOSE Circulating tumor DNA (ctDNA) in early-stage breast cancer (EBC) is a prognostic biomarker for disease recurrence. ctDNA dynamics in the low shedding EBC subtype, hormone receptor-positive (HR+) human epidermal receptor 2-negative (HER2-) EBC are challenging to track, motivating the use of ultrasensitive ctDNA assays. In this study we used an ultrasensitive tumor-informed ctDNA assay to detect molecular residual disease (MRD) and its association with disease recurrence among patients with HR+/HER2-EBC who were receiving oncological treatment. METHODS Using a whole-genome sequencing tumor-informed assay that tracks up to 1800 mutations we assessed for MRD and ctDNA dynamics among patients with stage II-III HR+/HER2- EBC who provided serial plasma samples at varying intervals while receiving oncological therapy. MRD was defined as a plasma sample that tested positive for ctDNA with this assay. We then examined the association of ctDNA dynamics with disease recurrence. We defined lead time as the time interval between the first plasma sample that tested positive for ctDNA and the clinical detection of disease recurrence. RESULTS Of 28 patients who provided two or more serial plasma samples, 26 (93%) had tumor tissue suitable for DNA sequencing. The median follow-up time was 5.0 years from surgery (range, 2.3-10.8 years). Patient-specific ctDNA assays were designed to test 118 plasma samples (2-9 samples per patient). Eleven patients (42%) had MRD detected at one or more time points, with 58% of detections in the ultrasensitive range (<100 parts per million). Six patients (23%) eventually experienced distant disease recurrence, all of whom had MRD detected prior to standard clinical detection of disease recurrence, with a median lead time of 1.5 years (range, 1.0-2.5 years). All 15 patients who remained persistently ctDNA-negative and 5 patients who experienced ctDNA clearance (from ctDNA-positive to ctDNA-negative) while receiving oncological treatment remained clinically recurrence-free throughout the follow-up of the study. CONCLUSION Using an ultrasensitive tumor-informed ctDNA assay, we found that all patients with HR+/HER2-EBC who eventually experienced distant disease recurrence had MRD detected prior to standard clinical detection, with a median lead time of 1.5 years, and ctDNA dynamics showed a strong correlation with oncological outcomes with 100% of patients that persistently or eventually cleared ctDNA remaining disease free, and 100% of patients that persistently or became ctDNA positive experiencing recurrence, with 100% sensitivity and specificity of this assay for disease recurrence. This assay appears to have the high sensitivity needed for this application and is being tested in large prospective trials.
| Recurrence | Persistently ctDNA+ | Persistently ctDNA-neg | ctDNA+ then Cleared | ctDNA-neg then ctDNA+ | Total | ||||||||||||
| Yes | 3 | 0 | 0 | 3 | 6 | ||||||||||||
| No | 0 | 15 | 5 | 0 | 20 | ||||||||||||
| Total | 3 | 15 | 5 | 3 | 26 |
Presentation numberPS2-10-24
Impact of HER2 status dynamics on fam-trastuzumab deruxtecan-nxki treatment outcomes in metastatic breast cancer: real-world insights
Yuanqing Ye, Eisai Inc., Nutley, NJ
Y. Ye, A. Potdar, A. Apfel, V. Devanarayan, P. Sachdev, Y. Zhang; Clinical Research, DHBL, Eisai Inc., Nutley, NJ.
Background: Metastatic breast cancer (MBC) remains a significant clinical challenge. Fam-trastuzumab deruxtecan-nxki(T-DXd) has demonstrated notable efficacy in clinical trials such as DESTINY-Breast01 and DESTINY-Breast09. T-DXd has shown promising results in treating not only HER2-positive but also HER2-low and HER2-ultralow subtypes. However it is still unclear how HER2 status and its changes over time affect treatment outcomes with T-DXd. Moreover, there is emerging evidence that HER2 status can change during or after T-Dxd treatment. This study uses real-world data (RWD) from a large, clinically annotated cohort to evaluate the impact of HER2 status and its dynamics on treatment outcome with T-DXd as well as T-DXd treatment on HER2 status. Methods: A retrospective analysis was conducted using RWD from 300 MBC patients treated with T-DXd, sourced from the Tempus database1. Key endpoints included real-world best overall response (rwBOR; non-responder vs. responder), real-world progression free survival (rwPFS), real-world time to next treatment (rwTTNT), and real-world overall survival (rwOS). Patient outcomes were curated by Tempus based on the physician assessment. Patients with complete response (CR) or partial response (PR) were classified as responders while those with stable disease (SD) or progressive disease (PD) were classified as non-responders. HER2 status was defined using immunohistochemistry (IHC; 0, 1+, 2+, 3+) and in-situ hybridization (ISH; negative, positive). HER2 low was defined as IHC 1+ or IHC 2+ with a negative ISH, HER2 positive was defined as IHC 2+ with a positive ISH or IHC 3+. HER2 status change was assessed in patients with both pre- and post-T-DXd HER2 status data, with post-treatment IHC conducted at least six weeks after treatment initiation. Patients with multiple pre-T-DXd treatment measurements showing no change in HER2 status over time were considered HER2 IHC Stable. Logistic regression was used for rwBOR, and Cox proportional hazards models with log-rank tests were applied for rwPFS, rwTTNT, and rwOS while adjusting for age at T-DXd treatment, ER/PR status, care plan, sampling time, tissue location. Results: Among 300 patients, 298 subjects had HER2 data, 223 subjects had pre-T-DXd HER2 IHC Stability data. Compared to HER2-positive, HER2-low patients had significantly worse outcomes: rwBOR (OR=1.87, p=0.035), rwPFS (HR=1.60, p=2.32X10-3), rwTTNT (HR=1.75, p=4.30X10-3), and rwOS (HR=2.35, p=2.02X10-4). Patients with stable HER2 IHC status had more favorable outcomes: rwBOR (OR=0.27, p=1.23X10-4), rwPFS (HR=0.58, p=0.0014), rwTTNT (HR=0.61, p=0.025), and rwOS (HR=0.48, p=2.98X10-3). Among 62 out of 67 patients with pre- and post-treatment HER2-status available, all were classified as either HER2-positive or HER2-low before receiving T-DXd treatment. Following treatment, 21 (34%) of these 62 patients exhibited a change in HER2 status with T-DXd treatment. Specifically, among 31 HER2-positive patients, 5 (16%) dropped to HER2-negative and 7 (22%) to HER2-low and among 31 HER2-low patients, 9 (29%) dropped to HER2-negative. A change in HER2 status post-T-DXd treatment was associated with increased risk for progression (rwPFS HR=1.89, p=0.041). However, due to the relatively small sample size of 67, these findings should be interpreted with caution. Conclusions: This study highlights the clinical relevance of monitoring HER2 status and the dynamics of HER2 status and its impact on treatment outcome with a HER2-targeting ADC, T-DXd in MBC. Changes in HER2 status post-treatment with T-DXd were associated with poorer progression-free survival, underscoring the importance of ongoing HER2 monitoring. These real-world insights can inform personalized treatment strategies for MBC. References: 1. www.tempus.com
Presentation numberPS2-10-25
Combined T and B cell signatures as prognostic biomarkers in triple-negative breast cancer (TNBC)
Shipra Gandhi, Winship Cancer Institute of Emory University, Atlanta, GA
S. Gandhi1, S. Kumar Deshmukh2, S. Wu2, J. Xiu2, G. Sledge Jr.2, E. Roussos Torres3, M. B. Lustberg4, M. Opyrchal5, S. Yao6, K. Takabe7, M. Ernstoff8, M. Abdelbary1, A. T Ruffin1, A. C Cole1, H. M. Knochelmann1, K. Kalinsky1, G. B. Lesinski1, C. M. Paulos1; 1Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 2Oncology, Caris Life Sciences, Phoenix, AZ, 3Oncology, University of Sothern California, Los Angeles, CA, 4Oncology, Yale University, New Haven, CT, 5Oncology, Indiana University, Bloomington, IN, 6Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 7Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 8Developmental Therapeutics Program, National Institute of Health, Bethesda, MD.
Background: CD8⁺ T cells infiltration is a known predictor of improved outcomes in patients with triple-negative breast cancer (TNBC). However, the contribution of coordinated T cell and B cell immune interactions within the tumor microenvironment (TME), remains to be fully explored. We posit that dual expression of T cell (IFNy, CD40LG) and B cell (CD19, CD40) markers would associate with patient survival and more favorable immune landscape in TNBC. In this study, we evaluate the prognostic relevance of dual expression of IFNy/CD19 and CD40/CD40LG in TNBC and its association with TME. Methods: 3,662 TNBC samples were analyzed by NGS (592, NextSeq; WES/WTS, NovaSeq; Caris Life Sciences, Phoenix, AZ). Dual expression groups IFNy/CD19-high (n=1275), IFNy/CD19-low (n=1272), IFNy-high/CD19-low (n=558), IFNy-low/CD19-high (n=557), CD40LG/CD40-high (n=1303), CD40LG/CD40-low (n=1303), CD40LG-high/CD40-low (n=529) and CD40LG-low/CD40-high (n=529) were classified by RNA expression above or below the 50th percentile. Immune cells were estimated using WTS deconvolution (Quantiseq). Real-world median overall survival (mOS) was derived from insurance claims and calculated from biopsy to last contact using Kaplan-Meier. Statistical significance was assessed using chi-square and Mann-Whitney U with multiple comparison adjustments (q<0.05). Results: Patients with TNBC with IFNy/CD19-high tumors exhibited longer mOS (28 months (m) vs 16.1 m, HR 0.6, 95% CI 0.55- 0.66, p<0.00001) compared to those with IFNy/CD19-low. Dual high-expression (IFNy/CD19-high) had higher mOS compared to the other subgroups, namely IFNy-high/CD19-low (28 m vs 20 m, HR 0.76, 95% CI 0.67-0.86, p<0.0001), IFNy-low/CD19-high (28 m vs 19.1 m, HR 0.7, 95% CI 0.62-0.79, p<0.00001). A similar pattern was observed for the CD40/CD40LG-high that exhibited longer mOS (27 m vs 17.9 m, HR 0.68, 95% CI 0.61-0.74, p<0.00001) compared to those with CD40/CD40LG-low. Also, dual CD40/CD40LG-high had higher mOS compared to CD40-high/CD40LG-low (27 m vs 17.2 m, HR 0.7, 95% CI 0.62-0.8, p<0.00001), CD40-low/CD40LG-high (27 m vs 19 m, HR 0.7, 95% CI 0.62-0.8, p<0.00001). These dual high-expression tumors were enriched for diverse immune cell populations, including B cells, dendritic cells (DC), M1 MΦ, M2 MΦ, natural killer (NK) cells, CD8⁺ T cells, all q<0.05. Moreover, dual high group had higher levels of immune checkpoint genes CD274, PDCD1, CTLA4, HAVCR2 (TIM3), LAG3, TIGIT and CD47, all q<0.05 (Table), suggesting a highly active yet potentially exhausted immune microenvironment. Conclusions: Coordinated expression of T and B cell markers identifies a subset of TNBC with enhanced immune infiltration and improved survival, underscoring the prognostic significance for T-B cell crosstalk. These findings highlight the potential of combined immune signatures to inform prognosis and guide immunotherapeutic strategies in TNBC.
| IFNy/CD19-high | IFNy/CD19-low | q-value | CD40/CD40LG-high | CD40/CD40LG-low | q-value | ||||||||
| B cell (median %) | 4.92 | 3.25 | <0.01 | 4.9 | 3.19 | <0.01 | |||||||
| DC (median %) | 3.21 | 2.78 | <0.01 | 3.26 | 2.74 | <0.01 | |||||||
| M1 MΦ (median %) | 3.41 | 2.62 | <0.01 | 3.9 | 2.39 | <0.01 | |||||||
| M2 MΦ (median %) | 3.41 | 2.69 | <0.01 | 3.92 | 2.21 | <0.01 | |||||||
| NK cell (median %) | 3.04 | 2.73 | <0.01 | 3.06 | 2.64 | <0.01 | |||||||
| CD8+ T cell (median %) | 1.61 | 0 | <0.01 | 1.59 | 0 | <0.01 | |||||||
| CD274 (Median TPM) | 8.31 | 2.25 | <0.01 | 8.44 | 2.11 | <0.01 | |||||||
| PDCD1 (Median TPM) | 1.25 | 0.21 | <0.01 | 1.19 | 0.21 | <0.01 | |||||||
| CTLA4 (Median TPM) | 4.03 | 0.59 | <0.01 | 4.08 | 0.6 | <0.01 | |||||||
| HAVCR2 (Median TPM) | 27.33 | 11.48 | <0.01 | 32.43 | 9.76 | <0.01 | |||||||
| LAG3 (Median TPM) | 7.71 | 1.81 | <0.01 | 7.86 | 1.91 | <0.01 | |||||||
| TIGIT (Median TPM) | 3.11 | 0.4 | <0.01 | 3.22 | 0.4 | <0.01 | |||||||
| CD47 (Median TPM) | 70.65 | 38.34 | <0.01 | 76.49 | 34.03 | <0.01 |
Presentation numberPS2-10-26
Tumor genomic profiling results in breast cancer patients: A comprehensive analysis from a laboratory research registry.
Gregory A. Vidal, West Cancer Center and Research Institute, Germantown, TN
G. A. Vidal1, S. Benn2, A. Martin1, K. Hayes1, R. Gilmore1, R. Fine1, L. Brzeskiewicz3, T. McKamie3, S. Cummings3; 1Medical Oncology, West Cancer Center and Research Institute, Germantown, TN, 2Medical Oncology, West Cancer Center and Research Institute, Southaven, MS, 3Oncology, Myriad Genetics, Inc., Salt Lake City, UT.
Background: Precision oncology has transformed breast cancer care, with comprehensive genomic profiling (CGP) playing a key role in identifying somatic alterations and biomarkers that guide personalized treatment. Separately, germline testing helps uncover inherited pathogenic variants relevant to therapy and familial risk. Despite the growing use of CGP and germline testing, real-world data on the distribution and clinical relevance of results across diverse breast cancer populations is limited. To better understand the molecular landscape of breast cancer in a real-world setting, we analyzed CGP results from a clinical laboratory research registry, including somatic variant classifications, biomarker profiles, and germline findings. Methods: A retrospective analysis was conducted on 279 breast cancer patients who underwent CGP utilizing a clinical laboratory research registry. Variables assessed included age at diagnosis, sex at birth, histological subtype, ancestry, microsatellite instability (MSI), tumor mutational burden (TMB), PD-L1 expression, tumor variant classifications based on their clinical significance (IA, IB, IIC, IID), and gene-specific alterations. Germline testing rates and pathogenic variant (PV) findings were also assessed. Ordering provider specialties and family history were reviewed to contextualize clinical decision-making. Results: The majority of patients, 253/279 (90.4%), had invasive ductal carcinoma, followed by 22 (7.9%) with invasive lobular carcinoma, 3 (1.1%) with male breast cancer, and 1 (0.36%) with ductal carcinoma in situ (DCIS). The median age at diagnosis was 61 years (range 24-90). Most patients were female (273, 98%). This cohort was predominantly White (156, 53.6%), with Black (32, 11.5%) and Hispanic/Latino (20, 6.9%) individuals comprising smaller proportions. Tumor variant classification showed that 96.8% (270) of patients had at least one classified tumor variant in categories IA, IB, IIC, or IID. IA variants, defined as alterations with an FDA-approved therapy for the patient’s tumor type, were present in 126 patients (45%), where 16 patients harbored 2 variants. The most frequently altered IA genes were PIK3CA (75), ERBB2 (27), ESR1 (10), PTEN (7), AKT1 (6), BRCA1 (5), BRCA2 (4), and gene fusions (4). Microsatellite instability (MSI) was high in only 2 (0.7%) patients. High tumor mutational burden (TMB) was observed in 37 (13.2%) cases. PD-L1 analysis was performed in 204 (73.1%) patients, with 54 (26.5%) having positive PD-L1 expression. Germline testing was conducted in 163 (58%) patients. 14 pathogenic variants were identified, including in MUTYH (5), CHEK2 (2), and one each of BRCA1, BRCA2, RAD51C, TP53, PALB2, APC, and MSH3. Thirteen of these variants (93%) were detected in tumor profiling. Most genomic tests were ordered by medical oncologists (218, 76.8%), followed by genetics providers (51, 18%). There were 499 relatives with reported cancer. 111 (22.2%) had a history of female breast cancer. Half of the patients reported no family history of cancer. Conclusion: This analysis highlights the genomic complexity and heterogeneity of breast cancer. The high prevalence of actionable tumor variants, particularly in PIK3CA and ERBB2, demonstrates the clinical utility of CGP. Importantly, 93% of germline PVs were also detected in tumor profiling; however, one PV was missed, consistent with prior data showing that 8-10% of germline PVs may not be captured by tumor-only testing. Despite guideline recommendations, only 58% of patients underwent germline testing, and just half reported a family history of cancer. These findings underscore the importance of integrating both germline and somatic testing to ensure comprehensive genomic assessment and optimize patient care.
Presentation numberPS2-10-27
Unmasking Sampling Artefacts in Early Breast Cancer: Molecular Shifts Between Diagnostic and Surgical Specimens in the Absence of Therapy
Milana Arantza Bergamino Sirvén, Hospital Clinic de Barcelona, Barcelona, Spain
M. A. Bergamino Sirvén1, A. Pous2, S. Cabrero3, A. Castillo4, G. Martinelli5, L. Pons4, C. Perelló6, X. Zhu7, M. Pascual8, E. Riveira9, P. Rodriguez4, E. Felip2, M. Luna10, I. Teruel11, B. Cirauqui2, L. Blay12, A. López-Paradís2, J. Antón2, F. Schettini1, M. González-Rodríguez1, L. Boronat13, A. Mariscal Martínez14, M. Margeli2, C. Ríos10, E. Ballana15, P. Fernández-Ruíz16; 1Medical Oncology, Hospital Clinic de Barcelona, Barcelona, SPAIN, 2Medical Oncology, Institut Català d’Oncologia de Badalona, Badalona, SPAIN, 3AIDS Research Institute-IrsiCaixa, Health Research Institute Germans Trias i Pujol, Barcelona, SPAIN, 4Pathological Anatomy, Hospital Germans Trias i Pujol, Badalona, SPAIN, 5Medical Oncology, Ospedale Policlinico Di Modena, Modena, ITALY, 6Pathology Anatomy, Hospital Germans Trias i Pujol, Badalona, SPAIN, 7ICR-CTSU Integrative Genomic Analysis in Clinical Trials, The Institute of Cancer Research, London, UNITED KINGDOM, 8General Surgery, Hospital Germans Trías i Pujol, Badalona, SPAIN, 9AIDS Research Institute-IrsiCaixa, Health Research Institute Germans Trias i Pujol, Badalona, SPAIN, 10Gynaecology, Hospital Germans Trías i Pujol, Badalona, SPAIN, 11Medical Oncology, Intitut Català d’Oncologia de Badalona, Badalona, SPAIN, 12General Surgery, Hospital Germans Trias i PUjol, Badalona, SPAIN, 13Medical Oncology, Hospital Germans Trías i Pujol, Badalona, SPAIN, 14Radiology, Hospital Germans Trias i Pujol, Badalona, SPAIN, 15AIDS Research Institute-IrsiCaixa, Health Research Institute Germans Trias i PujolHospital Clinic de Barcelona, Badalona, SPAIN, 16Pathological Anatomy, Hospital Germans Trías i Pujol, Badalona, SPAIN.
Background: Transcriptomic analyses using patient-matched tumour samples collected before and after treatment enable the assessment of treatment-induced molecular changes by minimizing inter-individual biological variability. However, the absence of untreated control groups often limits the ability to distinguish true treatment-induced changes from sampling-related artefacts. This issue was highlighted in the POETIC window-of-opportunity trial. To overcome this limitation, we systematically evaluated gene expression dynamics (GED) in a cohort of patients with invasive breast cancer (BC) undergoing primary surgery without prior systemic treatment, to provide a reference for treatment-independent GED.Methods: Within the SEOM-ARTEFACT Spanish study (2023-2024), we prospectively collected paired tumour samples from 30 patients with early BC at three timepoints: diagnostic core biopsy (TP1), immediate biopsy from the surgical specimen (TP2), and the routine surgical excision sample after the usual standard fixation delay of 30 minutes to 2 hours (TP3). All patients were treated at the Institut Català d’Oncologia – Hospital Germans Trias i Pujol (Badalona) and received no neoadjuvant or window-of-opportunity therapy. Time from excision to fixation was documented for adjustment. RNA sequencing was used to assess intrinsic subtypes using the PAM50 RNAseq-adapted algorithm and GED. Here, we report transcriptomic findings from the first ten patients with high-quality material available across all three timepoints. Results: Median age was 74 years (range 24-94). Most tumours were ductal (80%) and grade 2 (80%). Surrogate IHC-based subtypes included Luminal B (60%), Luminal A (30%), and triple-negative (10%). Intrinsic and surrogate subtypes matched in 60% of cases (6/10). Only 40% of patients received adjuvant chemotherapy, while 80% were treated with adjuvant endocrine therapy. Despite the absence of preoperative treatment, we observed consistent GED changes between TP1 and TP3: 28 genes were upregulated and 20 downregulated (|log₂FC| = 1.5-4.8; FDR <0.05), independent of subtype. Upregulated genes included stress-response and hypoxia-related markers (FOSB, DUSP1, EGR1 involved in FOS-JUN pathways), as well as inflammatory chemokines (IL6, CCL20, CCL4). Downregulated genes were associated with mechanical stimulus response and glucagon metabolism pathways (GNGT2, SLC9A5, TRPV3). No significant differences were found between TP1 and TP2, supporting a sampling-related rather than biological origin. Due to limited material, TP2-TP3 comparisons were not feasible. Subtype classification shifted across timepoints in 3 patients, all with borderline Luminal A/B profiles, indicating only minimal shifts in proliferation and endocrine-related genes. Conclusions: This prospective analysis shows that tumour sampling methodology significantly alters gene expression, likely reflecting transient hypoxia and wound-related immune responses. These changes, driven by upregulation of stress-response genes (FOS-JUN) and local cytokines (IL6, CCL20), highlight the biological impact of biopsy-induced microenvironmental remodelling. Such artefactual alterations represent a key confounding factor in neoadjuvant and window-of-opportunity trial designs and Log2FC values from these untreated controls may serve as correction factors in future studies. Ongoing analyses in the full ARTEFACT cohort will further define the extent and implications of these artefacts.
Presentation numberPS2-10-29
Tumor emboli in inflammatory breast cancer reveals macrophage-TNF-a signaling networks as targetable pathways
Pritha PAI, Duke University School of Medicine, Durham, NC
P. PAI1, C. Van Berckelaer2, S. Van Laere2, A. Bennion1, T. Charity3, J. Yang4, F. Bertucci5, P. Van Dam2, G. M. Palmer3, S. J. McCall6, L. Y. Dirix7, N. T. Ueno8, G. R. Devi1; 1Surgical Sciences, Duke University School of Medicine, Durham, NC, 2Center for Oncological Research, Integrated Personalized and Precision Oncology Network, University of Antwerp, Antwerpen, BELGIUM, 3Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, 4Program in Medical Physics, Duke University School of Medicine, Durham, NC, 5Predictive Oncology team, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, Inserm, CNRS, AMU, Marseille, FRANCE, 6Department of Pathology, Duke University School of Medicine, Durham, NC, 7Department of Medical Oncology, ZAS, Campus Sint Augustinus, Wilrijk-Antwerp, BELGIUM, 8Translational and Clinical Research Program, University of Hawaiʻi Cancer Center, Honolulu, HI.
Background: Despite multidisciplinary treatment strategies, overall survival rates for patients with inflammatory breast cancer (IBC) remain dismal. IBC tumors are characterized by diffuse clusters of cells found in dermal tissue and lymphatic vessels, known as tumor emboli, believed to contribute to disease lethality. Yet, the mechanisms behind tumor embolus formation and their interaction with the immune-rich microenvironment (TiME) remain poorly understood. We hypothesized that the presence of tumor-associated macrophages (TAMs) in the TiME of IBC tumor emboli leads to the activation of the TNFα-NFκB-XIAP survival signaling, causing immune evasion and therapy resistance. Methods: Ex vivo tumor emboli/organoids were generated from patient-derived cell lines cultured in lymphatic-like conditions for transcriptomic and proteomic analysis. To validate these preclinical datasets, spatial immunophenotyping was performed on clinical IBC samples. A transgenic CX3cr1GFP murine model was generated for visualization of macrophage infiltration and tumor emboli within the TiME via a surgically implanted window chamber, enabling intravital imaging and targeting. Results: Firstly, to overcome the scarcity of annotated tumor emboli biospecimens, a novel technique was established to culture tumor emboli organoids recapitulating the physiological conditions of the dermal lymphatics. Using these organoids, transcriptomic and proteomic profiling were performed, revealing differentially expressed genes and proteins compared to monolayer cell cultures, which resembled drug resistance (JI = 0.402, P < 0.001). Gene Set Enrichment Analysis of organoid versus monolayer culture revealed significant activation of TNFα and NF-κB signaling, as well as upregulation of inflammatory cytokines and chemokines, including IL-6, CCL20, and CXCL10, implicating enhanced leukocyte chemotaxis. Next, to validate the ex vivo data, 24 biospecimens with tumor emboli underwent spatial immunohistochemical analysis, for immune cell infiltration, revealing TAM enrichment (19/23). Notably, the ratio of cell densities between emboli and stroma was the highest for TAMs (P = 0.1). Other immune cell types less frequently observed in emboli included cytotoxic T cells (12/24), activated B cells (11/24), and regulatory T cells (6/23). Moreover, In the World IBC Consortium cohort (n = 78), 73% of patients with dermal lymphatic emboli failed to achieve pathological complete response (pCR) to neoadjuvant chemotherapy (Odds Ratio = 2.093; Chi-square P = 0.145) suggesting a potential association between the presence of tumor emboli and a drug-resistant phenotype in IBC patients. Finally, to further study macrophage infiltration, a novel murine model with GFP-tagged macrophages (CX3cr1GFP Nu/Nu) was developed. Intravital imaging of these mice using a dorsal skin window chamber revealed rapid macrophage recruitment to the implanted tumor embolus stroma (P < 0.05), suggesting active macrophage infiltration in TiME. Additionally, to target TNFα-XIAP signaling, birinapant-treated IBC cell clusters were implanted in mice, resulting in significant inhibition of tumor spread (P < 0.01), despite unchanged macrophage migration, indicating potential benefit of TNFα-XIAP blockade in IBC. Conclusion: Using both preclinical models and patient-derived biospecimens, our research showed that IBC tumor emboli contribute to TAM infiltration in TiME, driving pro-survival TNFα-NFκB-XIAP signaling leading to drug resistance. We identified birinapant as a potential therapeutic agent to disrupt this signaling axis, which paves a path towards the development of new treatments for IBC. Funding: W81XWH2010153, MBG-20-141-01-MBG, R01CA264529.
Presentation numberPS2-10-30
Tumor-associated macrophage (TAMs) and cancer-associated fibroblasts (CAFs) profiles in invasive lobular carcinoma (ILC) vs no special type (NST)
Jason Mouabbi, MD Anderson Cancer Center, Houston, TX
J. Mouabbi1, P. pohlmann1, B. Lim1, R. A. Mukhtar2, D. Grachev3, B. Baranov3, C. Chalabyan3, D. Goncharova3, P. Turova3, K. Chernyshov3, V. Kushnarev3, F. Paradiso3, J. Litton1, R. Layman1, F. Meric-Bernstam4, J. Rodon4; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Division of Surgical Oncology, University of California San Francisco, San Francisco, CA, 3NA, BostonGene, Boston, MA, 4Invest. Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX.
Background: ILC exhibits unique tumor microenvironment (TME) characteristics with abundant stroma and immune evasion. TAMs and CAFs are critical non-lymphoid TME components modulating immune response and progression. We aimed to compare macrophage polarization and CAF subtypes in ILC vs NST (previously known as invasive ductal carcinoma or IDC) using cellular deconvolution platform to generate large-scale transcriptomic data, and to correlate these profiles with immunotherapeutic targets, identifying potential mechanisms of immune evasion and therapeutic opportunities.Methods: 617 breast cancer samples were reviewed by central pathologist for confirmation of histologic diagnosis of ILC or NST. Based on transcriptomic results, they were subsequently classified as histomolecular ILC (hmILC) based on CDH1 inactivation/low expression; or histomolecular NST (hmNST). We analyzed bulk RNA-seq from 126 hmILC (105 Luminal, 21 non-Luminal) and 491 hmNST tumors (274 Luminal, 217 non-Luminal). The Cancer Genome Atlas (TCGA) cohort served as independent validation set. TME composition was determined using Kassandra deconvolution algorithm, quantifying M1/M2 macrophages and CAF subtypes (inflammatory-iCAF, myofibroblastic-myCAF, perivascular-like-PVL).Results: In the discovery set, predicted proportions of macrophage and CAF cell populations were assessed in the experimental cohort across molecularly defined breast cancer groups. Among Luminal hmILC tumors, median proportions were as follows: Macrophages M2 – 0.04, Macrophages M1 – 0.02, iCAF – 0.15, myCAF – 0.07, and PVL – 0.03. In Luminal hmNST tumors, values were comparable: Macrophages M2 – 0.05, Macrophages M1 – 0.03, iCAF – 0.14, myCAF – 0.06, and PVL – 0.02. In the non-Luminal subset, hmILC showed Macrophages M2 and M1 proportions of 0.05 and 0.03, respectively, with iCAF – 0.10, myCAF – 0.03, and PVL – 0.04, whereas hmNST displayed values of 0.05 (Macrophages M2), 0.03 (Macrophages M1), 0.13 (iCAF), 0.05 (myCAF), and 0.02 (PVL). The TCGA validation set showed similar trends to the discovery set; the only statistically significant observation across both cohorts was a modest enrichment of PVL cells in Luminal hmILC compared to Luminal hmNST tumors (p < 0.05, U-test).Conclusions: This large-scale comparative analysis reveals that hmILC harbors an immunosuppressive, stromal-rich TME similar to that of hmNST, characterized by TAM accumulation and FAP^+ myCAF dominance and an equal distribution of M1/M2-polarization ratio. These findings suggest that multiple immune evasion mechanisms are present in ILC, including TAM-mediated suppression. CAF-driven barriers in ILC may contribute to poor immune cell penetration, warranting investigation into CAF-driven modulation and drug delivery.
Presentation numberPS2-11-01
Lmtk2: a novel driver of endocrine resistance in ER+ breast cancer
Anna M Detry, Mayo Clinic, Rochester, MN
A. M. Detry1, S. Marigliano1, J. Heinen1, A. Bass1, C. Jones1, R. S. Muthyala1, M. P. Goetz2, J. R. Hawse1; 1Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 2Oncology, Mayo Clinic, Rochester, MN.
Breast cancer is the most common female malignancy in the U.S. and the leading cause of cancer-related deaths in women worldwide. Nearly 80% of breast tumors express estrogen receptor alpha (ERα) which drives tumor progression. While CDK4/6 inhibitors in combination with endocrine therapy improve overall survival in the metastatic setting and disease-free survival in the adjuvant setting, many patients eventually relapse/progress with resistant forms of disease. In the post-CDK4/6 inhibitor setting, second line endocrine therapy responses are limited, highlighting the need for alternative treatment approaches. Through a genome-wide CRISPR knockout screen, we identified Lemur Tail Kinase 2 (LMTK2) as the top hit exhibiting synthetic lethality in the presence of the tamoxifen metabolite, endoxifen, in both endocrine-sensitive and -resistant cell line models. We further studied multiple other endocrine resistant models and identified that LMTK2 mRNA and protein expression were both increased in ERα cell lines resistant to estrogen deprivation, tamoxifen, fulvestrant, abemaciclib and palbociclib, as well as in endocrine resistant patient derived models. We then studied LMTK2 in patient samples and identified copy number gain/amplification in 67% of ERα+ human metastatic breast tumors, while only 1% of primary tumors harbored these genomic alterations. From a prognostic standpoint, elevated tumoral LMTK2 expression was associated with worse recurrence-free survival and overall survival in early-stage breast cancer. Through proteomic studies, we identified the MET-PI3K-AKT pathway as overactivated in LMTK2 overexpressing cells. Unlike activating PIK3CA mutations that drive AKT signaling, LMTK2 was found to employ an alternative mechanism to activate AKT signaling by inhibiting PP1α, an AKT-inhibitory phosphatase. Further, LMTK2 overexpression was sufficient to drive endocrine and CDK4/6 inhibitor resistance in vitro and resulted in dramatic increases in AKT and ERα phosphorylation and activity. While knockout of LMTK2 in treatment naïve ERα+ breast cancer cells neither altered AKT/ERα phosphorylation/activity nor endocrine/CDK4/6 inhibitor responsiveness, attempts to knockout LMTK2 in endocrine/CDK4/6 inhibitor-resistant models failed, further confirming its essentiality in this setting. In summary we have uncovered LMTK2 as a novel biomarker of endocrine/CDK4/6 inhibitor resistance that is mechanistically linked to the AKT pathway. LMTK2 may be a novel drug target for the effective treatment of resistant and advanced forms of ERα+ breast cancer.
Presentation numberPS2-11-02
Chemotherapy-educated macrophages promote dormant tumor cell reactivation by shaping a pro-fibrotic niche
Dalit Barkan, University of Haifa, Haifa, Israel
V. Zubenko, S. Michaeli-Ashkenasi, K. Weidenfeld-Barenboim, C. Ezra, D. Barkan; Department of Human Biology, University of Haifa, Haifa, ISRAEL.
Recurrence of metastatic breast cancer (BC) after neoadjuvant chemotherapy remains a major clinical challenge and is often driven by dormant disseminated tumor cells (DTCs) that escape initial treatment and later reawaken in distant organs. While cytotoxic therapies target proliferating tumor cells, they may also alter the tumor microenvironment in ways that promote metastatic progression. However, the mechanisms linking chemotherapy to DTCs reactivation remain poorly understood. We hypothesized that macrophages engulfing chemotherapy-treated tumor cells are reprogrammed into a pro-metastatic phenotype, secreting factors that contribute to the formation of a fibrotic niche permissive to DTCs outgrowth. To test this, we performed RNA sequencing on macrophages that had phagocytosed Her2+ breast cancer cells (D2A1 cells) pre-treated with either doxorubicin (DOX) or paclitaxel (PTX). Transcriptional profiling revealed distinct signatures: DOX-exposed macrophages (Mφ-DOX) upregulated immune defense genes, whereas PTX-exposed macrophages (Mφ-PTX) showed a strong pro-fibrotic signature. Both macrophage populations shared enrichment in pathways associated with extracellular matrix remodeling, angiogenesis, and wound healing, hallmarks of metastatic niche activation. Secreted mediators of Mφ-DOX and Mφ-PTX promoted the escape of dormant DTCs from quiescence in a 3D dormancy model and in vivo. Previously, we demonstrated that a fibrotic niche supports DTCs outgrowth. Likewise, our results suggest that secreted mediators of these chemo-educated macrophages contribute to fibrotic niche formation, accelerating DTCs reactivation via beta1 integrin. Significance: These findings uncover a novel mechanism by which chemotherapy may inadvertently promote recurrence by educating macrophages into pro-metastatic effectors. Identifying the pro-metastatic mediators secreted by these chemo-educated macrophages may uncover novel therapeutic targets. This approach holds promise for developing complementary treatments to prevent BC recurrence and improve long-term patient outcomes.
Presentation numberPS2-11-03
Fish-positive circulating tumor cells persist after effective therapy in estrogen receptor positive breast cancer: insights into genomic mechanisms of endocrine resistance
Kathryn Baldwin, LSUHS Shreveport, Shreveport, LA
K. Baldwin1, K. von Maltzan1, A. Zulli1, M. Richard1, J. Abdulsattar2, D. Maroni3, P. T. Greipp4, R. Knudson4, Mayo Cytogenetics Core, S. P. Thayer1; 1General Surgery, LSUHS Shreveport, Shreveport, LA, 2Pathology, LSUHS Shreveport, Shreveport, LA, 3Neurosurgery, University of Nebraska Medical Center, Omaha, NE, 4Mayo Clinic Cytogenetics Core Laboratory, Mayo Foundation, Rochester, MN.
Background: Estrogen receptor (ER) positive breast cancer is commonly treated with endocrine therapy, yet resistance to estrogen deprivation remains a significant clinical challenge. Using custom FISH probes designed to detect tumor-specific genomic alterations, we identified circulating tumor cells (CTCs) of true tumor origin in early-stage luminal breast cancer patients. Longitudinal analysis revealed that 80% of patients had FISH-positive CTCs after completing therapy and while on antiestrogen treatment, despite no clinical evidence of disease. This finding suggests that standard endocrine therapy is insufficient to eradicate all malignant clones, indicating the persistence of therapy-resistant tumor cells. Methods: Primary tumor DNA was analyzed using the Oncoscan microarray platform to identify somatic copy-number alterations (SCNAs). Custom FISH probes targeting these alterations were applied to peripheral blood samples before and after therapy. Immunofluorescence characterization of CTCs was performed to assess epithelial and mesenchymal phenotypes as well as ER expression status. Genomic profiling focused on identifying drivers of proliferation, known ER resistance genes, and ER transcriptional regulators. Results: Immunofluorescence revealed that CTCs were highly heterogeneous, displaying both epithelial and mesenchymal phenotypes. ER expression among CTCs was variable: while some CTCs were ER-positive, a significant proportion showed reduced or absent ER staining. This heterogeneity suggests that loss or downregulation of ER is a key mechanism of resistance, allowing tumor cells to persist despite antiestrogen therapy. Genomic profiling of parent tumors demonstrated multiple, overlapping mechanisms supporting both proliferation and endocrine resistance. All tumors harbored several genomic alterations known to drive proliferation, most commonly involving MCL1, NTRK1, SDHC, SDHD, CHEK1, ATM, and BCL6. Every tumor exhibited at least one established mechanism of ER resistance, with frequent alterations in ATM and PIK3CA, and additional recurrent changes in AKT3, MTOR, and ARID1A. Several tumors also had alterations in genes directly involved in ER transcriptional regulation, notably ARID1A and SPEN. Conclusions: The persistence of FISH-positive CTCs in the majority of patients after therapy—despite no clinical evidence of disease—highlights the insufficiency of current endocrine regimens. The underlying genomic complexity, characterized by multiple drivers of proliferation, redundant ER resistance mechanisms, and disruption of ER regulatory pathways, underpins the survival and persistence of CTCs. These findings underscore the need for therapeutic strategies that address the multifaceted and overlapping nature of endocrine resistance in luminal breast cancer.
Presentation numberPS2-11-04
Transcriptional Plasticity in Early Evolving Endocrine Therapy Resistance
Yash N Agrawal, University of North Carolina at Chapel Hill, Chapel Hill, NC
Y. N. Agrawal, S. Haase, A. A. Whitman, P. M. Spanheimer; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.
Introduction: 70% of breast cancers express estrogen receptor (ER) alpha. Endocrine therapies (ET) initially form the backbone of their treatment, but more than one-third develop ET resistance. Most research on ET resistance focuses on pathways cooperating with high ER activity, but less on cellular diversity within ER-positive (ER+) tumors. We hypothesize that a fraction of cells with low ER expression in untreated ER+ tumors can resist tamoxifen, which is selected by treatment and can evolve towards adapted resistance.
Methods: We performed single-cell RNA expression and chromatin accessibility analysis on ER+/HER2-negative patient-derived xenografts (PDX) treated with tamoxifen (50 mg/kg PO daily) or control for 6 weeks. Cell clusters were characterized using gene set enrichment analysis and trajectory analysis.
Results: ESR1 expression correlated inversely with PIM3, PLK3, and IRAK2 kinase activity and positively with RET kinase activity in untreated tumors in multiple datasets (PMID: 34493872, 23000897, 34739872) and two PDX models (Figure 1A). Trajectory analysis of the PDX1 model defined the closest trajectory between untreated and tamoxifen-treated cells through ESR1-low cell clusters, suggesting that tamoxifen resistance arises from an untreated ESR1-low population (Figure 1B). The untreated ESR1-low cluster was again characterized by greater PIM3, PLK3, and IRAK2 kinase activity expression.
In the PDX1 model, the untreated ESR1-low cluster also had higher expression of kinase signatures related to MAPK, PIK3CA, and NF-κB. The trajectory from untreated to tamoxifen-treated ESR1-low clusters was associated with lower activity of kinase signatures related to MAPK and NF-κB pathways, and increased HER2 kinase activity signature. Late-resistant cells at the endpoints of diverging trajectories regain expression of ESR1 but not ESR1-associated genes, and have decreased PIM3 kinase activity signature and increased BRAF and PIK3CA kinase activity signature expression (Figure 1C).
Conclusions: ESR1-low cells in untreated ER+ tumors may represent a funnel point in the development of ET resistance and consistently demonstrate enrichment of specific kinase signatures. In persisting on ET, these ESR1-low cells might seed ET-resistant trajectories with heterogeneous potential acquired driver kinase pathways. Early targeting of pathways enriched in ESR1-low cells in ER+ tumors in combination with ET could prevent or delay ET resistance. Further study is needed to determine the reproducibility of resistant trajectories and the efficacy of selectively targeting preexisting ESR-low cells.
![[d771d6c8-5fc7-420e-91b5-448656a825e9saa34b.html_g�0002.gif]](d771d6c8-5fc7-420e-91b5-448656a825e9saa34b.html_g2.gif)
Presentation numberPS2-11-05
Nuclear HER2-ER crosstalk in HR+/HER2+ breast cancer
Stanley Tam, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC
S. Tam, S. Bahnassy, L. Jin, D. Kung, R. Rahhal, S. M. Swain, R. B. Riggins; Oncology, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC.
Approximately half of all HER2+ breast cancers are hormone receptor positive (HR+) and express estrogen and/or progesterone receptors (ER, PR). These HR+/HER2+ tumors are less responsive to neoadjuvant anti-HER2 therapy and exhibit lower rates of pathologic complete response (pCR). While HER2 primarily functions as a transmembrane receptor that mediates cytoplasmic signaling, a subpopulation of HER2 resides in the nucleus. Prior studies have shown that chromatin-associated HER2 can serve as a transcriptional co-regulator, and its presence in tamoxifen-treated, ER+ (non-HER2-amplified) breast cancer correlates with shorter disease-free survival. We therefore hypothesized that nuclear HER2 may reprogram ER function in HR+/HER2+ breast cancer, thereby contributing to reduced pCR rates. In multiple HR+/HER2+ cell line models, low baseline levels of nuclear HER2 are rapidly increased following treatment with the monoclonal antibodies trastuzumab and pertuzumab (TP). While TP does not alter nuclear ER levels, it enhances ER recruitment to promoter and enhancer regions of TFF1 and PGR (two classical ER target genes) and upregulates their mRNA levels in HR+/HER2+, but not in ER+ only breast cancer cells. To define the global impact of TP treatment on the ER cistrome, we performed Cleavage Under Targets & Release Using Nuclease (CUT&RUN) for ER in the presence and absence of TP in the HR+/HER2+ cell line BT474. TP markedly expands the ER cistrome, promoting ER binding to novel sites on genomic regions enriched for cut-like homeobox 2 (CUX2) and goosecoid (GSC) motifs. These TP-induced ER binding sites are enriched in genes involved in the Wnt, NOD, and receptor for advanced glycation end-products (RAGE) signaling pathways. Expression of a 39-gene signature of unique genes from these pathways that exhibit increased ER binding is associated with reduced distant metastasis-free and overall survival (DMFS, OS) in HER2+ breast cancer, but not in ER+ or triple negative breast cancer in the METABRIC cohort. In publicly available data from 151 HER2+ breast cancers across several clinical studies, expression of this signature is also significantly increased in patients who did not achieve pCR following anti-HER2 neoadjuvant therapy. Finally, multispectral immunofluorescence staining of an in-house tissue microarray (TMA) of 50 HER2+ breast cancers (18 HER2+ only, 32 HR+/HER2+) shows significantly higher nuclear HER2 intensity in HR+/HER2+ vs HER2+ only, with the strongest nuclear HER2 signal observed in cells positive for both ER and Ki67. Together, these data suggest that TP induces reprogramming of the ER cistrome via nuclear HER2, and that these events may ultimately play a role in reducing the rate of pCR in HR+/HER2+ breast cancer. Ongoing CUT&RUN analyses of primary and recurrent HR+/HER2+ tumors will further define the ER cistrome in clinical samples.
Presentation numberPS2-11-06
Quiescent-like phenotype characterizes treatment-resistant early-stage HER2-negative breast cancers and suggests novel therapeutic targets; analysis of over 2000 patients from the I-SPY2 trial
Tam Binh Vinh Bui, University of California San Francisco, San Francisco, CA
T. Bui1, D. M. Wolf1, R. W. Sayaman1, C. Yau2, A. M. Glas3, J. Coppé4, L. Brown Swigart1, G. L. Hirst1, P. R. Pohlmann5, L. Pusztai6, J. M. Rosenbluth7, L. J. Esserman2, E. F. Petricoin8, L. J. Van ‘t Veer1; 1Laboratory Medicine, University of California San Francisco, San Francisco, CA, 2Surgery, University of California San Francisco, San Francisco, CA, 3Biomarkers and Innovation, Quantum Leap Healthcare Collaborative, San Francisco, CA, 4Radiation Oncology, University of California San Francisco, San Francisco, CA, 5Department of Breast Medical Oncology and Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 6Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, 7Medicine, University of California San Francisco, San Francisco, CA, 8Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA.
Introduction The I-SPY2 neoadjuvant platform trial (NCT01042379) evaluates new therapies in high molecular risk breast cancer patients. From 2010-2022, 24 therapies were tested in 2117 patients. One-third of these patients had HER2-negative tumors with low expression of immune genes and no DNA repair deficiency (DRD) (HER2-/Immune-/DRD- subtype). These patients had very low pathologic complete response (pCR) rates (~10% on average) across all treatment and control arms, whereas those patients with Immune, DRD, or HER2 targetable tumors had higher pCR rates (with the most successful treatments achieving pCR rates between 60-100%), associated with long-term survival benefit. Here, we focus on characterizing the biology of HER2-/Immune-/DRD- tumors, with the aim of identifying novel targetable vulnerabilities to increase treatment sensitivity in these treatment-resistant tumors. Methods We analyzed pre-treatment transcriptomic data of HR+HER2- (n=889) and triple-negative (TN) (n=739) patients across 20 treatment arms in I-SPY2 for enrichment of canonical metabolic, signaling, and regulatory pathways from the Molecular Signatures Database (n=2274) including Hallmark, BioCarta, Reactome, and Pathway Interaction Database (PID) gene sets. We compared 1) HR+HER2-/Immune-/DRD- (n=617) vs. other (Immune+ and/or DRD+) HR+HER2- tumors (n=272) and 2) TN/Immune-/DRD- (n=177) vs. other (Immune+ and/or DRD+) TN tumors (n=562). Comparisons were made using Wilcoxon rank-sum tests with Benjamini-Hochberg adjustment. Results with adjusted p-values <0.05 were considered significant. Results Patients with HR+HER2-/Immune-/DRD- and TN-/Immune-/DRD- tumors had an overall pCR rate across all treatments, including chemoimmunotherapy, of 9% and 16%, respectively. Both subtypes had lower expression of the majority of canonical signaling pathway signatures, particularly of proliferation signatures, including those reflecting cell cycle checkpoints, Myc, and E2F targets, compared with other tumors, suggesting a quiescent-like phenotype. However, both HR+HER2-/Immune-/DRD- and TN-/Immune-/DRD- tumors had significantly enriched expression of signatures related to PI3K and FGFR signaling and fatty acid metabolism, which have previously been associated with treatment resistance. We identified subtype-specific signature enrichment with increased expression of WNT signaling and epigenetic modulators (e.g., HDAC) in HR+HER2-/Immune-/DRD- tumors, and enrichment of signatures reflecting coagulation events in TN/Immune-/DRD- tumors, which could potentially induce remodeling of the tumor microenvironment and/or mediate immune evasion, leading to resistance. Additional analysis of 87 epigenetic modulator genes in HR+HER2- tumors showed that 48 genes were significantly up- or downregulated in HR+HER2-/Immune-/DRD- compared with other tumors, suggesting that the quiescent-like phenotype may be driven by epigenetic silencing. Conclusion HR+HER2-/Immune-/DRD- and TN/Immune-/DRD- resistant tumors are characterized by low response to all therapies and low expression of many cancer-relevant biological pathways, including proliferation. Both had increased expression of PI3K and FGFR signaling signatures and fatty acid metabolism relative to more treatment-sensitive tumors that were Immune+ or DRD+. Moreover, receptor subtype-specific biology was identified. Our findings, which will be validated using RPPA functional signaling data from the rich I-SPY biomarker resource, suggest novel targetable vulnerabilities. These include pro-apoptotic and epigenetic targets, with already promising hits in drug screens with patient-derived organoids, supporting rapid translation to clinical trials.
Presentation numberPS2-11-07
Linc00152-regulated pde4d is a mediator of drug resistance and metastasis in highly aggressive breast cancer
Ozge Saatci, Medical University of South Carolina, Charleston, SC
O. Saatci1, R. Alam1, K. Huynh-Dam1, A. Isik2, M. Uner2, N. Belder3, P. G. Ersan3, U. M. Tokat3, B. Ulukan1, M. Cetin1, K. Calisir1, M. E. Gedik1, H. Bal3, O. Sener Sahin1, Y. Riazalhosseini4, D. Thieffry5, D. Gautheret6, B. Ogretmen1, S. Aksoy7, A. Uner2, A. Akyol2, O. Sahin1; 1Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, 2Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, TURKEY, 3Department of Molecular Biology and Genetics, Bilkent University, Ankara, TURKEY, 4Department of Human Genetics, McGill University, Montreal, QC, CANADA, 5Département de biologie de l’Ecole normale supérieure, PSL Université, Paris, FRANCE, 6Institute for Integrative Biology of the Cell, Université Paris-Saclay, Gif-sur-Yvette, FRANCE, 7Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, TURKEY.
Resistance to anti-cancer therapies is one of the major challenges against effective cancer treatment that is frequently accompanied by metastatic recurrence, the major cause of cancer-related death. Here, we identified the Phosphodiesterase 4D (PDE4D) and its novel upstream long non-coding RNA (lncRNA) LINC00152 as critical mediators of drug resistance and metastasis in aggressive breast cancers. We showed that LINC00152 interacts with and stabilizes PDE4D mRNA, thus driving resistance to standard of care endocrine therapy. Mechanistically, LINC00152/PDE4D deactivates the downstream cAMP/PKA/CREB axis, leading to reduced intracellular calcium and iron accumulation, preventing drug-induced ferroptosis. Inhibiting LINC00152 restores sensitivity by activating cAMP/PKA/ferroptosis axis which is reversed by PDE4D overexpression. In addition, we demonstrated that PDE4D promotes migration and metastasis via triggering luminal-to-basal transition in highly aggressive drug resistant models. Inhibiting LINC00152 or PDE4D reduces cancer cell migration upon PKA-mediated blockage of TGF-β signaling and reduction of mesenchymal gene expression. Targeting PDE4D using the clinically-tested PDE4D selective inhibitor, BPN14770 significantly reduces the number of circulating tumor cells and spontaneous metastasis in the highly aggressive, endocrine resistant-mimicking MMTV-PyMT model in vivo. Importantly, we showed that high levels of PDE4D mRNA is associated with worse metastasis-free survival in endocrine-treated ER+ breast cancer patients. Overall, we identified PDE4D and its highly oncogenic upstream lncRNA, LINC00152 as mediators of endocrine resistance and metastasis in the highly aggressive ER+ breast cancer.
Presentation numberPS2-11-08
Stemness-targeted therapies to enhance the efficacy of AURKA inhibitors in endocrine resistant er+ breast cancer
ANTONINO BONAVENTURA D’ASSORO, Mayo Clinic, ROCHESTER, MN
A. B. D’ASSORO1, M. Drago1, T. Haddad1, M. GOETZ1, K. Giridhar1, K. Kalari1, L. Manzella2, P. Vigneri2, F. Drago3, C. Vancheri4, C. Lange5, J. Ingle1; 1Medical Oncology, Mayo Clinic, ROCHESTER, MN, 2Experimental and Clinical Medicine, University of Catania, Catania, ITALY, 3Biomedical and Biotechnological Sciences, University of Catania, Catania, ITALY, 4Experimental and Clinical Medicine, Catania, Catania, ITALY, 5Hematology and Oncology, University of Minnesota, Minneapolis, MN.
Significance: Emergence of resistance to endocrine therapies represents a major challenge in theclinical management of estrogen receptor positive (ER+) breast tumors. CDK4/6 inhibitors(palbociclib, ribociclib, abemaciclib) have improved survival when combined with endocrine therapy.Nonetheless, resistance to CDK4/6 inhibitors eventually occurs. One of the emerging mechanisms ofresistance toward endocrine therapy and CDK4/6 inhibitors is characterized by the enrichment ofbreast cancer stem cells (CSCs) that show stem-like features such as self-renewal capacity, highCD44 expression and ALDH activity. We have demonstrated that Aurora-A kinase (AURKA) has apivotal role in promoting cancer cellular plasticity and clonal expansion of endocrine resistantCD44high/ALDHhigh CSCs. Our preclinical studies on AURKA-driven tumor stemness provided thetherapeutic foundation for a Phase-II clinical trial (TBCRC041) of the AURKA inhibitor, alisertib, whichdemonstrated clinically meaningful activity in patients with endocrine and CDK4/6 inhibitors resistantER+ metastatic breast cancer (MBC). Despite such activity, alisertib resistance finally occurred.Therefore, we hypothesize that up-regulation of IL6R/STAT3 and PR stemness oncogenic pathwaysmay sustain the clonal expansion of CD44high/ALDHhigh CSCs after tumor progression on AURKAinhibitors.Experimental Design: We used variant MCF-7 breast cancer models that show resistance toendocrine therapy (letrozole and fulvestrant) and alisertib (MCF-7 LR/FR, MCF-7 LR/FR-AlisR) todefine in vitro and in vivo the efficacy of stemness-targeted therapies to enhance alisertib activity byimpairing the enrichment of hormone refractory CD44High/ALDHHigh CSCs. Parental MCF-7 cells wereused as control. We employed exclusive PDX-derived 3D-Mammospheres (MPS) that wereestablished from the Phase-II clinical trial TBCRC041 to test the efficacy of stemness-targetedtherapies in enhancing the therapeutic activity of alisertib. In Vitro Studies: Immunofluorescence andimmunoblot assays were used to assess the expression of breast cancer stemness biomarkers.Enrichment of ALDHHigh CSCs was measured using Red-Aldefluor kit. Cell proliferation and Red-Annexin-VHigh apoptotic cells were quantified in real-time using IncuCyte S3. In Vivo Studies: 1x106breast cancer cells were injected into the mammary fat pad of NSG-female mice and treated withfulvestrant, alisertib and tocilizumab as monotherapy or in combination. Control groups were treatedwith saline solution placebo. Drug Treatments: Tocilizumab and Onapristone were used to inhibit theIL6R/STAT3 and PR stemness pathways, respectively.Results: Our study demonstrates for the first time that nuclear STAT3/PR complex mediatesAURKA-induced expression of KLF4 stemness gene and enrichment of endocrine resistantCD44high/ALDHhigh CSCs. Alisertib resistant MCF-7 LR/FR cells showed increased expression ofIL6R, T288-AURKA, S727-STAT3 and S294-PR compared to alisertib naïve cells. MCF-7 LR/FR-AlisR-derived 3D-MPS had increased KLF4 and CD44 expression. Alisertib resistant PDX modelsalso showed high levels of CD44, IL6R/STAT3 and KLF4 expression. Remarkable, pharmacologicalblockade of PR and STAT3 transcriptional regulatory networks enhanced the efficacy of alisertib byreducing the enrichment of CD44High/ALDHHigh CSCs.Conclusions: These findings provide the strong preclinical rationale for the development of novelcombinatorial therapeutic strategies targeting IL6R/STAT3 and PR stemness pathways to enhancethe efficacy of AURKA inhibitors that will result in the selective eradication of CD44High/ALDHHighCSCs with consequent benefits on the overall survival of patients with advanced ER+ breast tumorsrefractory to endocrine therapies and CDK4/6 inhibitors.輀
Presentation numberPS2-11-09
Pan RTK negative regulator LRIG1 as a therapeutic target for endocrine resistance in estrogen receptor-positive breast cancer
Brittany D Jenkins, Johns Hopkins University, Baltimore, MD
B. D. Jenkins, K. S. Ingram, A. Hokulani Pajimola, J. Morgan, E. Liu, U. Nayar; Biochemistry and Molecular Biology, Johns Hopkins University, Baltimore, MD.
Background: Endocrine resistance affects 30-40% of estrogen receptor-positive (ER+) breast cancer patients within 5 years, often leading to metastatic disease responsible for >90% of breast cancer deaths. Resistance commonly emerges through hyperactivation of receptor tyrosine kinase (RTK) pathways such as HER2, EGFR, FGFR, and IGF-1R. Current clinical approaches targeting individual RTKs have limited durability due to compensatory pathway activation. LRIG1 (Leucine-rich Repeats and Immunoglobulin-like Domains Protein 1) is a pan-negative regulator of RTKs that simultaneously downregulates multiple RTK families through c-CBL-mediated ubiquitination and proteasomal degradation. We hypothesized that LRIG1 restoration could overcome endocrine resistance by targeting multiple RTK pathways simultaneously. Methods: We analyzed LRIG1 gene expression in publicly available data from METABRIC ER+ breast cancer patients who received endocrine therapy (n=1,079). We also evaluated LRIG1 expression and function in T47D and MCF7 ER+ breast cancer cell lines harboring HER2 (S653C, L869R, L755S, V777L) and FGFR2 (K660N, M538I) activating mutations associated with endocrine resistance. LRIG1 modulation was assessed via siRNA knockdown and recombinant LRIG1 protein supplementation. Functional outcomes were measured through proliferation assays (EdU incorporation) and western blot analysis of RTK pathways. Results: Clinical analysis of METABRIC data revealed that ER+ breast cancer patients with higher LRIG1 expression had significantly longer progression-free survival after endocrine therapy (hazard ratio = 0.65, p<0.0001). Patients whose cancer recurred had significantly lower LRIG1 expression compared to those without recurrence (p<0.0001). In endocrine-resistant cell lines with activating RTK mutations, LRIG1 was consistently downregulated compared to controls. HER2-activating mutants showed decreased LRIG1 protein expression and increased phosphorylated HER2. Treatment with RTK inhibitors (Neratinib for HER2 and FIIN3 for FGFR) significantly restored LRIG1 expression in resistant cells. Recombinant LRIG1 supplementation (0-20 μg/mL) in LRIG1-knockdown T47D cells significantly decreased proliferation in a dose-dependent manner, with greater than 50% reduction at the highest concentration tested. Conclusions: LRIG1 represents a clinically actionable biomarker and potential therapeutic target in endocrine-resistant ER+ breast cancer. The inverse relationship between LRIG1 expression and RTK pathway activation, coupled with the restoration of LRIG1 by RTK inhibitors, suggests a therapeutically exploitable bidirectional relationship. Exogenous LRIG1 supplementation effectively suppresses proliferation in endocrine-resistant models, supporting its development as a first-in-class pan-RTK therapeutic adjuvant. This approach could address the fundamental limitation of current targeted therapies by simultaneously blocking multiple resistance pathways, potentially offering new treatment options for patients with limited therapeutic alternatives.
Presentation numberPS2-11-10
Mct4 drives triple-negative breast cancer progression via lactate-induced histone lactylation and mapk pathway activation
Yang Peng, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
Y. Peng, Q. Luo, S. Liu; Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, CHINA.
Background: Triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype with limited therapeutic options and poor prognosis. Monocarboxylate transporter 4 (MCT4) is a key lactate efflux transporter involved in cancer metabolism. However, the mechanistic role of MCT4 in TNBC progression and its potential as a therapeutic target remain unclear.Methods: MCT4 expression was analyzed in TNBC patient tissues and correlated with clinical outcomes. MDA-MB-231 cells were subjected to hypoxia treatment and paclitaxel (PTX) exposure. MCT4 was knocked down using siRNA. Cell proliferation, invasion, migration, and PTX sensitivity were assessed. In vivo xenograft experiments were performed to evaluate tumor growth and drug response. RNA-sequencing and KEGG pathway analysis were conducted. Lactate secretion and glucose uptake were measured. Western blot analysis examined MAPK pathway proteins and histone modifications. CUT&Tag sequencing analyzed histone H3K9 lactylation changes and associated gene transcription.Results: MCT4 was significantly overexpressed in TNBC tissues and high expression correlated with poor patient prognosis. Both hypoxia and PTX treatment upregulated MCT4 expression in MDA-MB-231 cells. MCT4 knockdown markedly inhibited cell proliferation, invasion, and migration while enhancing PTX sensitivity. In vivo xenograft studies confirmed that MCT4 knockdown significantly retarded tumor growth and increased PTX sensitivity. RNA-sequencing revealed that MCT4 knockdown dramatically downregulated MAPK signaling pathway. Metabolically, MCT4 knockdown reduced both lactate secretion and glucose uptake. Western blot analysis confirmed that MCT4 knockdown decreased MAPK pathway-related proteins. Under hypoxic conditions, RNA-sequencing showed MAPK pathway activation, which was accompanied by increased histone lactylation levels and H3K9 lactylation. MCT4 knockdown reversed these histone modifications. CUT&Tag analysis revealed that hypoxia-induced H3K9 lactylation promoted transcription of MAPK pathway genes. Notably, MCT4 was not identified among the genes with increased H3K9 lactylation-mediated transcription, suggesting that MCT4 functions as an upstream regulator of histone lactylation rather than a downstream target, establishing a direct mechanistic link between lactate metabolism and gene regulation.Conclusions: This study demonstrates that MCT4 promotes TNBC progression through a novel lactate-histone lactylation-MAPK axis. MCT4 facilitates lactate production and efflux, which drives histone H3K9 lactylation and subsequent activation of MAPK signaling genes, ultimately enhancing cancer cell aggressiveness and chemoresistance. Our findings are validated both in vitro and in vivo, providing strong mechanistic evidence for MCT4 as a therapeutic target in TNBC.Clinical Relevance: MCT4 represents a promising biomarker for TNBC prognosis and a novel therapeutic target. Targeting MCT4-mediated lactate metabolism could enhance chemotherapy efficacy and improve clinical outcomes in TNBC patients. This metabolic-epigenetic crosstalk mechanism opens new avenues for combination therapy strategies.
Presentation numberPS2-11-11
Mechanisms of Acquired Resistance to PI3K Inhibitors in Breast Cancer: The Central Role of Autophagy
Stefania Cocco, INT IRCSS Fondazione G. Pascale, naples, Italy
B. Pucci1, R. Lombardi2, T. Moccia1, R. Migliorino1, C. Ciardiello3, E. Di Gennaro1, A. Budillon4, M. De Laurentiis5, A. Leone1, S. Cocco5; 1Experimental Pharmacology Unit, Laboratory of Naples and Mercogliano (AV), INT IRCSS Fondazione G. Pascale, naples, ITALY, 2Experimental Animal Unit, INT IRCSS Fondazione G. Pascale, naples, ITALY, 3Neoplastic Progression Unit, INT IRCSS Fondazione G. Pascale, naples, ITALY, 4Scientific Directorate, INT IRCSS Fondazione G. Pascale, naples, ITALY, 5Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, INT IRCSS Fondazione G. Pascale, naples, ITALY.
The PI3K pathway plays a pivotal role in regulating cell proliferation and survival of cancer cells including breast cancer. This pathway is frequently hyperactivated in cancer due to genetic mutations, amplifications, or functional alterations in key components such as PIK3CA, PTEN, and AKT. Over the years, several PI3K inhibitors have been developed and approved for the treatment of breast cancer. However, drug tolerance and adaptive responses often emerge, leading to reactivation of the PI3K pathway and ultimately limiting the clinical efficacy of these inhibitors. This study aims to elucidate the mechanism underlying resistance to PI3K inhibitors, and to understand how tumor cells evade PI3K inhibition. At this purpose, we developed a model of acquired resistant to taselisib, a selective PI3KCA inhibitor, by generating resistant subline (MCF7 Tas-r) from MCF7 human breast cancer cells through stepwise exposure of MCF7 parental cells (MCF7-Par) to increasing concentrations of taselisib (ranging from 50 nM to 500 nM). Then, since phosphorylation is one of the major posttranslational modification of proteins that regulates several signaling pathways, including PI3K signaling, we performed phosphoproteomic profiling followed by functional enrichment analysis in order to identify the molecular alterations associated with acquired resistance. Our analysis identified 253 differentially phosphorylated proteins (196 upregulated and 57 downregulated) in resistant cells compared to parental cells. Functional enrichment analysis revealed significant enrichment of pathways related to autophagy regulation, as “mTOR signaling,” and “AMPK signaling,” highlighting autophagy as a key process altered in resistant cells. Notably, SQSTM1/p62, a critical cargo adaptor in autophagy, emerged as a central hub, together with AMPK, reinforcing the hypothesis that autophagy activation contributes to taselisib resistance. These findings were validated by Western blot analysis, which confirmed the upregulation of key proteins involved in PI3K and autophagy signaling in MCF7 Tas-r cells compared to parental cells, including p-ULK1, SQSTM1/p62, ATG5, and Beclin-1. Furthermore, we observed increased levels of phosphorylated AMPK (p-AMPK) along with downregulation of phosphorylated mTOR (p-mTOR), suggesting that AMPK activation may contribute to mTOR suppression and promote autophagy induction in resistant cells. Given the central role of autophagy in resistance, we tested the therapeutic potential of chloroquine (CQ), an anti-malarial agent known to block autophagy by inhibiting autophagosome-lysosome fusion. CQ enhanced the antitumor activity of taselisib in vitro, as demonstrated by cell viability and clonogenic assays. Taken together, our findings suggest that autophagy activation serves as a key adaptive mechanism driving acquired resistance to PI3K inhibitors such as taselisib in breast cancer. Targeting autophagy-related pathways may represent a promising therapeutic strategy to overcome or delay resistance and enhance the clinical efficacy of PI3K-targeted treatments.
Presentation numberPS2-11-12
Ccl5 and endoglin as a therapeutic target in er positive breast cancer
Kideok Jin, Albany College of Pharmacy and Health Sciences, Albany, NY
D. Lomonaco, K. Jin; Pharmaceutical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY.
Estrogen receptor (ER) drives approximately 75% of all breast cancers and serves as the primary target for endocrine therapies. However, resistance to these therapies remains a major clinical hurdle, especially in metastatic ER-positive breast cancers harboring activating ESR1 mutations (e.g., Y537S, D538G) within the ligand-binding domain. These mutations are commonly detected in metastatic lesions and circulating tumor DNA (ctDNA) from patients undergoing endocrine treatment. While combination therapies with CDK4/6 inhibitors have shown promise, the mechanisms driving resistance in ESR1-mutant tumors and their associated therapeutic vulnerabilities remain poorly defined. To address this gap, we investigated the interaction between endocrine-resistant breast cancer (ERBC) cells and the tumor microenvironment. Using co-culture systems involving seven ERBC cell lines—including genome-edited ESR1 mutants—and four stromal cell types, we profiled 28 tumor-stroma secretome combinations via cytokine antibody arrays. This approach identified CCL5 and endoglin as significantly upregulated in resistant tumor-stroma interactions. We hypothesized that CCL5 promotes endocrine resistance and metastatic progression through paracrine signaling with stromal components. To test this, we generated CCL5 knockout EO771 cells using CRISPR-Cas9 and confirmed CCL5 loss through qRT-PCR and ELISA. CCL5-deficient cells exhibited markedly reduced proliferation and migration in vitro. In vivo, orthotopic mouse models demonstrated that CCL5 knockout tumors had significantly decreased growth and metastasis. Moreover, treatment with maraviroc, a CCR5 antagonist, selectively impaired the viability of wild-type but not CCL5-deficient cells. Together, these findings identify CCL5 as a key driver of endocrine resistance and metastatic potential in ERBC, highlighting it as a promising target for therapeutic intervention. This study lays the groundwork for future clinical strategies aimed at overcoming resistance in ESR1-mutant breast cancers by disrupting CCL5 signaling.
Presentation numberPS2-11-13
Resistance to tucatinib associated with hyperactive EGFR could be overcome by co-targeting HER2 and EGFR in HER2-positive breast cancer models
Fu-Tien Liao, Baylor College of Medicine, Houston, TX
F. Liao1, L. Qin1, S. Nanda1, M. J. Shea1, C. Liu1, C. M. Sabotta2, A. Elkhanany3, S. G. Hilsenbeck1, C. Gutierrez1, M. F. Rimawi1, C. K. Osborne1, J. Veeraraghavan1, R. Schiff1; 1Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 2Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 3Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX.
Background: Tucatinib (Tuca) is a highly HER2-selective small molecule tyrosine kinase inhibitor (TKI) approved for use in HER2+ metastatic breast cancer (MBC), including in patients with brain metastasis. In current practice, despite the expanding use of other anti-HER2 agents like TDXd in the 2nd and potentially the 1st line setting, Tuca continues to be a key treatment choice for HER2+ MBC, mostly after TDxd. We previously showed that EGFR amplification leading to HER pathway reactivation is associated with acquired resistance to Tuca in HER2+ BC models, which could be overcome using dual/pan-HER TKIs or the combination of potent EGFR and HER2 inhibitors (Liao et al, Can Res, 2022). Building on these key prior in vitro findings, here we sought to validate the efficacy of this treatment strategy in the in vivo setting. Materials and Methods: Our recently developed HER2+/ER+ BT474/AZ cell model with acquired resistance to Tuca (TucaR) generated through long-term exposure to gradually increasing doses of Tuca was used. NSG mice bearing GFP/Luc-tagged TucaR xenografts grown with estrogen supplementation were randomized to vehicle, Tuca (100mg/kg, once daily gavage, 7 days/week), or EGFR-specific inhibitors: 1) the TKI gefitinib (Gef 100 mg/kg, once daily gavage, 5 days/week) or 2) the monoclonal antibody cetuximab (Cetux, 30mg/kg, IP twice a week)), either alone or in combination, and monitored for primary tumor growth (caliper measurement) and spontaneous metastasis (by bioluminescence). Results: We first observed that our TucaR model grew effectively as xenografts in the presence of Tuca thereby confirming its Tuca resistant phenotype. EGFR inhibition, using Gef or Cetux, in combination with Tuca achieved complete regression of the primary TucaR tumors. Interestingly, although complete tumor regression was also observed in all mice treated with single-agent Cetux, within 10-14 days post randomization, the tumors recurred in all mice, starting at around day 55, suggesting the potential need for simultaneous HER2 blockade. Importantly, though the combination of Tuca with Gef or Cetux achieved complete primary tumor regression (i.e., no palpable/measurable tumor) within 10-14 days post randomization, bioluminescence imaging at the end of the experiment (around day 70-80) revealed minimal/microscopic residual disease in all mice at the subcutaneous injection site. This finding was further confirmed through histological analysis. Monitoring the metastatic propensity, our TucaR model was highly metastatic, including to the lungs, lymph nodes, and potentially bone. Specifically, lung metastasis was observed in almost all mice in the vehicle (6/6), Tuca (6/7), and Cetux (5/6) treatment arms. However, these lung metastatic lesions were completely abolished in all mice treated with Gef+Tuca (6/6) and Cetux+Tuca (5/5), further accentuating the promise of this strategy in the metastatic setting. Ongoing histochemical and molecular analyses of the primary tumors, metastatic lesions, and residual disease harvested at the end of the experiment are focused on gaining additional mechanistic insights. Conclusions: Our novel findings have crucial therapeutic implications and suggest that HER2+ primary and metastatic tumors with high EGFR (~30% of HER2+ MBC) may not benefit from Tuca or EGFR inhibitors alone and need dual/pan-HER inhibitor therapy, a strategy that we intend to translate via a prospective trial. Our data also suggest the need for imaging-based endpoints in order to capture the minimal/microscopic residual disease that may otherwise be missed through routine tumor measurements, as well as the potential need for additional targeted therapies such as endocrine therapy or CDK4/6 inhibitors, beyond HER2-targeted treatment, to achieve complete tumor eradication.
Presentation numberPS2-11-14
Residual disease after trastuzumab deruxtecan treatment of HER2+ breast tumors is promoted by leucyl tRNA synthetase 2 (LARS2) in HER2-low tumor cells.
Sheheryar Kabraji, Roswell Park, Buffalo, NY
V. Kurisetty, M. Homsi, S. Kabraji; Medicine, Roswell Park, Buffalo, NY.
Background: HER2+ breast cancer is routinely treated with neoadjuvant HER2-targeted therapy but the molecular mechanisms driving residual disease (RD) remain unclear. We previously reported that RD after HER2 genetic inhibition in an inducible mouse model is associated with reduced proliferation and upregulation of the leucyl tRNA synthetase (LARS2). We also found that LARS2 was negatively correlated with ERBB2 expression and originated in a ‘HER2-low’ subpopulation in primary HER2+ mouse tumors. Here we describe a preliminary mechanism by which LARS2 promotes RD after trastuzumab deruxtecan treatment (T-DXd) in vitro and characterize the expression of LARS2 in HER2 high and HER2 low patient tissues. Methods: We used HER2+ve and HER2-ve cell lines, SK-BR-3 and HCC-1806, respectively treated with T-DXd at 3 concentrations of 1mM, 100mM and 10mM for 21 days. Total protein lysates were collected at five different time points – day 1, 3, 7, 14, and 21 post shRNA transfection (ERBB2, LARS2, HEXB, non-targeting control) as well as post-tDXD drug treatment. For the cell proliferation / survival assay (Cell-Titer Glo) cells were plated in 24 well plates and assayed at three different time points – day 1, 7, 14 To examine LARS2 and HER2 expression at single-cell resolution in patient tissues we used cyclic immunofluorescence on a tissue a microarray of 41 patients (81 cores). 48 cores were HER2+ (IHC 3+) and 33 were HER2-low (30 HER2 2+ FISH negative, 3 HER2 IHC 1+). We used anti-human HER2 (29D8), which has been previously validated against clinical grade antibodies. We also examined the expression of LARS2 between proliferating/quiescent cancer cells using our previously described multivariate proliferation index (MPI). Results: We found that T-DXd treatment reduces ERBB2 expression at doses >10 nM starting at day 1 by immunoblotting in SK-BR-3 but not HCC1806 cells. T-DXd treatment increased LARS2 expression by immunoblotting in SK-BR-3 cells between day 1-7 that returned to baseline by day 14. This change was not seen in HCC-1806. We saw a similar transient increase in LARS2 expression by immunoblotting over day 1-7 in response to shRNA mediated ERBB2 knockdown. Compared to control, T-DXd treatment, ERBB2 knockdown and LARS2 knockdown reduced cell proliferation in SK-BR-3 but not HCC1806 cell lines, as measured by Cell-Titer-Glo. Experiments to test whether T-DXd-mediated reduction in cell numbers can be abrogated by LARS2 knock down will be reported at the conference. In our TMA, mean LARS2 intensity per tumor cell as measured by t-CyCIF was higher in HER2-low tumors (0.62 ± 0.07 in HER2 1+ and 0.61 ± 0.09 in HER2 2+) compared to HER2+ (IHC 3+) tumors (0.59 ± 0.09, p<1.00e-04, paired t-tests), consistent with our mouse data showing a negative correlation between Erbb2 and Lars2 at the mRNA level. Mean LARS2 intensity in tumor cells from our TMA of HER2 expressing tumors was higher in MPI 0 (QCC) cells than MPI 1 (proliferating) tumor cells (0.41 ± 0.15 vs 0.38 ± 0.14, p ≤ 1.00e-04). Conclusion: LARS2 expression is higher in HER2-low vs HER2+ human tumors and in quiescent vs proliferating tumor cells at baseline. Genetic (ERBB2 knockdown) or pharmacologic (T-DXd) HER2 inhibition increases LARS2 expression in vitro in HER2+ but not HER2- cell lines, while LARS2 knockdown reduced proliferation in HER2+ but not HER2- cell lines. These data suggest that LARS2 is specifically upregulated after HER2 inhibition and may promote RD after T-DXd by regulating the quiescence to proliferation switch. Further functional studies will be reported at the conference.
Presentation numberPS2-11-15
Etv4 mediates fgf1-induced metabolic reprogramming in obesity-driven endocrine-resistant breast cancer
Barbara Mensah Sankofi, University of Oklahoma Health Sciences Center, Oklahoma City, OK
B. M. Sankofi, N. S. Thomas, W. L. Berry, E. A. Wellberg; Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK.
Breast cancer remains the most commonly diagnosed malignancy among women globally. Obesity significantly worsens patient outcomes and contributes to therapy resistance, particularly in estrogen receptor-positive (ER-positive) subtypes, which account for approximately 70% of all breast cancer cases. In women with obesity, adult weight gain, characterized by adipose tissue expansion, can drive breast cancer risk and progression. In a preclinical model, we previously showed that weight gain promotes ER-positive tumor growth after endocrine therapy through adipose-derived fibroblast growth factor 1 (FGF1). Inhibition of fibroblast growth factor receptor (FGFR) signaling restored sensitivity of tumors to estrogen deprivation in obese mice. To determine how FGFR signaling promoted endocrine resistance, we treated ER-positive breast cancer cells (MCF7, tamoxifen-resistant MCF7, and UCD12) with FGF1 and performed transcriptomic and metabolic profiling. ETS variant 4 (ETV4), a transcription factor known to regulate estrogen signaling and glycolysis, emerged as the most upregulated gene across multiple ER-positive cell lines. Others have shown that ETV4 regulates breast cancer stemness and metabolic reprogramming, implicating it in disease progression. We hypothesized that ETV4 mediates FGF1-induced progression of ER-positive breast tumors in obesity. ETV4 expression was upregulated in human patient-derived xenograft tumors grown in obese versus lean mice. In invasive human breast cancer specimens, high ETV4 expression predicted a shorter recurrence-free survival for patients with ER-positive tumors. Consistent with these findings, analysis of ER-positive breast tumor data revealed a significant positive correlation between FGF1 and ETV4 expression specifically in patients with obesity, and in treatment-resistant tumors at both baseline and in progressive disease, suggesting a clinically relevant FGF1-ETV4 axis in driving obesity-linked disease progression. Mechanistically, ETV4 knockdown suppressed FGF1-induced proliferation and glycolytic gene expression, while ETV4 overexpression increased glycolysis and cell growth. Transcriptomic profiling revealed that ETV4 regulates key tumor-promoting pathways, including glycolysis, epithelial-mesenchymal transition, hypoxia, estrogen signaling, and inflammation. In vivo, ETV4 overexpression enhanced mammary tumor growth in lean mice with low FGF1 levels, indicating it can drive tumor progression independently of FGF1. Taken together, our data suggest a mechanism by which FGF1 supports breast cancer endocrine therapy resistance in the context of obesity through ETV4 induction and glycolytic metabolic reprogramming. Understanding this process may aid in designing effective treatments, especially for patients resistant to current ER-targeted therapies. Furthermore, ETV4 may serve as both a mechanistic driver and a biomarker of poor outcomes, offering a potential therapeutic target for patients with obesity-driven resistance to endocrine therapies
Presentation numberPS2-11-17
Engineered EVs-mediated miR-222 targeting PTEN/FN1 axis reverses anthracycline resistance in HER2-negative breast cancer
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen1, Q. Shao1, Z. Wang1, B. Zhu1, S. Yang2; 1Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA, 2Department of Breast Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, CHINA.
Anthracycline resistance represents a critical therapeutic challenge in breast cancer treatment, wherein alterations in tumor immune microenvironment and enhanced cellular resistance mechanisms facilitate chemoresistance progression. Transcriptome analysis of 142 HER2-negative breast cancer patients undergoing anthracycline-based chemotherapy revealed four distinct tumor-infiltrating cell subtypes, with subtype D exhibiting elevated M1 macrophage infiltration and superior prognostic outcomes. Differential expression analysis identified miR-222 as the predominantly upregulated microRNA in adriamycin-resistant cells, while Tandem Mass Tag mass spectrometry-based quantitative analysis elucidated PTEN as its direct target and FN1 as a crucial downstream mediator. Engineered extracellular vesicles (EVs) carrying miR-222 inhibitor reversed adriamycin resistance via PTEN/FN1 signaling modulation. Molecular docking analysis found specific PTEN-FN1 protein interactions characterized by stable hydrogen bonds at ARG142-ASP23 and ARG15-GLU95. In xenograft models, EVs-mediated delivery of miR-222 inhibitor significantly attenuated MCF-7/ADR tumor progression through miR-222 suppression and PTEN restoration, with concordant molecular alterations observed in serum derived EVs. Our findings establish a novel mechanism of EVs-mediated drug resistance through the microRNA-222/PTEN/FN1 axis and present engineered EVs as a promising therapeutic strategy for anthracycline resistance in breast cancer, while highlighting circulating EVs profiles as potential treatment monitoring biomarkers.
Presentation numberPS2-11-18
Acetyl coA-fuels lipid storage to support triple negative breast cancer chemoresistance
Katherine E Pendleton, Baylor College of Medicine, Houston, TX
K. E. Pendleton; Breast Center, Baylor College of Medicine, Houston, TX.
Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype in which neoadjuvant chemotherapy (NACT) is the backbone of standard of care. Unfortunately, ~45% of patients have residual tumor burden following NACT, strongly associated with poor prognoses. Our group previously demonstrated that mitochondrial oxidative phosphorylation (oxphos) is upregulated and is a therapeutic vulnerability in chemo-refractory TNBC. We used metabolomic flux tracing to show a heightened contribution of glucose oxidation to the tricarboxylic acid (TCA) cycle in residual human TNBC cells refractory to several conventional chemotherapies (PMID: 36813854). Subsequently, we found significantly elevated abundance of citrate and acetyl-coA (AcCoA) in residual cells, and glucose-derived heavy carbon was more strongly incorporated into both metabolites in residual relative to naïve cells. Both citrate and AcCoA are TCA cycle intermediates and can both contribute to fatty acid synthesis (FAS). Indeed, we observed drastic lipidomic remodeling, largely characterized by elevation of triglycerides, long chain fatty acids, and poly unsaturated fatty acids (PUFAs) in cultured human cells as well as in orthotopic PDX tumors following chemotherapy relative to their treatment naïve counterparts. This was accompanied by significant increase in the number of lipid droplets (LDs) in residual cells. Our analyses of publicly available human TNBC proteomic and transcriptomic data (PMID: 36001024) affirmed the significant association of fatty acid metabolism with TNBC chemoresistance, as well as its upregulation in treated tumors relative to their treatment naïve counterparts. Mining those data led us to ATP citrate lyase (ACLY), the rate limiting enzyme for cytosolic AcCoA production from citrate. Given these data, we hypothesized that ACLY converts excess citrate, generated by heightened mitochondrial respiration, to AcCoA to promote lipid production and storage to support TNBC chemoresistance. Western blotting revealed protein levels of ACLY and its activating phosphorylation mark Ser455 increased in human TNBC cells surviving treatment with doxorubicin and carboplatin. ACLY knockdown or inhibition with BMS-303141 potently reduced chemotherapy-induced accumulation of AcCoA and LDs and elicited substantial lipidomic changes consistent with increased PUFAs. Cell growth experiments examining TNBC cells treated with conventional NACT agents and ACLY inhibition or knockout showed significantly impaired growth in combination-treated cells compared to cells given either of the single agents. These data indicate that NACT can cause accumulation of AcCoA, heightened LDs, and lipidomic rewiring through ACLY. ACLY is a novel functional dependence of chemo-refractory TNBC cells and may represent a useful therapeutic target. Notably, perturbation of ACLY in nontumor mammary epithelial cells did not alter ACLY expression, ACLY phosphorylation at Ser455, or increase LDs and mice treated with BMS-303141 exhibited no overt signs of toxicity. We posit that TNBC cells adapt to the stress of NACT by upregulating lipid synthesis and storage in conjunction with glucose catabolism, enhancing metabolic flexibility and cell survival.
Presentation numberPS2-11-19
Intratumoral Lactobacillus iners Drives Chemoresistance in Breast Cancer: A Ferroptosis-Evasion Mechanism via NSUN2 Lactylation
Yuanxiang Lu, Henan provincial people’s Hospital, Zhengzhou, China
Y. Lu, S. Pilei; Breast department, Henan provincial people’s Hospital, Zhengzhou, CHINA.
Abstract Chemoresistance remains a significant obstacle in breast cancer therapy, while the role of intratumoral microbiota in this process has yet to be fully elucidated. Here, we identify Lactobacillus iners (L. iners) as a critical microbial factor contributing to resistance against standard anthracycline-taxane-based neoadjuvant chemotherapy. Through integrated multi-omics analyses of longitudinal patient cohorts and 4T1 murine models, we demonstrate that L. iners promotes chemoresistance by reprogramming tumor GSH metabolism and inhibiting ferroptosis. Mechanistically, L. iners-derived L-lactate induces lysine lactylation of the RNA methyltransferase NSUN2, thereby enhancing m⁵C modification and post-transcriptional stabilization of glutathione synthetase (GSS) mRNA. This subsequently upregulates GSS, augments GSH biosynthesis, and reinforces redox homeostasis, ultimately protecting tumor cells from ferroptotic cell death. Clinically, high intratumoral abundance of L. iners correlates with diminished treatment response and shorter recurrence-free survival. Notably, selective depletion of L. iners using a tumor-targeted antibiotic nanoplatform restores chemosensitivity, suppresses NSUN2 lactylation, and reactivates ferroptosis without disrupting systemic microbiota homeostasis. Collectively, these findings uncover a microbiota-NSUN2 lactylation-GSS regulatory axis underlying chemotherapy resistance and provide a translational strategy for microbial-targeted therapy in breast cancer.
Presentation numberPS2-11-20
Recurrent ciliary alterations under neoadjuvant selection are associated with inferior breast cancer outcomes
Julia Dory Ransohoff, Stanford University, Palo Alto, CA
J. D. Ransohoff, M. Carleton, S. Miron Barroso, N. Phillips, J. Demeter, J. Goldstein, A. Maltos, M. Khodadoust, J. Ford, A. Kurian, G. Bean, M. Diehn, P. Jackson, M. Telli, D. Kurtz, A. Alizadeh; Medicine, Stanford University, Palo Alto, CA.
Introduction: Failure to achieve a pathologic complete response (pCR) to neoadjuvant treatment informs risk-adapted adjuvant therapies, which reduce recurrence risk and improve outcomes. However, pCR-based assessments are limited given most patients with residual disease do not relapse and tissue visualization cannot define molecular residual disease (MRD). While liquid biopsies can define MRD, circulating tumor DNA is undetectable in most patients with pathologic residual disease, even with the most sensitive technologies. Methods: Motivated by the critical need to define which patients with residual disease are at risk of relapse and characterize their post-neoadjuvant genomic landscapes, we developed a novel, personalized, ultra-sensitive, tissue-based strategy to define MRD. We characterized the resection tissues of 29 diverse breast cancers using a joint tumor-informed and tumor-naive approach, capturing both persistent and emergent lesions. To interrogate genomic alterations under neoadjuvant selection at the level of genes and cytobands, we derived a statistical framework ranking each lesion’s position in a clonal hierarchy based on relative pre- and post-neoadjuvant abundance and associated alterations with selection and survival. We focused on recurrent ciliary alterations and evaluated their enrichment and survival associations, which we validated in external breast cancer datasets. Results: We identified recurrent alterations in genes and pathways involved in cell migration, adhesion, and tumor invasion under selection. Across the cohort, we also identified alterations in ciliary genes with diverse roles, including intraflagellar transport (IFT88), ciliary vesicle recruitment (CEP89), microtubule stabilization (CSPP1), and structural components of the ciliary base (EHD1), distal appendage/centriole (CEP350), and dynein arm (DNAAF4, DNAH14, DYNC2H1). We found enrichment of a broader panel of ciliary alterations in the post-neoadjuvant tissues of relapsing patients and that these lesions were under selection. In public breast cancer datasets, we validated ciliary alterations as under neoadjuvant selection and associated with relapse (Table 1). Conclusions: Loss of the cilium may not only be an early breast cancer event, but also potentially a novel mechanism of taxane chemoresistance. We hypothesize that cilia, cellular antennae that are the primordial remnants of motile flagella, promote physical dispersion and metastatic potential of invading tumor cells through aberrant cellular signaling, polarity, and mechanosensation. Because hedgehog and PI3K pathway signaling rely on cilia, adjuvant targeting of these pathways in tumors bearing ciliary lesions may represent a novel approach to eradicate micrometastatic disease in the post-neoadjuvant window. We suggest the need for functional study of ciliary alterations in breast tumors.
| Study cohort: Association of ciliary lesions with relapse | Ciliary alteration (N [# alterations]) | Non-ciliary alteration (N) | P-value (Chi-square) | ||||
| Relapse | 70 | 647 | < 0.00001 | ||||
| No relapse | 42 | 1045 | |||||
| Study cohort: Association of ciliary lesions with selection | Ciliary alteration (N [# alterations]) | Non-ciliary alteration (N) | P-value (Chi-square) | ||||
| Selected | 58 | 68 | < 0.00001 | ||||
| Non-selected | 53 | 924 | |||||
| External cohort: Association of ciliary lesions with relapse | Ciliary alteration (N [# patients with alteration]) | Non-ciliary alteration (N) | P-value (Chi-square) | ||||
| Inferior relapse-free survival | 125 | 48 | < 0.00001 | ||||
| Non-inferior relapse-free survival | 631 | 755 | |||||
| External cohort: Association of ciliary lesions with selection and relapse | Selected (N [# patients with alteration]) | Non-selected (N) | P-value (Chi-square) | ||||
| Inferior relapse-free survival | 36 | 84 | < 0.00001 | ||||
| Non-inferior relapse-free survival | 46 | 540 |
Presentation numberPS2-11-21
The mechanism research on polyamine targeting ZNF133-L1CAM axis to suppress malignant progression of breast cancer
chunhua xiao, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Medical, tianjin, China
c. xiao, l. Li, y. Tu, y. Zhu; breast oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer Medical, tianjin, CHINA.
Polyamines, which have been found to accumulate in several kinds of body samples in breast cancer patients, are closely related to malignant progression of breast cancer and chemotherapy resistance. However, the downstream molecular mechanism of polyamines is still unclear. We have previously reported that ZNF133 is associated with a favorable prognosis for breast cancer; ZNF133 can inhibit the transcriptional activity of L1CAM gene promoter, inhibit the proliferation and invasion of breast cancer cells in vitro, and inhibit the growth and metastasis of breast cancer in vivo. Polyamines can reduce the level of ZNF133 protein and weaken the inhibitory effect of ZNF133 on the proliferation and invasion of breast cancer cells. Polyamine synthesis inhibitors can increase the level of ZNF133 protein and promote its transcriptional inhibition of L1CAM, thus inhibiting the proliferation and invasion of breast cancer cells. Therefore, we propose a scientific hypothesis: polyamines promote the transcription of L1CAM by inhibiting ZNF133, thus promoting the malignant progression of breast cancer. This project intends to further elaborate the molecular mechanism of polyamine regulation of ZNF133 protein level in breast tumor cells and ZNF133 inhibition of L1CAM transcription, and verify the effect of polyamine metabolism on regulation of ZNF133-L1CAM axis in the malignant progression of breast cancer, providing a new target for the diagnosis and treatment of breast cancer.
Presentation numberPS2-11-22
Understanding resistance to the SMAC mimetic birinapant in triple-negative breast cancer
Elisabeth Brown, University of Utah, Salt Lake City, UT
E. Brown, C. Yang, E. Cortes-Sanchez, Z. Chu, A. Welm; Oncological Sciences, University of Utah, Salt Lake City, UT.
Title: Understanding resistance to the SMAC mimetic birinapant in triple-negative breast cancer Background: Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with limited targeted therapies and high metastatic potential. Thus, it is crucial to identify new therapies for TNBC patients in both the primary and metastatic settings. SMAC (Secondary Mitochondrial Activator of Caspases) mimetics represent a potential new therapeutic avenue for TNBC patients. Previously, we found that approximately twenty-five percent of our TNBC patient-derived xenograft (PDX) and xenograft-derived organoid (PDxO) models are exquisitely sensitive to the SMAC mimetic birinapant; response profiles also remained consistent across multiple SMAC mimetics. However, the majority of our TNBC patient-derived models are resistant to birinapant through unknown means. Despite promising pre-clinical results in several cancers, SMAC mimetics have not reached FDA approval. This may be explained by a dichotomy of patient responses, as observed in our birinapant-treated preclinical models, and highlights the need to understand the mechanism of resistance to SMAC mimetics to better stratify patients for treatment. The goal of this work is to uncover this mechanism of resistance and identify biomarkers of response and co-treatment strategies to sensitize more TNBC patients to SMAC mimetics. Results: SMAC mimetics, such as birinapant, function by targeting and degrading Inhibitor of Apoptosis (IAP) proteins in the Tumor Necrosis Factor Receptor (TNFR) and intrinsic apoptosis pathways, leading to apoptosis signaling and tumor cell death. We found that both birinapant-sensitive (BS) and –resistant (BR) TNBC PDxOs (n=2 lines each) show uniform IAP degradation after birinapant treatment and have similar levels of apoptotic proteins. Therefore, disrupted apoptotic signaling downstream of IAP degradation is likely the culprit of birinapant resistance. To interrogate this further, we tested the functionality of multiple cell death pathways, including intrinsic, TRAIL-mediated and TNFR-mediated apoptosis, as well as TNFR-mediated necroptosis. BR-PDxOs remained resistant to cell death exclusively in TNFR-mediated pathways, whereas BS-PDxOs died via all treatments, indicating a modality of resistance specific to TNFR signaling. Conclusions: Using PDxOs, inability to execute TNFR-mediated apoptosis signaling was identified as a mechanism of birinapant-resistance in TNBC. This regulation protects TNBC tumors from birinapant-induced cell death and is likely protective against all SMAC mimetics. These data suggest that identifying ways to overcome the failure to execute TNFR-mediated apoptosis signaling may expand the patient population that will benefit from birinapant treatment.
Presentation numberPS2-11-23
Targeting APT-2 against therapy resistance in Breast Cancer
Kanjoormana Aryan Manu, Amala Cancer Research Centre, Thrissur, India
A. Jose K, A. Mariya Davis, S. Prabha, K. Dhanura, K. Manu; Immunology, Amala Cancer Research Centre, Thrissur, INDIA.
Breast cancer (BC) is the most dangerous cancers among women. Therapy resistance to thechemotherapy is the most concerning for the breast cancer management.Protein S-palmitoylation is a post-translational reversible modification of some specific proteins whichhelps them to attach to the cell membrane. Depalmitoylation reverses palmitoylation and is catalyzed byenzymes belonging to the a/b hydrolase family of serine hydrolases called acyl protein thioesterases 1/2(APT 1/2). SCRIB, STAT3, TNFR1, ZDHHC6, and MC1R are the known targets of APT-2 and many of themhave established role in cancer progression.APT-2 inhibitor ML349 inhibited soft agar colony formation, 2D clonogenicity of breast cancer cells. ML349 treatment sensitized breast cancer cells toward Doxorubicin both in vitro and invivo. Both treatment with ML349 and siRNA knock down of APT-2 showed suppression of cell cycle regulators, cancer stem cell markers, YAP/TAZ target genes in breast cancer cells. Patient sample data from TCGA shows that lower expression of SCRIB is correlated with increased Overall and Progression free survival of Breast Invasive Ductal Carcinoma, in doxorubicin treated patients. In conclusion, APT-2 inhibition may suppress cancer stemness and therapy resistance in breast cancer cells.
Presentation numberPS2-11-25
Paracrine FGFR activation by stromal signals and its association with immune infiltration in breast cancer
Lise Mangiante, School of Medicine, Stanford University, Stanford, CA
L. Mangiante1, Z. Ma1, C. Weiss1, M. Shaw1, B. Simon2, S. Mouron3, G. Bean4, M. Quintela-Fandino3, C. Curtis1, J. Caswell-Jin5; 1Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, CA, 2Cancer Biology, School of Medicine, Stanford University, Stanford, CA, 3Clinical Research, Centro Nacional de Investigaciones Oncológicas, Madrid, SPAIN, 4Pathology Clinical, School of Medicine, Stanford University, Stanford, CA, 5Department of Medicine (Oncology), Stanford University, School of Medicine, Stanford, CA.
Background: Fibroblast growth factor receptor (FGFR) alterations occur in up to 20% of estrogen receptor-positive (ER+) breast cancers and are associated with endocrine therapy resistance and poor prognosis. These tumors often classify as high-risk integrative cluster subtypes, which account for approximately one-quarter of ER+ tumors, are characterized by complex genomic rearrangements, and are associated with increased and persistent risk of relapse. In prior work, we found that high-risk integrative subtypes of ER+ breast cancer were significantly more likely to exhibit immune depletion compared to typical-risk subtypes, an observation especially pronounced among tumors with putative FGFR pathway alterations. Despite the clinical rationale, therapeutic efforts to directly target FGFR signaling in breast cancer have largely not been successful. To better understand the role of FGFR in shaping the tumor microenvironment and to identify potential therapeutic vulnerabilities, we employed multiplex single-molecule RNA imaging on the Bruker CosMx platform to profile stromal interactions and immune contexture in primary and metastatic ER+ breast tumors. Methods: We constructed a single-cell spatially resolved transcriptomic meta-cohort of ER+ breast cancer, consisting of primary pre-treatment tumors (208 primaries), 69 patients treated per standard of care at Stanford University (27 primaries, 66 metastatic sites), and 4 patients treated on an investigator-sponsored trial of the FGFR inhibitor infigratinib (3 primaries, 6 metastatic sites). We deployed state-of-the-art machine learning algorithms for cell segmentation (CellPose), cell type classification (singleR), and inference of intercellular communication (CellChat). We identified stromal-tumor interactions enriched in high vs. typical-risk primary tumors and compared the microenvironmental features associated with those interactions. Results: We identified recurrent interactions between tumor-expressed FGFR1 and fibroblast-derived FGF1 or FGF7, significantly enriched in high-risk (17/65) compared to typical-risk tumors (2/87, P<0.001). Tumors exhibiting this interaction, designated FGF/FGFR1+, demonstrated increased epithelial tumor cell density (P=1.3×10-6) and reduced lymphocyte density (P=0.002). FGFR1 amplification was present in 10 of the 19 FGF/FGFR1+ tumors. In mixed linear models, FGF/FGFR1+ status was a stronger predictor than FGFR1 amplification status of lymphocyte depletion. Within the lymphocyte populations, FGF/FGFR1+ tumors demonstrated an increased ratio of regulatory T-cells relative to B-cells, plasma cells, NK cells, and CD8+ T-cells (P<0.001), indicating that lymphocyte density was not only lower but its composition shifted to a pro-tumoral phenotype. Co-localization analysis revealed that immune cells in FGF/FGFR1+ tumors were more likely to cluster with each other and less likely to colocalize with tumor cells, suggesting immune exclusion in addition to lower lymphocyte content. Notably, the FGF/FGFR1 interaction was not restricted to the breast tissue microenvironment and its resident fibroblasts: it was observed in 12 metastatic samples, including 5 of 8 primary FGF/FGFR1+ tumors with matched metastases. Conclusion: We identified a recurrent interaction between tumor-expressed FGFR1 and fibroblast-derived FGF ligands that is enriched in high-risk ER+ breast cancers and associated with an immunosuppressive, immune-excluded microenvironment. Our results indicate that tumor-stromal crosstalk can shape the immune landscape to promote tumor progression and immune evasion. The FGF/FGFR1 interaction represents a potentially targetable axis, with implications for therapeutic strategies that aim to reprogram the tumor microenvironment in high-risk ER+ breast cancer.
Presentation numberPS2-11-27
Whole-genome doubling promotes immune evasion in TNBC by epigenetically silencing antigen presentation pathways through PRC2 activity.
Pierre Foidart, Dana-Farber Cancer Institute, Boston, MA
P. Foidart1, Z. Li1, X. Can1, M. Seehawer1, E. Rojas-Jimenez1, P. Baldominos2, J. Nishida1, T. Bui1, B. Diciaccio1, S. Parvin1, M. Goyette1, P. Yan1, X. Qiu3, R. Li3, Y. Jiang3, Y. Xie3, X. Huang1, L. E. Stevens1, P. Cejas3, L. Mangiante4, C. I. Sotomayor Vivas4, K. Houlahan4, T. Scales5, I. S. Harris5, C. Curtis4, A. G. Letai1, H. W. Long3, J. Agudo2, K. Polyak1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 3Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 4Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 5Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY.
Whole-genome doubling (WGD) is a frequent yet poorly understood genomic event linked to adverse clinical outcomes. Multiple distinct mechanisms can lead to tetraploidization and aneuploidy, including defects in the mitotic checkpoint, chromosome cohesion, and disruptions of microtubules, spindle assembly, or centromere function. In addition, cell-cell fusion events can also result in polyploidization. Polyploid cells often exhibit chromosomal instability, which contributes to increased intratumoral heterogeneity and accelerates the evolution of the cancer genome. Although chromosomal instability and the presence of tetraploid or hyperdiploid cancer cells have been linked to treatment resistance and enhanced phenotypic diversity, the molecular mechanisms driving these associations remain poorly understood. We began by assessing the frequency of WGD in breast cancer across tumor subtypes and stages of disease progression. To do this, we performed WGD calling or reanalyzed publicly available genomic sequencing datasets with annotated WGD status. Analysis of three datasets revealed that WGD occurs more frequently in HER2+ and triple-negative breast cancers (TNBC) compared to hormone receptor-positive tumors. Furthermore, WGD frequency was consistently higher in distant metastases than in primary tumors. We then established through homofusion several murine mammary tumor models of WGD of TNBC to investigate how WGD drives tumor evolution. We observed that WGD increases transcriptomic and epigenetic heterogeneity and identified the survivin inhibitor YM155 as a selective suppressor of WGD+ tumors. Importantly, WGD promoted immune evasion by enabling escape from CD8⁺ T cell-mediated immune responses, rendering WGD+ tumors more sensitive to anti-PD-L1 therapy. Through single-cell profiling, we found that WGD+ cancer cells exhibit impaired antigen presentation, driven in part by a reduced response to IFN-γ and epigenetic silencing of MHC class I transcriptional regulators via the PRC2 complex. Treatment with a PRC2 inhibitor preferentially inhibited WGD+ tumor growth, restored antigen presentation, and enhanced CD8⁺ T cell infiltration. These findings suggest that combined inhibition of PRC2 and PD-1 represents a promising therapeutic approach for WGD+ breast cancers. Keywords: Whole genome doubling, Breast Cancer, single cell profiling, immune escape
Presentation numberPS2-11-28
Gdc-4198, a next-generation CDK4/2 inhibitor, induces durable cell cycle arrest and overcomes CDK2-driven adaptation to CDK4 inhibition
Marc Hafner, Genentech, South San Francisco, CA
M. Hafner1, S. Vartanian2, G. Luca1, N. Kosaisawe3, M. Hwang4, E. Lin2, L. Wang4, J. Oeh5, J. Vijay6, K. N. Islam7, A. Zheng7, K. Samy8, U. Segal5, J. Moffat3, D. Zingg2, A. Collier4, I. Sanidas7, J. Xie9, S. A. Wander7; 1gRED Computational Sciences, Genentech, South San Francisco, CA, 2Discovery Oncology, Genentech, South San Francisco, CA, 3Biochemical Cellular Pharmacology, Genentech, South San Francisco, CA, 4Translational Medicine, Genentech, South San Francisco, CA, 5Translational Oncology, Genentech, South San Francisco, CA, 6Data & Analytics, Roche Informatics, F. Hoffmann-La Roche Ltd., Mississauga, ON, CANADA, 7Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, 8Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, CA, 9Oncology and Immunology, Regor Therapeutics Group, Cambridge, MA.
While CDK4/6 inhibitors (CDK4/6i) have changed the therapeutic landscape of hormone receptor-positive (HR+) breast cancer, resistance to these therapies is a major challenge limiting their clinical benefit. Recent studies have shown that CDK2 activity is a key mechanism of resistance to CDK4/6i. Adaptation to CDK4 inhibition can arise from the overexpression or amplification of cyclin E1 or E2, leading to increased CDK2 activity that phosphorylates Rb and bypasses CDK4/6i-induced G1 arrest. Additional alterations, such as p53 loss of function, can activate CDK2. It has been proposed that targeting CDK2, in addition to CDK4, can lead to more durable cell cycle arrest in HR+ cancer cells. In this study, we evaluated the preclinical activity of GDC-4198, a next-generation CDK4/2 inhibitor currently being tested in clinical trials, and its potential to address CDK2-driven resistance to CDK4 inhibition. In biochemical kinase activity assays, GDC-4198 has sub-nanomolar potency against CDK4 and is a more potent inhibitor of CDK2 than CDK6/CycD3, unlike approved CDK4/6i. Immunofluorescence data from HR+ breast cancer cells revealed that the shift in cell cycle distribution induced by GDC-4198 was differentiated from the effects of CDK4/6i and can most closely be reproduced by a combination of atirmociclib (a CDK4 inhibitor) and tagtociclib (a CDK2 inhibitor). This observation was most pronounced in cell line models that have developed resistance to CDK4/6i. RNA-seq data collected after 96 hours of treatment confirmed that the effects of GDC-4198 on gene expression are differentiated from CDK4 inhibition alone. In cell viability assays, GDC-4198 induced growth inhibition and sustained cell cycle arrest in a diverse panel of HR+ breast cancer cell lines. Notably, time-course assays demonstrated that parental T-47D cells can reenter the cell cycle within two days after initial arrest under treatment with CDK4/6i, whereas GDC-4198 led to sustained growth inhibition over five days. Across CDK4/6i resistant HR+ cells, engineered via either long-term exposure to palbociclib or CCNE1/2 overexpression, GDC-4198 was substantially more effective at inhibiting growth than approved CDK4/6i or atirmociclib. Notably, the activity of GDC-4198 was comparable to the combination of CDK4 and CDK2 inhibitors. Additional studies in patient-derived cell lines obtained in the setting of metastatic HR+ breast cancer following progression on letrozole/ribociclib, as well as in patient-derived cell lines with acquired in vitro resistance to palbociclib, confirmed the enhanced antitumor activity of GDC-4198 compared to current CDK4/6i or atirmociclib. In xenograft studies of HR+ breast cancer, GDC-4198 demonstrated dose-dependent tumor growth inhibition. Pharmacokinetic and pharmacodynamic analyses indicated favorable drug exposure and pharmacodynamic modulation in tumor tissues consistent with the effects observed in in vitro experiments. Taken together, these preclinical findings demonstrate that GDC-4198 can induce more durable cell cycle arrest than approved CDK4/6i by overcoming CDK2-driven adaptive and intrinsic resistance to CDK4 inhibition. These results suggest that GDC-4198 is a promising investigational agent with the potential to provide benefit to those who have progressed on approved CDK4/6i while prolonging benefit in earlier disease settings.
Presentation numberPS2-11-29
Deciphering the tumor microenvironment related to immune regulation in triple-negative breast cancer
Yoon Jin Cha, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, Korea, Republic of
Y. Cha1, S. Bae2, J. Kim1, K. Kim3, Y. Kook2, A. Kim2, S. Ahn2, J. Jeon2; 1Pathology, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Surgery, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 3Institute for Breast Cancer Precision Medicine, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF.
Background: Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive subtype of breast cancer, with the features of tumor microenvironment and the underlying molecular mechanisms of immune-regulation remaining largely elusive. This study aimed to explore the immune landscape of TNBC by assessing tumor-infiltrating lymphocytes (TILs) level, programmed cell death ligand 1 (PD-L1) expression, and the degree of TIL infiltration within the tumor. Methods: Utilizing the GeoMX Digital Spatial Profiler platform, we also performed spatially resolved transcriptomic profiling of tumor, immune, and stromal cells, focusing on the distinctions between immune-activated and immune-suppressed phenotypes. Results: Immune-activated phenotypes, characterized by high TILs, PD-L1 positivity, and close proximity between tumor cells and TILs, were found to correlate with the basal-like immune-activated subtype based on transcriptomic alterations derived solely from tumor cells. These phenotypes were also associated with the upregulation of known immune-related pathways. In contrast, immune-suppressed phenotypes were linked to pathways involved in tumor progression, such as epithelial-mesenchymal transition, TGF-beta signaling, and angiogenesis. Notably, our transcriptomic analysis suggests that tumor cells play a more dominant role than immune cells in shaping the immune phenotype, as indicated by the greater number of differentially expressed genes in tumor cells rather than immune cells. Stratifying patients based on TILs and PD-L1 status revealed that the TIL+PD-L1+ subtype exhibited the most favorable prognosis, a finding further supported by TIL+PD-L1+ signature with excellent survival outcomes in public gene datasets. Conversely, the TIL-PD-L1- subtype displayed characteristics of an “cold” tumor, potentially driven by desmoplastic changes within the tumor microenvironment. Interestingly, the TIL+PD-L1- subtype, despite a high TILs level, was associated with poorer prognosis, likely due to a higher proportion of myeloid cells, decreased activity of immune cells, and increased activation of the adipogenesis pathway across cell types. This finding underscores that in the TIL+PD-L1- subtype, immune cell activity remains low even when TILs infiltrate the intra-tumoral region, highlighting PD-L1 as a critical marker reflecting the immune system status in patients with high TILs. Conclusion: In summary, this study emphasizes the clinical significance of integrating TILs and PD-L1 expression as prognostic biomarkers in TNBC and underscores the central role of tumor cells in dictating immune phenotypes. Furthermore, our findings suggest potential underlying mechanisms and target genes regarding the immune system regulation in TNBC. Based on these insights, future research is warranted to develop targeted therapies, especially for immune-suppressed subtypes, aiming to improve clinical outcomes for patients with TNBC.
Presentation numberPS2-11-30
Functional analysis of gedatolisib combined with fulvestrant and/or palbociclib in breast cancer cell models adapted to estrogen receptor and/or CDK4/6 inhibitors
Stefano Rossetti, Celcuity Inc., Minneapolis, MN
S. Rossetti1, A. Broege1, L. Davis1, M. Seibel1, S. Stokke1, J. Molden1, I. Gorbatchevsky2, B. Sullivan3, L. Laing1; 1R&D, Celcuity Inc., Minneapolis, MN, 2Clinical Development, Celcuity Inc., Minneapolis, MN, 3G&A, Celcuity Inc., Minneapolis, MN.
Background: The estrogen receptor (ER), cyclin-dependent kinase (CDK), and PI3K-AKT-mTOR (PAM) pathways are interconnected and drive hormone receptor (HR)+/HER2- advanced breast cancer (ABC). In the first line setting, endocrine therapy (ET) combined with a CDK4/6 inhibitor is the standard of care regimen for most patients with HR+/HER2- ABC. For patients in the second line setting, several inhibitors targeting components of the PAM pathway (e.g. PI3Kα, AKT, mTORC1) are approved in combination with ET. By only addressing a single target of the PAM pathway, the currently approved PI3Kα, AKT, mTORC1 inhibitors do not prevent adaptive resistance induced by the other uninhibited PAM targets. In a phase 1b clinical trial, gedatolisib, a multi-target PAM inhibitor, combined with ET and palbociclib showed promising efficacy and safety in patients with HR+/HER2- ABC, regardless of PIK3CA mutation status. Here we investigated the effects of gedatolisib and single-target PI3Kα and AKT inhibitors in combination with fulvestrant and/or palbociclib in breast cancer (BC) cell models with and without PAM pathway mutations and with varying levels of sensitivity to fulvestrant and palbociclib. Methods: Gedatolisib (pan-PI3K, mTORC1/2 inhibitor), inavolisib (PI3Kα inhibitor), capivasertib (AKT inhibitor) were tested as single agents or in combination with fulvestrant and/or palbociclib in treatment-naïve BC cell lines with or without PIK3CA mutations as well as in BC cell lines adapted to long-term treatment with fulvestrant and/or palbociclib. Cell viability, growth rate, cell cycle, DNA replication, apoptosis, and cell death were evaluated by luciferase-based and flow cytometry-based functional assays. Results: Gedatolisib reduced growth rates of treatment-naïve or fulvestrant/palbociclib-adapted BC cell lines, regardless of PIK3CA mutational status, and the addition of fulvestrant and/or palbociclib increased growth-inhibitory effects. From a functional standpoint, the gedatolisib/palbociclib/fulvestrant triplet inhibited DNA replication and/or induced apoptotic cell death more effectively than treatment with single drugs or drug doublets. The gedatolisib/palbociclib/fulvestrant triplet was more effective than PI3Kα and AKT inhibitors (inavolisib, capivasertib) combined with palbociclib and/or fulvestrant in the BC cell lines tested, regardless of PIK3CA mutational status or previous adaptation to palbociclib and/or fulvestrant. Conclusions: These results indicate that gedatolisib plus fulvestrant, with and without palbociclib, effectively controls the growth of treatment-naïve BC cells as well as BC cells adapted to palbociclib/fulvestrant treatment. Moreover, in contrast to the currently approved PI3Kα and AKT inhibitors, the gedatolisib/palbociclib/fulvestrant triplet is effective in BC cells with or without PIK3CA mutations. The combination of gedatolisib with ET and a CDK4/6 inhibitor is being evaluated in two ongoing Phase 3 clinical trials as first-line (VIKTORIA-2) or second-line (VIKTORIA-1) treatment of HR+/HER2- ABC with or without PIK3CA mutations. The non-clinical results presented here provide a strong mechanistic rationale for these clinical studies.
Presentation numberPS2-12-01
Washout Efficiency in Cancer Drug Development: A Strategy to Minimize Toxicity
Mamoru Takada, Chiba University, Chiba, Japan
M. Takada1, M. Yu1, Y. Chen2, M. Otsuka1, A. Suzuki2; 1Medicine, Chiba University, Chiba, JAPAN, 2Oncology, Wisconsin University, Madison, WI.
Introduction Cytotoxic anticancer drugs, such as microtubule-targeting agents (MTAs), are widely used in cancer treatment due to their ability to inhibit cell division. However, these drugs often cause significant side effects because they lack specificity for cancer cells, leading to toxicity in normal tissues. This challenge limits the therapeutic potential of MTAs. Enhancing drug selectivity for cancer cells while minimizing toxicity to normal cells is crucial. One promising strategy to achieve this is improving washout efficiency, where cancer cells retain the drug’s effects longer than normal cells. CMPD1, a dual-target inhibitor that disrupts the p38-MK2 signaling pathway and microtubule dynamics, has shown potential to selectively target cancer cells, offering a novel approach to improve MTA efficacy with reduced toxicity. Methods To evaluate the efficacy and safety of CMPD1, we conducted a series of in vitro and in vivo experiments. In vitro, we used live-cell imaging, cell cycle analysis, and apoptosis assays to compare the effects of CMPD1 on several breast cancer cell lines (MDA-MB-231, CAL-51) and normal cell lines (MCF10A, RPE1). We specifically assessed how CMPD1 impacts mitotic progression and cell viability in cancer cells versus normal cells. Additionally, we performed washout assays to study the difference in drug retention and the subsequent effects on cell cycle progression between cancer and normal cells. In vivo experiments involved xenograft mouse models of breast cancer where we evaluated tumor growth, weight loss, and organ toxicity, comparing the effects of CMPD1 with those of the standard chemotherapeutic agent, Taxol. Blood markers and histological analysis of liver and kidney tissues were performed to assess any adverse effects caused by treatment. Results CMPD1 demonstrated a striking ability to preferentially induce mitotic defects in cancer cells while sparing normal cells. This selective activity was further confirmed through washout assays, where CMPD1-treated cancer cells exhibited prolonged mitotic arrest even after the drug was removed, whereas normal cells quickly resumed mitosis following drug washout. This differential washout efficiency between cancer and normal cells underpinned CMPD1’s enhanced specificity for cancer cells. In animal models, CMPD1 effectively suppressed tumor growth at doses much lower than those required for Taxol, with minimal adverse effects on the mice. Notably, CMPD1-treated animals did not experience significant weight loss, and no liver or kidney damage was observed, contrasting sharply with the Taxol group, which showed marked organ damage and significant weight loss. Blood analysis revealed that CMPD1 treatment did not induce a significant drop in white blood cell count, a common side effect seen with Taxol. These findings suggest that CMPD1’s superior washout efficiency may contribute to its ability to selectively target cancer cells, while minimizing systemic toxicity and adverse side effects. Conclusion CMPD1’s differential washout efficiency in cancer versus normal cells enhances its specificity and reduces off-target toxicity. This mechanism makes CMPD1 a promising candidate for cancer therapy, offering potent efficacy with minimal side effects. These findings suggest that improving washout efficiency could be a key strategy in developing safer and more effective anticancer drugs.
Presentation numberPS2-12-02
An Innovative Postbiotic Product Based on Sequential Fermentation of L.paracasei NPB01 and L.rhamnosus GG Demonstrates Antitumor Activity against Breast Cancer
Roberto Berni Canani, University of Naples “Federico II”, Naples, Italy
R. Berni Canani1, C. Luongo1, L. Pisapia2, R. Di Santillo1, A. Gaeta2, C. Scocco1, M. Michelini1, V. Mauriello1, A. Cadavere1, C. Messuri1, S. Chumsri3, D. Mussallem3; 1Department of Translational Medical Science, University of Naples “Federico II”, Naples, ITALY, 2Institute of Genetics and Biophysics, Italian National Council of Research, Naples, ITALY, 3Mayo Clinic Breast Disease Group, Mayo Clinic Cancer Center, South Jacksonville, FL.
Backgroud. Breast cancer (BC) is the most common and deadly cancer affecting women globally. While traditional treatments are used, factors limit their effectiveness, such as tumor heterogeneity, drug resistance, and non-targeted actions on cancer cells. Emerging evidence highlights the role of gut microbiota in modulating breast cancer risk, treatment response, and recurrence through various mechanisms, including the production of biologically active compounds and metabolites. These non-viable microbial products and metabolic byproducts from probiotic fermentation, known as postbiotics, exert beneficial effects on the host without the need for live microorganisms. Postbiotics derived from Lacticaseibacillus species represent a novel class of therapeutics with potential anti-cancer properties. In this study, we investigated the therapeutic effects of an innovative postbiotic product (iPB), developed through sequential fermentation of L.paracasei NPB01 and L.rhamnosus GG, in BC experimental models. Methods. The following cell lines were used to represent main BC subtypes: MCF7 cells (HR+/HER2-), MDA-MB-231 (triple negative breast cancer), SKBR3 (HER2+). MCF-10A (normal breast epithelial cells) and Caco-2 (enterocytes) cell lines were used as control. Cell viability was assessed by colony formation assay. Apoptosis was evaluated using Annexin V and 7-AAD staining and HLA class I surface expression was quantified by flow cytometry. Wound healing assay was applied to investigate the inhibitory effect of iPB on cell migration. Results. The iPB exposure resulted in >50% colony formation reduction in MCF7, MDA-MB-231 and SKBR3 cells starting from 0.5 mg/ml for 72 h. Furthermore, the iPB significantly increased early apoptotic cell rate in MCF7 cells starting at 0.5 mg/ml for 96 h. A similar trend was observed in MDA-MB231 and SKBR3 cells exposed to 5 mg/ml for 96 h iPB, as well as significant increase in late apoptotic cells detected by 7-AAD. On the contrary, control cells (MCF-10A, Caco-2) viability remained unaffected by the iPB exposure up to 5 mg/ml for 96 h. Evaluation of HLA class I surface expression revealed up to 30% increase in HLA class I expression in MCF7 cells induced by iPB starting at 0.5 mg/ml for 96 h, MDA-MB-231 cells at 1 mg/ml for 72 h, and SKBR3 cells at 5 mg/ml for 72 h, but there was no significant changes in MHC class I expression in control cells. Finally, the results of wound healing analysis revealed that iPB induced an up to 30% reduction in cell migration in all BC cell models, compared to the untreated cells where it induced a full wound closure.Conclusions. The iPB product reduces cell viability and cell migration in all main BC subtypes. These effects parallel with HLA class I surface expression increase in all BC cell phenotypes, potentially enhancing immunogenicity. These findings suggest that this iPB product may represent a promising novel agent in BC treatment and prevention.
Presentation numberPS2-12-04
Promoter Hypermethylation-Induced Silencing of FXYD1 Drives Breast Cancer Metastasis via DDX5-Mediated Wnt/β-Catenin Pathway Activation
Ping Wen, Chongqing University Cancer Hospital, Chongqing, China
P. Wen, G. Wang, F. Qu, Q. Shao, L. Wang, N. Zhang, X. Zeng; Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, CHINA.
Background: Epigenetic dysregulation, particularly promoter hypermethylation, plays a critical role in the silencing of tumor suppressor genes during cancer progression. Although FXYD1 has been associated with ion transport regulation, its epigenetic regulation and functional significance in breast cancer metastasis remain to be elucidated.Methods: FXYD1 expression was evaluated using public datasets (TCGA, GEO), breast cancer cell lines, and patient-derived tissues. Functional analyses, including proliferation assays, Transwell invasion assays, and murine xenograft metastasis models, were conducted following FXYD1 overexpression or knockdown. Mechanistic studies involved RNA sequencing, co-immunoprecipitation, DNA methylation analysis, ubiquitination assays, and genetic rescue experiments.Results: FXYD1 expression was significantly downregulated in breast cancer tissues compared with adjacent normal tissues (p<0.001, Student’s t-test), and low expression was associated with advanced TNM stage and decreased overall survival (HR=1.83, 95% CI 1.2-2.8). Methylation analysis demonstrated that promoter hypermethylation was a critical mechanism underlying FXYD1 silencing. Functional restoration of FXYD1 suppressed tumor proliferation (50% reduction in colony formation, p<0.01), migration (70% decrease in wound healing, p<0.001), and lung metastasis (4-fold reduction in nodules, p<0.005). Mechanistically, FXYD1 facilitated Lysine 63 (K63)-linked polyubiquitination of DDX5 at residue K470 by recruiting the E3 ligase MAEA, leading to proteasomal degradation. This resulted in inhibition of Wnt/β-catenin signaling, as evidenced by reduced nuclear β-catenin (60% decrease) and downstream target expression (c-Myc, Cyclin D1). Rescue experiments confirmed that DDX5 re-expression (1.5-fold increase) or β-catenin stabilization (via CHIR99021) reversed FXYD1-mediated tumor suppression.Conclusions: Our findings establish FXYD1 as a previously unrecognized tumor suppressor that inhibits breast cancer metastasis via the DDX5-β-catenin axis. This study not only clarifies a novel ubiquitination regulatory mechanism but also highlights FXYD1 as a potential therapeutic target for metastasis intervention.
Presentation numberPS2-12-05
Programmed death-ligand 1expression in triple negative breast cancer: insights from an African ancestry-enriched multinational cohort
Brian Stonaker, Weill Cornell Medicine, New York, NY
B. Stonaker1, E. Adjei2, S. Demaria3, R. Martini4, M. B. Davis4, L. Newman1; 1Department of Surgery, Weill Cornell Medicine, New York, NY, 2Directorate of Pathology, Komfo Anokye Teaching Hospital, Kumasi, GHANA, 3Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY, 4Institute of Translational Genomic Medicine, Morehouse School of Medicine, Atlanta, GA.
Programmed death-ligand 1 (PD-L1) is an immune checkpoint protein expressed on tumor and immune cells that promotes immune evasion by inhibiting T-cell activation. In advanced triple-negative breast cancer (TNBC), a subtype with aggressive behavior and limited targeted treatment options, PD-L1 expression has emerged as a predictive biomarker for response to immune checkpoint inhibitors. However, most studies have focused on predominantly European-ancestry populations, leaving gaps in understanding PD-L1’s expression and clinical relevance among women of African ancestry, who are disproportionately affected by TNBC. Given TNBC’s molecular heterogeneity across ancestral groups, examining PD-L1 expression in diverse populations is critical for equitable immunotherapy and understanding ancestry-specific tumor biology.The International Center for the Study of Breast Cancer Subtypes (ICSBCS) biobank provides a unique ancestrally diverse tissue repository, enabling biomarker studies across historically underrepresented populations. Leveraging this resource, we are conducting a large-scale immunohistochemical (IHC) analysis of PD-L1 on formalin-fixed paraffin-embedded (FFPE) TNBC tissue sections. The study aims to evaluate PD-L1 in 1,000 TNBC samples, equally distributed among Ghanaian, Ethiopian, African American (AA), and European American (EA) patients. Here, we report initial findings.To date, 301 cases have been processed (AA n = 47, Ethiopian n = 12, Ghanaian n = 205, EA n = 37). PD-L1 expression, measured as percent positive tumor cells by IHC, showed means (± SD, 95% CI) of 7.25% (± 6.28, 3.34-11.16) in Ethiopian, 6.55% (± 6.28, 4.36-8.73) in EA, 5.32% (± 6.28, 3.39-7.25) in AA, and 1.13% (± 6.28, 0.20-2.06) in Ghanaian cases. Tukey-Kramer HSD comparisons revealed significant differences between Ethiopian and Ghanaian (p=0.0156), EA and Ghanaian (p<0.0001), and AA and Ghanaian (p=0.0008). Corresponding medians (IQR) were 9% (9.13%) for Ethiopian, 1% (4.5%) for EA, 2% (9.5%) for AA, and 0% (0.5%) for Ghanaian cases, reflecting heterogeneity and skewed PD-L1 expression. Initial analyses suggest differences in PD-L1 expression across self-reported race and ancestry groups. Comprehensive clinical data and additional tumor microenvironment measures, including tumor-infiltrating lymphocyte (TIL) scores, are being collected for integrative assessment of PD-L1 and its clinical significance. Continued analysis will advance understanding of population-specific tumor immunobiology and inform equitable immunotherapy approaches in TNBC.These preliminary findings underscore the value of the ICSBCS biobank in enabling ancestry-informed biomarker research in TNBC. By systematically evaluating PD-L1 across Ghanaian, Ethiopian, AA, and EA populations, this study aims to uncover differences in immune checkpoint activation to inform tailored therapies. These data are preliminary, with limitations including small sample sizes in some ancestry groups, particularly Ethiopian and EA cohorts, which may affect precision. As analysis progresses toward the full 1,000-case cohort, continued integration of clinical, pathological, and molecular data will be essential for advancing population-specific tumor immunobiology and improving equity in breast cancer outcomes.
Presentation numberPS2-12-07
Metabolic and immune reprogramming via SCD1 inhibition enhances chemo-immunotherapy response in TNBC
Pooja P Advani, Mayo Clinic, Jacksonville, FL
P. P. Advani1, S. K. Deshmukh2, R. Whetten3, M. Redig3, M. Pawlush3, S. Wu2, J. Xiu2, J. P. Leone4, S. Gandhi5, M. Lustberg6, G. Sledge Jr2, J. Copland3; 1Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 2Medical Affairs, Caris Life Sciences, Tempe, AZ, 3Cancer Biology, Mayo Clinic, Jacksonville, FL, 4Breast Oncology Center, Dana Farber Cancer Institute, Boston, MA, 5Hematology and Oncology, Emory University, Atlanta, GA, 6Hematology and Oncology, Yale School of Medicine, New Haven, CT.
Background: Triple-negative breast cancer (TNBC) is highly aggressive subtype with poor prognosis and there is an urgent need to develop novel therapies. Deregulated cellular energetics is a known hallmark of cancer. Stearoyl-CoA desaturase-1 (SCD1), is a key enzyme in fatty acid (FA) synthesis, converting saturated to monounsaturated FA. It is upregulated in TNBC, and its expression correlates with poor survival. Hence, targeting SCD1 is a novel strategy for cancer therapy. We have developed an SCD1 inhibitor, MTI-301 and shown anti-tumor activity in murine TNBC cell lines with MTI-301 alone and in combination with paclitaxel. Here, we study the efficacy of MTI-301 in TNBC mouse model and, molecular and immunological association of SCD1 in TNBC.Methods: MTI-301 activity (10 mg/kg oral gavage daily) was evaluated in E0771 immunocompetent TNBC mouse models alone and with immune checkpoint inhibitors (ICI) and paclitaxel (anti-PD-L1 Ab 200ug/mouse; IP and 25 mg/kg IP both given 2/week). Immune markers such as CD4+ T cell and CD8+ T cell in mice tumor tissue were assessed by immunohistochemistry (IHC). Tumor volume was measured by calipers 2/week and compared between groups by ANOVA test. Molecular and immune characterization was performed in 1,581 invasive ductal (ID) TNBC patient (pt) samples tested by NGS (592-gene panel, NextSeq; WES/WTS, NovaSeq; Caris Life Sciences, Phoenix, AZ). ID-TNBC SCD1-high(H) and SCD1-low(L) RNA expression were classified as above or below the 50 percentiles, respectively. Immune cell fractions were calculated by deconvolution of WTS: Quantiseq. Pathway enrichment was determined by GSEA (Broad Inst). Statistical significance was determined using chi-square and Mann-Whitney U test with p-values adjusted for multiple comparisons (q<0.05).Results: Compared to vehicle control, MTI-301 showed significant reduction in mean tumor volume and was associated with macrophage phagocytosis and intratumor infiltration of CD4 and CD8 T cells. We have also showed that MTI-301 + anti-PD1 Ab + paclitaxel has significant antitumor activity compared to either agent alone. IHC analysis is ongoing. In real-word pt data, SCD1-H TNBC (N=791) had higher immune cell infiltration of M2 MΦ (2.95% vs. 2.58%) and neutrophils (4.5% vs. 4.1%) compared to SCD1-L (N=790), all q<0.05. SCD1-H had decreased IFNγ score (-0.29 vs -0.24), but increased MAPK activation score (-0.57 vs -1.77), all q<0.05. SCD1-H had higher levels of metabolic regulator genes (IL10, IL10RA, SOCS3, HIF1A, DUSP6, FC: 1.4-1.8), immune checkpoint genes (PDCD1, CTLA4, LAG3, TIGIT, VISTA, CD244, CD160, BTLA, HAVCR2, FC: 1.3-1.6), stem cell related genes (CD44, ALDH1A1, ALDH1A2, ALDH1A3, KLF4, NANOG, FC: 1.4-1.6) and markers of exhausted T cells (CXCL13, CD38, CD101, ENTPD1, CD69, CD103, KLRG1, PRF1, GZMB, FC: 1.2-1.7), all q<0.05. SCD1-H had enrichment of pathways related to oxidative phosphorylation, FA metabolism, PI3K/AKT/MTOR, EMT, DNA repair, TGF beta, P53 and inflammatory response (NES: 1.7-3.8, all FDR<0.25). SCD1-H had higher frequency of PIK3CA (18.1% vs 8.8%), PTEN (10.7% vs 5.2%), PIK3R1 (6.4% vs 2.3%), AKT1 (3.3% vs 1.3%) and NF1 (8.3% vs 2.7%) mutation compared to SCD1-L, all q<0.05. Conclusion: SCD1 high TNBC tumors are associated with immune suppressive TME, FA metabolism, and dysregulated PI3K pathway, positioning SCD1 as a metabolic driver of therapy resistance and immune evasion and support its targeting to enhance chemo-ICI efficacy. SCD1 inhibitor, MTI-301 significantly reduced tumor growth and enhanced effector immune cell infiltration in TNBC murine models with combinatorial activity with chemo-ICI, indicating that pharmacologic modulation of SCD1 is a promising strategy for cancer therapy. We will be conducting Phase I clinical trial with MTI-301 in pts with advanced tumors including TNBC (NCT NCT06911008).
Presentation numberPS2-12-08
Obi‑201: a bispecific trop2 × her2 antibody-drug conjugate to expand therapeutic reach beyond her2-low limitations
Chi-Huan Lu, OBI Pharma, Inc., Taipei, Taiwan
C. Lu, S. Wang, Y. Chen, Y. Wu, T. Huang, W. Chan, L. Chen, H. Wu, C. Wei; Pharmacology, OBI Pharma, Inc., Taipei, TAIWAN.
OBI‑201: A Bispecific TROP2 × HER2 Antibody-Drug Conjugate to Expand Therapeutic Reach Beyond HER2-Low Limitations Background: While Enhertu® has delivered meaningful clinical benefits in HER2-positive and HER2-low cancers, many patients still experience suboptimal outcomes, particularly in HER2-low tumors that account for over 50% of the HER2-positive pan cancer population. Resistance to HER2-targeted therapies remains a critical challenge, including HER2 downregulation or loss, which diminishes antibody binding and payload delivery. OBI Pharma is the first to discover that TROP2 and HER2 form heterodimers (Data not published) in cancer cells. This TROP2 and HER2 crosstalk provides a strong rationale for dual targeting. In response, we developed OBI-201—a bispecific antibody-drug conjugate (ADC) engineered to simultaneously engage both TROP2 and HER2, aiming to enhance tumor specificity and overcome resistance associated with single-target therapies. Method: OBI-201 is a next-generation bispecific ADC that co-targets HER2 and TROP2, using site-specific GlycOBI® glycan-based conjugation technology to attach an exatecan payload at a DAR of 4 for precise delivery. To assess the efficiency of internalization, pHAb-labeled OBI-201 was applied to HER2/TROP2 co-expressing cells by quantifying fluorescence signals. To evaluate its antitumor activity, OBI-201 was tested in both cell line-derived xenograft (CDX) models and patient-derived xenograft (PDX) models. Results: The bispecific ADC format of OBI-201 was designed to enhance tumor targeting and promote efficient internalization, which was confirmed by strong internalization signals observed in cells co-expressing HER2 and TROP2. OBI‑201 demonstrated superior antitumor activity compared to Enhertu®, Datroway®, and Trodelvy® in HER2-low colorectal, pancreatic, and NSCLC models, as well as in Enhertu-resistant NSCLC CDX models. Remarkably, in a HER2-low TNBC PDX model with established multi-drug resistance, including resistance to Trodelvy®, OBI-201 achieved complete tumor regression in 3 out of 6 mice, highlighting its potential to overcome treatment resistance and address unmet needs in difficult-to-treat tumors. Conclusion: OBI-201 is a next-generation bispecific ADC targeting HER2 and TROP2, designed to overcome the limitations of single-target therapies. Its dual-targeting approach, enabled by GlycOBI®, proprietary ADC enabling technologies for precise payload delivery, offers strong potential to overcome HER2-low expression and resistance, addressing significant unmet needs across a broad spectrum of solid tumors.
Presentation numberPS2-12-09
Global expression of small DNA damage-associated RNAs (sdRNAs) in breast cancers
Jennifer M Rosenbluth, University of California, San Francisco, San Francisco, CA
D. K. DeConti1, J. Z. Yu2, S. Warhadpande2, F. Abderazzaq3, A. Desbuleux3, E. Hatchi3, J. M. Rosenbluth2; 1Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 2Medicine, University of California, San Francisco, San Francisco, CA, 3Cancer Biology, Dana-Farber Cancer Institute, Boston, MA.
Background: BRCA1 is a large protein that regulates many cellular tumor suppressive processes and maintains genome integrity, at least in part through DNA repair and euploidy maintenance. Interestingly, multiple studies suggest that the function of BRCA1 and/or the DNA damage repair mechanisms it regulates are altered in a substantial subset of breast cancers (BCs). Recently, we deciphered the molecular basis for a new process of BRCA1-driven, small RNA-mediated repair of R-loop-associated DNA damage. These previously undescribed, 30- 35 nt long small RNAs, called sdRNA (single-stranded, DNA damage-associated small RNA), are synthesized by BRCA1/RNAi-containing complexes. sdRNAs catalyze the repair of ssDNA breaks by promoting the formation of an atypical DNA repair complex composed of PALB2 (a bona fide BC suppressor gene product) and RAD52 (recently shown to be involved in R-loop processing at double-strand breaks, and also associated with BC). We sought to determine the levels and role of these sdRNAs in breast tumor formation using human specimens and patient-derived models of breast tissue. Methods: A total of 30 normal breast samples and 96 BC samples were collected. To ensure a sufficient amount of tissue available for RNA sequencing, some samples were expanded into organoids, which are three-dimensional tissue cultures that mimic the characteristics of the parent tissues. Whole-genome small RNA sequencing was used to determine the expression profiles of sdRNAs in BCs across all major subtypes. Abundance profiling and computational modeling methods were used to identify key BC-associated sdRNAs. Key sdRNAs, highly expressed in BCs, were overexpressed in organoid culture to determine the impact of sdRNAs on the growth of normal breast cells. Results: BC samples expressed very high levels of multiple sdRNAs compared to normal breast cells, as determined by focused RNA measurements (qRT-PCR) and by small RNA sequencing. sdRNAs were particularly increased in triple-negative BCs, compared to ER+ BCs, including sdRNAs associated with genes that play roles in DNA damage and repair, cell growth, survival, proliferation, and metastasis. We were able to identify a select few of these differentially expressed sdRNAs in an independent published study on circulating small RNAs in urine and blood in BC patients that underwent neoadjuvant therapy and surgery. The circulating sdRNAs were identified as differentially expressed throughout the study timeline. A dataset of small RNAs in BCs, specifically targeting the detection of sdRNAs, was generated across all major subtypes and linked to clinical annotations to identify sdRNAs for further evaluation in vitro. From this, 770 key BC-associated sdRNAs were identified, including for example sdRNAs associated with the AKT1 and SOD1 genes. Conditions were successfully optimized to transfect sdRNAs into breast organoids in 3-dimensional culture with high efficiency. After select sdRNAs were transfected in normal breast organoids, organoid growth rates were observed to be higher compared to non-specific control RNAs. Conclusions: BC samples have higher levels of multiple sdRNAs compared to normal breast cells. Our initial studies have identified specific sdRNAs associated with BC subtypes that increase the growth of normal breast tissue, consistent with a potential role in breast tumor development. Our data further suggest that sdRNAs highly expressed in BCs can be detected in liquid biopsies, and future studies will assess whether they can be used as a biomarker of therapy efficiency. Ongoing efforts are focused on further assessing the roles of sdRNAs in cancer initiation and exploring the expression of BC-associated sdRNAs in other cancer types.
Presentation numberPS2-12-10
Mediterranean Diet and Breast Cancer Biology: Novel Insights into the Role of Extracellular Vesicles
Luca Gelsomino, University of Calabria, Rende (Cs), Italy
L. Gelsomino1, D. Cristofaro2, G. Filice2, F. Giordano2, I. Barone1, D. Bonofiglio1, G. Arpino3, G. Salvatore4, S. Andò1, S. Catalano5, C. Giordano5; 1Pharmacy and Health and Nutrition Sciences; Health Center, University of Calabria, Rende (Cs), ITALY, 2Pharmacy and Health and Nutrition Sciences, University of Calabria, Rende (Cs), ITALY, 3Department of Clinical Medicine and Surgery, University of Naples” Federico II”, Naples, ITALY, 4Department of Medical, Human Movement and Well-being Sciences, University Parthenope, Naples, ITALY, 5Pharmacy and Health and Nutrition Sciences; Health Center; Clinical Laboratory Unit, A.O. “Annunzia, University of Calabria, Rende (Cs), ITALY.
A number of risk factors have been identified in relation to breast cancer (BC). Among these, environmental factors, particularly those related to nutritional status, have been shown to play a significant role in the biology of BC. Thus, there is a growing interest in assessing the potential benefits of diet and life style in the prevention and treatment of BC. Among dietary interventions, the Mediterranean Diet (MD) has been suggested to be the most efficacious in reducing the risk of BC and enhancing patient outcomes. Numerous studies have attempted to address the relationship between diet and circulating Extracellular Vesicles (EVs), small membrane-bound particles that represent a novel axis of intercellular communication in BC. It has been reported that dietary compounds may influence the biogenesis, release, and composition of the endogenous EVs. However, there is still a knowledge gap regarding the accurate phenotyping of diet-induced EVs and their potential role in mediating the effects of nutritional status on BC. Here, we investigated the impact of MD adherence on the composition and function of serum circulating EVs isolated from BC patients. BC patients have been classified by their level of MD adherence (Low: ≤5; Medium: 6-9; High: ≥10), as assessed by the MEDAS questionnaire. We isolated and fully characterized EVs from the serum of BC patients, in accordance with the MISEV 2023 guidelines, by Transmission Electron Microscopy, Nanotracking particle analysis (NTA) and immunoblot. The biological effects of BC patient-derived EVs on cell, proliferation, stemness, motility and metabolism in two different ERα-positive BC cell lines, MCF-7 and T-47D, were evaluated. The expression profiles of EV-packaged miRNAs were investigated by small RNA-sequencing analysis. NTA analysis revealed a correlation between elevated MD adherence and alterations in the number and dimensions of circulating EVs in BC patients. In addition, we demonstrated that the EVs isolated from BC patients with High MD adherence were less effective in sustaining cell viability and growth in both MCF-7 and T-47D as evidenced by anchorage-dependent (clonogenic assay) and anchorage-independent (soft-agar) assays, respectively. The boyden-chamber transmigration assay revealed that EVs isolated from BC patients with High MD adherence were less effective in promoting the migration of BC cells in the two employed experimental models. Finally, given that migratory potential is among the most significant characteristics of BC stem cells, the mammosphere forming efficiency assay was conducted. We found that EVs isolated from BC patients with High MD adherence exhibited a reduced capacity to form mammospheres in ER+ BC cells compared to EVs isolated from BC patients with Low MD adherence. Accordingly, BC cells treated with high MD adherence EVs showed a reduced cellular bioenergetics as measured by oxygen consumption rate, in terms of basal and maximal respiration, compared to cells treated with low MD adherence EVs. The analysis of EV-miRNA profile revealed 172 miRNAs, 18 of which were differentially expressed (∣fold change∣≥1.5) in Low vs High MD adherence EVs. These deregulated miRNAs are mainly involved in controlling proliferation, migration and metabolism in BC. These data demonstrated that High MD adherence modulates the EV composition (e.g. the miRNA profile) and exerts beneficial effects on BC cell development, growth and progression. Although these results are still preliminary, they underscore the importance of healthy dietary patterns in modulating BC biology and open new avenues for identifying potential therapeutic targets, to be exploited in patients with poor dietary habits.
Presentation numberPS2-12-11
Tgfβ-associated transcription factors induce metabolic and chromatin remodeling in breast cancer cells
Mohamad Moustafa Ali, Uppsala University, Uppsala, Sweden
M. M. Ali1, C. Tsirigoti1, H. Yasmin2, S. S. Ali2, A. Moustakas1; 1Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, SWEDEN, 2Research Department, Children’s Cancer Hospital, Cairo, EGYPT.
The transforming growth factor β (TGFβ) family encodes pleiotropic cytokines that modulate cellular identity, primarily through the epithelial-mesenchymal transition (EMT) and the promotion of cancer stem cell populations, which accounts for elevated tumor heterogeneity. In breast cancer cells, such heterogeneity can be manifested at both transcriptional and epigenetic levels, with direct implications for metabolic rewiring. Using CRISPR/Cas9 gene editing technology, we ablated the EMT-related transcription factor SNAI2 in triple-negative breast cancer cells. Whole-transcriptome profiling revealed significant downregulation in cell cycle-related processes associated with the induction of apoptosis and cell death signatures. Strikingly, knockout (KO) cells exhibited significant alterations in various genes involved in central metabolic pathways, including oxidative phosphorylation, fatty acid metabolism and mTOR signaling. Functional analyses of mitochondrial respiratory activities showed enhanced complex I and complex II activities as well as increased ATP-linked, maximum, and reserve respiratory capacities in SNAI2 KO cells. Feul-dependence studies indicated a significant reduction in fatty acid dependence, attributed to the account of pyruvate metabolism, while glutamiolysis and glycolytic metabolism remained unchanged. The KO cells exhibited elevated mitochondrial density, accompanied by accumulation of intracellular and mitochondrial reactive oxygen species. Phenotypically, the KO cells presented an intermediate epithelial-mesenchymal state with decreased proliferation capacity and migration potential. Additionally, the KO cells showed enhanced sensitivity to the CDK4/6 inhibitor palbociclib, compared to parental cells. Seminal studies in various breast cancer models have revealed the potency of TGFβ in promoting cell resistance to palbociclib through transcriptional modulation of EMT factors, including SNAI2, and subsequent metabolic remodeling. Therefore, the present study demonstrates the pivotal role of SNAI2 in mediating transcriptional and metabolic rewiring, leading to increased cancer heterogeneity.
Presentation numberPS2-12-12
Targeting RNA binding protein-deficiency associated neoepitopes for triple negative breast cancers
Michael Lee, Baylor College of Medicine, Houston, TX
M. Lee; Cancer & Cell Biology, Baylor College of Medicine, Houston, TX.
Neoantigen-based cancer vaccines are largely individualized treatment modalities based on a patient’s DNA mutations. However, tumor-specific neoantigen formation may also come about as the result of other downstream processes like splicing dysregulation and nonsense mediated RNA decay (NMD) that result in aberrant transcripts. Such processes can be regulated by RNA binding proteins (RBPs). We have shown previously that one such RBP, hnRNPM, preferentially binds to UG-rich motifs and suppresses cryptic splice sites. Knockdown of hnRNPM resulted in consistent upregulation of common aberrant cryptic transcripts across multiple cancer cell lines. We identified a peptide produced by translation of these hnRNPM-associated cryptic transcripts in triple negative breast cancer cell lines. This peptide is predicted to be a strong binder to human leukocyte antigen class I (HLA-I) and does not map to canonical protein sequences. In addition, we have identified several other RBP candidates through analysis of public omics datasets of patient gene expression and clinical response to immunotherapy. These preliminary results will be followed up with further characterization of the neoantigens produced by RBP dysregulations. The goal will be to develop a pool of clinically actionable neoantigens for individual patients by searching for RBP dysregulations that can predictably produce abnormal neoantigens.
Presentation numberPS2-12-13
Breast Cancer Mutation Detection via Liquid Biopsy Using cfDNA from 50 mL Urine Samples
Nafiseh Jafari, nRichDX, Irvine, CA
N. Jafari1, Mayer Saidian, Jason Saenz, Carlos Hernandez, Daniel Cedeno, Cameron Van-Dieren, Y. Lu2; 1R&D, nRichDX, Irvine, CA, 2R&D, Anchor Molecular, Pleasanton, CA.
Background: Urine cfDNA offers a promising, non-invasive option for molecular profiling in breast cancer. However, challenges with cfDNA stability, yield, and compatibility with preservatives have limited its widespread clinical adoption. This study evaluated the recovery and mutation detection performance of large-volume (50 mL) urine samples preserved with three common urine stabilizers and analyzed after storage for 0 and 3 days at room temperature. Methods: Pooled healthy donor urine was contrived with Anchor Molecular’s cfDNA reference standards containing four clinically actionable breast cancer mutations: IDH1 (R132H), PIK3CA (E545K), PTEN (R130G), and TP53 (R248Q). Three preservatives—Streck Urine preservative, UAS urine analyte stabilizer, and Tris-EDTA (TE) buffer pH (8.0)—were evaluated at time zero (T0) and after 3 days (T3) of room temperature storage. For each condition, three technical replicates were extracted from 50 mL urine samples using the nRichDX Revolution Max 50 cfDNA Kit. cfDNA yield and fragment integrity were measured using the Agilent cfDNA High Sensitivity ScreenTape assay. Mutation detection was performed using a multiplex qPCR assay targeting the four hotspot mutations. Results: cfDNA was successfully recovered in all conditions, with consistent detection of all four mutations across replicates, time points, and preservatives. The cfDNA fragment size profile was preserved from T0 to T3 across all preservatives, indicating minimal degradation during storage. Quantitative results showed that cfDNA yield varied by preservative, with average recovery of 88% (Streck), 75% (UAS), and 62% (TE) relative to input. Despite differences in yield, mutation detection was robust across all conditions, with no statistically significant shift in Ct values between T0 and T3 (p > 0.05). Inter-replicate variation remained low (CV < 5%), and all replicates consistently showed reliable detection of IDH1, PIK3CA, PTEN, and TP53 mutations. Conclusion: This study demonstrates that 50 mL urine inputs allow for consistent recovery and detection of clinically relevant breast cancer mutations across multiple preservative chemistries, even after 3 days at room temperature. Mutation standards from Anchor Molecular were reliably detected using qPCR in all conditions, and the nRichDX Revolution Max 50 cfDNA Kit was compatible with all three preservatives tested, showing variable but sufficient recovery to support downstream mutation detection. These findings support the feasibility of urine as a stable and scalable liquid biopsy sample type for breast cancer genotyping, longitudinal monitoring, and remote sample collection workflows.
Presentation numberPS2-12-15
Glucocorticoid receptor activation leads to remodeling of the chromatin landscape in infiltrating lobular carcinoma cells and changes in estrogen receptor-mediated gene expression
Amanda Briceno, UT Southwestern Medical Center, Dallas, TX
A. Briceno, B. Porter, L. Bennett, S. Conzen; Internal Medicine, UT Southwestern Medical Center, Dallas, TX.
Title: Glucocorticoid receptor activation leads to remodeling of the chromatin landscape in infiltrating lobular carcinoma cells and changes in estrogen receptor-mediated gene expression Invasive lobular carcinoma (ILC) is the second most common subtype of estrogen receptor (ER)-positive breast cancer. ILC relies, in part, on ER signaling for cell proliferation and tumor growth. Our laboratory is interested in understanding ER transcriptional activity in the context of glucocorticoid receptor (GR), the body’s stress response hormone. Previously, it was shown that ligand-dependent GR occupancy suppresses ER binding at shared enhancer regions of infiltrating ductal carcinoma (IDC) chromatin, and that GR activation inhibited ER-mediated proliferation gene expression (Tonsing-Carter et al., 2019). Similar to IDC, our lab observed that GR-activation of ER+ ILC cells also had an anti-proliferative effect on ER-mediated gene expression (Porter et al., 2023). To elucidate how ER and GR interact on chromatin in ILC models, we performed genome wide chromatin immunoprecipitation followed by sequencing (ChIP-seq) in ER+ ILC cell lines treated with vehicle, estradiol (E2), dexamethasone (Dex), or the combination (E2+Dex). ChIP-seq revealed that GR-driven chromatin remodeling reduces ER mediated proliferation. To measure the effect of liganded GR occupancy at cell cycle gene enhancers, directed ChIP-qPCR was performed at the CCND1 locus, a known ER-regulated gene. We found that activated GR recruitment to the ER-bound CCND1 enhancer element, suppressed CCND1 gene expression, suggesting that GR may inhibit ER-mediated cell cycle progression through direct chromatin interaction. To further understand how GR occupancy on chromatin impacts global gene expression we performed RNA-sequencing in parallel. RNA-seq analysis showed significant down regulation of cell-cycle-related genes supporting a model where GR recruitment to ER-bound chromatin contributes to reduced tumor cell proliferation. Additional genes that encode proteins involved in oxidative phosphorylation, mitochondrial function, and epithelial cell organization were also prominently regulated by GR activation. We hypothesize that GR activation leads to chromatin remodeling resulting in upregulation of gene expression pathways such as oxidative phosphorylation, while downregulating ER-mediated cell cycle progression and proliferation. Several genes including MTCO1P11, ZNF652, MT-CYP, and ITGA10 were upregulated upon treatment with a combination of 10nM estradiol + 100nM Dex and were linked to GR/ER co-regulated regions, suggesting functional relevance. Ongoing studies integrating GR and ER chromatin binding with gene expression data will uncover the hormone-responsive (and potentially modulatable) chromatin landscape that contributes to functional crosstalk between GR and ER. Future in vivo and gene expression studies using highly selective GR modulation (SGRMs) with and without systemic ER antagonism will define the physiological relevance of GR/ER crosstalk in ILC and therapeutic potential of SGRMs.
Presentation numberPS2-12-16
Tp53 mutant tnbc overexpresses serine biosynthesis genes psat1 and phgdh and may demonstrate vulnerability to dietary glycine supplementation
Jules Cohen, Stony Brook Cancer Center, Stony Brook, NY
J. Cohen; Hematology and Medical Oncology, Stony Brook Cancer Center, Stony Brook, NY.
INTRODUCTION Treatment of TNBC is limited by a lack of identifiable therapeutic targets. One of the biggest gene expression differences between TNBC and luminal breast cancer is upregulation of serine biosynthesis enzyme PSAT1. PSAT1 (and, from the same pathway, PHGDH) upregulation correlates with TP53 loss-of-function mutations, common in TNBC. PSAT1 upregulation is negatively correlated with luminal breast cancer genes including ESR1, GATA3 and FOXA1. Since serine deplete diets are likely impractical, glycine supplementation may be toxic to serine-dependent TNBC cells through its effect on one-carbon metabolism. A randomized trial of chemotherapy with or without glycine supplementation is warranted. METHODS We used Breast Cancer Gene-Expression Miner v5.2 (Integrated Center for Oncology), muTarget (Semmelweis University) and GEPIA (Peking University) to interrogate publicly available datasets including METABRIC, GTEx, SCAN-B and TCGA. We looked for serine biosynthesis genes and TP53-regulated genes on well characterized gene sets Oncotype DX, PAM50 and Mammaprint. RESULTS PHGDH is one of the PAM50 genes and its upregulation correlates with the TNBC (basal) subset and is inversely related to the luminal subset. Neither serine biosynthesis gene is present on Oncotype DX or Mammaprint. TP53 mutations are associated with upregulation of 10 genes and downregulation of 7 genes on the PAM50 gene set. PSAT1 is expressed 4-5 times higher in TNBC vs. luminal breast cancers (p<0.0001). PSAT1 is upregulated 2.64 times and PHGDH 2.37 times in TP53 mutated vs wildtype breast cancers. ESR1 (5.4 fold), GATA3 (3.33 fold) and FOXA1 gene expression is downregulated in TP53 mutated breast cancers. ESR1, GATA3 and FOXA1 gene expression is inversely correlated with PSAT1 gene expression (correlation coefficient -0.69 to -0.76). ESR1 gene expression is highly correlated with GATA3 (0.84) and FOXA1 (0.75) and inversely correlated with PSAT1 (-0.76). GATA3 mutations correspond to upregulation of GATA3 gene expression (2.45 times), ESR1 (2.39 times) and FOXA1 (1.86 times). GATA3 mutations correspond to downregulation of PSAT1 (1.65 times) and PHGDH (1.68 times). CONCLUSIONS TNBC is frequently characterized by mutations in TP53 and demonstrate upregulation of serine biosynthesis genes PHGDH and PSAT1. Luminal breast cancers harbor TP53 mutations less frequently and demonstrate downregulation of serine biosynthesis genes. At least 17 of the PAM50 genes are differentially regulated by TP53 mutations, including PHGDH. Glycine supplementation may target a particular vulnerability in TP53 mutant TNBCs.
Presentation numberPS2-12-18
The role of MARCKS in inflammation in inflammatory breast cancer using in vitro and in silico studies
Maroua Manai, Pasteur Institute of Tunis, Tunis, Tunisia
M. Manai1, N. Boughzala1, N. Bayou2, P. Finetti3, N. Akkari1, O. Bejar1, M. Selmi1, Y. Houcine4, R. Benhassen4, C. Reduzzi2, J. Donahue2, V. Fraser5, E. Nicolò2, M. Driss4, F. Bertucci3, H. Boussen6, M. Cristofanilli2; 1Immunology, Pasteur Institute of Tunis, Tunis, TUNISIA, 2Medicine, Weill Cornell Medicine, New York, NY, 3Predictive Oncology Laboratory, Institut Paoli-Calmettes, Marseille, FRANCE, 4Immunohistocytology, Salah Azaiz Institute, Tunis, TUNISIA, 5IBC, The Inflammatory Breast Cancer-International Consortium (IBC-IC), New York, NY, 6Immunology, Medical Oncology Service, Tunis, TUNISIA.
Title: The role of MARCKS in inflammation in inflammatory breast cancer using invitro and in silico studiesAuthors: Maroua Manai1,2,3, Nesrine Boughzala*2, Nadia Bayou1*, Pascal Finetti4*, NourAkkari2, Omar Bejar2, Manel Selmi2, Yoldez Houcine5, Rihab Benhassen4, Carolina Reduzzi1,Jeannine Donahue1,3, Valerie Fraser3, Eleonora Nicolò1, Maha Driss5, François Bertucci3,4,Hamouda Boussen3,6, Massimo Cristofanilli1,3Affiliations1Weill Cornell Medicine, Department of medicine, Division of hematology-oncology, 1300York Ave, 10021, NY, NY, USA2Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02 (LTCII),Institut Pasteur de Tunis, 13, place Pasteur, BP74, 1002, Tunis-Belvédère, Tunisia. Universityof Tunis El Manar, 1068, Tunis, Tunisia;3The Inflammatory Breast Cancer-International Consortium (IBC-IC);4Predictive Oncology Laboratory, Centre de Recherche en Cancérologie de Marseille(CRCM), Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Aix MarseilleUniversité, Marseille, F-13009, France;5Immunohistocytology Department, Salah Azaiz Institute, Bab Saadoun, Tunis 1006, Tunisia.6Medical Oncology Service, Hospital of Ariana, Ariana 2080, TunisiaInflammatory Breast Cancer (IBC), an aggressive and rare form of breast cancer, is characterized byhigh metastatic potential and an inflammatory microenvironment. The Myristoylated Alanine-Rich CKinase Substrate (MARCKS) protein is overexpressed in IBC, suggesting its involvement in theinflammatory activation linked to its aggressiveness. Our study explored the role of MARCKS andinflammatory signaling pathways in IBC, using both in vitro (siRNA-mediated inhibitionof MARCKS in SUM149, KPL4 and MDA-MB-231 cell lines) and in silico (differential geneexpression analysis, annotation, enrichment, identification and visualization of pathways containingthese genes) approaches. Our in vitro studies showed that the inhibition of MARCKS reducedmigration and pro-tumor activity of IBC cells vs. non-inflammatory breast cancer (nIBC) underinflammatory conditions following treatment with lipopolysaccharide (LPS). Furthermore, mRNAexpression analysis by qRT-PCR, NFkBIB and NFkBIE expression were upregulated followingMARCKS inhibition by siRNA and this only in IBC compared to non-IBC. Interestingly, for NF-κB-IE, a significant decrease and increase in expression was observed after transfection in all three ISCmodel lines compared to ISC. In KPL4 cells, expression increased from approximately 0.03419 to1.376 units (p = 0.007). In the SUM149 cell line, this increase was even more pronounced withexpression increasing from 0.8194 to 1.801 units (p < 0.001), while in MDA-MB-231 cells, it alsodecreased from -0.9488 to -0.4712 units (p = 0.007). We also assessed protein expression ofMARCKS and NFκB in a series of 40 samples from patients with IBC. Our results showed thatoverexpression of NFκB, a major transcription factor in chronic inflammation, was associated withthat of MARCKS in IBC patients (p = 0.003). In silico, we demonstrated different alterations in keypathways (PI3K/AKT, JAK/STAT) involved in inflammation and cell proliferation. These resultsconfirm the importance of MARCKS in IBC tumor inflammation and open up prospects for itspersonalized therapeutic targeting.Keywords: Inflammatory breast cancer, inflammation, MARCKS, signaling pathways
Presentation numberPS2-12-19
Single cell mutational fingerprints and the viral culprits: uncovering APOBEC3 dysregulation in breast cancer
Jake D Lehle, Texas Biomedical Research Institute, San Antonio, TX
J. D. Lehle, M. Soleimanpour, N. Haghjoo, D. Ebrahimi; Host-Pathogen Interaction, Texas Biomedical Research Institute, San Antonio, TX.
Viral infection plays a significant role in the development and evolution of many cancers. It is estimated that between 13% to 18% of all new cancer cases are caused by infection. It is known that viral infection can induce transcriptional changes in cancer, including altering the expression of a seven-membered DNA editing enzyme family known as APOBEC3 or A3. These cytosine deaminases have been implicated in infectious diseases and cancer for their ability to induce cytidine to uridine (C>U) mutations in both viral and host DNA. Indeed, A3-associated mutational signatures SBS2 and SBS13 are present in more than 70% of cancer types and around 50% of all cancer genomes. However, little is known about the interplay between viral infection and A3 dysregulation in cancer. Accurate profiling of A3 expressions and their dysregulations caused by viral infections in cancer has historically been challenging using bulk RNA-seq. To address this, we leverage existing single-cell RNA-seq (scRNA-seq) data sets and our custom single cell bioinformatics pipeline, which uses multiple machine learning models for cell type annotation and cancer cell risk prediction, statistical methods for detection of mutational signatures at the single cell resolution, and custom k-mer based viral reference alignment for viral read detection in order to identify the source of A3 and viral reads across individual cell types within the breast cancer tumor microenvironment. Our studies using this pipeline have provided unparalleled resolution within the tumor microenvironment required to address questions about the association between viral presence and A3 dysregulation in various cancers. Previous analysis of head and neck tumors (GEO: GSE173468) allowed our lab to identify the presence of HPV16 viral reads, which were associated with a significant increase in A3B expression in infected basal cells of the tumor. Our recent analysis of breast cancer tumors (GEO: GSE16529 and GSE243526) has given us critical insights into cell type-specific A3 expression dynamics, indicating that the source of A3A expression was solely found coming from macrophage cells in the tumor and that high-risk cancer cells had primarily high expression of A3B and lacked A3A expression. In this study, we analyzed publicly available scRNA-seq datasets from two normal tissues (NTs), three primary tumors (PTs), and three tamoxifen-treated recurrent tumors (RTs) (GEO: GSE240112) in order to investigate the association between the presence of viral reads, A3 expression, and mutational signatures SBS2/13. We are currently optimizing our pipeline to further improve our viral read identification and association with specific cell types within each tumor microenvironment. This work is supported by the Texas Biomedical Research Institute’s Postdoctoral Forum Fellowship Grant.
Presentation numberPS2-12-20
The Oncogenic Role of MitoNEET in Breast Cancer
Connor Kent, Louisiana State University Health Sciences Center, New Orleans, LA
C. Kent1, Z. Liang1, G. Vontz1, C. Li1, H. Ding2, Q. Shen1; 1Genetics, Louisiana State University Health Sciences Center, New Orleans, LA, 2Biological Sciences, Louisiana State University, Baton Rouge, LA.
The Oncogenic Role of MitoNEET in Breast Cancer Connor Kent1, Zhipin Liang1, Gabrielle Vontz1, Caiyue Li1, Huangen Ding2, Qiang Shen11 Department of Interdisciplinary Oncology, LSU-LCMC Health Cancer Center, LSU Health Sciences Center, New Orleans, LA2 Department of Biological Sciences, BMB Division, Louisiana State University, Baton Rouge, LA 70803, USABackground: Aberrant glucose and energy metabolism of cancer cells, a phenomenon referred as the Warburg Effect in which most cancer cells produce energy predominantly through aerobic glycolysis in the cytosol, has long been considered as the major metabolic process for energy production and anabolic growth of cancer cells and is documented to promote breast cancer (BC) development and progression. However, how dysregulated glycolysis promotes BC carcinogenesis largely remains undefined. We discovered that mitoNEET or CISD1, a mitochondrial outer membrane protein, has a novel function as a redox enzyme. We believe mitoNEET constitutes a previously unknown cytosolic route of NADH oxidation to enlarge the cytosolic NAD+ pool, leading to aberrantly increased glycolysis and ATP/energy production in the cytosol, rendering mitoNEET a potential energy metabolism regulator. This study aims to determine whether mitoNEET acts as an oncogene to promote breast cancer growth by increasing glycolysis via an increase in the cytosolic pool of NAD+, promoting the Warburg Effect, and to determine the conditions in which mitoNEET promotes BC growth. Methods: Pan-cancer mitoNEET mRNA expression levels were determined using the TCGA database and analyzed using GraphPad Prism. mitoNEET expression was determined via Western Blotting (Cell Signaling #D5M4C). Colony formations were performed via staining with 0.5% Crystal Violet. mitoNEET knockout was achieved utilizing mitoNEET CRISPR sgRNA lentivectors (ABM LA116200). mitoNEET overexpression was achieved using mitoNEET Lentiviral Vector (ABM LC116200). Cell Viability was determined using 0.5% Crystal Violet staining with absorbance measured at 590nm using Promega’s GloMax Detection System (E9032). Inhibition of glycolysis was achieved using 2-Deoxy-D-glucose (2DG) (Sigma #D8375-1G). OCR, ECAR, and ATP production rates were determined using the Aglient Seahorse XF Analyzer. Conclusions: mitoNEET is highly expressed in the majority of BC cell lines screened. Its expression is also upregulated in oncogene-transformed mammary epithelial cell lines compared to normal or immortalized controls. Overexpresion of mitoNEET in immortalized mammary epithelial cells significantly increased growth compared to control. Suppression of mitoNEET via CRISPR-CAS9 led to significantly reduced growth of BC cell lines. Stable overexpression mitoNEET in BC cell lines led to significantly increased growth. Control BC cells were found to be more sensitive to 2DG treatment than mitoNEET-KO BC cells. Funding: LSU Health Sciences Center – New Orleans: Startup FundLSU: LSU Interinstitutional Cancer Research Funding Initiative Seed Grant, LCRC: Louisiana Cancer Research Center Strategic Investment In Translational Research Award
Presentation numberPS2-12-21
Deubiquitinase USP16 stabilizes CYBA expression and promotes progression and chemoresistance in triple-negative breast cancer chemoresistance in triple-negative breast cancer
Ningning Zhang, Chongqing University Cancer Hospital, Chongqing, China
N. Zhang, Y. Huang, S. Zhang, Y. Qin, X. Zeng; Department of Breast Center, Chongqing University Cancer Hospital, Chongqing, CHINA.
BACKGROUND: Triple-negative breast cancer (TNBC) represents a significant challenge in global breast cancer prevention and treatment due to its aggressive behavior, poor prognosis, and the absence of molecular markers for targeted therapy. There is an urgent need to elucidate its progression mechanisms and identify effective therapeutic targets. This study aimed to investigate the role and mechanism of the tumor promoting factor CYBA (Cytochrome B-245 Alpha Chain) in TNBC progression. METHODS: The expression of CYBA in normal tissues, tumor tissues, and adjacent tissues, as well as its correlation with prognosis, was evaluated by analyzing public databases, performing qRT-PCR and Western blot asssays, and conducting immunohistochemistry (IHC). The impact of CYBA on the proliferation of TNBC was assessed using CCK-8 assays, EDU incorporation assays, and subcutaneous xenograft models in nude mice. The role of CYBA of TNBC metastasis was assessed by scratch assay, transwell assay and tail vein injection lung metastasis models. The mechanisms underlying the biological effects of CYBA were investigated through the integrated analysis of LC-MS/MS and RNA-seq data, combined with Western blot validation, immunofluorescence (IF) and immunoprecipitation (CoIP). RESULTS: In patients with breast cancer and TNBC subtypes, those with high CYBA expression exhibited significantly shorter overall survival (OS) and recurrence-free survival (RFS) compared to those with low expression. In vitro and in vivo studies have confirmed that elevated CYBA expression promotes TNBC cell proliferation and metastasis. Drug sensitivity assays revealed that CYBA -overexpressing tumor cells demonstrated resistance to paclitaxel and carboplatin. Mechanistically, CYBA is co-localized in the cytoplasm and interacts with the deubiquitinase USP16, which inhibits CYBA degradation and stabilizes its expression via the deubiquitination pathway, thereby exerting its oncogenic function. CONCLUSION: USP16 stabilizes the expression of the tumor promoter CYBA through deubiquitination. High expression of CYBA in TNBC is associated with tumor progression, poor prognosis, and resistance to chemotherapy. Targeting the USP16/CYBA axis or inhibiting CYBA expression may represent a therapeutic potential strategy for TNBC.
Presentation numberPS2-12-22
The Molecular Mechanism of PRAK Regulating DNA Damage Repair and Its Role in Breast Cancer
Liu Jingjing, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
L. Jingjing; Department of Breast Oncology III, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA.
The Molecular Mechanism of PRAK Regulating DNA Damage Repair and Its Role in Breast CancerAbstractBackground: DNA damage plays a critical role in breast cancer, induced by both endogenous and exogenous factors. Abnormal repair mechanisms, such as homologous recombination repair (HRR) and non-homologous end joining (NHEJ), result in genomic instability and the accumulation of mutations, thereby driving cellular malignant transformation. DNA damage also promotes tumor progression by activating inflammatory pathways and modulating the immune microenvironment, serving as a key basis for breast cancer development and treatment resistance. Core Findings: PRAK (p38-regulated/activated protein kinase) is highly expressed in breast cancer and significantly correlates with poor patient prognosis and reduced survival rates. In vitro and in vivo experiments demonstrate that PRAK deficiency sensitizes tumor cells to PARP inhibitors and impairs DNA double-strand break (DSB) repair capacity, leading to defects in the homologous recombination (HR) pathway. Molecular Mechanism: Through affinity chromatography, mass spectrometry, and co-immunoprecipitation, PRAK was confirmed to directly interact with FOXO3 in vitro and in vivo. DNA-damaging agents (PARP inhibitors) induce nuclear accumulation of FOXO3, promoting the interaction between PRAK and FOXO3 within the nucleus. RNA-seq analysis revealed that BRCA1 is a key target of PRAK. PRAK deficiency reduces FOXO3 nuclear localization and inhibits its binding to the BRCA1 promoter, thereby downregulating BRCA1 expression. In vivo validation demonstrated that PRAK deficiency enhances the sensitivity of mouse xenografts to DNA-damaging agents. Conclusions: PRAK is upregulated in multiple human tumor tissues. PRAK deficiency inhibits tumor cell proliferation and sensitizes cells to DNA-damaging agents. PRAK participates in HR-mediated DNA damage repair in an enzyme activity-independent manner. PRAK interacts with FOXO3, influences FOXO3 nuclear localization to regulate DNA damage responses, and affects FOXO3 binding to the BRCA1 promoter to modulate BRCA1 transcriptional activation. PRAK deficiency enhances tumor sensitivity to DNA-damaging drugs. Collectively, this study reveals that PRAK promotes DNA damage repair by transcriptionally regulating BRCA1 expression through FOXO3, representing a novel regulatory mechanism by which PRAK maintains genomic stability via DNA damage repair. Targeting the PRAK/FOXO3/BRCA1 axis may serve as a potential therapeutic strategy to enhance the efficacy of DNA-damaging agents in cancer treatment. Our findings uncover the functions and molecular mechanisms of PRAK in cell proliferation and DNA damage responses, with potential clinical implications for breast cancer therapy. Keywords: PRAK; DNA damage repair; FOXO3; BRCA1; breast cancer
Presentation numberPS2-12-23
Persistent Morphological and DNA Damage Changes Following Ultra-Low Dose and Dose Rate Radiation in a 3D Human Mammary Organoid Model with Wild-Type and p16-Mutant Isogenic Lines
Janice Pluth, UNLV, Las Vegas, NV
A. Antypiuk1, M. Kang2, F. Cucinotta1, J. Pluth1; 1Health Physics and Diagnostic Sciences, UNLV, Las Vegas, NV, 2Computer Science, UNLV, Las Vegas, NV.
Radiation exposure at ultra-low doses and dose rates—such as those encountered in medical imaging or by radiation workers—may still induce persistent biological effects in human tissue. Understanding these effects in genetically relevant models is essential for breast cancer risk assessment. We used a 3D human mammary acinar organoid system and isogenic epithelial cell lines, including one with a p16 mutation commonly found in normal women without cancer, to investigate long-term consequences of radiation. Isogenic human mammary epithelial cells (wild-type and p16 mutant) were cultured in Matrigel to form polarized acini. Organoids were irradiated with X-rays at a total dose of 10 mGy, delivered at 5 or 20 mGy/h. Morphology, DNA double-strand breaks (γH2AX), and preliminary markers of oxidative damage (ROS, 8-oxo-dG) were assessed 6 days post-exposure. Even at these extremely low doses, both wild-type and p16-mutant acini showed persistent changes in size and abnormal morphology. γH2AX foci were significantly elevated in irradiated samples. Preliminary ROS and oxidative DNA damage results are underway. The impact of the p16 mutation is being further investigated for each endpoint. This study demonstrates that 10 mGy of radiation at low dose rates can produce measurable long-term effects on tissue structure and DNA integrity in 3D mammary organoids. Inclusion of a common p16 mutation increases the relevance to population-level breast cancer risk modeling and highlights the need to consider low-dose exposures in genetically diverse individuals.
Presentation numberPS2-12-24
Ifitm1 influences natural killer cell-mediated cytotoxicity by modulation of hla-class i expression in triple-negative breast cancer cells
Hye Sung Won, Uijeongbu St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Gyeonggi-do, Korea, Republic of
H. Won1, H. Lee2, C. Park3, K. Kang3, S. Hong4, Y. Ahn5, Y. Ko6; 1Internal Medicine, Uijeongbu St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Gyeonggi-do, KOREA, REPUBLIC OF, 2Internal Medicine, Cancer Research Institute, College of Medicine, The Catholic University of Kore, Seoul, KOREA, REPUBLIC OF, 3Microbiology, Dankook University, Cheonan, KOREA, REPUBLIC OF, 4Pathology, Chung-Ang University College of Medicine, Seoul, KOREA, REPUBLIC OF, 5Molecular Medicine and Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul, KOREA, REPUBLIC OF, 6Internal Medicine, Eunpyeong St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, KOREA, REPUBLIC OF.
Interferon (IFN) activates the Janus kinase (JAK)/signal transducer and activator of transcription signaling pathway, resulting in induction of IFN-stimulated genes such as IFN-induced transmembrane protein 1 (IFITM1). Previous studies have shown that the overexpression of IFITM1 is associated with a poor prognosis in triple-negative breast cancer (TNBC); however, the mechanisms by which IFITM1 contributes to oncogenesis in TNBC are unclear. This study explored the mechanism by which IFITM1 induces tumorigenesis in TNBC. We established two stable cell lines: IFITM1-overexpressing cell line (MB-231-IFITM1-OE) and IFITM1 knockdown cell line (BT-20-IFITM1-KD). The transcriptional activity of β-catenin and the cytotoxic activity of natural killer (NK) cells were evaluated using the luciferase assay and the lactate dehydrogenase cytotoxicity assay. Knockdown of IFITM1 in BT-20 cells reduced cell proliferation and ectopically expressed IFITM1 in MB-231 cells increased cell proliferation. RNA sequencing analysis revealed that IFITM1 expression positively correlated with the Wnt/β-catenin signaling pathway and NK cell-mediated cytotoxicity. MB-231-IFITM1-OE cells showed increased β-catenin transcriptional activity and NK cell cytotoxic activity compared with controls, while transient knockdown of IFITM1 in MB-231-IFITM1-OE cells led to a decrease in β-catenin transcriptional activity and NK cell cytotoxic activity. MB-231-IFITM1-OE cells exhibited decreased HLA class I expression, which may have contributed to their increased susceptibility to NK cell-mediated lysis. β-catenin or JAK inhibitor reduced NK cell-mediated cytotoxicity via upregulation of HLA class I in TNBC cells. In conclusion, IFITM1 overexpression was associated with Wnt/β-catenin signaling and NK cell-mediated cytotoxicity via downregulation of HLA class I in TNBC cells, suggesting that IFITM1 might have immunoregulatory effects on the tumor microenvironment.
Presentation numberPS2-12-25
Edil3/del-1-induced phosphorylation of ampkβ facilitates progression and correlates with poor prognosis in triple-negative breast cancer
Lee Soo Jung, Kyungpook national university chigok hospital, Daegu, Korea, Republic of
S. Jeong1, L. Soo Jung2, L. Jeeyeon3, L. In Hee2, K. Byeongju3, M. Joon Suk3, P. Ho Yong3, P. Ji Young4, P. Nora Jee-Young4, K. Eun Ae1, K. Jieun1, Y. Jung Dug5, L. Yangsoo6, J. Tae-Du7, C. Yee Soo2; 1Breast cancer precision medicine institute, Kyungpook national university chigok hospital, Daegu, KOREA, REPUBLIC OF, 2Department of Oncology/Hematology, Kyungpook national university chigok hospital, Daegu, KOREA, REPUBLIC OF, 3Department of Breast Surgery, Kyungpook national university chigok hospital, Daegu, KOREA, REPUBLIC OF, 4Department of Pathology, Kyungpook national university chigok hospital, Daegu, KOREA, REPUBLIC OF, 5Department of Plastic and Recontructive Surgery, School of Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 6Department of Surgery, Kyungpook National University School of Medicine, Daegu, KOREA, REPUBLIC OF, 7Department of Rehabilitation Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF.
BackgroundTriple-negative breast cancer (TNBC) is an aggressive subtype lacking ER, PR, and HER2, with limited targeted therapies. EDIL3/Del-1, a secreted glycoprotein enriched in extracellular vesicles, has been detected at elevated levels in TNBC, but its mechanistic contribution remains unclear. AMPK functions as a key energy sensor, and its β subunit (AMPKβ) is often amplified in cancers. This study examines Del-1’s role in regulating AMPKβ and its effects on TNBC progression and patient outcomes. MethodsAMPKβ expression in TNBC patient samples (n = 100, KNUCH) was evaluated by immunohistochemistry on tissue microarrays, and its prognostic relevance was further assessed using web-based TNBC patient cohorts (n = 126, KM Plotter). For the invitro assays, expression of Del-1 and AMPK subunits in TNBC cell lines was measured by qRT-PCR and Western blot. CRISPR/Cas9 was used to generate Del-1 knockout cells. Additionally, pharmacologic activation of AMPKβ and overexpression of a phospho-mimetic AMPKβ mutant were employed. Proliferation and invasion were assessed via MTT, BrdU, and Matrigel assays. ResultsIn our institution KNUCH TNBC patient cohorts (n = 100), elevated AMPKβ protein levels by IHC showed a trend toward shorter distant disease-free survival (p = 0.067). Consistently, in a public TNBC cohort (n = 126), high PRKAB2 mRNA levels analyzed via KM plotter were significantly associated with reduced overall survival (p = 0.0009). In parallel, Del-1 and AMPKβ were found to be upregulated in TNBC cell lines, especially within the mesenchymal stem-like subtype, while AMPKα levels remained unchanged. Functional assays demonstrated that Del-1 overexpression or pharmacological activation of AMPKβ enhanced proliferation and invasion, whereas CRISPR/Cas9-mediated Del-1 knockout reduced AMPKβ expression and abrogated these phenotypes. Mechanistically, Del-1 promoted phosphorylation of AMPKβ at Ser108, and expression of a phospho-mimetic AMPKβ mutant recapitulated the tumor-promoting effects. ConclusionsHigh AMPKβ expression in TNBC patient samples correlates with shorter distant disease-free and overall survival, underscoring its prognostic value. Complementing these clinical data, Del-1–mediated Ser108 phosphorylation of AMPKβ in mesenchymal stem-like TNBC cells promotes aggressive phenotypes, establishing AMPKβ as a promising biomarker and therapeutic target.
Presentation numberPS2-12-26
Tp53-dependent and -independent transcriptomic responses to pan-14-3-3 inhibition under dna damage in breast cancer cells
EMIKO HIRAOKA, Hiroshima Prefectural Hospital, Hiroshima, Japan
E. HIRAOKA1, H. SHIGEMATSU2, T. MARINO2, T. MOMOKO2, S. KANAKO2, F. MUTSUMI2, I. HARUKA2, A. AI2, S. SHINSUKE2, O. MORIHITO2; 1Department of Breast Surgery, Hiroshima Prefectural Hospital, Hiroshima, JAPAN, 2Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, JAPAN.
Background: The 14-3-3 protein family comprises seven isoforms (γ, ζ, β, ε, η, τ, σ) and is highly expressed in various malignancies, including breast cancer. These scaffold proteins interact with p53 in response to DNA damage, thereby regulating apoptosis and cell cycle progression. Pan-14-3-3 inhibition using difopein has been shown to enhance apoptosis when combined with chemotherapeutic agents, primarily in tumors harboring wild-type TP53. However, its effects in TP53-mutant tumors remain poorly understood. TP53 mutations are present in approximately 30% of primary breast cancers and are associated with poor prognosis, but no mutation-based therapeutic strategies have been established. The current study investigated transcriptomic responses to pan-14-3-3 inhibition under DNA-damaging conditions using RNA sequencing in TP53 wild-type and mutant breast cancer cell lines. Methods: TP53 wild-type MCF-7 and TP53-mutant MDA-MB-231 breast cancer cells were treated with 1 μM difopein for 24 hours, followed by 3 μM doxorubicin for 2 hours to induce DNA damage-associated phosphorylation. Total RNA was extracted from biological triplicates and sequenced using the DNBSEQ-G400RSQ platform. Differential gene expression analysis was performed by comparing the difopein + doxorubicin group with the doxorubicin-only control. Differentially expressed genes (DEGs) were identified, and pathway enrichment analysis was conducted using Metascape to determine pathways significantly modulated by pan-14-3-3 inhibition under DNA-damaging conditions. Results: In TP53 wild-type MCF-7 cells, 112 DEGs were identified. Upregulated genes included TNS4, STBD1, PPARA, and ZSCAN26, whereas FHL1, HERPUD1, and TXNIP were downregulated. In gene ontology (GO) analysis “positive regulation of apoptotic process” was the most significantly enriched biological process. Protein-protein interaction (PPI) network analysis identified ATF3, ATF4, and CEBPB as central hubs using the MCODE algorithm. TRRUST transcription factor analysis highlighted KLF6 and TP53, and transcription factor target analysis identified STAT3 as the most significantly enriched regulator. In contrast, in TP53-mutant MDA-MB-231 cells, 43 DEGs were identified. Upregulated genes included PPIAP22, STXBP4, and MSI2, whereas SGK1, MIDN, and EPHB3 were downregulated. In GO analysis “NGF-stimulated transcription” was the most significantly enriched process. PPI network analysis identified RIPK4, RGS16, and RGS2 as central nodes. TRRUST analysis highlighted RBMX and TP53, and transcription factor target analysis identified GTF2A2 as the most significantly enriched regulator. Conclusion: Pan-14-3-3 inhibition under DNA-damaging conditions elicited distinct transcriptomic responses depending on TP53 status. In TP53 wild-type cells, the response was characterized by activation of pro-apoptotic transcriptional programs involving ATF3, ATF4, and CEBPB, consistent with enhanced apoptotic signaling. In contrast, TP53-mutant cells exhibited a survival-adaptive response involving ceRNA regulation (PPIAP22), SGK1 suppression, and reprogramming of transcriptional and proteasomal machinery. These findings highlight subtype-specific vulnerabilities and suggest that TP53 status may inform therapeutic strategies targeting 14-3-3 proteins. Further functional validation, including apoptosis assays, is currently underway to elucidate the mechanistic consequences of these transcriptomic changes.
Presentation numberPS2-12-27
Differential Responses to the Senolytic Activities of Navitoclax and Venetoclax in Murine Models of Triple Negative Breast Cancer
Moureq Alotaibi, King Saud University, Riyadh, Saudi Arabia
H. As Sobeai1, K. Alhazzani1, M. Almutairi1, T. Saleh2, B. Al-Ramadi3, D. Gewirtz4, H. Harada5, M. Alotaibi1; 1Pharmacology and Toxicology, King Saud University, Riyadh, SAUDI ARABIA, 2Pharmacology & Therapeutics, Arabian Gulf University, Manama, BAHRAIN, 3Medical Microbiology and Immunology, United Arab Emirates University, AlAin, UNITED ARAB EMIRATES, 4Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 5Massey Cancer Center, Virginia Commonwealth University, Richmond, VA.
Therapy-induced Senescence (TIS) can potentially influence breast cancer treatment outcomes, in part by contributing to disease recurrence; hence, the utilization of senescence-eliminating agents (i.e., senolytics) is considered as a possibly adjuvant to both antitumor drugs and radiation. However, one of the most effective senolytic agents, navitoclax (ABT-263), is limited by its associated toxicities of thrombocytopenia and neutropenia. In contrast, venetoclax (ABT-199) is currently considered standard of care in CLL and AML. This study compared the senolytic potential of the BH3 mimetics, navitoclax and venetoclax, in targeting doxorubicin-induced senescence in two models of triple negative breast cancer (4T1 and E0771 cells). Senescence was cytochemically confirmed via Senescence-associated β-galactosidase upregulation (and quantified by flow cytometry), TP53 and p21Cip1 (CDKN1A) induction, and the senescence-associated secretory phenotype (SASP) expression (using qRT-PCR). Cell viability and the percentage of apoptotic cells were determined using MTT and Annexin V/7AAD assays, respectively. Responses differed in the two cell lines. Both navitoclax and venetoclax were effective as apparent senolytics in the E0771 cells. In contrast, only navitoclax was effective against the 4T1 cells. The in vitro findings in E0771 cells were validated through studies conducted in vivo in immunocompetent mice implanted with E0771-derived tumors where both drugs reduced tumor progression in sequential combination with doxorubicin. These findings suggest that administration of venetoclax has the potential to enhance suppression of chemotherapy-induced senescent cancer cells, and that it may not be necessary to use senolytic agents that target Bcl-xL. However, given the variable outcomes in the two triple negative breast tumor cell lines, it becomes incumbent to identify the factors that confer susceptibility to Bcl-2 targeting senolytics in anticipation of their potential utilization in the clinic for combination therapy in solid tumors.
Presentation numberPS2-12-28
Marveld1 expression in breast cancer and the impact on the aggressive nature of breast cancer cells
Hong Yu Xiang, Cardiff University, CARDIFF, United Kingdom
H. Y. Xiang1, H. Y. Xiang2, T. A. Martin1, K. Mokbel1, Y. H. Liu2, W. G. Jiang1; 1School of Medicine, Cardiff University, CARDIFF, UNITED KINGDOM, 2Department of Thyroid and Breast Surgery, Peking University First Hospital, Beijing, CHINA.
Introduction MarvelD1 (MARVEL (MAL and related proteins for vesicle trafficking and membrane link) domain containing-1) is a protein with multiple transmembrane helix and potential microtubule binding properties. It has been indicated to have a role in cell division and locates in the tight junctional regions of cells. Some evidence suggests that MarvelD1 plays a role in the development and progression of solid cancer but this is conflicting, namely that it exhibits a potential oncogenic role in pancreatic cancer and a tumour suppressor in hepatocellular carcinoma. There has been no report on the role of MarvelD1 in human breast cancer.Methods The expression of the MarvelD1 gene transcript and MarvelD1 protein in breast cancer were investigated by quantitative gene transcript analysis and immunohistochemistry respectively. MarvelD1 expression was analysed against clinical outcome, pathological factors, hormone receptor status and molecular subtypes of the patients. We created breast cancer cell models by knockdown the MarvelD1 with anti-MarvelD1 shRNA delivered by lentiviral approach. The biological responses following MarvelD1 knockdown were assessed including the rate of cell growth, cellular migration, cell adhesiveness and potential paracellular barrier functions of the breast cancer cells.Results Breast tumour tissues had a significantly higher level of MarvelD1 than normal mammary tissues (p=0.023). ERBB2/HER2 positive tumours had higher levels of MarvelD1 than HER2 negative tumours (p=0.0129). There was no significant difference between hormonal receptor subtypes (oestrogen receptor (ER) and progesterone receptor (PGR) positive and negative tumours). Amongst the main molecular subtypes, triple negative breast cancer demonstrated the lowest levels of MarvelD1. Patients with high levels of MarvelD1 had a significantly shorter overall survival (OS) compared with patients with low MarvelD1 (p=0.019, hazard ratio (HR) 0.304 (95% confidence interval 1.113-8.18). It is noteworthy that the OS and MarvelD1 link is more prominent in PGR negative (p=0.006), ERBB3/HER3 positive (p=0.007) and ERBB4/HER4 positive (p=0.026) tumours. We successfully created MarvelD1 knockdown breast cancer cell models by way of lentiviral based shRNA. There had been significant biological responses in cells with MarvelD1 knockdown including a markedly reduced cell growth rate, weakened matrix adhesion and cellular migration.Discussion MarvelD1, a membrane and microtubule associated protein, has a significant clinical relevance in breast cancer in which the expression levels have a link with the survival of the patients, and the link is to a degree dependent upon the receptor status. Experimental evidence further supports the connection between MarvelD1 and the aggressiveness of breast cancer cells.
Presentation numberPS2-12-29
Helz-mediated r-loop resolution orchestrates brca2-dependent dna repair in hormone-driven breast cancers
Wenjing Li, UTHSCSA, San Antonio, TX
W. Li1, B. Wu1, B. Gao2, E. M. Irvin:3, A. Arijit Ghosh2, L. Eliaz4, Y. Huang1, Y. Kwon1, C. M. Stiefel5, T. T. Nguyen5, D. Zhao1, H. J. Suarez1, T. Ni1, S. Alejo6, O. Fitzgerald1, X. Song7, E. V. Wasmuth1, S. Zheng7, J. Leung5, X. Xue4, H. Wang3, J. Ji1, L. Lan2, W. Zhao1; 1Biochemistry & Structure Biology, UTHSCSA, San Antonio, TX, 2Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, 3Department of Physics, North Carolina State University, Raleigh, NC, 4Department of Chemistry & Biochemistry Materials Science, Texas State University, San Marcos, TX, 5Department of Radiation Oncology, UTHSCSA, San Antonio, TX, 6Department of Obstetrics & Gynecology, UTHSCSA, San Antonio, TX, 7Department of Population Health Sciences, UTHSCSA, San Antonio, TX.
R-loops, transcription-induced three-stranded nucleic acid structures, play essential roles in gene regulation but jeopardize genomic integrity when unresolved. BRCA2, a key tumor suppressor required for homologous recombination (HR) repair of DNA double-strand breaks (DSBs), also maintains R-loop homeostasis, though its mechanisms remain incompletely understood. To identify BRCA2-interacting factors involved in R-loop resolution during DNA repair, we employed biotin-based proximity labeling and mass spectrometry (BioID-MS), uncovering HELZ—a previously uncharacterized RNA-specific helicase—as a direct BRCA2 binding partner. BRCA2 enhances HELZ’s catalytic activity in vitro and promotes its recruitment to transcription-associated R-loops in vivo, particularly at DSB sites. Functionally, HELZ resolves co-transcriptional R-loops, facilitates DNA end resection, and promotes efficient HR. HELZ depletion results in R-loop accumulation, impaired HR capacity, and heightened sensitivity to genotoxic stress. Notably, HELZ activity is context-dependent and proves indispensable in hormone-driven malignancies such as estrogen receptor-positive (ER⁺) breast cancers, which exhibit transcriptional dysregulation and elevated R-loop burden. These findings establish HELZ as a critical mediator of BRCA2-dependent genome integrity in R-loop-rich chromatin environments. HELZ expression or function may serve as a biomarker for therapeutic response, and HELZ helicase activity offers a promising target to restore HR proficiency in transcriptionally dysregulated breast tumors. By integrating proximity proteomics with functional assays, our study identifies HELZ as a novel and therapeutically actionable regulator of R-loop resolution and BRCA2-mediated repair in breast cancer.
Presentation numberPS2-12-30
Distribution and prognostic significance of recurrence score by histologic sub-type among women with early-stage hormone receptor-positive invasive breast cancer
MacKenzie Mayo, Karmanos Cancer Institute, Detroit, MI
M. Mayo, H. Assad, J. J. Ruterbusch, J. Boerner, S. Karne, M. S. Simon; Oncology, Karmanos Cancer Institute, Detroit, MI.
Background The Oncotype Dx Recurrence Score (RS) has been validated in TAILORx and RxPONDER as both prognostic and predictive of chemotherapy benefit in women with hormone receptor positive (HR+) breast cancer with tumor size 1-5cm and 0-3 lymph nodes involved. It is unclear whether the ability of RS to predict breast cancer outcomes varies by histologic type because a large proportion of women in these studies had invasive ductal cancer (IDC). The goal of this analysis is to compare the clinical utility of RS by histologic type at diagnosis among women treated at an NCI designated comprehensive cancer center in Detroit, with particular attention to invasive lobular histology. Methods The study population included women diagnosed between 2013 and 2022, age 18 to 79 years, with invasive, HR+, HER2 negative, AJCC stage I or II breast cancer. All women with invasive lobular carcinoma (ILC) and mixed IDC/ILC were included, with a random sample of women with IDC selected. A medical record review was conducted to supplement cancer registry data obtained from the Metropolitan Detroit Cancer Surveillance System (MDCSS). Two abstractors performed the review, with a third reviewing a random sample of charts to assess accuracy. We compared the proportion of women with low risk (RS≤25) vs high risk (RS>25) stratified by histologic group using chi-square tests. Time to distant recurrence was the primary end point, with patients censored at death or last clinic visit. Results Of the 787 women identified, we included 366 for medical record review; 140 with IDC, 92 ILC, 48 mixed IDC & ILC, and 86 other histologies. Of these, 208 (57%) had documentation of quantitative RS in the medical record and were included in the analysis. There was a significant difference in high RS by histologic group, with 25.0% of IDC, 3.6% of ILC, and 4.4% of mixed IDC/ILC having RS >25 (p=0.001). There were a total of 14 distant recurrences after a mean 6.7 years of follow-up: five recurrences among women with IDC, four ILC, and five mixed IDC/ILC. Further analyses compared women with ILC or mixed vs. IDC. For women with low RS who did not receive chemotherapy, 4% (n=2/55) of women with IDC had distant recurrence compared with 9% (n=6/69) of women with ILC or mixed. Of the women with high RS who received chemotherapy, 13% (n=2/15) of women with IDC recurred compared to 100% (n=2/2) of women with ILC or mixed. Of the women with high RS who did not receive chemotherapy, 22% (n=2/9) of women with IDC recurred compared to 100% (n=1/1) of women with ILC or mixed. Conclusion High RS was significantly more common among women with IDC compared to tumors with lobular histology in this cohort of women with early-stage invasive breast cancer. There was a suggested higher rate of distant recurrence among women with lobular histology compared to those with IDC or mixed. All women with invasive lobular histology and RS > 25 developed a distant recurrence, regardless of whether they received chemotherapy, however the total number was small. Longer follow-up is needed for additional evaluation of differences in the predictability of RS by histologic type among women with early-stage breast cancer. We have received a grant from the Dynami Foundation to continue this work.
Presentation numberPS2-05-02
Feasibility of Tailored Axillary Surgery (TAS) in Patients with Clinically Node-Positive Breast Cancer in the Upfront Surgery Setting: Results of a Prospective, Single-Arm, Multicenter Phase II Trial
Kaori Terata, Akita University Hospital, Akita, Japan
K. Terata1, Y. Sagara2, S. Yamamoto3, T. Sakai4, S. Takayama5, D. Kitagawa6, M. Ogita7, N. Sanuki8, M. Yoshida5, T. Ueno4, R. Nakamura9, H. Shigematsu10, K. Watanabe11, H. Bando12, A. Yoshimura13, T. Onishi14, E. Tokunaga15, M. Takahashi16, T. Hayashi17, S. Nishimura18, M. Saimura19, H. Matsumoto20, M. Harao21, T. Yoshiyama22, T. Sangai23, T. Ohtake24, H. Tsuda25, F. Hara13, T. Fujisawa26, H. Iwata27, T. Shien28; 1Akita University Hospital, Akita, Japan, 2Social Medical Corporation Hakuaikai Sagara Hospital, Kagoshima, Japan, 3Shizuoka Graduate University of Public Health, Shizuoka, Japan, 4Cancer Institute Hospital of JFCR, Tokyo, Japan, 5National Cancer Center Hospital, Tokyo, Japan, 6National Center for Global Health and Medicine, Tokyo, Japan, 7The University of Tokyo Hospital, Tokyo, Japan, 8Keio University School of Medicine, Tokyo, Japan, 9Chiba Cancer Center, Chiba, Japan, 10Hiroshima University Hospital, Hiroshima, Japan, 11Hokkaido Cancer Center, Sapporo, Japan, 12University of Tsukuba Hospital, Ibaraki, Japan, 13Aichi Cancer Center, Nagoya, Japan, 14National Cancer Center Hospital East, Kashiwa, Japan, 15NHO Kyushu Cancer Center, Fukuoka, Japan, 16National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan, 17Nagoya Medical Center, Nagoya, Japan, 18Shizuoka Cancer Center, Shizuoka, Japan, 19Kitakyushu Municipal Medical Center, Kitakyushu, Japan, 20Saitama Cancer Center, Saitama, Japan, 21Jichi Medical University Hospital, Tochigi, Japan, 22Kure Medical Center, Kure, Japan, 23Kitasato University Hospital, Sagamihara, Japan, 24Fukushima Medical University Hospital, Fukushima, Japan, 25Chiba Medical Center, Chiba, Japan, 26Gunma Prefectural Cancer Center, Gunma, Japan, 27Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan, 28Okayama University Hospital, Okayama, Japan
Background: Axillary lymph node dissection (ALND), while historically the standard of care for clinically node-positive (cN+) breast cancer, is associated with impaired quality of life and has been progressively de-escalated. In the upfront surgery setting for cN+ disease, ALND remains the standard of care, and evidence supporting its omission is lacking. Tailored axillary surgery (TAS) is a novel approach that targets suspicious nodes while omitting ALND, aiming to reduce morbidity. We conducted a prospective phase II study to assess the feasibility of TAS, ensure procedural consistency, and evaluate associations between clinicopathologic factors and residual tumor burden. Patients and Methods: This single-arm, multicenter phase II trial (jRCTs061220113) was conducted across 42 institutions in Japan within the JCOG Breast Cancer Study Group. TAS was defined as the removal of marked lymph node (LN) with clip, wire, or tattoo, sentinel lymph nodes (SLNs), and palpable LNs, and was followed by completion ALND in accordance with the trial protocol. Eligible patients had invasive breast cancer undergoing upfront surgery, pathologically confirmed axillary metastasis, 1-3 suspicious nodes confined to level I axilla on imaging, and cT1-T3 tumors. The primary endpoint was the proportion of patients with positive non-TAS LNs (non-TAS LN positivity rate). Clinicopathologic factors (cT stage, the number of suspicious nodes on imaging, the number of TAS nodes retrieved, and the number of positive nodes among them) were analyzed using multivariable logistic regression to explore factors associated with non-TAS LN positivity. Secondary endpoints included the retrieval of marked nodes, the TAS identification rate (defined as≥1 metastatic node among TAS nodes), and complications. Results: Between 2023 and 2025, a total of 212 patients were enrolled. The median age was 53.5 years (range, 27-77). Clinical T stage: T1 27.4%, T2 66.0%, T3 6.6%. ER positive in 94.8%, HER2 positive in 3.3%. Histologic grade: G1-2 in 66.0%, G3 in 29.7%. Pathological nodal stage: N1 65.1%, N2 26.4%, N3 8.5%. Pathological stage: IIA 23.6%, IIB 47.6%, IIIA-C 28.8%. Marked node retrieval was achieved in 100% of patients, although 30% of the marked nodes lacked SLN tracer uptake. The median number of TAS nodes retrieved was 4 (IQR, 3-6; range, 1-11), including 2 metastatic nodes (IQR, 1-3). Median number of LNs retrieved by ALND was 18 nodes (IQR 14-24). The identification rate of TAS was 100%. The non-TAS LN positivity rate was 38.7% (82/212). In multivariable analysis, a smaller number of TAS LNs retrieved (OR 0.85; 95% CI 0.73-0.99) and more positive TAS LNs (OR 1.50; 95% CI 1.17-1.94) were associated with higher non-TAS LN positivity, while clinical T stage and number of suspicious nodes on imaging were not significant. The major complications included post-operative bleeding (4.3%) and infection (3.7%). Conclusions: TAS was feasible in cN+ breast cancer undergoing upfront surgery, with acceptable safety. The non-TAS LN positivity rate was slightly higher but comparable to the false-negative rates reported in cN0 SLN-positive trials, while the axillary tumor burden in the present cN+ cohort was higher than that in those trials. These findings support the evaluation of ALND omission as a rational next step, supported by systemic therapy and radiotherapy, and provide the basis for our planned Phase III trial within the framework of stepwise axillary de-escalation.
Presentation numberPS2-05-12
Axillary Sentinel Lymph Node Biopsy After Neoadjuvant Chemotherapy in Clinically Node-Negative Early Breast Cancer: Feasibility and Safety in Triple-Negative and HER2-Positive Breast Cancer Subtypes
Qijun Zheng, Peking University Cancer Hospital & Institute, Peking, China
Q. Zheng, X. Wang, Y. He, W. Cao, Y. Yang, C. Gu, L. Wang, X. Wang, J. Li, T. Ouyang, Z. Fan; Peking University Cancer Hospital & Institute, Peking, CHINA.
Background: Axillary sentinel lymph node biopsy (SLNB) is the standard procedure for axillary staging in patients with clinically node-negative (cN0) early breast cancer (BC). Neoadjuvant chemotherapy (NACT) achieves a pathologic complete response (pCR) in 30-50% of patients with biopsy-confirmed N1 disease. Targeted axillary dissection (TAD) has been shown to reduce false-negative rates in clinically node-positive (cN+) patients following NACT. While triple-negative breast cancer (TNBC) and HER2-positive BC exhibit high response rates to NACT (potentially leading to ypN0 status), the feasibility and safety of SLNB after NACT in cN0 patients with TNBC or HER2-positive BC remain insufficiently established by high-quality evidence. Objective: This study aimed to evaluate the feasibility of SLNB following NACT in patients with clinically node-negative TNBC and HER2-positive early BC, and to assess the oncologic safety of omitting axillary lymph node dissection (ALND) in SLNB-negative patients via long-term follow-up. Methods: A prospective single-arm trial was conducted on 404 patients with primary early breast cancer (BC, cT1b-2N0M0, age ≤70 years) who were enrolled and received neoadjuvant chemotherapy (NACT) at Peking University Cancer Hospital from October 2017 to May 2023. Inclusion criteria were: histologically confirmed invasive carcinoma, indication for chemotherapy, and eligibility for SLNB before NACT (including cases with suspicious lymph nodes but negative results on fine-needle aspiration [FNA] or core needle biopsy [CNB]). Exclusion criteria included: history of prior malignancies, contraindications to chemotherapy, prior axillary surgery, or refusal of NACT, evaluation, or study participation. Tumor subtypes were categorized as HER2-negative/hormone receptor < 10% (n=147) and HER2-positive (n=242). NACT regimens were heterogeneous, including protocols such as TCbH(P) and ddEC-T±H(P). SLNB was performed using a technetium-99m-labeled tracer. Results: The success rate of SLNB was 95.63% (95% confidence interval [CI]: 93.60%-97.66%). Among 372 successful SLNB cases (median number of lymph nodes retrieved: 2, range: 1-10), 9 patients had positive SLNs, with a SLN-positive rate of 2.42% (95% CI: 0.86%-3.98%). Univariate analysis showed that ultrasound (US) T response, magnetic resonance imaging (MRI) T response, number of NACT cycles, Ki-67 expression level, and breast tumor pCR were factors influencing lymph node status (p < 0.5). Multivariate analysis identified US T response as an independent significant factor (p=0.041). During a median follow-up of 43 months (range: 6-85 months), 1 patient developed ipsilateral axillary recurrence, with an ipsilateral axillary recurrence rate of 0.28% (95% CI: 0.05%-1.54%) among 363 SLN-negative patients who did not undergo ALND. Conclusion: SLNB following NACT in patients with clinically node-negative TNBC and HER2-positive early BC shows high success rates and low positivity rates. Omitting ALND in SLN-negative patients appears feasible, with a low risk of axillary recurrence, indicating potential oncologic safety. However, longer follow-up durations and larger-scale studies are required to validate the long-term outcomes of this approach.
Presentation numberPS2-05-13
Evaluation of Axillary Treatment in Patients with Early Breast Cancer According to Study Z0011 in a Public Tertiary Hospital in the Federal District, Brazil
Thais Karla Vivan, Rede Americas, Brasilia, Brazil
T. K. Vivan1, M. Passos2, V. X. Santana2, F. C. Salum2, C. Fuschino2, A. F. Esterl2, R. Pepe1; 1Rede Americas, Brasilia, Brazil, 2Hospital de Base Distrito Federal HbDF, Brasilia, Brazil
IntroductionThe radical mastectomy technique described by Halsted in 1894 was based on the concept that more extensive surgical resection increased the likelihood of patient cure. Over more than 120 years, medical knowledge about breast cancer has expanded, enabling less mutilating treatments with equal or better survival rates, especially following studies on tumor biology. Axillary surgical treatment has been a significant milestone, changing medical management and reducing the rates of axillary lymph node dissection after the publication of the American College of Surgeons Oncology Group Z0011 clinical trial, that demonstrated that patients with initial cT1-T2 tumors without palpable axillary adenopathy, undergoing breast-conserving surgery, systemic adjuvant treatment, and radiotherapy, maintained overall survival and distant recurrence-free survival rates, even with 1 or 2 metastatic axillary lymph nodes, compared to those who underwent axillary dissection.ObjectivesTo evaluate the applicability of the Z0011 protocol in the axillary treatment of early invasive breast cancer and its impact on reducing axillary lymph node dissection by omitting this surgical treatment.MethodThis is an observational, descriptive, retrospective study based on data collected from the medical records of patients who underwent surgical treatment at a public hospital, Hospital de Base, in the Federal District, Brazil.ResultsA total of 119 patients from the mastology service at Hospital de Base met the Z0011 eligibility criteria. Nine patients were excluded due to failure in patent blue migration, resulting in 110 patients included in the study. The average age of patients was 58 years (ranging from 35 to 83 years). The predominant immunohistochemical profile was luminal B (55%), followed by luminal A (30%), HER2 positive (9%), and triple-negative (6%). Among the HER2 positive patients, 80% also had positive hormone receptors (HR), while 20% were HR negative. Among the 110 patients, 27% had sentinel lymph node (SLN) metastasis. Specifically, 19% had metastasis in 1 SLN, 3% had metastasis in 2 SLNs, and 5% had metastasis in 3 or more SLNs. Consequently, 83% of patients with 1 or 2 metastatic SLNs did not undergo lymphadenectomy, representing a significant omission of axillary surgical treatment.ConclusionThe successful replication of the Z0011 protocol in our study demonstrates the significant potential for reducing axillary lymphadenectomy in patients treated at our hospital, with an observed 83% reduction in axillary dissection among patients with axillary lymph node metastasis. These findings are particularly important as they underscore the feasibility of implementing the Z0011 protocol in a public hospital setting, providing evidence that even in resource-constrained environments, it is possible to achieve substantial improvements in patient care. KeywordsBreast cancer; Sentinel lymph node; Axillary metastases; Lymphadenectomy.
Presentation numberPS2-05-14
Omitting Sentinel Lymph Node Dissection After Neoadjuvant Treatment in Early Breast Cancer: Interim Results from the VENUS Randomized Clinical Trial
Giuliano M Duarte, State University of Campinas – UNICAMP, CAMPINAS, Brazil
M. P. L. Kraft1, G. M. Duarte1, D. M. Araújo1, R. M. Jales1, J. Y. Shinzato1, R. Z. Torresan1, C. Cardoso Filho1, F. P. Brenelli1, S. B. Esteves1, H. K. Mantovani1, G. M. Tavares1, A. S. Detoni1, E. C. Pessoa2, C. K. C. Pessoa2, I. de Oliveira Júnior3, R. Freitas Júnior4, R. M. S. Rahal4, V. M. Budel5, L. R. Budel5, A. P. S. Damin6, L. R. Soares7, R. D. J. A. de Andrade8, F. P. Cavalcante9, M. Antonini10, R. S. Machado11, D. L. Ferreira Filho12, K. B. F. Diocesano13, S. Vieira14, A. Mattar15, T. Giuzio16, L. Z. Sarian1; 1State University of Campinas – UNICAMP, CAMPINAS, Brazil, 2São Paulo State University – UNESP, Botucatu, Brazil, 3Hospital de Amor, Barretos, Brazil, 4Federal University of Goiás – UFG,, Goiânia, Brazil, 5Federal University of Paraná – UFPR, Curitiba, Brazil, 6Federal University of Rio Grande do Sul, Hospital de Clinicas de Porto Alegre, Porto Alegre, Brazil, 7Dona Iris Maternity Hospital, Goiânia, Brazil, 8Hospital Liga contra o Câncer de Natal, Natal, Brazil, 9Fortaleza General Hospital, Fortaleza, Brazil, 10Hospital do Servidor Público do Estado de São Paulo, São Paulo, Brazil, 11Federal Hospital Lagoa, Rio de Janeiro, Brazil, 12Hospital Barão de Lucena, Recife, Brazil, 13Federal University of Piauí – UFPI, Teresina, Brazil, 14Oncocenter, Teresina, Brazil, 15Hospital da Mulher, São Paulo, Brazil, 16Pontificia Universidade Católica de Campinas, CAMPINAS, Brazil
Introduction: Sentinel lymph node dissection (SLND) omission after neoadjuvant therapy (NAT) in early-stage breast cancer has not yet been studied, because most SLND omission trials do not allow recruitment of patients who undergo NAT. The VENUS trial (ClinicalTrials.gov NCT05315154, ReBEC RBR-8g6jbf, Ethics approved: CAAE:06805118.2.0000.5404) is an ongoing study that aims to evaluate the safety of omitting sentinel lymph node dissection in clinically and ultrasound node-negative early breast cancer (BC). The trial permits neoadjuvant treatment (NAT). Objectives: Evaluate the characteristics of patients enrolled in the VENUS trial until September 25th 2025, who underwent NAT and compare them to the other patients enrolled, in order to determine if it is safe to maintain the recruitment of women with early-stage breast cancer and neoadjuvant treatment indication. Methods: Descriptive interim exploratory analysis of the patients enrolled on the VENUS trial until September 25th 2025. The VENUS trial is a multicenter, prospective, non-inferiority, phase 3 clinical trial that includes women with T1/T2, N0 (clinical/ultrasound), M0 breast cancer, randomized to SLND or no axillary surgery (NAS). Initial treatment could be NAT or upfront surgery, and the axillary ultrasound must be negative before and after the neoadjuvant treatment (analysis groups: NAT-SLND, NAT-NAS, up-front-SLND, up-front-NAS). This interim analysis focuses on patient and tumor characteristics, SLND positivity, and axillary recurrence.Results: So far, 788 women were enrolled, with 287 randomized to SLND and 273 to NAS. Patients’ epidemiological characteristics (body mass index, race and menopausal status) were similar in all randomization groups, but those who underwent NAT were younger (NAT-SLND and NAT-NAS mean ages were 53.4 and 53.7 years old versus up-front-SLND and up-front-NAS of 59.7 and 62.7 years old, p=0.01). Among all randomized patients, 45 (8%) underwent NAT, of which 25 had SLND performed and 20 no axillary surgery. They had more aggressive tumor profiles (37.7% HER2 positive tumors and 33.3% triple-negative tumors) and tumors’ mean size of 28 mm. SLND positivity seems to be lower after NAT compared to upfront surgery (4.2% vs. 19.9%, p=0.1048), though this difference did not reach statistical significance. No axillary recurrences were observed during the 22-month follow-up. Conclusions: This interim analysis suggests that SLND positivity is lower after NAT and that omitting SLND in this context may be oncologically safe, with no axillary recurrences observed over 22 months of follow-up. However, the small NAT sample limits statistical power, and continued data collection is needed to confirm safety and feasibility. At this stage, continuing the inclusion of patients with indication for NAT in the VENUS trial appears justified. Recruitment will proceed through December 2025.
Presentation numberPS2-05-15
Outcome of De-Escalation of Axillary Surgery in Locally Advanced Breast Cancer Patients
Brijesh Kumar Singh, All India Institute Of Medical Sciences New Delhi, New Delhi, India
B. K. Singh, M. Sangade, R. mukherjee, A. Krishna , S. Soren, V. Seenu, A. Batra, S. Mathur; All India Institute Of Medical Sciences New Delhi, New Delhi, India
Title: Outcome of De-Escalation of Axillary Surgery in Locally Advanced Breast Cancer PatientsBackground: Evidence is supporting de-escalation of axillary surgery in early breast cancer; however, the role of the same in locally advanced breast cancer (LABC) responding to neo-adjuvant chemotherapy (NACT) is unclear.Objective: To evaluate the outcome of cT2-4N1M0 patients undergoing sentinel lymph node biopsy (SLNB) alone who become ycT0-4N0M0 following NACT.Methods: Digital and medical case records of eligible patients registered at Breast Cancer Clinic were assessed retrospectively for the year 2022-2023 and prospectively for 2024. Clinical presentation, results of cytology, histology, treatment received and follow-up were noted in a predesigned proforma. After SLNB, patients with positive SLN showing macro metastasis or those in whom a sentinel lymph node (SLN) could not be identified were subjected to axillary lymph node dissection (ALND). The primary outcome of the study was composite outcome of mortality, locoregional, or distant recurrence rates, overall survival (OS) and disease-free survival (DFS) in overall population and their comparison between the patients undergoing SLNB alone vs SLNB followed by ALND. Data were analyzed by SPSS version 25.Results: A total of 82 breast cancer LABC patients with ycN0 after NACT were included. All the patients had a dual mapping procedure with either blue dye and indocyanine green [54 (67.5)] or blue dye with Technetium [26 (32.5%)] for SLNB. A sentinel lymph node was identified in 77 (93.9%) cases of which 24 (31.2%) were positive (Table 1). Of 82 cases, a total of 25 (30.4%) patients underwent ALND either because of positive SLN showing macrometastasis (n=20) or non-identification of SLNs (n=5). Median (IQR) lymph nodes removed during SLNB and ALND were 3(2–4) and 13(15–19), respectively. All these patients were followed-up for a median (IQR) duration of 21(15-29) and range (9-44) months. Composite outcome, locoregional recurrence and distant metastasis occurred in 7.0%, 3.5% and 1.8%, respectively in 57 patients undergoing SLNB alone. In the ALND group (n=25), 12.0%, 4.0% and 4.0% patients developed composite outcome, locoregional recurrence and distant metastasis, respectively. These outcomes remained comparable in SLNB and ALND groups. Mean (95%CI) OS was 42.7 (40.3–45.1) months and 37.8 (35.7–39.9) months in SLNB and ALND treated patients, respectively (p=0.617). There was no significant difference in DFS duration between the groups [DFS (months), SLNB vs ALND: 40.4(37.3–42.6) vs 35.3(31.6–39.0), respectively, p=0.414] also.Conclusions: The short-term outcomes suggest low rates of composite, locoregional/ distant recurrences, OS and DFS in LABC patients with ycN0 disease after NACT treated with SLNB followed by regional nodal irradiation supporting axillary nodal de-escalation surgery in this cohort.
Presentation numberPS2-05-27
Omission of Axillary Surgery in Early Breast Cancer with Negative Lymph Nodes: A Systematic Review and Meta-Analysis of Randomized Clinical Trials
Giuliano M Duarte, State University of Campinas – UNICAMP, CAMPINAS, Brazil
B. B. Castelo, L. O. Brito, R. Z. Torresan, C. Cardoso Filho, G. M. Duarte; State University of Campinas – UNICAMP, CAMPINAS, Brazil
Objective: To evaluate whether the omission of axillary surgery impacts clinical outcomes in patients with early-stage breastcancer and clinically negative lymph nodes. Methods: We conducted a systematic review and meta-analysis of randomized clinical trials (RCTs) comparing no axillarysurgery with standard axillary interventions (sentinel lymph node biopsy [SLNB] or axillary dissection [AD]). This study followed PRISMA guidelines and was registered in PROSPERO (CRD420250653779). Searches were conducted in PubMed,Web of Science, and Embase through June 2025. Outcomes assessed included overall survival (OS), disease-free survival (DFS),and axillary recurrence (AR). Meta-analyses were performed using RevMan 5.4. Risk of bias was assessed using the RoB 2 tool. Results: Out of 853 records, seven RCTs including 8,806 patients met the inclusion criteria. Among them, 2,915 patients underwent no axillary surgery, while 5,891 received surgical axillary treatment. Two trials compared no surgery with SLNB, and five compared no surgery with AD. No significant differences were found in OS (OR = 1.02; 95% CI, 0.86-1.20; p = 0.84; I² =36%) or DFS (OR = 0.80; 95% CI, 0.63-1.00; p = 0.05; I² = 63%). AR was significantly lower in the axillary surgery group (OR= 0.18; 95% CI, 0.10-0.31; p < 0.01; I² = 39%). Conclusion: The omission of axillary surgery in early-stage breast cancer with clinically negative lymph nodes does not negatively impact overall or disease-free survival. However, it is associated with a higher—though still low—risk of axillary recurrence.
Presentation numberPS2-05-28
High-resolution specimen PET-CT imaging for intraoperative margin assessment in early-stage breast cancer: the multicenter BrIMA study.
Menekse Göker, University Hospital Ghent, Gent, Belgium
M. Göker1, O. Gentilini2, G. Vergauwen1, H. Markus3, S. Kümmel4, G. Cisternino5, B. Lambert6, X. Kraemer6, M. Vanhoeij7, V. Vergucht6, R. Di Micco2; 1University Hospital Ghent, Gent, Belgium, 2IRCCS Ospedale San Raffaele, Milan, Italy, 3Universitätsklinikum Tübingen, Tübingen, Germany, 4Kliniken Essen-Mitte, Esse n, Germany, 5IRCCS Ospedale San Raffaele, Milan, Germany, 6Maria Middelares, Gent, Belgium, 7UZ Brussel, Brussel, Belgium
Goals: Approximately 12-30% of patients with breast cancer undergoing breast-conserving surgery (BCS) require additional surgery due to positive margins, delaying adjuvant therapy and increasing local recurrence risk. Therefore, finding a time- and cost-effective method for intraoperative margin assessment (IMA) is crucial to avoid reoperation. The prospective, multicenter, interventional BrIMA study evaluated the clinical value of intraoperatively addressing positive margins by specimen PET-CT imaging. Methods: Patients received a preoperative intravenous low-dose radiotracer injection (18F-FDG; 0.8 MBq/kg) at the nuclear medicine department and were subsequently transported to the operating room (OR). Immediately after tumor excision, the breast specimen was imaged in the OR using a compact and mobile high-resolution specimen PET-CT imager (AURA10 PET-CT, XEOS, Belgium). The operating surgeon evaluated the 3D PET-CT images during BCS. When positive margins were suspected on imaging, additional breast tissue had to be excised to achieve final negative margins (i.e. oriented cavity shaving). Routine IMA methods (e.g. specimen X-ray, gross pathology, specimen ultrasound, palpation) were permitted only after PET-CT image interpretation was completed. All specimens were then sent to the pathology department for routine histopathological evaluation. The clinical value of intraoperative specimen PET-CT imaging was quantified by the success rate. Surgery was considered successful when all margins of the main specimen were negative, or when positive margins were adequately addressed during BCS; whereas failure was defined as positive margins not addressed during BCS. Histopathology served as the gold standard. Results: The BrIMA study was conducted in six European breast centers. A total of 148 study patients were found eligible for analysis. The final analysis cohort consisted of patients with Invasive Ductal Carcinoma (IDC; n=84), IDC with preoperative aromatase inhibitor therapy (IDC-AI; n=15), IDC with neoadjuvant chemotherapy (NAT; n=13), Invasive Lobular Carcinoma (ILC; n=16), ILC with preoperative aromatase inhibitor therapy (ILC-AI; n=7) and Ductal Carcinoma In Situ (DCIS; n=13). Specimen PET-CT imaging visualized breast tumors across all subtypes, including less 18F-FDG-avid lesions (i.e. ILC, DCIS) and tumors from patient treated with neoadjuvant therapy. Resection margins were evaluable in all tumor specimens (148/148) during BCS. Across all study groups, considering both the invasive and the in situ component, the success rate improved from 76.4% (113/148) without IMA to 81.8% (121/148; p = 0.004) with the routine IMA technique and to 91.9% (136/148; p < 0.001) with specimen PET-CT imaging. For the invasive component of IDC, the success rate in addressing positive margins increased from 83.3% (70/84) without IMA to 86.9% (73/84; p = 0.125) with the hospital’s routine IMA technique and to 95.2% (80/84; p < 0.001) with specimen PET-CT imaging. Fifteen patients (15/148; 10.1%) required reoperation due to final positive margins. Five of these patients had positive margins in the cavity shaves, which were not assessed by specimen PET-CT imaging per study protocol and one patient required reoperation due to postoperative MRI findings. The other nine patients that underwent reoperation could be attributed to the failure of obtaining final negative margins (9/148; 6.1%). For these cases the positive margin of the main tumor specimen was neither addressed by intraoperative specimen PET-CT imaging, nor by the routine IMA methods. Conclusions: Specimen PET-CT imaging provides reliable intraoperative visualization across breast cancer subtypes and outperforms routine IMA methods, yielding clinically relevant improvements in margin assessment during BCS.
Presentation numberPS2-05-29
Feasibility and outcomes of premastectomy radiation therapy in locally advanced breast cancer
Risa Kiernan, Memorial Sloan Kettering Cancer Center, New York, NY
R. Kiernan, V. Sevilimedu, A. Xu, M. Corriddi, L. Hor, A. Pellecchia, M. Morrow, A. Khan, A. Tadros; Memorial Sloan Kettering Cancer Center, New York, NY
Introduction: There is growing evidence to support the oncologic feasibility of pre-mastectomy radiotherapy (PreM-RT) to allow immediate breast reconstruction (IBR). Most trials excluded patients with locally advanced breast cancer (LABC), cT3-4N0-3M0. Generally, LABC patients are recommended trimodality therapy with neoadjuvant chemotherapy (NAC) followed by surgery, and ultimately radiation therapy (RT). Patients who undergo mastectomy are recommended delayed reconstruction to decrease postoperative complications and prevent RT delays. We aimed to assess the feasibility of PreM-RT followed by mastectomy with IBR for LABC. Methods: This was a prospective, single arm, feasibility trial. Women age ≥18 with cT3-T4N0-N3M0 breast cancer with partial or complete response to NAC by RECIST criteria, and desired IBR were enrolled beginning 01/2022. All patients received NAC and PreM-RT. The primary endpoint was postoperative wound complication rate. Secondary endpoints reported here include physician reported RT toxicities at baseline and weekly during PreM-RT using the NCI CTCAE v5, as well as patient reported outcomes at baseline, during RT, preoperatively, and postoperatively using the complementary PRO-CTCAE. Results: Among 27 patients, median (IQR) age was 42 (37-57) years (1 Asian [3.8%], 5 Black [19], 16 White [62%], and 6 Hispanic [24%]). 48% (13/27) were ER+/HER2-, 30% (8/27) were ER-/HER2-, and 52% (14/27) had inflammatory breast cancer (IBC). 25/27 patients completed PreM-RT and surgery at this time, and all had autologous reconstruction. Median (IQR) time from end of NAC to start of RT was 38 days (32-42); median (IQR) time from end of RT to surgery was 34 days (30-40.5). Dosing of PreM-RT was 50 Gy in 25 daily 2-Gy fractions over 5-6 weeks (24/25, 96%) or 42.56 Gy in 16 daily 2.66-Gy fractions over 3-4 weeks (1/25, 4%). Most prevalent physician reported RT toxicities were dermatitis (25/25, 100%) and fatigue (25/25, 100%), with the majority grade 1-2 toxicities (Table). Grade 3-4 toxicities were only reported for dermatitis (7/25, 28%), fatigue (3/25, 12%), and breast lymphedema (1/2, 50%). The most common patient reported RT toxicities were pain, fatigue, skin breakdown, and breast enlargement (Table). 12% (3/25) of patients developed postoperative complications. 2 had flap skin necrosis; 1 required bedside debridement and 1 resolved without intervention by postoperative month 3. The third had a delayed hematoma at postoperative week 5 after resuming therapeutic anticoagulation and required washout. Conclusion: PreM-RT followed by mastectomy with IBR for patients with LABC appears safe, with postoperative complication rates lower than reported for delayed autologous reconstruction (~30%). This is among the first reports of PreM-RT in IBC patients. Patients tolerated PreM-RT with no surgical delay, and physician/patient reported RT toxicities mirror the postmastectomy RT population.
| Physician-reported radiation toxicities | ||||||
| Toxicity | Overall (n=25) | Grade I | Grade II | Grade III | Grade IV | |
| Dermatitis | 25 (100%) | 5/25 (20%) | 13/25 (52%) | 5/25 (20%) | 2/25 (8%) | |
| Fatigue | 25 (100%) | 19/25 (76%) | 3/25 (12%) | 2/25 (8%) | 1/25 (4%) | |
| Nausea | 1 (4%) | 1/1 (100%) | ||||
| Dizziness | 1 (4%) | 1/1 (100%) | ||||
| Fibrosis | 6 (24%) | 5/6 (83%) | 1/5 (20%) | |||
| Decreased ROM | 8 (32%) | 7/8 (88%) | 1/8 (13%) | |||
| Sensory neuropathy | 6 (24%) | 5/6 (83%) | 1/6 (17%) | |||
| Chest wall pain | 4 (16%) | 1/4 (25%) | 3/4 (75%) | |||
| Secondary malignancy | 0 (0%) | |||||
| Telangiectasia | 2 (8%) | 2/2 (100%) | ||||
| Lymphedema | 2 (8%) | 1/2 (50%) | 1/2 (50%) | |||
| Brachial Plexopathy | 1 (4%) | 1/1 (100%) | ||||
| Patient-reported radiation toxicities | ||||||
| Toxicity | RT week 4-5 | RT week 4-5 | Post RT weeks 1-2 | Post RT weeks 1-2 | Postoperative weeks 1-2 | Postoperative weeks 1-2 |
| Mild-moderate | Severe | Mild-moderate | Severe | Mild-moderate | Severe | |
| Pain | 6/9 (67%) | 2/9 (22%) | 9/16 (56%) | 6/16 (38%) | 5/10 (50%) | 2/10 (20%) |
| Fatigue | 5/9 (56%) | 3/9 (33%) | 13/16 (81%) | 2/16 (12.5%) | 10/17 (59%) | 2/17 (11%) |
| Skin breakdown | 5/9 (56%) | 1/9 (11%) | 7/16 (44%) | 5/16 (31%) | 5/6 (83%) | 1/6 (17%) |
| Breast enlargement | 8/11 (73%) | 1/11 (9%) | 8/12 (67%) | 1/12 (8%) | 4/4 (100%) | 0/4 (0%) |
Presentation numberPS2-05-30
Long-term Outcome Prediction with Residual Cancer Burden (RCB) post Neo-adjuvant Chemotherapy in Breast Cancer patients – LOPNeoRCB: An ambispective study
Brijesh Kumar Singh, All India Institute Of Medical Sciences New Delhi, New Delhi, India
A. P. Singh, B. K. Singh, A. Krishna , S. Mathur, V. Seenu; All India Institute Of Medical Sciences New Delhi, New Delhi, India
Aim: To study the utility of RCB in predicting outcomes of breast cancer after neoadjuvant chemotherapy Methodology: Patients of carcinoma breast (stage 1, 2 and 3) treated in the Surgery Department after neo-adjuvant chemotherapy (NACT) from Jan 2016 to December 2023 were followed up for event free survival (EFS), distant relapse free survival (DRFS) and overall survival (OS). Demographic details, clinical course, treatment records, histopathology reports and follow-up records were collected through available digitalised hospital data, breast cancer clinic files and phone calls. Results: From January 2016 through December 2023, 654 patients were diagnosed with primary breast cancer, who received NACT upon multi-disciplinary team meeting in the Department of Surgical Disciplines of All India Institute of Medical Sciences, New Delhi, India. 124 cases were excluded. Most common molecular subtypes were HR+/Her2 Neu negative 193(36.4%) followed by Triple Negative Breast Cancer (TNBC) 156(29.4%), Triple positive breast cancer (TPBC) 91(17.2%) and HR negative/Her Positive 83(15.7%). Most of the patients had T4 disease 241(45.5%). Out of 530 patients, 335 (63.2%) had N1 disease followed by N0, N2 and N3, respectively. Most of the patients, i.e, 242 (45.6%) received anthracycline/taxane-based NACT. Following NACT, 244(46%) patients had partial response followed by complete response in 160(30%), no response in 114(21.5%) and progressive disease in 12 (2.3%) patients. 461 (87%) patients underwent mastectomy and 69(13%) underwent breast conserving surgery. In our cohort, majority of patients were negative for LVI, PNI, ENE and DCIS. Out of 323 patients with complete final histopathology with RCB, 118(36.5%) showed pCR/RCB0, RCB-2 in 99(30.6%), RCB-3 in 90(27.9%) and RCB-1 in 16(4.9%) patients. Out of 446 patients, with Miller-Payne(MP) grading, Grade5 response was seen in 143(32.1%), Grade4 in 67(15%), Grade3 in 152(34.2%), Grade2 in 63(14.2%) and Grade1 in 20(4.5%) patients. 470 (90%) patients received adjuvant radiotherapy. Hormonal therapy was received by 288(54.3%). 6-year EFS for RCB-0 was 96.6% vs RCB-3(71.1%) with HR of 9.83 (p=0.000, CI, 3.33-29.04), however there was no statistically significant difference between 0/pCR and RCB-1(p=0.07). For the MP system, prognosis of G5 was good. Lower the grade, worse the prognosis. Patients with G5 showed 95.1% 6-year’s EFS in comparison to G4-83.6%, G3- 80.3%, G2 -84.1%, G1-90% with hazards ratio (HR) of 3.11 (p=0.019, CI, 1.20-8.03), 4.16(p=0.001, CI, 1.81-9.57), 5.11 (p=0.001, CI, 1.91-13.6) and 2.05 (p=0.299, CI, 0.53-7.97), respectively. On subgroup analysis, HR-/Her2 Neu positive and TNBC were associated with worse 6- year EFS of 79.8% and 85.3%, respectively, in comparison to HR+/Her2 Neu Negative (95.1%) with increased HR, 1.91(p=0.041, CI, 1.02-3.56) and 1.34 (p=0.307, CI, 0.76-2.39) respectively. Patients with TPBC were having good EFS rate in comparison to HR+/Her2 Neu negative with HR 0.54 (p=0.213, CI, 0.20-1.42), but it wasn’t statistically significant. In the whole cohort and all the molecular subtypes, the C-index was higher for RCB system in comparison to MP system except in HR-/Her2 positive in consideration of EFS and in HR+/Her 2 neu negative while considering for OS, which is suggesting that overall, the RCB was better than MP grading system in predicting long term survival outcomes in terms of EFS, DRFS and OS. RCB was better than MP in the whole cohort and HR+/Her2Neu negative while considering EFS and difference was statistically significant with p =0.04 on comparing the C- index of RCB and MP grading system with z-score test. Conclusion: The RCB score demonstrates superior prognostic accuracy over the MP grading system in predicting eEFS, DRFS and OS.
Presentation numberPS2-04-24
Factors Impacting Local Relapse Risk and Disease-Free Survival of Early Breast Cancer receiving Intraoperative Radiation Therapy in Taiwan
Shen Liang Shih, Division of Breast Oncology & Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast Surgery, Kaohsiung Medical University Gangshan Hospital; Center of Big Data Research, Kaohsiung Medical University, Kaohsiung, Taiwan
S. Shih1, H. Takahashi2, F.-M. Chen2, J.-Y. Kan2, L.-C. Kao2, P.-F. Yang3, C.-L. Li2, C.-N. Kao2, C.-Y. Kuo4, C.-H. Chuang4, M.-F. Hou2, J.-Y. Tang5; 1Division of Breast Oncology & Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast Surgery, Kaohsiung Medical University Gangshan Hospital; Center of Big Data Research, Kaohsiung Medical University, Kaohsiung, Taiwan, 2Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan, 3Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast Oncology and Surgery, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan, 4Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast oncology and Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan, 5Department of Radiation oncology, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung, Taiwan
Introduction: Intraoperative radiation therapy (IORT) delivered during breast-conserving surgery may streamline treatment and enhance cosmetic outcomes. However, optimal patient selection criteria remain uncertain, especially in Asian populations, where international guidelines may not be fully applicable. This study aimed to evaluate local recurrence rates, identify key prognostic factors, and assess the impact of supplemental external beam radiation therapy (EBRT) in Taiwanese patients receiving IORT. Methods: This retrospective analysis included 1,306 Taiwanese patients who underwent IORT between June 2014 and December 2021. Patients were stratified into low-risk and high-risk groups based on institutional criteria. Low-risk patients were those meeting all of the following: age ≥ 45 years, tumor size ≤ 3.5 cm, negative nodal involvement, Ki-67 ≤ 30%, hormone receptor (HR) positivity, and HER2 negativity. Patients who did not meet one or more of these criteria were classified as high-risk and were recommended to receive supplemental external beam radiation therapy (EBRT) following IORT. Results: In our cohort of 1,306 patients, the 3-year disease-free survival rate was 93.11% (95% CI: 91.6%-94.4%), with 87 patients (6.89%) experiencing recurrence. The median age was 52 years, and the median follow-up duration was 30.5 months. The overall local recurrence rate was 9.3%, which declined to 1.78% after excluding patients who did not receive indicated adjuvant therapies. Multivariable analysis identified Ki-67 >30%, omission of hormone therapy, and omission of recommended EBRT as significant predictors of local recurrence, whereas age <45 was not independently associated with increased risk. Among high-risk patients, the addition of EBRT was associated with a 48.8% reduction in recurrence, achieving outcomes comparable to those of low-risk patients. Conclusions: Age alone should not determine IORT eligibility. A multifaceted approach, including tumor biology and adherence to recommended therapies, is essential. Supplemental EBRT improves outcomes in high-risk patients, and adapting guidelines may enhance patient selection in Asian populations.
Presentation numberPS2-06-05
Comparing Cross-Toxicities and Efficacy of Everolimus and Alpelisib in Different Sequences in Metastatic Breast Cancer Patients
Ajay Dhakal, University of Rochester Medical Center, Rochester, NY
A. Dhakal1, A. Shrestha2, Z. Shah3, S. Chinniah4, A. Roy5, T. Smith1, S. Gandhi6, H. Moore4, A. Van Swearingen4, M. Strawderman1, D. Peterson1, C. Anders4, R. O’Regan1; 1University of Rochester Medical Center, Rochester, NY, 2University of Arkansas Medical Center, Little Rock, AR, 3Roswell Park Cancer Institute, Buffalo, NY, 4Duke Cancer Institute, Durham, NC, 5Ohio State University, Columbus, OH, 6Winship Cancer Institute, Atlanta, GA.
Background:Everolimus (EV) and alpelisib (AL), both approved for treating HR+/HER2- Metastatic Breast Cancer (MBC), inhibit different steps of the PI3K/AKT/mTOR (PAM) pathway. They share unique side effects (SE), many of which are considered class effects (e.g., hyperglycemia). It is unclear if the emergence of an SE during a PAM inhibitor (PAMi) treatment predicts the risk of the same SE during subsequent PAMi treatment. Such cross-incidence (CrIn) of SEs across PAMi is understudied and could help predict and prevent SEs. Similarly, the efficacy of a PAMi on a tumor resistant to a different PAMi hasn’t been well established, as these tumors are excluded from new PAMi clinical trials. Methods:The objectives of the study were to assess 1. CrIn of important AEs between AL and EV; 2. efficacies of AL after treatment with EV and vice versa, among patients with HR+/HER2- MBC. A retrospective study was conducted in 3 U.S. cancer institutes (Wilmot, Roswell Park, Duke). Patients with HR+/HER2-MBC with PIK3CA mutation who received both drugs were included. Groups (grps) were defined based on the PAMi sequence: EV followed by AL (EV-AL) or reverse, AL-EV. Progression-free survival (PFS) and Overall Survival (OS) were estimated using Kaplan-Meier method and compared between grps using log-rank test. Survival analyses were conducted for the 2nd PAMi in the sequence. Multivariable analyses were done using Cox Models. Results:Of 55 pts screened, 46 were eligible: 30 in EV-AL and 16 in AL-EV grp. Baseline (at start of 2nd PAMi) variables were similar between the grps. Median age (years) was 64 in EV-AL grp, 65 in AL-EV grp. 70% of EV-AL grp and 81% of AL-EV grp were white. 47% of EV-AL grp and 56% of AL-EV grp had visceral disease. 77% of EV-AL grp and 79% of AL-EV grp had ECOG performance status 0-1. 97% of EV-AL and 100% of AL-EV grp had prior CDK4/6 inhibitors. 23% of EV-AL grp and 12% of AL-EV grp had >2 lines of prior chemotherapy for MBC. Median AL use was 5.3 months (m) in EV-AL grp vs 4.5m in AL-EV grp (p= 0.15); Median EV use was 6.3m in EV-AL grp vs 3.8m in AL-EV grp (p=0.76). Fulvestrant was the most common endocrine therapy with AL (90% in EV-AL, 94% in AL-EV) vs exemestane with EV (67% in EV-AL, 75% in AL-EV). Overall, hyperglycemia (hypG) was the most common SE with AL (65%), followed by diarrhea (38%), rash (20%), and stomatitis (20%). On EV, patients had diarrhea (16%), stomatitis (16%), rash (14%) and hypG (11%). In EV-AL grp, among the 3 pts who had developed hypG on EV, all 3 developed hypG on AL (100% CrIn). CrIn of rash, diarrhea & stomatitis on AL after developing them on EV were 50%, 20% and 33% respectively. In AL-EV, CrIn of hypG, rash, diarrhea, & stomatitis on EV after having them on AL were 10%, 0%, 25%, & 33% respectively. Comparing the efficacy of 2nd PAMi among the grps, median PFS on AL was 6.9m (95% CI 4.2–10.8) in EV-AL grp vs 7.4m (95% CI 3.5–23.1) on EV in AL-EV grp (p=0.64). In multivariable analysis, risks of PFS event on 2nd PAMi were not significantly different between grps [Hazard ratio (HR) 0.95 (95% CI 0.37-2.4) p=0.91]. Median OS on AL was 15.9m (12.4-34.2) in EV-AL grp vs that on EV was 20.1m (13.2-46.7) in AL-EV grp (p=0.84). In multivariable analysis, the risks of death after starting 2nd PAMi were not significantly different between grps [HR 0.92 (95%CI 0.39-2.17), p=0.85]. Conclusion:This study has assessed CrIn of SEs across EV and AL use. Notably, all patients who developed hypG and half who developed rash on EV also developed them again on AL. These CrIn results could help clinicians use preemptive strategies (like metformin, topical steroids) while using 2nd PAMi. AL and EV therapy were each associated with mPFS >6m when used as 2nd PAMi, suggesting meaningful activity. No significant differences were observed in PFS or OS between these two drugs when used as the 2nd PAMi. Further validation of efficacy and CrIn of SEs is needed in a larger dataset.
Presentation numberPS2-06-07
The Necessity of Sentinel Lymph Node Biopsy for Ductal Carcinoma in Situ Patients: a Retrospective Analysis of a Single Institute in Taiwan
Yu-Hsuan Chen, Department of Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung City, Taiwan
Y. Chen1, S. Shih2, M. Hou3, F. Chen3, C. Li3, J. Kan3, C. Kao3, L. Kao3, H. Takahashi3, C. Chuang4, C. Kuo4, P. Yang2; 1Department of Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung City, TAIWAN, 2Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast Oncology and Surgery, Kaohsiung Medical University Gangshan Hospital, Kaohsiung City, TAIWAN, 3Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung City, TAIWAN, 4Division of Breast Oncology and Surgery, Kaohsiung Medical University Chung-Ho Memorial Hospital; Division of Breast Oncology and Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung City, TAIWAN.
Purpose: Ductal carcinoma in situ is the preliminary phase of invasive breast cancer, with lesions confined within the mammary ducts, and have limited extensiveness. Owing to its non-invasive feature, axillary lymph node involvement is typically non-existence. Therefore, under current guidelines and consensus adopted worldwide, lymph node management is not a mandated procedure. However, the risk of clinical diagnosis of in situ carcinoma upstaging to invasive carcinoma after definitive surgery is still menacing, with some reported upstaging rates exceeding 30%. Nowadays, sentinel biopsy for node staging is generally not recommended for ductal carcinoma in situ patients receiving breast conservative surgery, however, such procedure is sometimes carried out nevertheless for various individualized reasons. Our study aims to report our real-world clinical experience on such unmet need, hoping to aid in the precise assessment of sentinel lymph node biopsy indication for such patients. Materials and Methods: Patients with a clinical diagnosis of ductal carcinoma in situ admitted to a single institute in Taiwan from 2019 to 2023, who have received definitive surgery with final pathological outcomes were gathered. Various clinical features including patient characteristics, tumor palpability, sonographic tumor size, biopsy methods, ductal carcinoma in situ tumor grading, immunohistochemistry results were evaluated. Sentinel lymph node positive rate and cancer upstaging rate were our primary outcomes. We analyze the risk factors contributing to in situ carcinoma upstaging. Results: Within 203 enrolled patients with clinical biopsied diagnosis of ductal carcinoma in situ, 55 patients (27.1%) were upstaged to invasive carcinoma in surgical pathology. Concurrent sentinel lymph node biopsy during definitive surgery was performed in 172 patients (84.7%), and nodal metastasis was found in 4 cases (2.3%). Among the 31 enrolled patients who omitted sentinel lymph node biopsy, 3 patients (9.7%) had upstaged to invasive disease. Regarding characteristic features analyzed, age over 50 years (OR: 1.703, p=0.028), diagnosed by core needle biopsy rather than by vacuum-assisted or excisional biopsy (P<0.001), sonographic tumor size greater than 2cm (OR: 1.733, p=0.016), and HER2 receptor negativity (OR: 1.377, p<0.001), showed significant risk of upstaging. Conclusion: Sentinel lymph node biopsy is not routinely carried out for ductal carcinoma in situ patients according to current treatment consensus. Whereas cancer upstaging is the least desirable situation after definite surgery. We disclosed Taiwan’s real-world data to clarify the necessity of sentinel lymph node biopsy for certain patients. We believe, thorough discussion and shared decision making between surgeon and patients before surgery is crucial, and sentinel lymph node staging could be provided as an option to the highly selected individuals with significant upstaging risk, eliminating the distress of under-treatment and re-operation for the patient.
Presentation numberPS2-06-17
Olaparib Use and gBRCA Testing among HER2-Negative Early-Stage Breast Cancer Patients in the United States Community Oncology Setting
Kathryn Mishkin, Merck & Co., Rahway, NJ, MD
K. Mishkin1, M. Ru2, M. Mabel Mardones3, I. Sruti4, J. Murphy4, J. Guo4, Q. Li2, X. Xu2, K. Hirschfield5, J. A. Mejia5, P. Conkling4; 1Merck & Co., Rahway, NJ, MD, 2Astra Zeneca, Gaithersburg, MD, 3Rocky Mountain Cancer Centers, Denver., CO, 4Ontada, Boston, MA, 5Merck & Co., Rahway, NJ, NJ
Background: Long-term follow-up of the OlympiA trial confirmed the efficacy and safety of olaparib among high-risk HER2-negative (HER2-) early breast cancer (eBC) patients with a germline BRCA (gBRCA) mutation. This retrospective study focused on olaparib users and aimed to describe the contemporary real-world landscape for HER2- patients in the community oncology setting with regard to biomarker testing, treatment patterns, and olaparib uptake. Methods: Electronic medical record data from The US Oncology Network were used for this retrospective study among adult HER2- eBC patients who initiated systemic adjuvant treatment between 01 Jan 2020 – 31 Oct 2024 (adjuvant treatment initiation date as index date). Patient selection was conducted in a stepwise method, prioritizing patients who initiated olaparib, followed by non-olaparib patients who received BRCA and genomic risk testing (ODX or Mammaprint), followed by non-olaparib patients with a high genomic risk score (defined as ODX≥25 or High Mammaprint result). Patient and treatment characteristics, including biomarker and testing patterns, were assessed descriptively. Results: There were 251 HER2- eBC patients considered eligible for the study (222 HR+, 27 TNBC, 2 HR untested). Overall, 216 (97.3%) HR+ patients had a high genomic risk score, 42 (16.7%) patients had a gBRCA mutation, and 34 (13.5%) initiated olaparib. The median (IQR) time from gBRCA result date to olaparib initiation date was 8.8 (1.0, 23.1) months. gBRCA testing rates varied by biomarker status, with 64.4% (n=143) of HR+ patients and 85.2% (n=23) of TNBC patients tested. Overall, 34% (n=85) of patients did not receive gBRCA testing, and 12% (n=19) of patients received gBRCA testing after adjuvant therapy initiation. Compared to the overall cohort, olaparib users were younger (46.7 vs. 55.3 years), were more likely to have a family history of cancer (91.2 vs. 81.7%), and had a higher proportion of TNBC patients (47.1 vs.10.8%). Conclusion: Despite utilizing patient selection methods that prioritized olaparib users as well as patients tested for BRCA and genomic risk, olaparib uptake in the real-world is limited among HER2- eBC patients. Eligibility for olaparib is dependent upon having a BRCA mutation, but findings show that one-third of patients, especially among HR+ patients, did not receive gBRCA testing. Current guidelines recommend gBRCA testing for all patients with breast cancer, and as a result, there is an opportunity to improve gBRCA testing rates and timing to identify additional patients who may be eligible for and benefit from targeted treatment such as olaparib. As olaparib uptake increases, ongoing research to describe clinical outcomes is recommended.
Presentation numberPS2-06-18
Real-world second-line treatment use and clinical outcomes in patients with HR-positive/HER2-negative metastatic breast cancer (mBC) and an ESR1 mutation (ESR1m)
Massimo Cristofanilli, Weill-Cornell Medicine / New York-Presbyterian Hospital, New York, NY
S. L. James1, S. Vijapur2, S. Sreenivasan3, G. Long4, L. Robert5, C. Huang-Bartlett6, M. Cristofanilli7; 1Clinical Data Science, Oncology R&D, AstraZeneca, Barcelona, SPAIN, 2Tempus AI, Inc., New York, NY, 3Clinical Data Science, Oncology R&D, AstraZeneca, Gaithersburg, MD, 4Clinical Data Science, Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 5Late Development, Oncology R&D, AstraZeneca, Barcelona, SPAIN, 6Late Development, Oncology R&D, AstraZeneca, Gaithersburg, MD, 7Weill-Cornell Medicine / New York-Presbyterian Hospital, New York, NY.
Background: ESR1m lead to constitutive (estrogen-independent) activation of estrogen receptor alpha and are rare (<5%) at initial diagnosis of mBC. At disease progression following first-line (1L) treatment with an aromatase inhibitor (AI) plus CDK4/6i, ESR1m are detectable in the tumors of ~40% of patients and are recognized as the most common mechanism of acquired resistance to AI treatment. Current guidelines recommend ESR1m testing at the time of 1L treatment progression for informing treatment decisions. This retrospective cohort analysis aimed to characterize second-line (2L) treatment use and clinical outcomes (real-world progression-free survival [rwPFS] and overall survival [rwOS]) in mBC patients with a positive ESR1m test after 1L treatment with AI+CDK4/6i. Methods: Adult patients with a confirmed diagnosis of HR-positive/HER2-negative mBC after February 2015 were identified from electronic health records (EHR) in the US Flatiron Health database. Inclusion criteria included 1L treatment with AI+CDK4/6i (initiated before December 31, 2024), a positive ESR1m test before initiation of 2L therapy, and 2L therapy regimen adherent to NCCN Guidelines. Patients were followed until June 30, 2025. Descriptive statistics were used to summarize demographics, baseline clinical characteristics, and treatment patterns. rwOS and rwPFS were estimated using Kaplan-Meier survival analysis. The start of 2L therapy was the index date. Multivariable Cox proportional hazards regression was used to adjust for prespecified confounders, including age at 2L start, ECOG PS, metastatic site, and 1L treatment duration. Sensitivity analyses were conducted by duration of 1L AI+CDK4/6i. Similar analyses in a separate large US EHR database are ongoing and will be reported. Results: A total of 226 patients with a positive ESR1m test were included; median age at diagnosis was 63 years, and most were White (74.1%) with ECOG PS ≤1 (71.2%). Liver metastases were common (51.8%), and 25.2% of patients had bone-only metastasis. 2L treatments included ET+CDK4/6i (32.3%), elacestrant monotherapy (22.6%), PI3K/AKT/mTOR inhibitor+ET (19.0%), chemotherapy/ADC (11.9%), fulvestrant monotherapy (11.1%), or other (3.1%).In the overall population, median rwPFS was 4.8 (95% CI 4.0-5.6) months and median rwOS was 24.9 (95% CI 18.8-30.9) months; results varied by treatment (Table). Sensitivity analyses in patients with ≥6 months of 1L AI+CDK4/6i showed similar findings (Table). Conclusion: In patients with ESR1m HR-positive mBC, the most common real-world 2L treatments were a rechallenge ET+CDK4/6i, elacestrant, and PI3K/AKT/mTOR inhibitor+ET. ESR1m defines a difficult-to-treat subgroup, with rwPFS outcomes highlighting the need for more effective strategies that address resistance and improve patient outcomes.
| All patients | rwPFS median (95% CI), months | rwPFS hazard ratio (95% CI) | rwOS median (95% CI), months | rwOS hazard ratio (95% CI) | |||||
| All patients (n=226) | 4.8 (4.0-5.6) | — | 24.9 (18.8-30.9) | — | |||||
| Fulvestrant (n=25) (reference) | 3.0 (2.8-6.3) | 1.00 | 17.4 (11.8-30.9) | 1.00 | |||||
| ET+CDK4/6i (n=73) | 5.2 (3.6-6.1) | 0.54 (0.31-0.95) | 39.1 (19.79-NE) | 0.47 (0.24-0.93) | |||||
| Elacestrant monotherapy (n=51) | 5.3 (4.4-7.5) | 0.63 (0.34-1.16) | 17.8 (16.18-NE) | 0.92 (0.45-1.89) | |||||
| PI3K/AKT/mTOR inhibitor+ET (n=43) | 5.7 (4.0-10.2) | 0.61 (0.33-1.14) | 26.7 (15.12-NE) | 0.63 (0.29-1.34) | |||||
| Chemotherapy/ADC (n=27) | 4.8 (2.8-6.7) | 0.58 (0.30-1.14) | 17.6 (13.84-NE) | 0.53 (0.24-1.21) | |||||
| Other (n=7) | 2.5 (0.92-NE) | 0.75 (0.28-1.96) | 7.1 (0.92-NE) | 2.58 (0.90-7.40) | |||||
| Duration of 1L AI+CDK4/6i ≥6 months | |||||||||
| All patients (n=194) | 4.6 (3.7-5.6) | — | 23.5 (17.8-30.9) | — | |||||
| Fulvestrant (n=21) (reference) | 2.8 (1.61-NE) | 1.00 | 19.1 (11.84-NE) | 1.00 | |||||
| ET+CDK4/6i (n=52) | 4.6 (2.7-5.9) | 0.75 (0.39-1.42) | 39.1 (19.79-NE) | 0.43 (0.20-0.90) | |||||
| Elacestrant monotherapy (n=49) | 5.7 (4.5-7.5) | 0.64 (0.32-1.24) | 17.8 (16.18-NE) | 0.76 (0.35-1.65) | |||||
| PI3K/AKT/mTOR inhibitor+ET (n=42) | 5.4 (4.0-10.2) | 0.64 (0.32-1.25) | 26.7 (15.12-NE) | 0.48 (0.21-1.12) | |||||
| Chemotherapy/ADC (n=24) | 3.5 (2.8-7.6) | 0.54 (0.26-1.13) | 17.6 (13.84-NE) | 0.48 (0.19-1.20) | |||||
| Other (n=6) | 4.3 (2.07-NE) | 0.63 (0.22-1.81) | 19.2 (3.52-NE) | 1.71 (0.52-5.55) |
NE, not evaluable.
Presentation numberPS2-07-19
Proteomic subtyping of stage II/III TNBC from the I-SPY2 TRIAL reveals a druggable steroid hormone receptor signature (SRS) that is associated with poor outcome and enriched in non-pCR patients’ residual disease
Julia Wulfkuhle, George Mason University, Manassas, VA
J. Wulfkuhle1, P. Blas2, H. Williams2, G. Hirst3, L. Brown-Swigart3, R. I. Gallagher1, M. Pierobon1, C. Isaacs4, L. van ‘t Veer3, L. J. Esserman3, J. O’Shaughnessy5, E. F. Petricoin III1; 1George Mason University, Manassas, VA, 2Baylor Scott and White Research Institute, Dallas, TX, 3University of California San Francisco, San Francisco, CA, 4Georgetown University, Washington, DC, 5Baylor University Medical Center, Texas Oncology, Dallas, TX
Background: I-SPY Triple Negative Breast Cancer (TNBC) Response Predictive Subtypes (RPS) predict for benefit from immunotherapy and DNA damaging agents in Immune+ and DNA Repair Deficient (DRD) subgroups. We explored TNBC subtyping based on functional protein/phosphoprotein architecture and previously described a subset of TNBC that expresses high relative levels of estrogen receptor alpha (ERα)1. These ERα “high” TNBC express a steroid hormone receptor signature (SRS) characterized by co-incidentally increased expression of ERα, activated glucocorticoid receptor (pGR S211) and total androgen receptor (AR), present in ~30% of TNBC. We have now investigated the drug target activation architecture of SRS-positive (SRS+) TNBC and associations of SRS+ status with outcomes in I-SPY2 patients not responding to neoadjuvant therapy. Methods: Quantitative reverse phase protein array (RPPA) data from laser capture microdissection-enriched tumor epithelium comprised of 153 proteins/phosphoproteins, including ERα, AR, and pGR S211, were obtained from 253 I-SPY2 TNBC patients at pre-treatment baseline1 as well as in patient-matched surgical residual disease samples from 48 non-pCR TNBC patients. The SRS score was determined based on the equally weighted rank-sum addition of the 3 SRS analytes. SRS scores above and below the population median were considered SRS+ and SRS-negative (SRS-), respectively. Clinical information was provided by Quantum Leap Health Care Collaborative. For survival status and signaling pathway analyses, SRS scores were divided into tertiles and the top and bottom tertiles (N=84 each) were used as the SRS+ and SRS- populations. Statistical analyses were performed using GraphPad Prism v10 and JMP software v14. Uncorrected p-values <0.05 were used for determination of significance and results are descriptive. Results: SRS+ TNBC was found to be distinct from the luminal AR, Immune/DRD, PAM50, TNBC4, TNBC7, and MP1/2 subtypes. Analysis of the pre-treatment TNBC tumors revealed that patients with SRS+ tumors had worse survival status vs patients with SRS- tumors (p=0.05). Proteomic analysis revealed that SRS+ TNBC from both pre-treatment and residual disease surgical tumor samples were enriched for druggable targets including HARPS EGFR-HER21,2, AKT-mTOR and RAF-ERK activation signatures. Of interest, 58% (28/48) of the surgical samples from non-pCR patients were SRS+ based on the T0 population median cutpoint. SRS positivity in the pre-treatment samples did not correlate with worse outcomes from patients who achieved pCR. Conclusions: Proteomic data from I-SPY 2 showed that patients with SRS+ TNBC have a worse prognosis than SRS- TNBC. Surgical samples from non-pCR pts are enriched with SRS+ cancers. Proteomic analysis of SRS+ tumors obtained at pre-treatment baseline or in the residual disease surgical tumor samples from non-pCR SRS+ pts revealed activation of EGFR, HER2 and AKT-mTOR signaling networks. HER2/EGFR co-activated TNBC has been previously shown to have elevated levels of ER and were shown to be highly responsive to the EGFR-HER2 TKI inhibitor neratinib2. We hypothesize that inhibiting ER and AR, +/- GR, along with EGFR/HER2 may improve the poor prognosis of non-pCR SRS+ TNBC patients. 1.https://doi.org/10.1158/1538-7445.SABCS22-PD5-08 2. Wulfkuhle J et al. JCO Precis Oncol 2018
Presentation numberPS2-11-24
Collagen prolyl 4-hydroxylase promotes breast cancer endocrine resistance via regulation cellular stress
Gaofeng Xiong, The Ohio State Univeristy, Columbus, OH
D. Hironaka1, B. Kang1, R. Xu2, G. Xiong1; 1The Ohio State Univeristy, Columbus, OH, 2University of Kentucky, Lexington, KY
The luminal subtype accounts for approximately 70% of all diagnosed breast cancers cases. Endocrine therapy is the first-line therapy for luminal breast cancer patients. About 30%-50% of luminal breast cancer patients with five to ten years of treatment acquire resistance to endocrine therapy. Understanding the mechanisms of endocrine resistance and identifying new therapeutic targets remain urgent priorities. Here, we show that expression of the alpha subunit of Prolyl hydroxylation (P4HA1), an enzyme hydroxylates proline into 4-hydroxyproline at the Y position of the collagen -X-Y-Gly- triplet motif, is increased in endocrine therapy resistant luminal breast cancer cells compared with parental endocrine therapy sensitive cells. In luminal breast cancer patients, increased P4HA1 expression significantly correlates with short relapse-free survival. Knockdown of P4HA1 or P4HA1 small molecular inhibitors treatment restores endocrine-resistant luminal breast cancer cells sensitivity to hydroxytamoxifen (4OHT) or fulvestrant treatment, by enhancing cell apoptosis and inhibiting cell proliferation. These results indicate that P4HA1 is an important regulator for endocrine resistance of breast cancer. We also show that the inactivation of P4AH1 combined with endocrine treatment dramatically enhances endoplasmic reticulum (ER) stress-related gene expression and reactive oxygen species (ROS) level. ROS scavenger treatment rescues endocrine resistance in P4HA1-inactivated cells. These results indicate that P4HA1 is a key player that coordinates the crosstalk between endocrine resistance and cellular stress. These findings reveal P4HA1 as a novel therapeutic target and holds promising potential of developing new therapeutic strategies for luminal breast cancer patients with endocrine resistance.
Presentation numberPS2-07-09
Epigenetic Imprinting as a Novel Precision Biomarker for Early Breast Cancer and Precancer Diagnosis and Detection
Mitra Abdollahi Neisani, Mayo Clinic Florida, Jacksonville, FL
M. Abdollahi Neisani1, R. Guo1, T. Cheng2, M. K. Komforti1; 1Mayo Clinic Florida, Jacksonville, FL, 2LisenID, Wilmington, DE.
Introduction and rationale: There is a wide-spread urgent need for an objective reproducible diagnostic tool capable of precisely classifying atypical mammary lesions into cancer vs. benign. Current diagnostic methods in pathology rely on routine and highly subjective histomorphologic and immunophenotypic features. Some authors report a 35% error rate in pathologic interpretation of atypical lesions of the breast (Elmore, 2015). Diagnostic variability can drop to the flip-of-the-coin 50%, even amongst expert breast pathologists (Tozbekian, 2017; Samples, 2017) and is largely attributed to the lack of innovative tools to adequately distinguish biologic differences (Allison, 2016), resulting in diagnoses that are described as “murky” and “arbitrary” (Khoury, 2022). Genomic imprinting, a vital epigenetic regulatory mechanism, is critical in mammalian development and tumorigenesis (Barlow, 2014, Murrell, 2006). Typically, imprinted genes exhibit parent-specific allele silencing through differential methylation. However, in cancer, this regulation often breaks down, resulting in the aberrant activation of the typically silenced allele—a phenomenon known as loss of imprinting (LOI) (Jelinic, 2016). LOI is considered one of the earliest molecular changes in cancer development, occurs early in tumorigenesis, making it ideal for identifying precursor lesions. Objectives: We hypothesize loss of imprinting (LOI) can be developed as a diagnostic, predictive and prognostic novel epigenetic marker (Quantitative Chromogenic Imprinted Gene In-Situ Hybridization, QCIGISH) for breast precancerous lesions and cancer detection. Our objective is to initiate a proof of concept for atypical breast lesions with QCIGISH and develop a diagnostic model. Methods: We retrospectively identified benign, atypical, and carcinoma core needle specimens from year 2023 in the Department of Pathology at Mayo Clinic Florida. Inclusion criteria included female patients, ages 18-99, with archived breast material obtained through standard of care (n=20). All diagnoses were ascertained by a breast pathologist (MKK). Following a thorough literature review, we processed the material for in situ hybridization, and pretreated with RNA scope preparation procedure using probes targeting noncoding intronic regions of nascent RNAs (QCIGISH). Based on high Area Under the Receiver Operating Characteristics Curve (AUROC), 2 genes – HM13 and GRB10 were identified as the most promising candidates for classification with QCIGISH. Upon test completion, the detected gene-expression site appeared as a distinct red dot on the slide under the common bright-field microscope. Next, the gene expression signals were counted and further classified as bi-allelic (BAE), multi-allelic (MAE) and total expression (TE). Results: A decision tree model based on BAE, MAE, and TE metrics showed differential signal expression. High MAE was a strong predictor for malignancy. We achieved 100% sensitivity and specificity with the ground truth, thereby initiating proof of concept. Furthermore, one histomorphologically challenging atypical lesion, for which the positive LOI finding accurately classified as ductal carcinoma in situ (DCIS), was confirmed indeed DCIS by ground truth validation on the excision specimen. Conclusion: We have demonstrated a breakthrough molecular biomarker that is fully objective, reproducible, and practical. First of its kind, the innovative QCIGISH has superior performance than the current standard of care in breast cancer diagnostics allowing for single-cell resolution of epigenetic changes. Next, we seek to undergo full model validation.
Presentation numberPS2-08-27
Final Analysis of a Prospective Observational Study to Evaluate the Impact of Comprehensive Genome Profiling on Treatment Decision Making in Metastatic or Recurrent Breast Cancer in Japan: JBCRG-C07 REIWA Study
Hiroshi Tada, Tohoku Medical and Pharmaceutical University/Graduate School of Medicine, Tohoku University, Miyagi, Japan
H. Tada1, H. Masuda2, Y. Sagara3, Y. Adachi4, T. Iwatani5, Y. Uemoto6, O. Yoko7, Y. Kajiwara8, D. Kitagawa9, T. Kogawa10, T. Shien11, Y. Tanabe12, M. Tanioka11, F. Hara4, H. Yasojima13, K. Yoshimura14, H. Iwata15, S. Saji16, N. Masuda17; 1Tohoku Medical and Pharmaceutical University/Graduate School of Medicine, Tohoku University, Miyagi, Japan, 2Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan, 3Social medical corporation Hakuaikai, Sagara Hospital, Kagoshima, Japan, 4Aichi Cancer Center Hospital, Aichi, Japan, 5Tohoku Medical and Pharmaceutical University, Kanagawa, Japan, 6Nagoya City University Hospital, Aichi, Japan, 7JCHO Osaka Hospital, Osaka, Japan, 8Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan, 9National Center for Global Health and Medicine, Tokyo, Japan, 10The Cancer Institute Hospital Of JFCR, Tokyo, Japan, 11Okayama University Hospital, Okayama, Japan, 12Toranomon Hospital, Tokyo, Japan, 13National Hospital Organization Osaka National Hospital, Osaka, Japan, 14Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan, 15Nagoya City University, Graduate School of Medical Sciences, Aichi, Japan, 16Fukushima Medical University, Fukushima, Japan, 17Graduate School of Medicine, Kyoto University, Kyoto, Japan
Background: Comprehensive genome profiling (CGP) has been covered by insurance in Japan since June 2019. We conducted a prospective observational study (JBCRG-C07 (REIWA study), UMIN000038065) to determine the proportion of patients with stage IV or recurrent breast cancer (MBC) who received matched therapy (MT) identified by CGP since January 2020.Methods: A total of 602 patients with stage IV or recurrent MBC who consented to FoundationOne® CDx (F1CDx) or FoundationOne® Liquid CDx (F1LCDx) testing were prospectively enrolled. After excluding cases without valid testing or expert panel review, 576 patients comprised the per-protocol set (PPS). The primary endpoint was the rate of receiving MT, and the secondary endpoints included the clinical utility of panel testing, such as overall survival (OS).Results: Among the 576 patients in the PPS, 490 underwent F1CDx and 86 underwent F1LCDx. The median age was 56 years, and 25.7% of patients had de novo stage IV disease. The median number of pretreatment regimens before testing was five in the luminal subtype (n = 310), two in the triple-negative subtype (n = 173), and four in the HER2-positive subtype (n = 93). MT was recommended for 362 patients (62.8%) and administered to 93 patients (16.1%). Accordingly, the primary endpoint—the proportion of patients receiving MT among those for whom MT was recommended—was 25.7% (93/362). The most frequently administered MT within clinical trials was HER2 tyrosine kinase inhibitor therapy for ERBB2-mutated luminal breast cancer, whereas the most common off-trial MTs were immune checkpoint inhibitors (31.9%) primarily for TMB-high tumors and mTOR inhibitors (24.6%) primarily for tumors harboring PIK3CA mutations.During the follow-up period (median, 13.5 months), 412 deaths (71.5%) occurred. OS was significantly longer in the MT group than in the non-MT group (median 21.1 vs. 14.4 months; HR 0.651, 95% CI 0.494-0.857; P = 0.002).Subtype-specific analysis showed a significant OS benefit in the luminal subtype (22.3 vs. 15.1 months; HR 0.565, 95% CI 0.396-0.809; P = 0.002), whereas no significant benefit was observed in the triple-negative (14.0 vs. 11.8 months; HR 0.758, 95% CI 0.428-1.345; P = 0.342) or HER2-positive subtype (25.6 vs. 23.9 months; HR 1.057, 95% CI 0.530-2.107; P = 0.874).Conclusions: Among MBC patients recommended for targeted therapy through CGP, approximately one-quarter received MT and demonstrated an improved OS. An OS benefit was observed in the luminal subtype but not in the triple-negative or HER2-positive subtypes, highlighting the need for effective MT options and expanded early line clinical trials for these groups in Japan.
Presentation numberPS2-09-07
Ultra-sensitive Molecular Residual Disease Detection in Breast Cancer Using Whole-Genome Sequencing-Based Personalized ctDNA Panels: Preliminary Results from the MONSTAR-SCREEN-3 Project
Yoichi Naito, National Cancer Center Hospital East, Kashiwa, Japan
Y. Naito1, M. Nishimura2, Y. Hisamatsu3, M. Futamura4, T. Toyama5, T. Yamanaka6, M. Tsukabe7, S. Hashimoto8, E. Tokunaga9, M. Ishihara10, R. Nakamura11, H. Yasojima12, T. Hashimoto1, T. Shibuki1, M. Imai1, T. Fujisawa1, H. Bando1, S. Kobayashi1, S. Sakashita1, T. Kuwata1, A. Blackler13, G. Hogan13, K. J. Taber14, J. Jasper13, D. Muzzey13, E. Oki3, T. Yoshino1, H. Iwata5; 1National Cancer Center Hospital East, Kashiwa, Japan, 2Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan, 3Kyushu University, Fukuoka, Japan, 4Gifu University Hospital, Gifu, Japan, 5Nagoya City University, Nagoya, Japan, 6Kanagawa Cancer Center, Kanagawa, Japan, 7The University of Osaka, Suita, Japan, 8University of Tsukuba, Ibaraki, Japan, 9NHO Kyushu Cancer Center, Fukuoka, Japan, 10Osaka International Cancer Institute, Osaka, Japan, 11Chiba Cancer Center, Chiba, Japan, 12NHO Osaka National Hospital, Osaka, Japan, 13Myriad Genetics, Inc., Salt Lake City, UT, 14Myriad Genetics, Inc, Salt Lake City, UT
Background Circulating tumor DNA (ctDNA) has emerged as a promising biomarker for detecting molecular residual disease (MRD) and is increasingly integrated into both clinical practice and translational research in breast cancer. However, whole-exome sequencing (WES)-based approaches often lack sufficient sensitivity for MRD detection, particularly in low-shedding tumors such as luminal breast cancer. To address this limitation, the MONSTAR-SCREEN-3 study is evaluating an ultra-sensitive MRD assay based on whole-genome sequencing (WGS). Methods MONSTAR-SCREEN-3 is a prospective, multicenter study enrolling 1,100 patients with solid tumors receiving curative-intent therapy in the definitive cohort. Personalized ctDNA panels are generated using the Precise MRD platform (Myriad Genetics), incorporating up to 1,000 tumor-specific alterations—including short variants and indels—identified through WGS of matched tumor tissue. Serial plasma samples are collected at baseline, after neoadjuvant chemotherapy (if applicable), 1 month post-surgery, quarterly during the first year, and biannually thereafter for up to two years. Here we report preliminary results from the breast cancer cohort. Results As of August 2025, 117 patients with resectable breast cancer had been enrolled. Clinical data were available for 89 patients, and MRD results for 92 samples from 40 patients. Median age was 52.5 years (range, 30-89). Clinical stage distribution: IA (7.9%), IIA (47.2%), IIB (22.5%), IIIA (5.6%), IIIB (7.9%), and IIIC (9.0%). Subtypes: luminal 48.2%, HER2-positive 36.5%, and triple-negative (TNBC) 14.1%. Neoadjuvant chemotherapy was given to 55% (49/89). Among these, subtype distribution was luminal 18.8%, HER2-positive 60.4%, TNBC 20.8%. Median follow-up was 4.2 months (range, 2.0-8.3). Among 40 patients with MRD results, WGS was performed on 15 biopsy and 25 surgical samples, identifying a mean of 5,394 panel-eligible alterations per patient (range: 762-40,981). Customized panel creation succeeded in 40/40 patients (100%). The assay showed 97.5% baseline sensitivity (39/40), with 28.2% (11/39) detected at an ultra-sensitive level (tumor fraction <100 ppm; minimum 4.9 ppm). Sensitivity by subtype was luminal 95.8% (23/24), HER2-positive 100% (12/12), and TNBC 100% (4/4), with ultra-sensitive detection in 39.1% (9/23), 8.3% (1/12), and 25.0% (1/4), respectively. The one ctDNA-negative case was a pT2N0 pStage IIA luminal tumor with a ctDNA tumor fraction of 2 ppm, classified as MRD-negative based on the 99.615% specificity threshold. At 1 month post-surgery, MRD positivity was 14.8% (4/27), with tumor fractions of 11.2-142 ppm. Of the four MRD-positive cases, three had 3-month data: two receiving adjuvant chemotherapy and one not receiving any adjuvant therapy. MRD positivity persisted in all three patients. Extended follow-up and longitudinal ctDNA dynamics will be reported. Conclusions The WGS-based personalized ctDNA assay achieved ultra-sensitive MRD detection across breast cancer subtypes, including detection at ppm-level tumor fractions. Updated molecular and clinical outcome data will be presented.
Presentation numberPS2-11-26
Oncogenic er alpha as an rna-binding protein controls mrna stability and epitranscriptome of breast cancer
Nimmy Mohan, University of California, San Francisco, San Francisco, CA
N. Mohan, A. Dabrowska, V. Subramanyam, I. Liu, D. Kuzuoglu-Öztürk, D. Ruggero; University of California, San Francisco, San Francisco, CA.
Breast cancer is the most commonly diagnosed type of cancer worldwide in women. Estrogen receptor alpha (ERα), a well-studied transcription factor, is the major oncogene in over 70% of breast cancer cases. The majority of earlier studies have primarily focused on ERα transcriptional activity that promotes tumor progression. We discovered that ERα is a non-canonical RNA-binding protein that associates with over 1,000 mRNAs, mainly through their 3’-untranslated regions (UTRs), in breast cancer cells. Interestingly, recent studies have shown that a broad spectrum of transcription factors (TFs), including ERα, directly bind RNA; yet, how this association directly orchestrates gene expression remains largely unexplored. This led us to ask an outstanding question about the functional role of oncogenic ERα on RNA metabolism in breast cancer. Since the 3’-UTR is a molecular hub where cis and trans-regulatory elements converge to determine mRNA fate, we hypothesized that oncogenic ERα regulates the stability of a set of mRNAs by binding to its 3’-UTR, which in turn affects the downstream gene expression program that promotes breast cancer progression. To address this question, we performed SLAM-seq, a genome-wide approach for characterizing mRNA stability in MCF7 WT cells and MCF7 cells harboring an ER⍺ RNA binding domain mutation (ER⍺ RBDM). We found that the stability of a subset of ERα mRNA targets is decreased upon loss of ERα RNA-binding activity. We then focused on a critical oncogenic mRNA, NSUN2, an enzyme that catalyzes the 5-methylcytosine (m5C) modification on mRNA, which is an ERα target. NSUN2 is a m5C writer protein, a well-studied RNA methyltransferase enzyme, and is significantly upregulated in breast cancer. Interestingly, survival analysis shows that high NSUN2 correlates with poor prognosis in ER-positive breast cancer patients, but not in ER-negative patients. We show that ER⍺ WT strongly binds to the 3’UTR of NSUN2 mRNA, whereas the ER⍺ RNA binding mutant (ER⍺ RBDM) doesn’t bind to NSUN2 3’UTR. Strikingly, we observed that the loss of ERα RNA binding destabilizes NSUN2 mRNA and protein levels. Furthermore, we found that global RNA m5C methylation is significantly downregulated in breast cancer cells expressing the ERα RBD. Notably, ERα RBDM cells are impaired in forming tumors in vivo, and this phenotype is rescued by overexpressing NSUN2. This suggests the existence of ERα-mediated epitranscriptomic marks via NSUN2 in breast cancer progression.
Presentation numberPS2-12-03
A Comprehensive Transcriptional Atlas of Breast Cancer Reveals Clinically Relevant Molecular Subtypes
Xin Sun, Vanderbilt University Medical Center, Nashville, TN
X. Sun, P. I. Gonzalez-Ericsson, M. E. Sanders, H. An, H. Jin, Q. Sheng, J. M. Balko, J. A. Pietenpol, B. D. Lehmann; Vanderbilt University Medical Center, Nashville, TN
Background: Breast cancer comprises molecularly diverse subtypes associated with prognosis and treatment response. Currently, these subtypes are characterized by the transcriptional analyses of individual, large-scale datasets. A more thorough understanding of breast cancer transcriptional diversity has been hindered by the challenge of performing integrated analysis across multiple independent RNA-seq datasets without compromising their intrinsic biology. Methods: We applied single-cell RNA sequencing methods to integrate bulk RNA-seq data from 20 independent datasets, encompassing 6,661 breast tumors, 368 normal breast tissues, 708 cell lines, and 161 patient-derived xenografts/organoids. Unsupervised clustering of bulk transcriptomes identified molecular subtypes, which were annotated using clinical and molecular features, including histology and PAM50 classification. Multivariate analyses evaluated prognosis and treatment response across subtypes. Gene signatures were developed to validate subtype-specific features and predict therapeutic responses across multiple independent datasets. Results: We constructed a comprehensive, integrated transcriptomic map and identified 14 biologically distinct subtypes, including three novel subtypes; mucinous (MUC), neuroendocrine (NE) and mammary stem-like (STM), TNBC subtypes (luminal androgen receptor [LAR], basal-like 2 [BL2]) and intrinsic subtypes (LumA, LumB, LumB-A, basal, HER2 and normal-like/claudin-low). MUC and NE displayed increased transcript and protein levels of mucin and neuroendocrine markers. Breast cancer cell lines, organoids and PDXs shared similar molecular subtypes with corresponding tumors, including the newly identified NE DU4475 (SYP+) and MUC MDAMB134 (INSM1+) cell lines which express NE protein markers. Additionally, we identified three non-tumor subtypes, immune dominate (ID), fibroblast (FB) or normal (N) in which transcriptomes were driven by adjacent normal cells. Multivariate analysis, adjusted for age, stage, TMB, tumor size, histology showed significant differences in overall survival compared to LumA (reference): LumB (HR=1.76, CI:1.27-2.44, p<0.001), basal (HR=2.96, CI:2.21-3.97, p<0.001), HER2 (HR=1.83, CI:1.3-2.56, p<0.001), BL2 (HR=2.3, CI:1.42-3.73, p<0.001), LAR (HR=1.91, CI:1.17-3.12, p=0.009), and STM (HR=4.83, CI:2.97-7.85, p<0.001). Furthermore, molecular subtypes showed differing therapeutic responses. LumB-A, MUC, and NE patients benefited more from combined chemo-endocrine therapy than endocrine therapy alone. ER+ tumors outside the luminal clusters were frequently ER-low and had worse outcomes with endocrine therapy alone and significantly benefited from added chemotherapy. ER+HER2+ patients within luminal clusters did not respond to HER2-targeted therapy. The STM cluster contained a mixture of ER+, HER2+ and TNBC tumors that were enriched in metaplastic spindle morphology and stem markers and displayed the worst outcomes, suggesting current treatment strategies are ineffective for this subtype. To validate our findings, we developed an integrated breast transcriptome classification (iBTC) signature that reproduced prognosis and classification in multiple datasets, predicting therapy response to immunotherapy and HER2-targeted therapy. Conclusions: Our comprehensive integrated analysis of RNA-seq data refines the current understanding of molecular subtyping, particularly the relationship between molecular signatures and histology-based classifications, provides insight into cell origin and identifies matching preclinical models to study alternative treatment strategies. These findings provide a rationale for revisiting clinical management for patients with rare subtypes and histology-molecular subtype mismatched tumors.
Presentation numberPS2-12-06
BBO-10203, a first-in-class breaker of the RAS:PI3Kα interaction, inhibits tumor growth alone and in combination with fulvestrant or ribociclib in breast cancer models without inducing hyperglycemia
Kerstin W Sinkevicius, BBOT, South San Francisco, CA
K. W. Sinkevicius1, J. P. Stice1, C. Zhang1, S. Feng1, M. Chen1, E. Riegler1, C. Feng1, D. J. Czyzyk2, J.-P. Denson2, Y. Yang1, M. Dyba2, B. P. Smith2, S. Donovan1, L. Fu1, K. Lin1, F. C. Lightstone3, A. E. Maciag2, K. Wang1, D. V. Nissley2, E. M. Wallace1, D. K. Simanshu2, R. Xu1, F. McCormick4, P. J. Beltran1; 1BBOT, South San Francisco, CA, 2NCI RAS Initiative, Frederick National Laboratory for Cancer Research, Frederick, MD, 33Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, 4Helen Diller Family Comprehensive Cancer Center, University of California San Francisco and NCI RAS Initiative, Frederick National Laboratory for Cancer Research, San Francisco, CA
Aberrant activation of the PI3Kα pathway is one of the most frequent oncogenic events across human cancers and leads to promotion of tumor cell growth, survival, glucose metabolism, and acute resistance to numerous standard of care cancer therapies. Although PI3Kα inhibitors are approved, and mutant‑selective PI3Kα inhibitors are in development for treating HR+ HER2- breast cancers with PIK3CA mutations, a significant unmet need remains due to dose‑limiting, on‑target hyperglycemia. This toxicity can restrict target coverage, limit the number of eligible patients, and shorten the duration of treatment, highlighting the need for novel approaches to target the pathway. One such strategy is to block RAS-mediated activation of PI3Kα, a signaling event prevalent mostly in malignant cells. Here, we report on BBO-10203, a clinical-stage first-in-class small molecule which breaks the protein-protein interaction between RAS and PI3Kα and inhibits RAS-mediated activation of the PI3Kα pathway. Importantly, BBO-10203 does not induce hyperglycemia, as insulin signaling does not rely on RAS proteins to mediate glucose uptake. BBO-10203 covalently and selectively binds PI3Kα on cysteine 242 in the RAS binding domain, which prevents the activation of PI3Kα by KRAS, HRAS, and NRAS. BBO-10203 shows full cellular target engagement at 30 nM and potently inhibits RAS-driven pAKT in human breast cancer cell lines. BBO-10203 displays excellent drug-like properties and oral bioavailability. PK/PD studies show that a single dose significantly reduces tumor pAKT levels in a dose- and time-dependent manner, and efficacy studies show that BBO-10203 has monotherapy activity in a panel of ER+ HER2- breast cancer CDX and PDX models. Importantly, BBO-10203 does not induce hyperglycemia or hyperinsulinemia during an oral glucose tolerance test in fasted male C57BL/6 mice. Since activation of the PI3Kα pathway provides acute resistance to endocrine therapies and CDK4/6 inhibitors in HR+ HER2- breast cancer, BBO-10203 has the potential to enhance the long-term responses in combination with these therapies. In vivo studies show that BBO-10203 significantly enhances the anti-tumor activity of the SERD fulvestrant or the CDK4/6 inhibitor ribociclib in ER+ HER2- CDX and PDX breast cancer models harboring either kinase or helical domain mutations in PIK3CA or wild-type PIK3CA. These combinations induce tumor stasis or regressions through direct effects on tumor cells and are well tolerated. Lastly, we compare the activity of BBO-10203 against next-generation mutant-selective PI3Kα inhibitors. In the EFM-19 CDX model, which harbors a kinase domain PIK3CA mutation, the combination of BBO-10203 and fulvestrant show the same activity as the combination of STX-478 and fulvestrant. In conclusion, BBO-10203 blocks RAS-mediated activation of PI3Kα, strongly inhibits pAKT signaling in tumor cells without affecting glucose metabolism, and shows robust tumor activity alone or in combination with standard of care therapies in mutant or wild-type PIK3CA breast cancer models. BBO-10203 has entered phase 1 clinical trials (NCT06625775) and may provide clinical benefit without the limiting toxicities that have restricted the use of PI3Kα inhibitors.
Presentation numberPS2-13-04
Glucocorticoids promote metabolic reprogramming that underlies enhanced proliferation of breast cancer stem and progenitor cells under conditions of chronic stress
Jonathan Dowgielewicz, The University of Chicago, Chicago, IL
J. Dowgielewicz1, J. Caraveo1, B. Faubert1, M. McClintock1, S. Conzen2, M. Brady1; 1The University of Chicago, Chicago, IL, 2UT Southwestern Medical Center, Dallas, TX.
Glucocorticoids promote metabolic reprogramming associated with enhanced proliferation of rat mammary gland cancer cells Exposure to chronic stressors can have a significant role in both disrupting normal mammary gland development as well as potentially negatively impacting the breast cancer outcome. Glucocorticoids (GCs) are stress responsive steroid hormones that mediate cellular and physiological stress responses via activating the GC receptor (GR). While several studies have sought to understand how prolonged physiological GC exposure and GR stimulation negatively impact mammary gland health and breast cancer outcomes, much work remains to be done. We used the rat mammary gland (MG) adenocarcinoma LA7 cell line, a rat MG cancer cell line derived in vitro from an in vivo DMBA carcinogen-induced mammary tumor; to begin to uncover how chronic stress hormones might affect metabolism and energy production in MG epithelial cancer as well as elucidate the molecular mechanisms underlying these effects. LA7 cells were treated either with or without 140nM (high physiological level) of the rat GC corticosterone for 0-72 hours to mimic acute to chronic stress exposure. RNA-Seq results from the LA7 cells demonstrated that under conditions of corticosterone-induced GR stimulation there was a significant increase in expression of mitochondrial-encoded genes such as MT-ND1, MT-CO1, and MT-ATP6, suggesting an unexpected rise in mitochondrial biogenesis. In addition, several nuclear genes encoding proteins involved in mitochondrial metabolism including Isocitrate Dehydrogenase and Succinate Dehydrogenase were also upregulated. We then asked whether these metabolic gene expression changes accompanied expected cell phenotypic changes. MitoTracker Green and Red staining was used to measure mitochondrial number/mass and mitochondrial membrane potential (MMP), respectively. These experiments confirmed that both mitochondrial number/mass and MMP was significantly upregulated by chronic GR activation with GC exposure. In agreement with the MMP staining data, intracellular ATP abundance was shown to be significantly upregulated by chronic GR activation. Using a Seahorse XF Mito-Stress test we observed metabolic reprogramming showing that chronic GR activation promoted significant elevation in basal, ATP-linked, and spare capacity respiration/oxidative phosphorylation (OXPHOS). Because OXPHOS activity is integrally linked to the process of glucose metabolism, we sought to determine if chronic GR activation in the LA7 cells impacts glycolysis. To do this we performed a Seahorse XF Glycolysis Stress test, as well as measured glucose uptake and lactate secretion. The Seahorse XF Glycolysis Stress test revealed an increase in basal glycolysis and glycolytic capacity in the LA7 cells. In parallel, both glucose uptake and lactate production and secretion were also elevated. Lastly, a cellular proliferation assay showed increased cell division following chronic stress hormone exposure. Overall, these observations suggest that in addition to upregulating mitochondrial biogenesis, OXPHOS, and glycolytic activity, corticosterone can contribute to rat MG cancer cellular proliferation. These data also suggest that chronic stress exposure and the resulting GC exposure leads to increased overall cellular metabolism and energy production and promotes a shift to a highly energetic and more proliferative phenotype in a model of mammary cancer.
Presentation numberPS2-13-05
Transcriptomic regulation by pregnane x receptor drives pro-migratory signaling in human breast cancer cells
Catherine E Elliott, Kansas City University, Joplin, MO
C. E. Elliott, D. Brobst, V. Nguyen, C. Ballard, A. Youssef, W. Dye, R. M. Bodily, A. Ko, J. L. Staudinger, B. A. Creamer; Kansas City University, Joplin, MO.
The expression and activation of the nuclear receptor Pregnane X Receptor (PXR) has been linked with poor outcomes in human breast cancer. While PXR’s canonical role in drug metabolism is well defined in the liver and gastrointestinal tract, its contribution to tumor progression is less understood. We sought to define transcriptomic regulation mediated by PXR in breast cancer cells and its potential impact on metastasis. Triple-negative MDA-MB-231 cells were engineered to stably overexpress human PXR and treated with the agonist rifampicin. RNA-seq analysis revealed significant transcriptional changes compared with controls. KEGG and Gene Ontology enrichment identified pathways associated with cytoskeleton remodeling, adhesion complex components, and motility regulators. A panel of ten genes previously implicated in cancer cell migration was consistently up regulated (log2FC > 1.7; p <0.05). Post-hoc qPCR validation confirmed these results. Collectively, these data indicate that PXR expression enhances pro-migratory transcriptional pathways in breast tumor cells, providing mechanistic insight into its potential role as a driver of breast cancer metastasis and contributor to poor prognosis.
Presentation numberPS2-13-06
The M1C/MYC Axis Reprograms Alternative Splicing Networks in Triple Negative Breast Cancer
Atrayee Bhattacharya, University of Texas Health Science Center at Tyler, Tyler, TX
A. Bhattacharya1, L. Ding1, N. Yamashita2, S. Pitroda3, D. W. Kufe2; 1University of Texas Health Science Center at Tyler, Tyler, TX, 2Dana Farber Cancer Institute, Boston, MA, 3University of Chicago, Chicago, IL.
Advanced metastatic triple-negative breast cancer (TNBC) is a highly aggressive malignancy with limited treatment options. TNBCs are enriched in cancer stem cells (CSCs), which contribute to immune evasion and poor clinical outcomes. Approximately 22% of patients diagnosed with metastatic TNBC survive for 5 years, and the average duration of survival is only 3 years. Mucin 1 (MUC1) is a heterodimeric transmembrane glycoprotein that is aberrantly expressed in ~90% of TNBCs. MUC1 consists of two subunits: (i) an extracellular N-terminal subunit (MUC1-N) with glycosylated tandem repeats that are characteristic of the mucin family and (ii) a transmembrane C-terminal oncogenic subunit (M1C). The M1C cytoplasmic domain consists of 72 amino acids (aa) that include a CQC motif adjacent to the transmembrane domain, which is necessary for the formation of homodimers and interactions with binding partners, such as MYC. M1C regulates epigenetic remodeling and cancer stem cell states, yet its role in RNA splicing remains poorly defined. We performed Rapid Immunoprecipitation Mass Spectrometry of Endogenous proteins (RIME) analysis of M1C-associated nuclear proteins in BT549 TNBC cells. Our results demonstrate that M1C associates with RNA processing proteins, including splicing factors. Transcriptomic profiling of BT-549 and SUM149 models using long-read RNA-seq revealed that both inducible and stable silencing of M1C downregulates spliceosome gene signatures and suppresses expression of the SRSF, hnRNP, and RBM families of core splicing regulators. Studies with inducible MYC silencing further demonstrated overlapping regulation of these factors, supporting involvement of the M1C/MYC axis. Alternative splicing analysis using rMATS revealed a consistent shift in splicing subtype usage, with increased intron retention and reduced skipped exons upon M1C silencing. Genes affected by intron retention included regulators of cell cycle, RNA metabolism, and immune signaling, whereas skipped exon events were enriched in transcripts linked to stemness and stress adaptation. In 3D spheroid cultures, M1C and MYC silencing reinforced context-specific splicing alterations. Importantly, combined pharmacologic disruption of M1C and splicing produced synergistic inhibition of TNBC cell viability and suppressed oncogenic isoform expression. Of clinical significance, high expression of MUC1/MYC regulated splicing factors was enriched in basal-like TNBC tumors and correlated with poor survival. Together, these findings define a M1C/MYC axis that drives alternative splicing reprogramming as a hallmark of TNBC biology. Thus, the M1C/MYC axis is a master regulator of alternative splicing in TNBC. Inhibition of M1C and the splicing machinery demonstrates synergistic effects, highlighting a novel therapeutic opportunity in aggressive breast cancers. Future studies will assess whether these splicing events generate immunogenic neoantigens, offering opportunities for biomarker development and translational targeting.
Presentation numberPS2-13-01
Discrete Vulnerabilities to CDK2 Inhibition in Breast Cancer:Genetic Determinants and Therapeutic Opportunities
Erik Knudsen, Roswell Park Cancer Center, Buffalo, NY
E. Knudsen, V. Kumarasamy, A. Dommer, J. Wang, A. Witkiewicz; Roswell Park Cancer Center, Buffalo, NY.
Cyclin-dependent kinase 2 (CDK2) is a critical effector of cell-cycle progression, including during late G1, S, and G2 phases. While CDK4/6 inhibitors have become a backbone in HR+/HER2- breast cancer therapy, resistance frequently emerges through activation of alternative CDK/Cyclin complexes. Notably, the CDK2/Cyclin E axis is frequently associated with resistance to CDK4/6 inhibitors and has driven the development of catalytic CDK2 inhibitors for breast cancer therapy. Here, we sought to: 1, delineate the mechanistic determinants of response to pharmacologic CDK2 inhibition in breast cancer models; 2, define biomarkers predictive of response; and 3, interrogate rational combination strategies to maximize therapeutic efficacy. Using a panel of breast cancer models, it was found that CDK2 catalytic inhibitors induced a bimodal response with two fundamentally distinct phenotypic responses. A minority of cell lines underwent G1 arrest while most accumulated 4N DNA content consistent with a G2/M block. Co-expression of p16INK4A and high Cyclin E1 strongly correlated with the G1 arrest phenotype and marked sensitivity to CDK2 inhibition that was also observed in vivo. Models lacking this co-expression adopted the 4N arrest response and displayed upregulation of phospho-CDK1 (Y15) and Cyclin B1. Critically, this latter phenotype was observed in all HR+/HER2- models tested, including those with engineered resistance to CDK4/6 inhibitors. Using whole-genome CRISPR screens, loss of CDK2 was identified as a top hit conferring resistance to catalytic inhibitors, underscoring the concept that inhibitor binding to CDK2 (rather than simple loss) is essential for the cytostatic effect. Further, deletion of CDK2 reversed the G2/M arrest induced by the inhibitor and restored proliferation. Ahost of potential targets were identified that enhanced sensitivity to CDK2 inhibitors through CRISPR and drug screening approaches. These included CDK4/6 inhibitors that were broadly synergistic with CDK2 inhibitors irrespective of breast cancer subtype. The basis of this cooperation involved arrest in multiple phases of the cell cycle. Several additional combinatorial strategies have recently been defined that further credential CDK2 as a key therapeutic target moving forward. Together this work addresses the complexity of targeting CDK2 for the treatment of breast cancer and multiple opportunity areas for combinations.
Presentation numberPS2-13-02
Daxx is a novel predictive biomarker of response to anti-notch plus endocrine therapy to target cancer stem cells in ER-positive breast cancer
Clodia Osipo, Loyola University Chicago, Maywood Campus, Maywood, IL
C. Osipo1, D. Wyatt1, P. Tang2, L. Miele3, K. S. Albain4; 1Loyola University Chicago, Maywood Campus, Maywood, IL, 2Cook County Health, Chicago, IL, 3Louisiana State University Medical Center, New Orleans, LA, 4Loyola University Chicago Stritch School of Medicine, Maywood, IL.
Background: Notch promotes cancer stem cells (CSC) and resistance to endocrine therapy (ET) in ER+ breast cancer (BC). A Pre-surgical window study using ET plus a γ-secretase inhibitor (MK-0752) in ER+ BC identified novel Notch-regulated genes. Expression profiling showed more genes were downregulated by ET + GSI than ET alone. Of the few genes increased by ET plus the GSI, DAXX was the only gene required for GSI-mediated blockade of CSC survival in two distinct ER+ BC cell lines (MCF-7 and BT474) as measured by mammosphere-forming efficiency (MFE). Goals of the current research were to determine if DAXX is a 1) novel target gene of Notch1 or Notch4 and repressed by Notch signaling, 2) bona fide CSC suppressor, 3) predictive biomarker of anti-tumor efficacy of the GSI MK-0752 in a xenograft model, and 4) marker of response to ET in the neoadjuvant setting to inform if MK-0752 should be added to ET. Methods: Enrichment of Notch1 on CSL elements for validated genes was performed using ChIP-PCR. CSC survival was assessed by MFE when DAXX was knocked down. RT-PCR and western blotting measured DAXX RNA and protein expression under GSI treatment or Notch1 or Notch4 knockdown. RNA-sequencing was conducted from MCF-7 cells-expressing DAXX and depleted for DAXX. Tumor xenograft studies were performed using MCF-7 cells both expressing DAXX (DAXX+) and depleted for DAXX (DAXX-). Mice were treated with estrogen, estrogen deprivation, to mimic the use of an aromatase inhibitor, 75mg/kg GSI MK-0752, or estrogen deprivation plus MK-0752 for 80 days. Tumor volume was measured weekly. Survival of mice was monitored using Kaplan-Meier analysis. On day 80, tumors were excised and their masses weighed. To inform which patients would benefit from addition of MK-0752 to ET, DAXX and Ki67 protein expressions were measured by IHC and blindly scored in 35 untreated and then either ET- or chemotherapy-treated human ER+ tumors. Statistical calculations were performed using linear regression analysis, a T-test for two groups, or an ANOVA for multiple groups. Results: Notch1 enrichment on CSL elements increased with ET versus estradiol by 2-fold and attenuated with GSI treatment for most of the genes except for NOXA and PGR. The GSI eliminated CSCs from DAXX-expressing MCF-7 (P=9.4e-15) and BT474 cells (P<0.0001). DAXX-depleted cells were enriched for CSCs (MCF-7:P=0.004; BT474:P=0.0003). DAXX knockdown partially rescued the GSI-mediated inhibition of CSCs (MCF-7:P=0.003; BT474:P=0.0003). GSI or Notch1/Notch4 knockdown increased DAXX RNA and protein expression. RNA-sequencing of DAXX+ and DAXX- MCF-7 cells showed DAXX knockdown increased pluripotent stem cell genes (NANOG, SOX2, KLF4, and ALDH1A1) and decreased luminal genes (GATA3, CDH1, ESR2, and FOXA1). In mice, ET (estrogen deprivation) combined with MK-0752 GSI reduced tumor growth of MCF-7 xenografts compared to ET alone (P<0.05). DAXX was required for the anti-tumor efficacy of ET plus MK-0752 GSI (P=0.0084). Out of 35 patient tumors scored for DAXX and Ki67, 10/35 expressed less DAXX post-treatment (P=0.0052) and 25/35 retained expression. Lower DAXX was associated with less change in Ki67 levels (P=0.0097). High DAXX correlates with better response to ET as measured by Ki67. Tumors with decreased DAXX may potentially benefit from addition of MK-0752. Conclusions: This research in both cell lines and xenografts uniquely demonstrated that GSI-induced DAXX was necessary for the anti-CSC and anti-tumor efficacy of the GSI. DAXX is a potent CSC inhibitor and possible predictive biomarker of response to ET or ET plus the GSI MK-0752. In summary, ET plus a GSI modulated Notch and other CSC genes, in particular DAXX, a potential predictive biomarker of ET plus GSI efficacy in ER+ BC. Testing of anti-Notch or DAXX-promoting agents should be a clinical trial priority. Funding: BCRF
Presentation numberPS2-13-07
Disrupted epithelial differentiation and mitochondrial remodeling as early events in BRCA-associated carcinogenesis
Quentin Chartreux, UTSA Health Science Center, San Antonio, TX
Q. Chartreux1, M. Haro1, J. Brand2, A. Li3, B. J. Rimel3, S. Gayther1, H. Dinh2, F. Madeiros3, K. Lawrenson1; 1UTSA Health Science Center, San Antonio, TX, 2School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 3Cedars-Sinai Medical Center, Los Angeles, CA
Germline mutations in BRCA1 and BRCA2 dramatically increase the lifetime risk of developing both ovarian and breast cancers. While the fallopian tube epithelium has been recognized as the likely cell of origin for high-grade serous carcinoma (HGSC), the molecular mechanisms that initiate malignant transformation in BRCA1/2-associated tissues—whether in the fallopian tube or the breast—remain poorly defined. Understanding these early events is critical to identify shared pathways that predispose BRCA mutation carriers to cancer. To address this, we applied single-cell transcriptomic and epigenomic profiling to fallopian tube samples from BRCA1/2 mutation carriers undergoing prophylactic risk-reducing salpingo-oophorectomy (RRSO), aiming to uncover early cellular and molecular alterations that may also inform early breast tumorigenesis.We compared the cellular and transcriptomic landscapes of fimbrial epithelial brushings obtained via exfoliative cytology with those from conventional tissue specimens. Single-cell RNA sequencing was performed on 14 fimbrial brushings from high-risk BRCA1/2 mutation carriers, 3 brushings from non-carriers, and 12 tissue samples from non-carriers. In parallel, single-cell multi-omic analyses were conducted on short-term epithelial cultures derived from 4 high-risk and 2 average-risk individuals. This design enabled a high-resolution assessment of epithelial heterogeneity, transcriptional states, and gene regulatory differences between BRCA mutation carriers and controls.Fimbrial brushings primarily contained epithelial and immune populations, with a relative depletion of fibroblasts and endothelial cells compared to tissue samples, allowing detailed analysis of epithelial biology. Cells from BRCA mutation carriers exhibited marked transcriptomic alterations, including disrupted epithelial differentiation trajectories and upregulation of genes involved in mitochondrial respiration and oxidative phosphorylation. These findings suggest early mitochondrial and metabolic remodeling that may precede malignant transformation. In addition, transcriptional remodeling of immune-related pathways indicated subtle immunologic changes in the local microenvironment that could contribute to a permissive state for tumor initiation.By uncovering early transcriptomic and metabolic reprogramming in BRCA-mutated epithelia, this work provides new insights into the biological underpinnings of hereditary cancer predisposition. The identification of early molecular alterations in BRCA mutation carriers may inform the development of biomarkers and preventive strategies relevant to both ovarian and breast cancer, ultimately advancing risk prediction and early detection efforts.
Presentation numberPS2-13-08
Neoadjuvant Dalpiciclib Plus Letrozole Versus Standard Chemotherapy in High-Risk HR+/HER2- Negative Breast Cancer (DARLING-02): A Randomized Phase II Trial
Cuizhi Geng, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
C. Geng1, L. Zhang1, C. Yang1, T. Zhou1, L. Ma1, R. Luo1, Z. Song1, Y. Qi1, J. Yang2, X. Wang3, J. Han4, J. Ma5, Z. Zhang6, Y. Li1, C. Gao7, L. Yang8, J. Yu9; 1The Fourth Hospital of Hebei Medical University, Shijiazhuang, CHINA, 2HengShui People’s Hospital, Hengshui, CHINA, 3Affiliated Hospital of Hebei University, Baoding, CHINA, 4Affiliated Hospital of Hebei University of Engineering, Handan, CHINA, 5Tangshan People’s Hospital, Tangshan, CHINA, 6The First Affiliated Hospital of Hebei North University, Zhangjiakou, CHINA, 7Baoding NO.1 Central Hospital, Baoding, CHINA, 8Xingtai People’s Hospital, Xingtai, CHINA, 9Xingtai Central Hospital, Xingtai, CHINA.
Neoadjuvant Dalpiciclib Plus Letrozole Versus Standard Chemotherapy in High-Risk HR+/HER2– Negative Breast Cancer (DARLING-02): A Randomized Phase II TrialBackground: Dalpiciclib, a cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, has shown significant efficacy and a favorable safety profile when combined with endocrine therapy for advanced hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer. Building on this evidence, the present study evaluated the efficacy and safety of neoadjuvant dalpiciclib plus letrozole compared with standard chemotherapy in patients with high-risk HR+/HER2- early breast cancer.Methods: DARLING-02 was a multicenter, randomized, phase II non-inferiority trial (NCT06107673). Key eligibility criteria included women aged ≥18 years; Eastern Cooperative Oncology Group performance status 0-1; clinical stage T1c-2N1-2 or T3-4N0-2; estrogen receptor (ER) expression ≥50%; HER2- status; and Ki-67 level ≤30%. Patients were randomized 1:1 to receive 24-week treatment. The experimental arm received dalpiciclib 150 mg orally once daily (3 weeks on, 1 week off) plus letrozole 2.5 mg orally once daily. The control arm received epirubicin 100 mg/m² plus cyclophosphamide 500 mg/m² intravenously on day 1 every 3 weeks for 4 cycles, followed by docetaxel 100 mg/m² intravenously on day 1 every 3 weeks for 4 cycles. The primary endpoint was objective response rate (ORR), assessed per Response Evaluation Criteria in Solid Tumors, version 1.1. Results: Between August 2023 and April 2025, 170 patients were screened; 158 met the eligibility criteria and were randomized to the dalpiciclib-letrozole (n=80) or chemotherapy (n=78) group. At the data cut-off date (September 17, 2025), 9 patients in the experimental arm and 10 in the control arm were still on neoadjuvant therapy. Among patients with completed short-term efficacy assessment, the ORR was 69.0% (49/71) with dalpiciclib-letrozole and 70.5% (48/68) with chemotherapy; total pathological complete response (tpCR) rate was 5.6% (4/71) and 1.5% (1/68), respectively. The proportion of patients with Preoperative Endocrine Prognostic Index (PEPI) score ≥4 was higher in the chemotherapy group (74.6% [44/59]) than in the dalpiciclib-letrozole group (53.3% [32/60]). Grade ≥3 adverse events were predominantly hematological toxicities and occurred less frequently with dalpiciclib-letrozole than with chemotherapy (decreased white blood cell count: 36.4% [28/77] vs 64.0% [39/61]; decreased neutrophil count: 74.0% [57/77] vs 86.9% [53/61]).Conclusions: These findings support dalpiciclib plus letrozole as a promising neoadjuvant option for patients with high-risk HR+/HER2- early breast cancer and warrant confirmation in phase III trials.
Presentation numberPS2-13-10
Impact of Dose Intensity on Pathological Complete Response in Early-Stage HER2-Positive Breast Cancer
Shaheed K AlHumiued, Eastern health cluster, Dammam, Saudi Arabia
B. T. Albaqshi1, S. K. AlHumiued1, H. Abbas2, A. Alamer3, S. M. Alonaizi4, R. A. Alyousef4, F. R. Alqattan5, A. Salam6, F. A. Almulhim7; 1Clinical pharmacy, Eastern health cluster, Dammam, SAUDI ARABIA, 2Oncology MD, Eastern health cluster, Dammam, SAUDI ARABIA, 3Clinical pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, SAUDI ARABIA, 4Clinical pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, SAUDI ARABIA, 5Clinical pharmacy, King Faisal University, Alahsa, SAUDI ARABIA, 6Epidemiology and Biostatistics Administration, Esteran health cluster, Dammam, SAUDI ARABIA, 7population health management, eastern health cluster, Esteran health cluster, Dammam, SAUDI ARABIA.
Background: Chemotherapy dose intensity (DI), defined as the amount of drug given per unit of time, is a key determinant of efficacy in curable malignancies such as early-stage HER2-positive breast cancer. DI below 0.85 has been linked to worse survival outcomes and lower pathological complete response (pCR) rates. Factors contributing to reduced DI include myelosuppression, toxicities, drug shortages, and healthcare system barriers. This is among the first real-world studies to evaluate chemotherapy DI in early-stage HER2-positive breast cancer in a Middle Eastern population. Objective: This study aimed to evaluate the impact of chemotherapy dose intensity (DI) of the neoadjuvant TCHP regimen (docetaxel, carboplatin, trastuzumab, pertuzumab) on pathological complete response (pCR) rates in patients with early-stage HER2-positive breast cancer. A secondary objective was to identify patient- and system-related factors contributing to reduced DI, including the rate, frequency, and underlying causes of dose reductions and treatment delays. Methods: This retrospective cohort study included patients ≥18 years with stage I-III HER2-positive breast cancer treated with neoadjuvant TCHP at a tertiary center in Saudi Arabia from 2017 to 2023. Dose intensity (DI) was calculated as actual cumulative dose divided by intended dose ×100. The primary outcome was the association between DI and (pCR). Patients were stratified by DI (<85% vs. ≥85%) and pCR status. Secondary outcomes included factors contributing to dose reductions and delays. Analyses included descriptive statistics and multivariable logistic regression.Results: A total of 149 patients were included. Most (73.2%) had nodal involvement; 59.7% were hormone receptor (HR) positive. The pCR rate was 60.4%. No significant differences in pCR were observed between DI groups for docetaxel (60% vs. 61%; p = 1.00) or carboplatin (65% vs. 56%; p = 0.35). Median DI was highest for trastuzumab (100%) and lowest for pertuzumab (72.2%). Protocol deviation to weekly paclitaxel occurred in 43.6% of patients. Carboplatin DI was independently associated with higher odds of pCR (OR 1.01; 95% CI, 1.00-1.04; p = 0.029), while HR positivity was associated with lower pCR odds (OR 0.23; 95% CI, 0.09-0.61; p = 0.004). Switching to paclitaxel showed a non-significant reduction in pCR (OR 0.26; 95% CI, 0.05-1.37; p = 0.107). Docetaxel had the highest delay rate, affecting up to 30% of patients, mainly due to rescheduling. Conclusion: In this real-world cohort, chemotherapy DI reductions were common. DI ≥0.85 was associated with numerically higher pCR, but only carboplatin DI independently predicted pCR. HR positivity and docetaxel substitution were linked to lower pCR odds. Docetaxel and pertuzumab experienced the most delays, mainly from system-related factors. These findings highlight the importance of adherence to protocol and timely delivery. Addressing modifiable system-level barriers and adhering to guideline-based chemotherapy are essential. Larger multicenter studies are warranted to confirm these findings.
Presentation numberPS2-13-11
Enhancing Nurse Competence in Malignant Wound Management: A Multicenter Assessment of Knowledge, Attitudes, and Practices in Integrated Care Settings
Liang Xu, Peking University Cancer Hospital & Institute, Beijing, China
L. Xu, J. Tian, M. Zhu, H. Chen, H. Li, M. You; Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, CHINA.
Background: Malignant wounds present as ulcerative, cauliflower-like, volcano-like, and sinus tract-like lesions. Patients suffer not only from life-threatening cancer but also distressing symptoms (exudation, odor, pain, and bleeding), which significantly impact their quality of life. As specialized wound nursing advances, nurses play an increasingly vital role in malignant wound management. This study evaluated nurses’ knowledge, attitudes, and practices (KAP) in this field. Methods: The convenience sampling method was used to select nurses implementing the integrated medical care model in 42 Chinese medical centers. This study was approved by the Ethics Committee of Beijing Cancer Hospital (2022YJZ33). The general information questionnaire had 5 items: participant’s age, gender, work experience, education level, professional title, and wound care training status. Specialized wound care nurses were nurses with a certificate of completion in wound knowledge training. The Malignant Wound Management KAP Scale Questionnaire was self-designed and previously tested. Data analysis used SPSS 25.0. Inferential statistics employed the independent samples t-test and one-way analysis of variance (ANOVA) to compare scores. Multiple linear regression analysis was used to identify influencing factors. Results: From August 12, 2022 to June 27, 2024, 1002 nurses provided valid responses to the survey. The participants had a median age of 34 (21-56) years, and 86% had a bachelor’s degree. Nurses were divided into 4 groups based on training status, including non-wound (Group 1, 55.5%) and wound (Group 2, 10%) specialist nurses without training in malignant wound management and non-wound (Group 3, 17.4%) and wound (Group 4, 17.1%) specialist nurses with training in malignant wound management. Totally 34.5% (346/1002) had training for malignant wound management. In KAP score analysis, the participants showed a moderate level of knowledge, with a mean score of 15.24 ± 2.51 (72.6% of the maximum score) and had favorable attitudes, with a mean score of 59.02 ± 6.61 (90.8% of the maximum score). However, practical competency remained relatively low, with a mean practice score of 72.10 ± 19.58 (62.7% of the maximum score). Univariate analysis of KAP in malignant wound management showed age, years of experience, education level, professional title, and wound-related training significantly affected knowledge score (P<0.05); age, years of experience, professional title, and wound-related training significantly affected attitude score (P<0.001), while years of experience, city classification, and wound-related training significantly impacted practice score (P<0.01). Multiple linear regression analysis further revealed wound-related training as a factor affecting KAP (P10 years of experience had significantly higher scores (P=0.014), while those in second- and third-tier cities had lower scores (P=0.013 and P=0.003, respectively). Conclusion: While nurses had positive attitudes toward malignant wound management and adequate knowledge, their clinical practice required improvement. Training emerged as a critical factor affecting KAP but is rarely undertaken in China, with existing professional wound care training programs often lacking dedicated contents for malignant wound management. These findings highlight the critical roles of specialized training and professional experience in improving KAP outcomes in malignant wound care: training initiatives for junior nurses are required, as well as collaboration between primary and tertiary hospitals.
Presentation numberPS2-13-12
The BRAVE Study: Exploring the pathways to a breast cancer diagnosis from the perspectives of young survivors in California
Alice W Lee, California State University, Fullerton, Fullerton, CA
A. W. Lee1, M. Sami1, P. Fisher2, D. Aguilar-Cruz1, T. Do3, M. Vargas1, A. La Flair2, N. Wells2; 1Public Health, California State University, Fullerton, Fullerton, CA, 2The Young Breast Cancer Project, The Young Breast Cancer Project, Chula Vista, CA, 3Psychology, California State University, Fullerton, Fullerton, CA.
Background: Incidence of breast cancer is significantly increasing in young women. Unfortunately, young women often present with more aggressive, advanced stage tumors and have poorer prognoses relative to older women, making timely diagnosis imperative. However, studies have shown that young women often experience diagnostic delays when it comes to breast cancer. To date, no study has explored the underlying reasons for such delays in young women in the U.S., which could inform strategies for timely diagnosis. Methods: As part of the Breast Cancer in Young Women: Awareness, Views, Experiences (BRAVE) Study, we conducted two virtual semi-structured focus groups with a total of 16 women diagnosed with breast cancer under age 40 in California to better understand their pre-diagnostic experiences, including their awareness, attitudes, and behaviors leading up to their diagnoses. Each focus group was recorded, transcribed, coded, and analyzed using NVivo 15, and emergent themes were identified. A survey that included demographic, tumor, and risk factor-related questions was also administered. Results: Across the 16 focus group participants, the average age at time of survey was 39 years and the average age at diagnosis was 35 years. Three key themes emerged from the focus groups. First, participants consistently felt a need and a sense of urgency to self-advocate, with many describing the diagnostic process as burdensome and requiring persistent interactions with healthcare providers. Second, a recurring concern participants expressed was the perception that their initial symptoms were dismissed or minimized by providers, usually attributed to their young age. Third, participants identified obstetrics/gynecology (OB/GYN) providers as central healthcare figures during their reproductive years, suggesting their potential gatekeeping role when it comes to addressing young women’s breast health. Conclusions: Timely diagnosis of breast cancer in young women likely stems from a combination of factors, including patients’ persistence and ability to self-advocate as well as healthcare providers’ receptiveness to take further diagnostic steps despite patient’ risk profiles indicating otherwise (e.g., age), which may be especially important for OB/GYN providers who often serve as young women’s primary point of contact for health concerns. It is necessary to also explore the perceptions of a young woman’s pathway to a breast cancer diagnosis from the perspectives of healthcare providers to better understand the barriers and facilitators to a timely diagnosis in young women. Interviews with providers as well as additional focus groups with young breast cancer survivors are currently underway.
Presentation numberPS2-13-13
Mortality in HER2-positive male breast cancer between 2010 and 2022: A population-based study
Jose P. Leone, Dana-Farber Cancer Institute, Boston, MA
J. P. Leone1, D. Trapani2, M. J. Hassett1, D. L. Abravanel1, P. M. Spanheimer3, R. A. Freedman1, C. T. Vallejo4, S. M. Tolaney1, N. U. Lin1, J. Leone4; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Medical Oncology, European Institute of Oncology, Milan, ITALY, 3Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4Medical Oncology, Grupo Oncológico Cooperativo del Sur, Neuquén, ARGENTINA.
Background: Approximately 13% of all breast cancers (BC) in men are HER2 positive. Previous studies have suggested that men with HER2-positive BC have worse prognosis compared to women with HER2-positive disease. Despite advances in HER2-positive BC treatment over recent years, it remains unclear whether mortality rates have decreased in men as they have in women. The aims of this study were to compare breast cancer-specific mortality (BCSM) between men and women with HER2-positive BC and evaluate changes in BCSM in each sex over time. Methods: We evaluated men and women diagnosed with HER2-positive invasive BC between 2010 and 2022, reported in the Surveillance, Epidemiology, and End Results registry. Patients were categorized into two groups by year of diagnosis: 2010-2015, and 2016-2022. BCSM was estimated by cumulative incidence function and differences between groups were compared by Gray’s test. Fine and Gray model was used to evaluate differences in BCSM between men and women after adjusting for age, year of diagnosis, race, ethnicity, tumor grade, tumor histology, hormone receptor status, tumor stage, surgery type, chemotherapy, radiation, marital status, income, rurality, and region of residence. Stratified adjusted Fine and Gray models were fitted for each sex to assess whether there was an association between year of diagnosis and BCSM. For all analyses of BCSM, deaths from causes other than BC were treated as a competing event. Results: We included 483 men (2010-2015: N = 189, 2016-2022: N = 294) and 85,693 women (2010-2015: N = 36,578, 2016-2022: N = 49,115) diagnosed with HER2-positive BC. Comparing men vs women in the overall population, men were older (median age 62 vs 57 years), more often non-Hispanic white (68% vs 58%), presented more often with de-novo stage IV (14.7% vs 9.2%) and with hormone receptor positive BC (94.4% vs 69.7%), respectively (p<0.001 for each). The median follow-up for patients diagnosed 2010-2015 was 9.4 years in men and 9.5 years in women; for patients diagnosed 2016-2022 was 3.4 years in men and 3.3 years in women. Overall, BCSM at 5 years was 19.8% in men vs 11.6% in women, p<0.001. Men had higher risk of BCSM than women in the adjusted Fine and Gray model (adjusted subdistribution hazard ratio [aSHR], 1.3; 95% CI, 1.02-1.68). Among men, BCSM at 5 years was 20.0% and 20.5% in periods 2010-2015 and 2016-2022, respectively; p=0.9. In women, BCSM at 5 years was 12.5% in 2010-2015 and 10.4% in 2016-2022, p<0.001. In adjusted Fine and Gray models evaluating year of diagnosis as a continuous variable, each successive year of diagnosis had no significant association with BCSM in men (aSHR, 0.99; 95% CI, 0.91-1.08; p=0.9), but a significant association with BCSM in women (aSHR, 0.97; 95% CI, 0.96-0.98; p<0.001). Analysis of overall survival (OS) showed that men had significantly worse OS than women (5-year OS rate 65.3% vs 84.1%, respectively; p<0.001). Men had no significant differences in OS for those diagnosed 2010-2015 vs 2016-2022 (5-year OS rate 64.4% vs 64.7%, respectively; p=0.6). 5-year OS rate in women was 83.4% in 2010-2015 vs 85.1% in 2016-2022; p<0.001. Exploratory analyses revealed no significant improvement in BCSM across the two diagnosis periods in men with stage I-III and men with stage IV BC, whereas women had significant improvement in BCSM in both. Women showed improvements in BCSM in both the HR+/HER2+ and HR-/HER2+ subgroups, while men did not show any improvements. Conclusions: Men with HER2-positive BC exhibit significantly worse BCSM compared to women. Since 2010, BCSM has not improved in men with HER2-positive disease, contrasting with the observed reductions in BCSM among women. The lack of survival improvement in men with HER2-positive BC over time, coupled with the persistent higher mortality risk, underscores the substantial unmet medical needs for this population.
Presentation numberPS2-13-14
Transformation of ER/PR Positive Invasive Ductal Carcinoma to Triple-Negative Metastatic Breast Cancer Following Adjuvant Abemaciclib: A Case Report
Allison Poles, Rush University Medical Center, Chicago, IL
A. Poles, S. Atluri, R. Rao; Internal Medicine, Rush University Medical Center, Chicago, IL.
Background Abemaciclib is an oral CDK 4/6 inhibitor approved by the FDA in October 2021 for adjuvant treatment of HR-positive, HER-2 negative early breast cancer in combination with endocrine therapy (1). In the monarchE trial, 2 years of abemaciclib with endocrine therapy improved 5-year absolute invasive disease-free survival by 7.6% and distant relapse-free survival by 6.7% in high-risk, node positive patients (1). Findings A 45-year-old female was diagnosed with bilateral locally advanced IDC. Right breast biopsy showed ER 70%, PR 90%, HER2 0, Ki-67 90% (hot spots), and left breast biopsy showed ER > 90%, PR 5-10%, HER2 0, Ki-67 > 20%, both with Nottingham grade 3 (3+3+2=8) histology and bilateral axillary lymph node biopsies positive for macrometastatic carcinoma. Staging scans at diagnosis, including CT CAP and bone scan, were negative for distant metastasis. Genetic testing was negative. She was treated with 4 cycles of neoadjuvant dose dense AC-Taxol, then bilateral mastectomies, radiation, oophorectomy for ovarian suppression and letrozole for endocrine therapy. Given prior nodal involvement she began adjuvant abemaciclib six months after mastectomy. 8 months into abemaciclib treatment new bony metastases were incidentally found during imaging for pancreatitis. Bone scan confirmed new foci of increased uptake in C2/C3, T11, L4 and the left acetabulum concerning for osseous metastasis. Left acetabular core needle biopsy revealed metastatic adenocarcinoma consistent with known breast primary, ER positive (5%), PR negative (0%) and HER2 negative (1+). Given strong discrepancies between original strongly ER positive breast cancers and the bone metastases with low ER positivity of 5%, she underwent an FES PET which confirmed her osseous metastasis did not demonstrate any ER expression, further confirming change in the tumor’s ER status. Tempus testing revealed multiple inactionable pathogenic mutations, including two TP53 mutations (c.783-2A>C Splice region variant – LOF VAF: 59.8% and p.R267_N268delinsPD – c.800_802delinsCGG Missense variant – LOF VAF: 55%). Conclusions A study by Corti et al. (2025) evaluated changes in HR status and recurrence among high-risk, HR-positive, HER2-negative early breast cancer treated with adjuvant abemaciclib plus endocrine therapy. Of 163 patients, 15 (9.2%) had recurrence during or shortly after completing abemaciclib (2). The median time to recurrence was 8 months. While half the recurrent tumors had strong ER positivity at time of diagnosis, on recurrence the biomarkers showed statistically significant changes in HR status, with ER positivity ≤10% and PR positivity < 1% on first recurrence. Most common recurrence sites were liver (40%), bone (26.7%), and lung (13.3%). Interestingly, the tumors with recurrence showed alterations in the p53 pathway and of those that had next genomic sequencing completed (n=10), alterations in p53 were found in 90% of the subset. Our goal is to highlight the transition of HR status after adjuvant abemaciclib as our case illustrates a unique presentation of disease recurrence. Further study should evaluate the frequency of new p53 mutations in patients treated with abemaciclib and its role in the transition of HR status.
- Rastogi P, O’Shaughnessy J, Martin M, et al. Adjuvant Abemaciclib Plus Endocrine Therapy for Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative, High-Risk Early Breast Cancer: Results From a Preplanned monarchE Overall Survival Interim Analysis, Including 5-Year Efficacy Outcomes. J Clin Oncol 2024; 42:987-993
- Corti C, Martin AR, Kurnia PT, et al. Clinicopathological features and genomics of ER-positive/HER2-negative breast cancer relapsing on adjuvant abemaciclib. ESMO Open. 2025 Jun 3;10(6):105126. doi: 10.1016/j.esmoop.2025.105126
Presentation numberPS2-13-15
Systemic knowledge gaps in IBC recognition: a barrier to timely diagnosis and care
Tse Yen Tan, Duke University School of Medicine, Durham, NC
T. Tan1, A. Bennion, MS1, K. Lebron1, J. Barbour1, G. Devi2, A. N. Tran1; 1Duke University School of Medicine, Durham, NC, 2Hollings Cancer Center Medical University of South Carolina, Charleston, SC
Background: Inflammatory Breast Cancer (IBC) and other rare, aggressive breast cancer subtypes disproportionately impact younger women and is often misdiagnosed or diagnosed at later stages due to atypical presentation, limited awareness, and lower detection rate on screening mammograms While IBC incidence is low at around 2%, it accounts for up to 10% of all breast cancer deaths. Further, racial disparities in IBC incidence are extensively documented, with literature documenting black women as having significantly higher rates than white patients. Primary care providers (PCPs) are generally the first point of contact for patients, making their ability to recognize IBC critical. However, diagnostic decisions can be influenced by cognitive shortcuts and system-level constraints, which may contribute to delayed recognition. Given known urban-rural disparities of health care access and outcomes for various conditions, this study examined whether PCP awareness and confidence in diagnosing IBC differ by practice setting in North Carolina (NC). Methods: The PCPs recruited in this survey included physicians, nurse practitioners or certified nurse midwives and physician assistants, and majority practiced family medicine. Rural PCPs (N=30) and urban PCPs (N=78) within NC were recruited through institutional patient portals via convenience sampling to complete an online, structured survey that included confidence scales and open-ended questions. Survey items assessed participant demographics, IBC awareness, confidence levels, and recognition of 18 IBC-specific knowledge items (11 symptoms, 7 risk factors). Quantitative data was analyzed using descriptive statistics and bivariate comparisons to identify similarities and differences between rural and urban PCP responses. Results: Only 63% (19/30) of rural PCPs and 59% (46/78) of urban PCPs had heard of IBC prior to the survey (p = 0.85), highlighting a similar limited lack of awareness surrounding IBC regardless of practicing setting. Likewise, rural and urban PCPs reported similar levels of confidence on their ability to detect IBC. The mean confidence score (0-7 scale) was similar among rural and urban PCPs (3.39 vs 3.35), highlighting a low to moderate confidence in recognizing IBC. A chi-squared test of independence found no statistically significant difference in distribution of confidence ratings between rural and urban PCPs (χ² = 4.65, df = 6, p = 0.59). Urban and rural PCPs demonstrated high recognition rates of 9 clinically characteristic IBC symptoms, with statistically similar rates for 8 of the 9 symptoms: breast swelling, peau d’orange, breast tenderness/pain/itching, redness, skin thickening, nipple discharge, inverted nipple, palpable breast mass (all p > 0.05). Rural PCPs were significantly more likely than urban PCPs to recognize lymph node change as a symptom of IBC (90.0% vs. 69.2%, χ² = 6.75, p = 0.0094). When asked about initial follow-up steps for a hypothetical case of IBC, most rural (25/30; 83%) and urban PCPs (59/78; 75.6%) indicated they would refer for breast imaging (i.e. mammogram or ultrasound) after a failed course of antibiotics. A majority expressed interest in additional training to better diagnose and care for patients with IBC, with online CME being the preferred format (rural 22/30, 73.3%; urban 44/78, 56.4%). Conclusion: Awareness and confidence in diagnosing IBC remain limited among PCPs across of practice setting, suggesting that challenges extend beyond geography and reflect broader systemic gaps. These preliminary findings highlight an urgent need and demand for scalable, evidence-based strategies – such as targeted education and decision support tools– to strengthen early recognition and reduce delays in care for patients with IBC.
Presentation numberPS2-04-01
Early Adoption of Molecular Residual Disease Testing in Breast Cancer Patients using Real World Data
Ning Yan Gu, Exact Sciences, Madison, WI
N. Gu1, M. Kohli2, S. J. Noga3, Q. A. Le1, J. Rodriguez-Silva2, Y. Barhoush4, L. Batchu4, G. C. Carter1; 1Health Economics and Outcomes Research, Exact Sciences, Madison, WI, 2Research/RWE, STATinMED, Dallas, TX, 3Medical Affairs, Exact Sciences, Madison, WI, 4Programing/Analytics, STATinMED, Dallas, TX.
Early Adoption of Molecular Residual Disease Testing in Breast Cancer Patients using Real World DataAuthors: Ning Yan Gu1, Kohli M2, Stephen J Noga1, Quang A. Le1, Rodriguez-Silva J2, Barhoush Y2, Batchu L2, Gebra Cuyun Carter11Exact Sciences, Madison, WI, USA; 2STATinMED LLC., Dallas, TXWord count: 2765/3400 characters OBJECTIVE: Testing for molecular residual disease (MRD) via circulating tumor DNA (ctDNA) biomarker is an emerging practice in cancer treatment and disease recurrence detection. This study aims to assess the early adoption of MRD for patients diagnosed with breast cancer (BC) using real-world data. METHODS: A national multi-payer medical and pharmacy claims data (STATinMED RWD Insights) with nearly 80% of the US healthcare system was used. Data were extracted for female patients aged ≥18 years between January 1, 2015, and November 30, 2023. Index date was defined as the first date of BC diagnosis. Included patients were required to have continuous enrollment for a minimum of 12 months pre-index, 30 days post-index, and had ≥1 inpatient or ≥2 outpatient BC claims (ICD-9-CM: 174x, 175x; ICD-10-CM: C50x). Patients were excluded if they had metastasized disease, other cancers (not including non-melanoma skin cancers), any BC treatment, or had evidence of pregnancy during the 12-month baseline period. Proxies were used to determine BC subtype and cancer stage. MRD utilization was identified from the index date to study end time and/or patients’ death. Multivariate logistic regression was used to identify predictors of the MRD test adoption. RESULTS: Among 652,463 eligible patients (mean age: 65.4 years), 815 (0.12%) had evidence of MRD testing, the first one was in 2021, increased in 2022, and continued to the study end date of November 30th, 2023, with most of the MRD testing occurred in 2023 (n=748, 91.8%). The mean follow-up time per patient was 38.76 months (SD: 26.57) with a median follow-up per patient of 29.70 months. The mean age of MRD tested patients was 62.7 years; 67.7% were White; 60.1% were non-Hispanic; 36.0% had a college education; 51.4% had Medicare; 27.3% had an income below $25,000 and 55.5% resided in the Southern United States. Categorizing the subtype, 65.2% had HR+/HER2- BC, 23.3% had triple-negative BC (TNBC), and 9.5% had HR+/HER2+ BC. During the study time frame, 60.9% of patients received only one MRD test, 23.1% received two, 8.3% received three and the remaining 7.7% of patients received four or more MRD tests, for up to 10 MRD tests. The mean number of MRD tests per patient was 1.72 (SD: 1.27). Among the 815 MRD-tested patients, 392 (48%) had evidence of breast-conserving surgery (BCS). Of those 392 patients, 5 (1.3%) received MRD testing before the BCS and 387 (98.7%) received MRD test after the BCS. For every BC subtype, the average time between tests was > 80 days between the 1st and 2nd tests, > 70 days between the 2nd and 3rd tests, with shorter intervening times for additional MRD tests conducted during study period. Based on the multivariate logistic regression, key factors associated with the receipt or adoption of MRD test included younger age, advanced BC stage, BC subtype, and patients’ residence region. CONCLUSION: This descriptive analysis using real-world data offers insights into the early adoption of the MRD testing among BC patients in the United States. The real-world evidence showcased an upward adoption trajectory during the study period and there were variations in early utilization patterns. Findings herein provide hypothesis generating opportunities for future investigations. Key Words: 1.Molecular residual disease 2.MRD testing3.ctDNA4.Breast cancer 5.Real world data
Presentation numberPS2-06-29
Chemotherapy-free neoadjuvant strategy in HR+/HER2- breast cancer: CDK4/6 inhibitors plus endocrine therapy show comparable efficacy to chemotherapy-sequenced approach
Ang zheng, The First Hospital of China Medical University, Shenyang, China
A. zheng1, Z. Dong1, D. Song2, F. Fang3, J. Li4, D. Zhang5, G. Zhu6, B. Guo7, J. Li8, Y. Zhao9, N. Zhang10, F. Jin1, B. Chen1; 1Breast surgery, The First Hospital of China Medical University, Shenyang, CHINA, 2Breast surgery, The First Bethune Hospital of Jilin University, Changchun, CHINA, 3Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, CHINA, 4Breast surgery, Liaoning Cancer Hospital and Institute, Shenyang, CHINA, 5Breast and Thyroid Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, CHINA, 6Breast oncology surgery, The Fifth People’s Hospital Of Shenyang, Shenyang, CHINA, 7Breast surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, CHINA, 8Breast surgery, Shenyang the Fourth Hospital of People, Shenyang, CHINA, 9Breast surgery, Shengjing Hospital Of China Medical University, Shenyang, CHINA, 10Breast surgery, Chaoyang Central Hospital, Chaoyang, CHINA.
Background: The neoadjuvant treatment landscape for HR+/HER2- breast cancer is rapidly evolving. Although NACT demonstrates efficacy, its toxicity limits utility, whereas neoadjuvant endocrine therapy (ET) offers superior tolerability but inferior efficacy. Currently, in most Chinese centers, NACT remains the primary neoadjuvant approach for HR+/HER2- patients, with ET playing a supplementary role. However, the introduction of CDK4/6 inhibitors has transformed clinical practice, as their breakthrough efficacy in adjuvant therapy has brought revolutionary changes to disease management. The CORALLEEN trial demonstrated that preoperative CDK4/6 inhibitor combined with an aromatase inhibitor yields comparable efficacy to NACT. The WSG-ADAPT series of studies is also exploring whether CDK4/6 inhibitors can replace chemotherapy in intermediate-to-high risk HR+/HER2- early breast cancer. Nevertheless, whether patients can omit NACT remains controversial. This study aims to compare the objective response rate (ORR) between ET + CDK4/6 inhibitor alone and chemotherapy followed by ET + CDK4/6 inhibitor, while comprehensively evaluating treatment tolerability and adverse event profiles, to explore the feasibility of chemotherapy-sparing precision strategies. Methods: A total of 70 patients with HR+/HER2- breast cancer were enrolled across 11 tertiary hospitals. Of these, 45 received ET + CDK4/6 inhibitor (ribociclib [n=21], abemaciclib [n=16], dalpiciclib [n=7], palbociclib [n=1]). The remaining 25 patients underwent chemotherapy followed by ET + CDK4/6 inhibitor. ORR was defined as complete/partial radiologic response according to RECIST 1.1 criteria. For statistical analysis, the Mann-Whitney U test was used to analyze ranked data. The χ2 test or Fisher’s exact test was used to analyze categorical data. Logistic regression analysis was performed to identify significant factors for treatment response. All tests were two sided with significance level set at 0.05. All data cleaning and analysis were conducted using R statistical software (Version 4.5.0). Results: Baseline Characteristics and ORR Analysis by Treatment Groups: Median age of ET + CDK4/6 inhibitor vs chemotherapy-sequenced groups was 60 vs 55 years (P=0.029); Ki-67 was 20% vs 30% (P=0.023); other baseline characteristics (menopausal status (P =0.253), T stage (P =0.444), and N stage (P=0.721)) were balanced between treatment groups. The overall ORR was 63%, with higher responses observed in patients with Ki-67≤20% (68%) compared to those with Ki-67>20% (58%). The ET+CDK4/6i group achieved a 69% ORR (72% for Ki-67≤20% vs 63% for Ki-67>20%). No significant ORR difference between ET + CDK4/6 inhibitor (69%) and chemotherapy-sequenced groups (52%) (OR: 0.49, 95% CI: 0.18-1.34; P=0.164). The ribociclib-treated cohort achieved an ORR of 76% across all treatment strategies. Baseline Characteristics and ORR Analysis of Abemaciclib vs Ribociclib in non-Chemotherapy Patients: Median age of abemaciclib vs ribociclib groups was 61 vs 58 years (P=0.679); other baseline characteristics (menopausal status (P =1.000), T stage (P =0.641), N stage (P =0.191), or Ki-67 (P =0.709)) were balanced. Among non-chemotherapy patients, abemaciclib demonstrated comparable efficacy to ribociclib (ORR 62.5% vs 76.2%, OR: 1.82, 95% CI: 0.46-8.36; P=0.370).Conclusions: Primary ET + CDK4/6 inhibitor demonstrated comparable efficacy to chemotherapy-sequenced therapy in HR+/HER2- breast cancer, particularly in patients with low Ki-67. These results support the feasibility of chemotherapy-free neoadjuvant strategies for select HR+/HER2− patients, potentially minimizing chemotherapy-related toxicity without compromising treatment efficacy.