Poster Session 5
Session Details
Presentation numberPS5-01-01
Concordance of genomic alterations between paired tissue (T) and plasma (P) samples analyzed with next generation sequencing (NGS) panels from patients (pts) with HER2-positive metastatic breast cancer (mBC)
Santiago Escrivá-de-Romaní, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
S. Escrivá-de-Romaní1, L. Ollé-Monràs2, L. Joval-Ramentol3, E. Álvarez Colomé4, M. Arumí1, A. Rezqallah1, A. Suñol4, J. Jimenez5, E. Castillo6, R. Fasani5, P. Martinez5, A. Baizán4, M. Oliveira1, M. Bellet1, E. Zamora1, I. Pimentel1, M. Borrell1, N. Gómez1, A. Rodríguez1, M. Perachino7, V. Barberi7, M. Gaudio7, C. Salvà de Torres8, E. Cimbro4, R. Dienstmann3, J. Seoane2, P. Nuciforo5, A. Vivancos6, C. Saura1; 1Medical Oncology, Breast Cancer Group, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 2Computational Cancer Biology Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 3Oncology Data Science Group (ODysSey), Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 4Medical Oncology, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 5Molecular Oncology Group, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 6Cancer Genomics Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 7Medical Oncology, Breast Cancer Group, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 8Medical Oncology, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN.
BACKGROUND: Analyses of P circulating tumor DNA by NGS panels is an emerging useful tool for identifying tumor genomic alterations in BC. There is scarce data about the concordance of genomic alterations found at the same time point in T and P samples of pts with HER2-positive mBC. MATERIALS AND METHODS: This is a single institution study of a consecutive cohort of HER2-positive mBC pts receiving anti-HER2 therapies. Tumor T and P samples were collected prospectively after obtaining consent. Genomic alterations were analyzed retrospectively using NGS with the VHIO300 panel for T (includes 421 genes) and the VHIO360 panel for P samples (includes 74 genes). HER2 expression in T samples was evaluated using Immunohistochemistry (IHC) and in situ hybridization (ISH) following the 2013 ASCO/CAP guidelines. To test the concordance of paired T/P samples between the NGS panels, only the 73 common genes were included. General linear models, Pearson and Spearman correlations were used to associate numerical variables. RESULTS: 107 consecutive pts were included in the study, of which 39 pts had synchronous paired T/P samples. Out of these 39 pts, a total of 42 T/P pairs were available. T samples were analyzed with VHIO300 panel and IHC/ISH, while P samples were tested with VHIO360. Concordance in the presence or absence of ERBB2 amplification status between P and T was coincident in 30 out of 42 pairs (71%) showing a statistically significant correlation with copy number. As a reference, in T samples alone, the concordance between ERBB2 amplification detected by two different methodologies, NGS and by IHC/ISH, resulted positive in 35 out of 42 samples (83%). T and P shared at least one pathogenic or likely pathogenic mutation in 32 out of 42 pairs (76%) where these variants were present. The overall agreement between genotype status (mutant/ wild-type) for all genes was 78.5%. The number of detected mutations per sample varied from 0 – 6. For variants of uncertain significance (VUS), 8 of 38 pairs (21%) shared altered variants in both samples. The average percentage of altered shared genes was 35%. The range of VUS variants detected per sample varied from 0 – 14. For amplifications 26 pairs (65%) out of 40 shared at least one altered gene in both samples. The average percentage of concordant T/P pairs with regards to amplified genes was 60.8%. The range of copies detected per sample varied from 0 – 9. Concordance of genomic alterations found in paired T/P samples is summarized in the table. CONCLUSIONS: A high concordance rate for ERBB2 amplification was detected by NGS in T and P paired samples, as well as in T samples using different approaches, NGS and IHC/ISH. Concordance was high between T and P with regards to pathogenic and likely pathogenic mutations, moderate for amplifications other than ERBB2, and overall low for VUS.
| Genomic alteration | Pathogenic and likely pathogenic mutations | VUS | Amplifications | |||
| Genes | Samples (%) | Genes | Samples (%) | Genes | Samples (%) | |
| Shared | TP53 | 59,5 | APC | 10,5 | ERBB2 | 57,5 |
| PIK3CA | 38 | PIK3CA, CDK12, RB1 | 5,2 | CCND1 | 12,5 | |
| GATA3 | 9,5 | ARID1A, CDK6, EGFR, NTRK1 | 2,6 | CDK6, MYC, PIK3CA | 2,5 | |
| ARID1A | 7,1 | |||||
| PTEN | 4,8 | |||||
| BRCA2, ESR1, RHOA, SMAD4 | 2,4 | |||||
| Present in T (not in P) | TP53 (1 double mutation) | 19 | ERBB2 | 23,7 | ERBB2, MYC | 20 |
| BRCA2, CDH1, CDKN2A | 4,8 | EGFR | 15,8 | FGFR1 | 17,5 | |
| APC | 2,4 | BRCA2, CDK12, KIT, PDGFRA | 13,1 | CCNE1 | 7,5 | |
| NF1 (double mutation) | 2,4 | FGFR2, MYC, PTPN11, RAF1, RB1 | 7,9 | AR, CCND1, PIK3CA, RET | 5 | |
| Present in P (not in T) | TP53 | 11,9 | NF1 | 15,8 | CDK6, PIK3CA | 15 |
| ERBB2, ESR1, KRAS | 7,1 | ATM | 13,1 | BRAF, CCND1, CDK4 | 12,5 | |
| ARID1A, CDKN2A, PIK3CA | 4,8 | APC, CDK12, RAF1 | 10,5 | ERBB2 | 10 | |
| APC, BRCA1, PTEN | 2,4 | MTOR, NTRK1, PDGFRA | 7,9 | KRAS | 7,5 |
Presentation numberPS5-01-02
Pyrotinib or placebo in combination with trastuzumab and docetaxel for untreated HER2-positive metastatic breast cancer: long-term survival results from the phase 3 PHILA study
Binghe Xu, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
B. Xu1, F. Ma1, M. Yan2, W. Li3, Q. Ouyang4, Z. Tong5, Y. Teng6, Y. Wang7, S. Wang8, C. Geng9, T. Luo10, J. Zhong11, Q. Zhang12, Q. Liu13, X. Zeng14, T. Sun15, Q. Mo16, S. Zhou17, Y. Cheng18, J. Cheng19, X. Wang20, J. Nie21, J. Yang22, X. Wu23, X. Wang24, H. Li25, G. Yao26, Y. Fan27, J. Lin27, X. Zhu27; 1Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 2Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, CHINA, 3Department of Oncology, The First Hospital of Jilin University, Changchun, CHINA, 4Breast Internal Medicine Department, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, CHINA, 5Department of Breast Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA, 6Department of Breast Internal Medicine, The First Hospital of China Medical University, Shenyang, CHINA, 7Breast Surgery, Shandong Cancer Hospital & Institute, Jinan, CHINA, 8Department of Internal Medicine, Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 9Breast Center, The Fourth Hospital of Hebei Medical University and Hebei Tumor Hospital, Shijiazhuang, CHINA, 10Department of Medical Oncology of Cancer Center, West China Hospital, Sichuan University, Chengdu, CHINA, 11Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, CHINA, 12Ward One of Mammary Department, Harbin Medical University Cancer Hospital, Harbin, CHINA, 13Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA, 14Breast Cancer Center, Affiliated Cancer Hospital of Chongqing University, Chongqing, CHINA, 15Breast Internal Medicine Department, Liaoning Cancer Hospital & Institute, Shenyang, CHINA, 16Breast Surgery, Guangxi Medical University Affiliated Tumor Hospital, Nanning, CHINA, 17Department of Breast Oncology, The First Affiliated Hospital of USTC West District, Hefei, CHINA, 18Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, CHINA, 19Oncology Center Breast Department, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, CHINA, 20Breast Medicine, Zhejiang Cancer Hospital, Hangzhou, CHINA, 21Breast Surgery, Yunnan Cancer Hospital, Kunming, CHINA, 22Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, CHINA, 23Department of Breast Oncology, Hubei Cancer Hospital, Wuhan, CHINA, 24Department of Medical Oncology, Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, CHINA, 25Key Laboratory of Carcinogenesis and Translational Research, Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, CHINA, 26Department of Breast, Southern Medical University Nanfang Hospital, Guangzhou, CHINA, 27Department of Clinical Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, CHINA.
Background: PHILA, a randomized, double-blind, placebo-controlled phase 3 trial (NCT03863223), assessed the efficacy and safety of pyrotinib plus trastuzumab and docetaxel (PyroHT) as first-line therapy in HER2-positive metastatic breast cancer. PyroHT significantly prolonged progression-free survival (PFS) and improved overall survival (OS) compared with placebo plus trastuzumab and docetaxel (HT), with manageable safety (Xu et al., BMJ 2023; Xu et al., SABCS 2024). Here, we report updated survival results after a median follow-up of 45.5 months.Methods: Patients (pts) with untreated HER2-positive metastatic breast cancer were enrolled across 40 centers in China and randomized (1:1) to receive either oral pyrotinib (400 mg once daily) or placebo plus intravenous trastuzumab (8 mg/kg loading dose, then 6 mg/kg) and docetaxel (75 mg/m2) on day 1 of each 21-day cycle. The primary endpoint was investigator-assessed PFS.Results: From May 2019 to January 2022, 590 pts were randomized and received treatment (PyroHT n=297; HT n=293). As of May 30, 2025, the median follow-up was 46.5 months (IQR 36-55) for PyroHT and 44.6 months (IQR 31-53) for HT. In the PyroHT group, 75 pts (25.3%) were still receiving study treatment, compared with 19 (6.5%) in the HT group. Following treatment discontinuation, 174 pts (58.6%) in the PyroHT group and 229 pts (78.2%) in the HT group received post-discontinuation therapies. Although cross-over was not allowed, the proportions of pts receiving pyrotinib, trastuzumab, pertuzumab, or anti-HER2 antibody-drug conjugate (incl. T-DM1, T-DXd, SHR-A1811) after study treatment discontinuation in each group were 18.5% vs. 51.9%, 26.3% vs. 25.6%, 15.5% vs. 14.0%, and 22.6% vs. 28.0%, respectively. At data cutoff, 85 deaths (28.6%) occurred in the PyroHT group and 108 (36.9%) in the HT group. OS was superior in the PyroHT group compared to the HT group (HR 0.74, 95% CI 0.56-0.98; nominal 1-sided P=0.0188). The median OS was not reached in either group. OS rates were 79.6% in the PyroHT group and 72.1% in the HT group at 3 years, 72.9% and 63.5% at 4 years, 65.7% and 58.5% at 5 years, respectively. As reported previously, consistent and prolonged benefit of investigator-assessed PFS in the PyroHT group was observed (HR 0.44, 95% CI 0.36-0.54; nominal 1-sided P<0.0001). The PFS rates were 40.4% in the PyroHT group and 9.7% in the HT group at 3 years, 31.9% and 7.6% at 4 years, 29.2% and 4.3% at 5 years, respectively. Additionally, the benefits in PFS with PyroHT were consistent across nearly all analyzed subgroups. For pts with prior (neo)adjuvant trastuzumab, median PFS was 62.8 months (95% CI 19.2-not evaluable) in the PyroHT group (n=46) vs 10.4 months (95% CI 6.4-13.2) in the HT group (n=42). Among pts who developed brain metastases, the median time to onset was 16.6 months (IQR 13-28) with PyroHT (n=34) compared to 9.1 months (IQR 8-14) with HT (n=30). Safety data cutoff was April 30, 2024. The incidence of all adverse events decreased considerably after docetaxel discontinuation. In the PyroHT group, the incidence of any grade diarrhea decreased from 98.4% (245/249) to 79.9% (199/249), and grade 3 diarrhea incidence declined from 46.2% (115/249) to 16.1% (40/249). No grade 4 or 5 diarrhea was reported.Conclusions: The long-term analysis confirms that the previously observed improvements in OS and PFS with PyroHT compared to HT in the first-line treatment of HER2-positive metastatic breast cancer were maintained after 45.5 months of median follow-up. Notably, with PyroHT, 5-year OS rate remained at 66% and nearly 30% of pts remained progression-free at 5 years. PyroHT treatment delayed time to onset of brain metastasis. It reinforces dual anti-HER2 regimen (pyrotinib plus trastuzumab) as an established therapeutic strategy for this patient population.
Presentation numberPS5-01-03
Jbcrg-m06/emerald post-hoc analysis by physician’s choice of docetaxel or paclitaxel: efficacy and safety of eribulin mesylate vs taxanes combined with trastuzumab and pertuzumab as first-line for her2-positive locally advanced or metastatic breast cancer
Norikazu Masuda, Graduate School of Medicine, Kyoto University, Kyoto, Japan
N. Masuda1, S. Saji2, Y. Kojima3, M. Kitada4, H. Tada5, T. Iwasa6, K. Koizumi7, H. Shigematsu8, Y. Sagara9, T. Okuno10, Y. Hasegawa11, E. Tokunaga12, T. Shien13, U. Toh14, T. Kuwayama15, M. Shimoda16, H. Hasegawa17, S. Morita18, T. Yamashita19; 1Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, JAPAN, 2Department of Medical Oncology, Fukushima Medical University, Fukushima, JAPAN, 3Division of Breast Surgical Oncology, Department of Surgery, Showa Medical University Hospital, Tokyo, JAPAN, 4Department of Breast Disease Center, Asahikawa Medical University Hospital, Hokkaido, JAPAN, 5Division of Breast and Endocrine Surgery, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, JAPAN, 6Department of Medical Oncology, Kindai University Hospital, Osaka, JAPAN, 7Department of Breast Surgery, Hamamatsu University Hospital, Shizuoka, JAPAN, 8Department of Breast Surgery, Hiroshima University Hospital, Hiroshima, JAPAN, 9Department of Breast and Thyroid Surgical Oncology, Social Medical Corporation Hakuaikai, Sagara Hospital, Kagoshima, JAPAN, 10Department of Breast Surgery, Kobe City Nishi-Kobe Medical Center, Hyogo, JAPAN, 11Department of Breast Surgery, Hachinohe City Hospital, Aomori, JAPAN, 12Department of Breast Oncology, NHO Kyushu Cancer Center, Fukuoka, JAPAN, 13Department of Breast and Endocrine Surgery, Okayama University Hospital, Okayama, JAPAN, 14Department of Breast Surgery, Kurume University Hospital, Fukuoka, JAPAN, 15Department of Breast Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, JAPAN, 16Department of Breast and Endocrine Surgery, Hyogo Medical University Hospital, Hyogo, JAPAN, 17Medical HQs, Eisai Co., Ltd., Tokyo, JAPAN, 18Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, JAPAN, 19Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Kanagawa, JAPAN.
Background The randomized open-label phase 3 JBCRG-M06/EMERALD study (NCT03264547) was the first to show the non-inferiority of eribulin (E) to taxane (T) (docetaxel [DTX] or paclitaxel [PTX]) when combined with dual HER2 blockade as first-line chemotherapy for HER2+ locally advanced or metastatic breast cancer (LABC/mBC) (doi: 10.1200/JCO-24-01888). In the primary analysis results, median progression-free survival (PFS) was 14.0 and 12.9 months (mo) in the E and T groups, respectively (hazard ratio, 0.95). Although median treatment duration was longer with E (28.1 weeks) than with DTX or PTX (18.1 and 20.5 weeks, respectively), incidences of adverse events (AEs) were similar between the E and T groups. While DTX and PTX have some pharmacological similarities, their distinct properties may result in differences in therapeutic outcomes. We report the results of a post-hoc analysis in which outcomes were analyzed by type of T used and compared with those when E was used. Methods Patients (pts) with HER2+ LABC/mBC were randomized to receive E (1.4 mg/m2 on days 1 and 8) or T (DTX 75 mg/m2 on day 1 or PTX 80 mg/m2 on days 1, 8, and 15), intravenously in a 21-day cycle, each with trastuzumab + pertuzumab. In the T group, the choice between DTX and PTX was determined by the attending physician, but it had to be declared in advance at the time of enrolment. The Kaplan-Meier method was used to assess survival outcomes, and the log-rank test for intergroup comparisons. Waterfall and swimmer plots were used to assess outcomes based on type of T used. Results Of the total 446 pts, 224 received E and 222 received T (DTX, n = 186; PTX, n = 36) (intention-to-treat population); 224 in the E group and 218 in the T group (DTX, n = 184; PTX, n = 34) received at least one dose of the study drug (safety analysis population). Baseline characteristics were well balanced between the groups. In the T group, pts treated with DTX or PTX had no differences in age or ECOG performance status. Survival outcomes were similar between pts who received DTX or PTX (Table 1A). Incidences of AEs were generally similar between the E and T groups, regardless of type of T used; however, a higher incidence of peripheral sensory neuropathy was noted with PTX whereas it was lower with DTX, compared with E. Additionally, the incidence of neutropenia tended to be lower with DTX (Table 1B). Conclusions In our trial comparing T and E combined with dual HER2 blockade as first-line treatment for HER2+ LABC/mBC, DTX and PTX were used to treat 84% and 16% of pts, respectively. No differences in efficacy (i.e. non-inferiority results) were observed based on type of T used. While AEs were generally similar between the E and T groups, regardless of type of T used, a higher incidence of peripheral sensory neuropathy was noted with PTX compared with E or DTX.
| E vs DTX | E vs PTX | |||
| A. Survival outcomes, median (95% confidence interval), mo | ||||
| E (n = 184) | DTX (n = 186) | E (n = 40) | PTX (n = 36) | |
| PFS |
14.0
(11.2–16.2)
|
13.1
(11.2–15.8)
|
14.1
(10.6–20.3)
|
10.8
(9.0–17.5)
|
| Overall survival (OS) |
Not reached (NR)
(64.3–NR)
|
NR |
75.5
(45.9–NR)
|
NR
(45.9–NR)
|
| B. Drug-related AEs (all grades), incidence, % | ||||
| E (n = 184) | DTX (n = 184) | E (n = 40) | PTX (n = 34) | |
| All | 94.6 | 95.7 | 87.5 | 100.0 |
| Infusion reaction | 14.1 | 22.3 | 10.0 | 38.2 |
| Neutropenia | 62.0 | 28.8 | 60.0 | 41.2 |
| Peripheral sensory neuropathy | 63.0 | 47.3 | 52.5 | 82.4 |
| Peripheral motor neuropathy | 5.4 | 2.7 | 5.0 | 8.8 |
| Edema | 9.8 | 45.1 | 2.5 | 26.5 |
| Diarrhea | 37.5 | 53.3 | 32.5 | 58.8 |
| Febrile neutropenia | 5.4 | 10.3 | 2.5 | 0.0 |
| Cardiac events | 7.6 | 6.0 | 5.0 | 5.9 |
| Skin-related events (including nail disorders) | 42.9 | 63.0 | 30.0 | 58.8 |
Presentation numberPS5-01-04
Clinical predictors for first-line treatment duration in HER2-positive metastatic breast cancer Results from the AGMT_MBC-Registry
Gabriel Rinnerthaler, Medical University Graz, Graz, Austria
G. Rinnerthaler1, S. P. Gampenrieder2, A. Pichler3, W. Herz4, R. Pusch5, C. Dormann5, C. Suppan1, M. Sandholzer6, T. Winder7, S. Heibl8, L. Scagnetti9, C. A. Schmitt10, A. F. Zabernigg11, D. Egle12, C. Hager13, P. Pichler14, F. Roitner15, J. Andel16, K. Strasser-Weippl17, R. Bartsch18, V. Castagnaviz2, M. Hubalek19, M. Knauer20, C. F. Singer21, R. Greil22; 1Division of Oncology, Department for Internal Medicine, Medical University Graz, Graz, AUSTRIA, 2Department of Internal Medicine III, Paracelsus Medical University Salzburg, Salzburg, AUSTRIA, 3Internal Medicine – Department for Haemato-Oncology, LKH Hochsteiermark, Leoben, AUSTRIA, 4Department of Surgery, Breast Health Center, LKH Hochsteiermark, Leoben, AUSTRIA, 5Internal Medicine I, Ordensklinikum Linz Barmherzige Schwestern – Elisabethinen, Linz, AUSTRIA, 6Department of Internal Medicine II, Academic Teaching Hospital Feldkirch, Feldkirch, AUSTRIA, 7Department of Internal Medicine III, Academic Teaching Hospital Feldkirch, Feldkirch, AUSTRIA, 8Department of Internal Medicine I, Klinikum Wels-Grieskirchen GmbH, Wels, AUSTRIA, 9Department of Internal Medicine IV, Klinikum Wels-Grieskirchen GmbH, Wels, AUSTRIA, 10Department for haematology and internal oncology, Kepler University Hospital Linz, Linz, AUSTRIA, 11Department of Internal Medicine, County Hospital Kufstein, Kufstein, AUSTRIA, 12Department of Gynaecology, Medical University Innsbruck, Innsbruck, AUSTRIA, 13Breast Center, Breast Center Dornbirn, Dornbirn, AUSTRIA, 14Department for Internal Medicine 1, University Hospital St.Pölten, St. Pölten, AUSTRIA, 15Department of Internal Medicine II, Hospital Braunau, Braunau am Inn, AUSTRIA, 16Department of Internal Medicine II, Pyhrn-Eisenwurzen Klinikum Steyr, Steyr, AUSTRIA, 17Department of Medicine I, Clinic Ottakring, Vienna, AUSTRIA, 18Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, AUSTRIA, 19Breast Health Center Schwaz, Department of Gynecology, Schwaz, AUSTRIA, 20Breast Center, Tumor and Breast Center Eastern Switzerland, St. Gallen, SWITZERLAND, 21Department of Obstetrics and Gynecology and Comprehensive Cancer Center, Medical University of Vienna, Vienna, AUSTRIA, 22Austrian Group Medical Tumor Therapy, AGMT gemeinnützige GmbH, Vienna, AUSTRIA.
Background: Based upon the CLEOPATRA trial, a combination of docetaxel plus trastuzumab and pertuzumab (THP) has been the standard treatment for patients with metastatic HER2-positive breast cancer. However, this standard has recently been challenged by the results of the DESTINY-Breast09 study, which compared trastuzumab-deruxtecan (T-DXd) plus pertuzumab with THP. In this analysis, we aimed to develop a risk model based on known parameters that identifies patients with a long first-line (1L) therapy duration, as these patients may not need 1L T-DXd therapy. Patients and Methods: The AGMT_MBC-Registry is a nationwide, multicenter, ongoing retrospective and prospective registry for metastatic breast cancer (MBC) patients in Austria. Patients with HER2-positive breast cancer treated with a 1L chemo, trastuzumab plus pertuzumab combination were included in this analysis. Time to next treatment (TTNT) was defined as the interval from initiation of 1L to the initiation of the next subsequent treatment line. To identify prognostic factors associated 1L TTNT a multivariable Cox proportional hazards model was performed. Clinical, demographic, and tumor-related covariates – including metastatic sites, age, hormone-receptor (HR) status, and tumor grade – were included in the analysis. Variable selection was performed using a bidirectional stepwise approach based on the Akaike Information Criterion. Patients with missing values were excluded. A risk score was derived from the linear predictor of the Cox model. Survival-optimized cut-off points were identified using maximally selected rank statistics to stratify patients into low-, intermediate-, and high-risk groups. Hazard ratios (HRs) with 95% confidence intervals (CIs) are reported. Time-to-event analyses were performed using Kaplan-Meier estimates, with group-specific survival probabilities reported at 12, 18, and 36 months. Results: As of March 11, 2025, the registry included 3,094 patients, and 218 patients with HER2-positive MBC were available for this analysis. Median age at diagnosis of MBC was 58 years (range 26-85) and 62% had hormone receptor (HR) positive disease. Bone (57%), liver (43%), and lung metastases (35%) were the most common metastatic sites; brain, pleural, and peritoneal metastases were rare (<5%). At diagnosis, 63% had de novo MBC, 31% had recurrence ≥24 months, and 6% <24 months after surgery. The final multivariable Cox model identified the presence of brain (HR 2.22, p = 0.038), pleural metastases (HR 3.41, p = 0.001), disease-free survival < 24 months (HR 1.98, p=0.046), and older age (HR 1,01 per year, p = 0.043) as significant predictors for a shorter TTNT. Lung metastasis (HR 1.36, p = 0.089) showed a trend toward shorter treatment duration. Based on the risk score, patients were stratified into low- (27%, n = 56), intermediate- (62%, n = 127), and high-risk groups (10%, n = 21). Compared to the low-risk group, TTNT was shorter in the intermediate-risk group (HR 1.79, 95% CI 1.17-2.74; p = 0.007) and in the high-risk group (HR 5.39, 95% CI 2.95-9.86; p < 0.001). In the high-risk group, treatment continuation rates were 45% (95% CI 28-73%) at 12 months, 17% at 18 months, and 6% at 36 months, respectively. In the intermediate-risk group, corresponding rates were 74% (95% CI 67-82%), 63%, and 36%; and in the low-risk group, 84% (95% CI 74-94%), 65%, and 50%, respectively. Conclusion: This real-world analysis identified distinct clinical subgroups of patients with HER2-positive MBC who are at low risk for early 1L treatment discontinuation under the current standard CLEOPATRA regimen. The proposed risk model enables prognostic stratification and may help identify patients who do or do not need an alternative 1L approach, such as T-DXd-based combinations. External validation of the score in independent cohorts is warranted.
Presentation numberPS5-01-05
Unraveling KAdcyla (Trastuzumab emtansine; T-DM1) resistance in HER2+ advanced breast cancer geicam/2017-04-KATIA study: biomarker analyses
Jose Ángel García-Sáenz, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC); CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, Spain
J. Albanell1, J. García-Sáenz2, B. Bermejo3, A. Stradella4, Y. Izarzugaza5, A. Barnadas6, E. Alba7, J. Alonso-Romero8, S. López-Tarruella9, F. Henao-Carrasco10, S. Zazo11, F. Moreno12, M. Martínez-Martínez13, A. Vethencourt4, H. Callata-Carhuapoma5, J. Herranz14, R. Caballero15, M. Portela15, F. Rojo16; 1Medical Oncology, Hospital del Mar-Hospital del Mar Research Institute ; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 2Medical Oncology, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC); CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 3Medical Oncology, Hospital Clínico Universitario de Valencia;Biomedical Research Institute INCLIVA; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 4Medical Oncology, Breast Unit, Catalan Institute of Oncology-Hospital Universitari de Bellvitge-Bellvitge Research Institute (IDIBELL); GEICAM Spanish Breast Cancer Group, Hospitalet de Llobregat, SPAIN, 5Medical Oncology, Hospital Universitario Fundación Jiménez Díaz; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 6Medical Oncology, Hospital de la Santa Creu i Sant Pau; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 7Medical Oncology, Hospital Universitario Virgen de la Victoria; GEICAM Spanish Breast Cancer Group, Málaga, SPAIN, 8Medical Oncology, Hospital Clínico Universitario Virgen de la Arrixaca, (IMIB-Arrixaca), Universidad de Murcia; GEICAM Spanish Breast Cancer Group, El Palmar, SPAIN, 9Medical Oncology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 10Medical Oncology, Hospital Universitario Virgen Macarena; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 11Medical Oncology, Hospital Universitario Fundacion Jimenez Diaz; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 12Medical Oncology, ospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC); GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 13Medical Oncology, Hospital Clínico Universitario de Valencia. Biomedical Research Institute INCLIVA; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 14Statics department, GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 15Translational, GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 16Pathology, Hospital Universitario Fundacion Jimenez Diaz; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: T-DM1 remains a relevant therapeutic option for patients with HER2-positive metastatic breast cancer (HER2+ MBC), particularly for those who have previously received trastuzumab and taxane-based therapies. However, despite its clinical utility, the mechanisms underlying both primary and acquired resistance to T-DM1 remain largely uncharacterized, limiting the ability to optimize treatment strategies and identify patients most likely to benefit from this therapy. The aim was to characterize the mechanisms of resistance to T-DM1 in the KATIA study.Methods: The KATIA study (NCT03829306) was a prospective, multicenter study in progressive/recurrent HER2+ MBC patients treated with second-line T-DM1. Tumor and plasma samples were collected from 32 (100%) patients. Genomic profiling was performed on pre-treatment baseline (BL) metastatic tumors (n=18, 56%), and on liquid biopsies at BL (n=20, 62,5%) and at disease progression (n=12, 37,5%) using the FoundationOne CDx and FoundationOne liquid CDx assays (324-gene NGS panels; Foundation Medicine). Additionally, we analyzed a 77-gene NGS panel on circulating tumor DNA (AVENIO ctDNA Expanded Kit V2, Roche, n=31, 97%). HER2, Ki67, Estrogen and Progesterone Receptors (ER and PR) status and the tumor immune microenvironment markers (PD-L1, PD1, CD3, CD8, tumor-infiltrating lymphocytes) were analyzed on BL tumors (n=26, 81%) by immunohistochemistry (IHC). The Kaplan-Meier method estimated median progression free survival (PFS) and overall survival (OS). Hazard ratios (HR) with 95% confidence intervals (CIs) were calculated using Cox proportional hazards regression models and the p-values adjusted for multiple comparisons by the Bonferroni method.Results: Tumor BL mutations in CCDN1, FGF4 or FGF19 predicted better mPFS (CCDN1: 8.61 vs. 3.65 m., HR 0.19; 95% CI 0.04-0.92, p=0.039; FGF4: 8.77 vs. 3.84 m., HR 0.1; 95% CI 0.01-0.79, p=0.0298; FGF19: 8.61 vs. 3.65m., HR 0.19; 95% CI 0.04-0.92, p=0.039). On the contrary, GNAS mutations showed a trend towards worse mOS (6.67 vs. 17.45 m., HR 4.63; 95% CI 0.42-51.09, p=1). Tumor burden ctDNA analyses revealed that the presence of ≥3 BL mutations showed a trend towards worse mOS (14.23 vs. 22.87 m., HR 2.85; 95% CI 0.91-10.01, p=0.1032). BL ctDNA mutations in PIK3CA/TP53 (the most frequent mutated genes) also showed a trend towards worse mOS (14.55 vs. 22.87 m., HR 4.78; 95% CI 0.56-40.89, p=1), as well as mutations detected in the following signaling pathways: receptor tyrosine kinases (RTKs: 14.55 vs. 22.87 m., HR 3.15; 95% CI 0.62-16, p=1) and DNA damage response/TP53 (DDR/TP53: 14.55 vs. 22.87 m., HR 4.98; 95% CI 0.59-42.32, p=1). Similar results were observed for mutations detected in these pathways at the time of disease progression. Conclusions: Baseline tumor mutations in CCDN1, FGF4 or FGF19 genes were significantly associated with longer PFS in HER2+ MBC patients treated with second line T-DM1, suggesting their potential role as predictive biomarkers of treatment benefit.
Presentation numberPS5-01-06
Contribution of Sociodemographic Characteristics to Treatment Delays in Patients with de novo Metastatic HER2-Positive Breast Cancer
Apoorva Ravichandran, University of Chicago Medicine, Chicago, IL
A. Ravichandran1, M. Hennessy2, J. Q. Freeman3, S. Poland2, W. Guo2, R. Nanda2; 1Internal Medicine Residency Program, University of Chicago Medicine, Chicago, IL, 2Section of Hematology & Oncology, Department of Medicine, University of Chicago, Chicago, IL, 3Department of Public Health Sciences, University of Chicago, Chicago, IL.
Background: Outcomes for patients with de novo metastatic human epidermal growth factor receptor 2 (HER2)-positive breast cancer have dramatically improved due to advances in HER2-directed therapy, yet significant disparities persist, with Black and Hispanic patients presenting with more advanced stage disease and experiencing worse survival outcomes compared to White patients. Studies suggest racial and ethnic minorities experience longer delays in treatment initiation, however large, comprehensive analyses of the impact of sociodemographic and clinical factors at the national level in the U.S. remain limited. Methods: We retrospectively identified patients diagnosed with de novo metastatic HER2-positive breast cancer between 2010 and 2022 using the National Cancer Database (NCDB). Clinical and sociodemographic characteristics were collected and compared across racial and ethnic groups. We categorized systemic therapy as chemotherapy alone or chemotherapy plus HER2-directed therapy. Treatment receipt and time to treatment initiation were evaluated by race/ethnicity. Multivariable linear regression was performed to examine sociodemographic differences in time to initiation of systemic and localized (i.e., radiotherapy) therapies, respectively. Beta coefficients reflected adjusted differences in treatment timing, with 95% confidence intervals (95% CI) reported. Results: A total of 27,972 patients with de novo metastatic HER2-positive breast cancer were included. Mean age was 58.8 years; 69.0% were White, 17.5% Black, 7.8% Hispanic, and 4.6% Asian/Pacific Islander (per self-report). At initial presentation, 9.5% had brain metastases, and 58.2% had visceral metastases. At the time of diagnosis, Black and Hispanic patients were younger, more often uninsured or Medicaid-insured, and more likely to reside in lower-education, lower-income areas (p<0.001). While overall 88.5% of patients received a combination of chemotherapy and HER2-directed therapy, significant delays in treatment initiation were observed across all treatment modalities in minority groups. For systemic therapy, mean initiation times for Asian/Pacific Islander, White, Black, and Hispanic patients were 46.2 days, 47.2 days, 54.6 days, and 57.2 days, respectively (p<0.001). Time to systemic therapy was significantly reduced in patients with visceral metastases, while brain metastases were associated with earlier radiation initiation (median 28 days to initiation in patients with brain metastases vs. 116 days in patients without brain metastases, p<0.001). Adjusted analyses revealed that on average, Hispanic ethnicity was associated with a 7.3 day (95% CI: 3.2-11.4) delay in starting systemic therapy and 23.0 day (95% CI: 9.0-36.9) delay for radiotherapy initiation. Compared to White patients, Black patients had a 5.7 day (95% CI: 2.9-8.6) delay in starting systemic therapy and Asian/Pacific Islander patients had a 21.0 day (95% CI: 4.9-37.2) delay in starting radiotherapy. Lower education levels, lack of insurance, or having Medicaid/Medicare were also independent predictors of delayed initiation of systemic therapy, but interestingly not radiotherapy. Conclusion: In this U.S. national sample of patients with de novo metastatic HER2-positive breast cancer, substantial disparities persist in timely treatment initiation, particularly among Black and Hispanic patients, even after controlling for sociodemographic and clinical factors. Delays in this setting may adversely affect survival outcomes, highlighting the urgency of improving equitable access to care. Efforts must focus on enhancing health literacy, expanding insurance coverage, and improving care coordination infrastructure in disadvantaged communities.
Presentation numberPS5-01-08
Development of the HER2DX Predictor of Brain Metastasis in Advanced HER2-Positive Breast Cancer Treated with First-Line THP
Rodrigo Sanchez-Bayona, Hospital Universitario 12 de Octubre. Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
R. Sanchez-Bayona1, S. Cobo2, M. Kubeczko3, J. Soberino4, M. Rey5, B. Pycinski6, E. Chmielik7, J. Montes1, F. Pardo8, V. Sirenko5, P. Galván9, A. Lesniak3, M. Oczko-Wojciechowska10, I. Monge11, M. Gonzalez12, I. Garcia-Fructuoso12, F. Schettini12, G. Villacampa13, T. Pascual12, L. Paré8, F. Brasó-Maristany9, E. Ciruelos1, A. Prat14, M. Jarzab3; 1Medical Oncology, Hospital Universitario 12 de Octubre. Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, SPAIN, 2Bioestatistics, Fundació de Recerca Clínic Barcelona – IDIBAPS, Barcelona, SPAIN, 3Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 4Medical Oncology, IOB- QuirónSalud, Barcelona, SPAIN, 5Medical Oncology, Fundació de Recerca Clínic Barcelona – IDIBAPS, Barcelona, SPAIN, 6Medical Oncology, Silesian University of Technology, Faculty of Biomedical Engineering, Zabrze, POLAND, 7Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 8Diagnostic, REVEAL Genomics, Barcelona, SPAIN, 9Translational Genomics and Targeted Therapies in Solid Tumors Lab, Fundació de Recerca Clínic Barcelona – IDIBAPS, Barcelona, SPAIN, 10Department of Clinical and Molecular Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 11Department of Hospital Pharmacy, Hospital Clinic of Barcelona, Barcelona, SPAIN, 12Medical Oncology, Hospital Clinic of Barcelona, Barcelona, SPAIN, 13Statistics Unit, REVEAL Genomics, Barcelona, SPAIN, 14Medical Oncology, Hospital Clinic of Barcelona. Translational Genomics and Targeted Therapies Group, IDIBAPS, Barcelona, SPAIN.
Background: The Central nervous system (CNS) is a frequent and clinically significant site of progression in advanced HER2-positive (HER2+) breast cancer. Identifying patients at high risk for CNS progression could inform more personalized surveillance and therapeutic strategies. Prior work has demonstrated the prognostic value of the HER2DX ERBB2 mRNA score in this setting (NPJ Breast Cancer 2025; SABCS 2024). In addition, the HER2DX metastatic prognostic score—an integrated model combining ERBB2 with additional molecular signatures—was developed to predict overall survival (SABCS 2025, submitted). Here, we report the development and validation of the HER2DX CNS progression score, a distinct genomic tool specifically designed to predict the risk of brain metastasis.Methods: Transcriptomic data were analyzed from 215 patients with HER2-positive advanced breast cancer treated with first-line trastuzumab, pertuzumab, and chemotherapy (THP), including 93 cases from Spain and 122 from Poland. All tumor samples were profiled using the standardized HER2DX gene expression platform. Biological features associated with CNS progression were explored in the combined cohort, and a genomic score was developed using key signatures. Coefficients were derived in the Spanish cohort, and the final score was validated in the Polish cohort. Associations with CNS progression and overall survival were estimated using Cox models and Kaplan-Meier curves.Results: In the combined cohort (n=215), the median follow-up was 37.1 months. A total of 53 patients (24.8%) developed brain metastases. No significant differences in CNS progression rates (27.2% and 23.0%) or time to CNS progression (hazard ratio [HR] 0.90, 95% CI 0.51-1.56, p=0.70) were observed between cohorts. Clinical variables associated with CNS progression included younger age and having ≥3 metastatic sites. In contrast, hormone receptor status, HER2 IHC (3+ vs other), menopausal status, biopsy site (primary vs. metastatic), bone disease, and best overall response to THP were not associated with brain metastasis.HER2DX assay identified 3 main genomic patterns associated with CNS progression: (1) HER2DX ERBB2 expression and lower time to CNS progression, (2) HER2DX luminal score and longer time to CNS progression, and (3) a new HER2DX immune-related signature associated with longer time to CNS progression. In the Spanish set, the 3-year cumulative incidence of CNS progression was 42.8% in the HER2DX CNS progression high-risk group vs. 24.9% in the intermediate-risk group vs. 15.5% in the low-risk groups (HR between low vs high 0.32, 0.11-0.90, p=0.032); in the Polish set, the 3-year cumulative incidence of CNS progression was 39.2% in the HER2DX CNS progression high-risk group vs. 13% in the intermediate-risk group vs. 4.3% in the low-risk groups (HR between HER2DX low vs high groups of 0.11, 0.02-0.83, p=0.033). Importantly, the HER2DX CNS progression score was not associated with PFS or OS, confirming its specificity for CNS progression. Conversely, the HER2DX metastatic prognostic score (submitted to SABCS 2025) was not associated with CNS progression, and the two scores were uncorrelated (Pearson r=-0.049).Conclusions: The HER2DX CNS progression score is a novel, biology-informed genomic tool that independently predicts brain metastasis in HER2+ advanced breast cancer treated with first-line THP. These findings support its potential integration into clinical workflows to personalize CNS surveillance and treatment strategies.
Presentation numberPS5-01-09
Proactive Brain Screening using contrast-enhanced (CE) brain CT scans in HER2+ metastatic Breast Cancer (mBC)
Gaia Griguolo, Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
G. Griguolo1, M. Bottosso1, G. Landa1, G. Bonomi1, F. Miglietta1, M. Guarascio1, M. La Commare1, F. Zanghì1, C. Giorgi2, C. Falci2, C. Hodgdon3, M. Dieci1, V. Guarneri1; 1Department of Surgery, Oncology and Gastroenterology – University of Padova, Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, ITALY, 2Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, ITALY, 3GRASP, GRASP, Baltimore, MD.
Background: According to recent EANO-ESMO guidelines, proactive brain imaging can be considered in asymptomatic patients (pts) with HER2+ metastatic breast cancer (mBC), due to high risk of developing brain metastases (BMs), with the aim of detecting brain involvement before the development of impactful symptoms. However, no clear evidence to guide the use of a specific radiological technique and timing for brain screening is currently available. After changes in EANO-ESMO guidelines, several oncologists in Italy have started to pragmatically include brain evaluation in CE CT scans performed to re-evaluate HER2+ mBC (traditionally only including thorax/abdomen). However, we lack data regarding the potential impact of this strategy in reducing the frequency of symptomatic BM diagnosis. We here retrospectively assess the impact of brain screening using CE CT scans on incidence of symptomatic BMs in HER2+ mBC pts. Methods: All consecutive pts newly diagnosed with HER2+ mBC and treated with trastuzumab-pertuzumab plus taxane at a single center (2014-2024) were retrospectively identified. Pts with symptomatic BMs at first mBC diagnosis or lacking complete imaging data regarding the first 2 years from mBC diagnosis were excluded. Pts were classified as having undergone brain screening if at least one CE brain CT scan per year was performed in absence of neurological symptoms during the first 2 years following mBC diagnosis, by clinical practice of treating oncologist. Results: Overall, 148 pts were identified: 73 underwent brain screening during the 2-year observation period and 75 did not. Both subgroups represented similar clinicopathological features at mBC diagnosis, including Karnofsky Performance Status (KPS). As expected, patients who underwent brain screening were diagnosed more recently and had a significantly shorter median follow-up from mBC diagnosis (3.0 versus 5.9 years, p<0.001). Median number of brain CT scans per year in asymptomatic pts (during the first 2 years) was 2.0 (IQR 1.2-2.5) and 0.0 (IQR 0.0-0.5) in the screening and non-screening groups, respectively. Thirty pts (20.3%) developed BMs during the first 2 years. Cumulative incidence of BMs at 2-years was significantly higher in pts undergoing screening (vs not) (30.6% vs 12.3%, Gray’s p=0.004). However, cumulative incidence of symptomatic BMs at 2-years was significantly lower in pts undergoing screening (vs not) (0% vs 9.5%, Gray’s p=0.012). Moreover, pts undergoing screening presented a significantly more conserved performance status (KPS 90-100 for 71.4% of pts vs 11.1%, respectively; p=0.002) and a numerical trend toward a lower number of BMs at BM diagnosis (52.4% diagnosed with >3 BMs vs 88.9%, p=0.057). No significant difference in locoregional and systemic treatments administered after BM diagnosis was observed, although whole-brain radiotherapy (WBRT) was less frequently used in pts undergoing screening (14.3% vs 44.4%, p=0.073). Median OS from BM diagnosis was numerically longer in pts undergoing screening (28.6 vs 7.5 months, p=0.192). Conclusions: Brain screening with CT scans is associated with a significantly lower incidence of symptomatic BMs and with a significantly more conserved KPS at BM diagnosis in this retrospective real-world cohort of HER2+ mBC pts treated with pertuzumab-trastuzumab and taxane. These findings support the use of proactive brain imaging for pts with HER2+ mBC. Confirmatory prospective studies are needed to optimize surveillance timing and radiological techniques in this setting.
Presentation numberPS5-01-10
Estimating the effect of initiating early maintenance endocrine therapy on brain metastases-free survival and other clinical outcomes in patients with HER2+/HR+ mBC without brain metastases: A target trial emulation
Thibaut Sanglier, F. Hoffmann-La Roche Ltd, Basel, Switzerland
T. Sanglier1, I. Gravestock1, J. Leone2, M. Secrest3, C. Tchakoute3, E. Restuccia4, F. Montemurro5, M. Shivhare6, A. Knott7, P. Lambert3, D. Martinez8, P. Luhn9, X. Garcia de Albeniz Martinez10, S. Tolaney2, N. Lin2, S. Sammons2; 1Real World Data Science, F. Hoffmann-La Roche Ltd, Basel, SWITZERLAND, 2Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 3Real World Data Science, Genentech, Inc., South San Francisco, CA, 4Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, SWITZERLAND, 5Product Development Oncology, HER2 positive breast cancer franchise, F. Hoffmann-La Roche Ltd, Basel, SWITZERLAND, 6HER2+ Franchise, Roche Products Limited, Welwyn Garden City, UNITED KINGDOM, 7Product Development, Clinical Science Oncology, Roche Products Limited, Welwyn Garden City, UNITED KINGDOM, 8RTI-HS, RTI Health Solutions, Barcelona, SPAIN, 9Real World Data Oncology, Genentech, Inc., South San Francisco, CA, 10Epidemiology, RTI Health Solutions, Barcelona, SPAIN.
Background: Maintenance endocrine therapy (mET) is increasingly used as part of the post-chemotherapy treatment for HER2-positive, hormone receptor-positive metastatic breast cancer (HER2+, HR+ mBC). However, the impact of mET on delaying brain metastases (BM) and other clinical outcomes is unclear. As the time from taxane completion to mET initiation can vary considerably, standard cohort methods with fixed index dates can lead to immortal time and selection bias. This study estimates the effect of different mET treatment strategies (TS) on BM-free survival (BMFS), overall survival (OS), and real-world progression-free survival (rwPFS) in patients (pts) with HER2+, HR+ mBC without BM. Methods: The study used the Flatiron Health database (January 2011-September 2024). mET included aromatase inhibitors, selective estrogen receptor degraders or modulators, and ovarian function suppression. Our target trial compared four TS in pts with HER2+, HR+ mBC who had completed ≥5 cycles of 1L HER2-targeted therapy + taxane (baseline was last taxane administration): Arm 1: mET initiation within 3 months (mo) of baseline; Arm 2: no mET initiation within 3 mo of baseline (initiation was possible afterwards); Arm 3: no mET initiation within 60 mo of baseline; and Arm 4: mET initiation between 4-6 mo of baseline. In all TS, CDK4/6 inhibitor use was not permitted and pts who initiated 2L treatment/had a BMFS/OS/rwPFS event were excluded from the TS. In the target trial emulation (TTE), pts were assigned to all TS with which their baseline data were compatible, creating four clones (TS were not distinguishable at baseline), and were censored upon deviation from the assigned TS. BMFS, OS, rwPFS were measured up to 60 mo from last taxane administration (LTA); risk differences and risk ratios were estimated via pooled logistic regression adjusted for baseline confounders (de novo vs. recurrent disease, disease-free interval, treatment at an academic center, Roche Prognostic Score, last HR test being negative despite prior positive tests). Estimates were bounded with percentile-based 95% confidence intervals (CI) estimated via bootstrapping. Sensitivity analyses included changes in grace period, time discretization, functional form of time, censoring of early death events, and simulation of uniform initiation of TS. Results: The TTE (N = 784) evaluated 391 pts who received ET within 3 mo and 393 pts who did not receive ET within 3 mo. At the time of taxane completion, baseline characteristics were generally balanced between pt groups, except ET pts were more likely to be treated in an academic center (25.8% vs. 17.3%) and less likely to have a negative latest HR test (3.6% vs. 12.7%). The difference in the risk of death (95% CI) comparing Arm 1 vs. Arm 3 was -3.2% (-5.6, -0.8) at 12 mo and -6.5% (-10.9, -1.7) at 24 mo (negative estimates favor Arm 1). The corresponding risk differences for rwPFS were -6.8% (-16.0, 0.9) and -10.1% (-20.3, -1.9), and for BMFS, -2.0% (-8.9, 4.7) and -4.7% (-13.0, 2.8). Treatment effect estimates were less precise beyond 36 mo due to loss to follow-up. Similar estimates were found against other TS, although the limited number of pts with data compatible with Arm 4 yielded imprecise estimates. Conclusions: Early mET within 3 mo of LTA was associated with improved clinical outcomes in pts with HER2+, HR+ mBC (although the limited number of BMFS events and modest estimates prevent making any firm conclusions). The TTE framework for comparative effectiveness studies with flexible treatment initiation helps align real-world data with randomized clinical trials, supporting better design of real-world data studies including comparisons of TS that can inform clinical practice.
Presentation numberPS5-01-11
Impact of antiemetic strategy on overall survival in patients with metastatic HER2-positive breast cancer treated with trastuzumab deruxtecan: A real-world retrospective study
Sowjanya Reganti, Cancer Care Specialists, Reno, NV
S. Reganti1, R. Choksi2, F. Kudrik3, D. Patt4, S. Reddy1, S. Rosenfeld5, D. Parris6, G. Rahimi6, A. Rui6, M. Gart6, C. Wall6, B. Wang6, P. Varughese6, J. Donegan6, L. Morere6, R. Geller6, J. Scott6, V. Gorantla2; 1Medical Oncology, Cancer Care Specialists, Reno, NV, 2Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 4Medical Oncology, Texas Oncology, Austin, TX, 5Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 6PrecisionQ, IntegraConnect, West Palm Beach, FL.
Background: Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) approved for the treatment of metastatic HER2-positive (HER2+) breast cancer in the second-line of therapy or later and has shown significant improvements in progression-free and overall survival (OS) in this population. However, T-DXd is also associated with gastrointestinal toxicities, including nausea and vomiting, which can adversely affect quality of life and treatment adherence. While antiemetic guidelines recommend the use of combination prophylaxis for agents with moderate to high emetogenic potential, there is limited evidence on whether antiemetic strategies themselves impact treatment duration and survival outcomes in oncology patients (pts) receiving T-DXd. This real-world study investigates the potential association between antiemetic regimen choice and OS in metastatic HER2+ breast cancer pts initiating T-DXd therapy. Methods: The IntegraConnect PrecisionQ de-identified electronic health record database, consisting of >3 million cancer pts across >500 care sites, was used to conduct a retrospective cohort study of 540 pts with metastatic HER2+ breast cancer who initiated trastuzumab deruxtecan between December 20, 2019 and October 31, 2024. Pts were stratified by their antiemetic regimen within 7 days of T-DXd initiation: Group 1 (n=251) received 5HT3 receptor antagonist + dexamethasone and Group 2 (n=289) received NK1 receptor antagonist + 5HT3 receptor antagonist + dexamethasone. Real-world overall survival (rwOS) and time to treatment discontinuation or death (TTD) were assessed using Kaplan-Meier analysis starting from the time of T-DXd initiation. Multivariable Cox proportional hazards models were used to adjust for potential confounders including age at diagnosis, race, Eastern Cooperative Oncology Group (ECOG) performance status, and line of therapy. Results: Baseline demographics were balanced between groups. Median age at diagnosis was 54 years for Group 1 and 55 years for Group 2; median age at T-DXd initiation was 61 years for Group 1 and 62 years for Group 2. Both groups were predominantly White (60.2% in Group 1 and 66.4% in Group 2). Median TTD (16.0 vs. 11.0 months (log-rank P< 0.01) and median rwOS (35.7 vs. 19.2 months (log-rank P<0.01) were significantly longer in Group 2 (NK1-based regimen) compared to Group 1 (5HT3 + dexamethasone). The probability of remaining on treatment at 12, 24, and 36 months was higher in Group 2: 12 months: 63% vs. 48%, 24 months: 38% vs. 20%, 36 months: 27% vs. 8%. Similarly, estimated survival at 12, 24, and 36 months was higher in Group 2: 12 months: 83% vs. 71%, 24 months: 60% vs. 42%, 36 months: 48% vs. 29%. On multivariable analysis, receipt of the NK1-containing regimen was associated with a 40% reduced risk of discontinuation or death (hazard ratio: 0.60, 95% CI: 0.45-0.80; P<0.01) compared to the 5HT3 + dexamethasone group, independent of other covariates. Poorer ECOG performance status was associated with increased likelihood of discontinuing T-DXd and worse survival. Conclusion: Among pts with metastatic HER2+ breast cancer treated with T-DXd, the use of a 3-drug antiemetic regimen including a NK1 receptor antagonist + 5HT3 receptor antagonist + dexamethasone was associated with significantly longer treatment persistence which seems to translate to improved OS compared with a 2-durg antiemetic regimen of a 5HT3 receptor antagonist + dexamethasone. These findings suggest that optimization of supportive care strategies may have survival implications and merit further evaluation/prospective validation.
Presentation numberPS5-01-12
Real-world time to next treatment in HER2+ breast cancer patients with brain metastases: trastuzumab deruxtecan vs tucatinib based therapy as second-line treatment
Vikram Gorantla, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA
V. Gorantla1, R. Choksi1, F. Kudrik2, D. Patt3, S. Reddy4, S. Reganti4, S. Rosenfeld5, G. Cioffi6, T. Sura6, D. Parris6, A. Rui6, M. Gart6, C. Wall6, B. Wang6, P. Varughese6, J. Donegan6, L. Morere6, R. Geller6, J. Scott6, R. Mahtani7; 1Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 2Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 3Medical Oncology, Texas Oncology, Austin, TX, 4Medical Oncology, Cancer Care Specialists, Reno, NV, 5Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 6PrecisionQ, IntegraConnect, West Palm Beach, FL, 7Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Brain metastases (BrM) are a frequent and clinically challenging manifestation of HER2-positive (HER2+) metastatic breast cancer (mBC), affecting up to 50% of patients (pts) over the course of their disease. Current guidelines recommend trastuzumab deruxtecan (T-DXd) as the preferred second-line therapy for HER2+ mBC overall, while the combination of tucatinib, trastuzumab, and capecitabine (TTC) remains an important option for pts with active or progressing BrM disease due to its proven intracranial activity. However, there is a lack of real-world data comparing treatment outcomes with these regimens specifically among pts with BrM involvement. This study evaluates real-world time to next treatment (TTNT) in pts with prior HER2+ therapy and documented BrM treated with second-line T-DXd vs TTC. Methods: We used the IntegraConnect PrecisionQ de-identified electronic health record database, which includes data on over 3 million cancer pts across more than 500 care sites, to identify individuals with HER2+ mBC who received either T-DXd or TTC as second-line therapy on or after January 1, 2020. Eligible pts had documented BrM prior to initiating second-line treatment. HER2+ status was inferred based on receipt of at least one HER2-targeted therapy during first-line metastatic treatment. Individuals who received T-DXd in their first-line of metastatic therapy were excluded from the analysis. The index date was defined as the start of second-line therapy with either TTC or T-DXd. Descriptive statistics were used to summarize demographic and clinical characteristics by treatment group, including age at index date, race, ethnicity, health insurance, BMI, ECOG performance status, estrogen receptor status, first-line metastatic regimen (grouped as ado-trastuzumab emtansine, trastuzumab (H) +pertuzumab (P) + taxane, endocrine therapy +/- HP, or other regimens), and presence of liver, bone, or lung metastases. TTNT was analyzed using Kaplan-Meier survival curves, with median estimates reported. Multivariable Cox proportional hazards regression was used to evaluate the association between treatment and TTNT, adjusting for the aforementioned demographic and clinical covariates. Hazard ratios (HRs) and corresponding 95% confidence intervals (CIs) are presented. Results: A total of 255 pts met study eligibility, of whom 138 (54%) received T-DXd as second-line therapy. Pts treated with T-DXd were slightly older than those who received TTC (median age: 58 vs 55 years; Wilcoxon p=0.026). A lower proportion of pts in the T-DXd group were classified as overweight (BMI 25-30) compared to the TTC group (17% vs 29%; chi-square p=0.026). No other significant differences in demographic or clinical characteristics were observed between treatment groups. Median TTNT was significantly longer among pts who received T-DXd compared to TTC (17 vs 11 months; log-rank p=0.006). This difference remained significant after adjustment for demographic and clinical covariates, with T-DXd associated with a 48% reduced likelihood of requiring subsequent therapy (HR: 0.52; 95% CI: 0.37-0.74; p<0.001). Conclusions: In this real-world analysis of pts with prior HER2+ therapy and documented BrM, second-line treatment with T-DXd was associated with significantly longer TTNT compared to TTC among pts with BrM. These findings may help inform treatment decisions in this high-risk population. However, interpretation is limited by the retrospective design, potential selection bias, and the use of treatment patterns to infer HER2 status. Most importantly, granular clinical data on BrM, including prior local therapies (radiation and/or surgery) and CNS-specific outcomes, were unavailable. Further prospective studies are needed to validate these findings.
Presentation numberPS5-01-13
Utidelone plus bevacizumab for the treatment of HER2-positive breast cancer brain metastases (U-BOMB-HER): A multicenter, single-arm phase IIstudy
Min Yan, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
H. Lv1, S. Wang2, Y. Song3, L. Niu1, M. Zhang1, Z. Liu1, J. Wu1, X. Sun1, Y. Cui1, J. Wang1, Y. Feng1, H. Sun1, J. Huang2, J. Zhang2, L. Wang1, M. Lv3, H. Zeng1, S. Chen1, M. Yan1; 1Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, CHINA, 2Cancer Center, Sun Yat-sen University, Guangzhou, CHINA, 3Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao, CHINA.
Background: While novel HER2-targeted agents (including TKIs and ADCs) have demonstrated intracranial activity in HER2 positive (HER2 +) metastatic breast cancer (MBC) with active brain metastases (BM), effective management of TKI- and/or ADC-refractory disease remains an ongoing clinical challenge. Building on preclinical evidence of utidelone’s superior blood-brain barrier penetration and bevacizumab’s established anti-edema effects, we conducted this phase II study (NCT05357417) evaluating the combination in TKI-progressed HER2+ BM patients. Methods: This multicenter, two-cohort, single-arm phase II trial (NCT05357417) enrolled adult patients (≥18 years) with breast cancer who had brain metastases, either radiotherapy-naïve or with documented progression following prior radiotherapy. The results from HER2-negative cohort (U-BOMB study) have been completed and published [Yan M, et al. JAMA Oncol, 2025 Jun 26; e251694]. Here we present the preliminary findings from HER2-positive cohort (U-BOMB-HER) of patients with HER2-positive breast cancer who developed progressive brain metastases following treatment failure with trastuzumab and TKI . Patients received utidelone (30 mg/m² IV on days 1-5) plus bevacizumab (15 mg/kg IV on day 1) every 3 weeks until disease progression or intolerance. The primary endpoint was central nervous system (CNS) objective response rate (CNS-ORR) as assessed by Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1), with secondary endpoints including CNS disease control rate (CNS-DCR), progression-free survival (PFS), CNS-PFS, overall survival (OS) and safety.Results: From May 2022 to April 2025, 50 evaluable patients were enrolled (median age 52.0 years; range 32-68) with a median of 3 prior lines of therapy (range 0-9). Baseline characteristics included: 1 brain metastasis (n=14), 2 metastases (n=10), and ≥3 metastases (n=26). The cohort comprised 56.0% (28/50) radiotherapy-naïve patients and 44.0% (22/50) with post-radiotherapy progression, with 52.0% (26/50) having prior anti-HER2 ADC exposure (including 4 patients treated with ≥2ADCs with different payloads).Among 50 response-evaluable patients, the CNS objective response rate (CNS-ORR) was 54.0% (95% CI 39.3-68.2%), with a disease control rate of 92.0% (46/50; 95% CI 80.8-97.8%). For all 50 patients, median progression-free survival (PFS) was 8.6 months (95% CI 7.0-10.2) at data cutoff (June 30, 2025; median follow-up 14.0 months), with median overall survival (OS) of 11.0 months (95% CI 8.4-13.6).Treatment emergent adverse events (TEAEs) of any grade included peripheral neuropathy (96.0%), leukopenia (58.0%), ALT/AST elevation (52.0%), neutropenia (50.0%), anemia (42.0%), hypertension (42.0%), and proteinuria (38.0%). Grade 3 AEs occurred in peripheral neuropathy (6.0%), neutropenia (16.0%), leukopenia (8.0%), thrombocytopenia (4.0%), hypertension (12.0%), anemia (2.0%), and GGT elevation (2.0%). No grade ≥4 treatment-related AEs were observed. Conclusions: Preliminary findings from the U-BOMB-HER study indicate that utidelone plus bevacizumab demonstrates clinically meaningful CNS activity, along with a manageable safety profile, in patients with HER2-positive breast cancer brain metastases, refractory to trastuzumab and TKI. ClinicalTrials.gov: NCT05357417.
Presentation numberPS5-01-14
Trastuzumab deruxtecan (T-DXd) monotherapy and T-DXd + pertuzumab in patients (pts) with previously untreated HER2+ unresectable/metastatic breast cancer (mBC): final results from DESTINY-Breast07
Komal Jhaveri, Memorial Sloan Kettering Cancer Center, New York, NY
F. André1, E. Hamilton2, S. Loi3, C. Anders4, P. Schmid5, E. Artamonova6, R. Villanueva-Vázquez7, J. Pedrini8, D. Doval9, S. C. Chen10, S. Boston11, A. Konpa12, M. Markowska13, G. Fabbri14, K. Jhaveri15; 1Department of Medical Oncology, Gustave Roussy, Paris-Saclay University, Villejuif, FRANCE, 2Breast Cancer and Gynecologic Cancer Research, Sarah Cannon Research Institute, Nashville, TN, 3Breast Unit, Peter MacCallum Cancer Centre, Melbourne, VIC, AUSTRALIA, 4Division of Medical Oncology, Duke Cancer Institute, Durham, NC, 5Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 6Department of Chemotherapy, N N Blokhin National Medical Research Center of Oncology, Moscow, RUSSIAN FEDERATION, 7Breast Cancer Unit – Early Drug Development Unit, ICO L’Hospitalet, Barcelona, SPAIN, 8Department of Mastologia, Nossa Senhora da Conceição Hospital, Porto Alegre, BRAZIL, 9Breast and Thoracic Services, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, INDIA, 10Division of General Surgery, Department of Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan City, TAIWAN, 11Late Development Oncology, Oncology R&D, AstraZeneca, Gaithersburg, MD, 12Late Development Oncology, Oncology R&D, AstraZeneca, Warsaw, POLAND, 13Late Oncology Statistics, Oncology R&D, AstraZeneca, Warsaw, POLAND, 14Translational Medicine, Oncology R&D, AstraZeneca, Boston, MA, 15Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: T-DXd is approved for the treatment of adult pts with HER2+ mBC who have received a prior anti-HER2-based regimen. Recent results from the planned interim analysis of the Phase 3 DESTINY-Breast09 study demonstrated a statistically significant and clinically meaningful improvement in progression-free survival (PFS) by blinded independent central review for T-DXd + pertuzumab versus standard of care (taxane + trastuzumab + pertuzumab) in first-line (1L) HER2+ mBC (median 40.7 vs 26.9 months; hazard ratio 0.56 [95% CI 0.44, 0.71]). The T-DXd monotherapy module remains blinded until final PFS analysis. DESTINY-Breast07 (NCT04538742; initiated prior to DESTINY-Breast09) was a Phase 1b/2 multicenter, open-label, modular study exploring the safety, tolerability, and antitumor activity of T-DXd ± other anticancer agents in HER2+ mBC. Results presented are from the final analysis of the dose-expansion phase assessing T-DXd ± pertuzumab as 1L treatment in HER2+ mBC. Methods: Eligible pts had locally assessed HER2+ (immunohistochemistry [IHC] 3+ or IHC 2+ / in situ hybridization-positive) mBC. No prior therapy for mBC was allowed and a disease-free interval of ≥12 months from (neo)adjuvant therapy was required. Pts were stratified by hormone receptor status (positive [estrogen or progesterone receptor ≥1%] vs negative), disease status (recurrent vs de novo), and PD-L1 expression (positive [IHC ≥1%] vs negative). Pts received T-DXd 5.4 mg/kg intravenously (IV) every 3 weeks (Q3W) as monotherapy or in combination with pertuzumab 420 mg IV Q3W, with an 840-mg loading dose. Primary endpoints were safety and tolerability; secondary endpoints included objective response rate (ORR), duration of response, and PFS, per RECIST 1.1 by investigator, as well as time to progression on subsequent therapy (PFS2) by investigator, and overall survival (OS). Results: At final data cutoff (January 31, 2025), 75 pts in the T-DXd module and 50 pts in the T-DXd + pertuzumab module had received study treatment; demographics and disease characteristics were well balanced. Median follow up was 37.1 months with T-DXd and 38.6 months with T-DXd + pertuzumab. Median total treatment duration was 26.8 months in the T-DXd module, and 27.6 months for T-DXd and 26.0 months for pertuzumab in the T-DXd + pertuzumab module. Confirmed ORR (80% CI) was 78.7% (71.4, 84.7) with T-DXd and 84.0% (75.3, 90.5) with T-DXd + pertuzumab. At 36 months, 65.5% and 69.4% of pts remained in response in the T-DXd and T-DXd + pertuzumab modules, respectively. At study completion, median PFS and median OS were not reached for either module. The 80% CI lower limit for median PFS was 40.2 months for T-DXd + pertuzumab; all other CI limits were not evaluable. PFS rate (80% CI) at 24 months was 71.4% (63.5, 77.8) with T-DXd and 67.8% (58.2, 75.7) with T-DXd + pertuzumab. Grade ≥3 adverse events (AEs) occurred in 57.3% (n=43/75) and 62.0% (n=31/50), and serious AEs in 21.3% (n=16/75) and 28.0% (n=14/50), of pts in the T-DXd and T-DXd + pertuzumab modules, respectively. Adjudicated drug-related interstitial lung disease / pneumonitis events occurred in 11 (14.7%; Grade 1, n=3; Grade 2, n=8) pts who received T-DXd and 7 (14.0%; Grade 2, n=6; Grade 3, n=1) who received T-DXd + pertuzumab. Additional data by subgroup, including biomarker analyses, will be presented. Conclusion: In this Phase 1b/2 DESTINY-Breast07 study, safety profiles for T-DXd and pertuzumab were generally consistent with the known profiles of each agent. Encouraging clinical activity was demonstrated in pts who received either T-DXd monotherapy or T-DXd + pertuzumab as a 1L treatment for HER2+ mBC; results from the T-DXd + pertuzumab module are consistent with the interim findings from the Phase 3 DESTINY-Breast09 study.
Presentation numberPS5-01-15
Real-world effectiveness of trastuzumab deruxtecan in HER2+ metastatic breast cancer by racial and ethnic group – Data from US community practices
Ruchit Shah, Daiichi Sankyo, Inc., Basking Ridge, NJ
S. Mehta1, L. Stevens2, R. Shah1, M. Garretson1, S. Pasha2, C. Brown-Bickerstaff2, L. Herms2, M. Danso3; 1US Medical Affairs, Daiichi Sankyo, Inc., Basking Ridge, NJ, 2Real-World Research, Ontada, Boston, MA, 3Sarah Cannon Research Institute, Virginia Oncology Associates, Norfolk, VA.
Background: A novel antibody drug conjugate, trastuzumab deruxtecan (T-DXd), has demonstrated unparalleled clinical benefit in DESTINY-Breast clinical trials for HER2+ metastatic breast cancer (mBC). However, limited data exists on the clinical benefit of T-DXd in minority racial/ethnic patient groups in the US. This study assessed real-world patient characteristics, treatment patterns, and outcomes among patients (pts) with HER2+ mBC treated with T-DXd in the US community oncology setting by racial/ethnic group. Methods: This retrospective chart review study utilized the iKnowMed electronic health record and included pts who initiated T-DXd for HER2+ mBC (index event) between Jan 1, 2020 and Apr 30, 2024, had ≥1 post-index visit and had documented race/ethnicity as Black and/or Hispanic (BH cohort) or White and non-Hispanic (Wt cohort). Pts were followed until last patient contact or death on or before Apr 30, 2024. To balance key baseline characteristics across cohorts, BH pts were propensity score matched to Wt pts with exact matching according to line of therapy (LOT), adjustment for age category, and a caliper width of 0.25 standard deviations. Patient characteristics, treatment patterns, real-world Objective Response Rate (rwORR, complete/partial response) and incidence of adverse events of interest while on treatment were summarized descriptively for both cohorts. Time to treatment discontinuation (TTD), time to next treatment (TTNT), and real-world progression-free survival (rwPFS) from index were estimated using the Kaplan-Meier method. Results: A total of 162 pts were included, with 81 in each cohort. Mean age at index was 56.3 years (BH 56.3; Wt 56.2) and all pts all were female except for 1 BH male. A higher proportion of pts in the BH cohort were residing in the South region (59.3% vs. 37.0%), had visceral only metastasis (65.4% vs. 58.0%) and HR- status (27.2% vs. 22.2%) than in the Wt cohort. More pts in the BH cohort (49.4%) received T-DXd in 3L+ setting than in the Wt cohort (43.2%). The rwORR was 63.0% and 67.9% in the BH and Wt cohort, respectively. Among the BH pts (median follow-up: 15.2 months [mo]), median (95% CI) rwPFS was 16.6 mo (9.9-21.4), TTD was 12.5 mo (9.7-14.9), and TTNT was 18.3 mo (13.1-22.9). Similarly, among Wt pts (median follow-up: 13.6 mo), median (95% CI) rwPFS was 14.7 mo (9.8-17.4), TTD was 12.2 mo (9.7-16.8), and TTNT was 18.4 mo (13.2-21.1). The rates of adverse events of interest were as follows: nausea (BH 50.6%; Wt 70.4%), fatigue (BH 45.7%; Wt 64.2%), vomiting (BH 21.0%; Wt 27.2%), diarrhea (BH 24.7%; Wt 35.8%), neutropenia (BH 13.6%; Wt 7.4%), alopecia (BH 2.5%; Wt 11.1%), and interstitial lung disease/pneumonitis (BH 7.4%; Wt 4.9%). During follow-up, 30.9% of BH pts and 33.3% of Wt pts initiated subsequent treatment beyond index. Among these pts (BH n = 25; Wt n = 27), trastuzumab emtansine-based (BH 16.0%; Wt 18.5%) and T-DXd-based (BH 16.0%; Wt 14.8%) regimens were most frequently used in the LOT immediately following index. Conclusion: Among US minority racial/ethnic groups (Blacks and/or Hispanics) under-represented in prior HER2+ mBC clinical studies, T-DXd demonstrated real-world clinical benefit and tolerability similar to that observed in White non-Hispanic pts. Interpretation of findings should consider limitations inherent to the retrospective design, including residual and unmeasured confounding, and potential information bias stemming from differential symptom reporting, clinical monitoring, and documentation practices across racial/ethnic groups. Nonetheless, these findings support the effectiveness of T-DXd across diverse populations and underscore the importance of inclusive data to inform clinical decision-making and advance equitable breast cancer management and care delivery across racial/ethnic groups.
Presentation numberPS5-01-16
Phase II Trial of the FASN Inhibitor, Denifanstat (TVB-2640), Plus Trastuzumab in Combination with Paclitaxel or Endocrine Therapy (ET) in Patients with HER2+ Metastatic Breast Cancer (MBC) Resistant to Trastuzumab
Tufia C Haddad, Mayo Clinic, Rochester, MN
T. C. Haddad1, V. J. Suman2, R. R. Roa3, B. J. Ernst4, R. Lupu5, T. Vander Steen5, M. H. Solanki5, M. Keeney5, D. W. Northfelt4, K. S. Anderson4, T. J. Hobday1, S. Chumsri3, J. E. Hoppenworth1, J. L. Carroll1, K. V. Giridhar1, C. C. O’Sullivan1, R. A. Leon-Ferre1, M. P. Goetz1; 1Department of Oncology, Mayo Clinic, Rochester, MN, 2Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 3Division Hematology/Oncology, Mayo Clinic, Jacksonville, FL, 4Division Hematology/Oncology, Mayo Clinic, Phoenix, AZ, 5Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
Background: Fatty Acid Synthase (FASN) is a key lipogenic enzyme co-regulated with HER2 and highly expressed in ER+ and HER2+ BC. Its targeting downregulates HER2 expression. Our preclinical studies revealed that FASN inhibition can sensitize or restore sensitivity to trastuzumab, paclitaxel and ET in HER2+ BC models. Denifanstat (deni, formerly TVB-2640) is a novel FASN inhibitor that demonstrated promising clinical activity and safety in a prior phase I trial as either monotherapy or in combination with paclitaxel. The primary objective of this study was to evaluate the objective tumor response rate (ORR) of deni plus trastuzumab (H) in combination with paclitaxel (T) or ET. Methods: Two independent, phase II clinical trials were initiated to assess the efficacy and safety of deni plus H in combination with T (Cohort A) or ET (Cohort B). Key inclusion criteria included HER2+ MBC resistant to HER2-directed therapy, measurable disease, unlimited ET, ≤6 prior chemotherapy or antibody drug conjugate regimens, and confirmation of HER2+ MBC on a pre-registration biopsy. Deni was administered 100 mg/m2 orally once daily. HP or H plus ET (fulvestrant or aromatase inhibitor) was administered at standard dose and schedule. Blood was collected on Cycle 1 Day 1 (C1D1), C1D8 and C2D1 for serum FASN analysis. An additional MBC tumor biopsy was collected on C2D1 for correlative studies. A 2-stage Simon design with a safety-run in was used to test the null hypothesis that the true ORR is at most 5% against the alternative it is at least 20%. With a sample size of 32 patients (pts) per cohort, if ≥4 responses are seen, this design has a 90% chance of rejecting the true ORR is <5% when the true ORR is ≥20% at a 0.10 level of significance. The decision to close both trials to enrollment early was multifactorial. Results: From August 2017 – February 2024, 33 pts with HER2+ MBC pre-registered, among which 17 were eligible and 16 were evaluable (15 pts in Cohort A and 1 pt in Cohort B). The most common reason for ineligibility was HER2-negative status on central review of the pre-registration biopsy. Median age was 57 years (range 35-80). All pts were non-Hispanic and 13 (81.3%) were White. Median prior lines of therapy was 5 (range 0 – 11). In Cohort A, there were 5 partial responses; thus, the primary endpoint was met with the ORR estimated to be 33.3% (90% CI: 14.2-57.7%). The median PFS time was 7.0 months (95 %CI: 3.7 – 11.3), and median OS was 25.7 months (95%CI: 10.2 – NE). No dose limiting toxicities were observed during the C1 safety run-in (6 pts). In Cohort A, 7 (46.7%) developed a grade 3 or 4 adverse event (AE) attributed to combination therapy. Grade ≥2 AEs regardless of attribution occurring in ≥20% of all pts included anemia, fatigue, neutropenia, palmar plantar erythrodysesthesia (PPE), and peripheral neuropathy. Treatment was discontinued due to progression (12; 80%) or AE (3; 20% – pneumonitis; neuropathy; PPE). In Cohort B, the pt maintained stable disease through 8 cycles with grade 3 ALT increase, and she remained alive 31.2 months post-registration. Median C1D1 FASN level was 32.7 ng/mL (IQR: 18.7 – 49.6 ng/mL). There was minimal association between C1D8 and C2D1 FASN levels and time to disease progression (C1D8 n=9, Spearman rank correlation coefficient (rs) = 0.040; C2D1 n=13, rs = -0.019). Analyses of changes in MBC tumor expression of FASN, phospho-AKT, and phospho-S6 are in progress. Conclusion: Deni combined with HP provided clinical antitumor activity in pts with HER2+ MBC resistant to trastuzumab. Expected paclitaxel-associated AEs were observed. Nearly half of pts experienced a grade 3 or 4 AE attributed to the combination therapy, and 20% stopped treatment due to AE. Further study of deni, H plus ET is necessary to draw safety and efficacy conclusions.
Presentation numberPS5-01-17
Dna-based HER2 vaccine induces broader and more durable t-cell responses than peptide vaccine in advanced HER2-positive breast cancer
Ying Liu, University of Washington, Seattle, WA
Y. Liu, S. Huang, Y. Dang, M. L. Disis; Cancer Vaccine Institute, University of Washington, Seattle, WA.
DNA-Based HER2 Vaccine Induces Broader and More Durable T-Cell Responses Than Peptide Vaccine in Advanced HER2-Positive Breast CancerYing Liu, Sydney Huang, Yushe Dang, and Mary L. Disis. UW Cancer Vaccine Institute, University of Washington, Seattle, WABackground: Patients with HER2-overexpressing breast cancer often exhibit reduced immunity to the HER2 antigen. Therapeutic vaccines can enhance cytotoxic T cell responses against tumor-associated antigens, enabling selective tumor destruction. Both peptide- and DNA-based platforms have been explored for vaccine delivery, but direct comparisons of their safety and immunogenicity remain limited. We hypothesize that DNA-based vaccines may generate more robust and durable HER2-specific immunity than other vaccine platforms due to prolonged antigen expression from persistent plasmid presence in host tissues.Methods: We retrospectively analyzed 66 patients from a phase 1 DNA-vaccine trial and 38 from a phase 2 peptide-vaccine trial, all with stage III or IV HER2-positive breast cancer and either no evidence of disease or stable bone-only disease at enrollment. Both vaccines targeted HER2 intracellular domain (ICD) epitopes. Immunogenicity was assessed via IFN-γ ELISpot assays on cryopreserved PBMCs stimulated with HER2 ICD or extracellular domain (ECD) peptides as an indicator of epitope spreading. Changes in T-cell responses from baseline to peak post-vaccination were evaluated using the Wilcoxon test, and between-trial differences using the Mann-Whitney test. Patients were stratified by baseline HER2 immunity and immunologic responder status. Adverse events were recorded per NCI CTCAE v4.0. Results: The magnitude of T-cell immunity achieved to the HER2 ICD varied between the two modes of vaccination and was dependent on whether patients had preexisting immunity to HER2 when entering the study. Patients receiving the DNA vaccine showed significant increases in HER2 ICD immunity regardless of preexisting HER2 immunity (p=0.005 with, p<0.001 without), while those receiving the peptide vaccine showed elevated responses only in patients without preexisting immunity (p<0.001; p=0.76 with immunity). Both vaccines induced intramolecular epitope spreading, that is a broadening of the immune response to the HER2 ECD, representing the induction of endogenous immunity. DNA-vaccine recipients showed significant ECD immune responses both in those with (p=0.03) and without (p<0.001) baseline HER2 immunity. In contrast, peptide-vaccine recipients showed increased ECD immunity only in those without baseline immunity (p=0.04; p=0.33 with immunity). The DNA group had higher ECD immunity at baseline (p=0.02) and post-vaccination (p=0.005). The elevation in T-cell immunity from baseline to peak post-vaccination was comparable between vaccine types for both HER2 ICD (p=0.37) and ECD (p=0.13), including among responders (p=0.35 for ICD, p=0.44 for ECD). However, among non-responders, DNA-vaccine recipients demonstrated greater increases in both HER2 ICD (p=0.005) and ECD (p=0.04) immunity. One grade 3 ALT elevation occurred in the DNA-vaccine trial; no grade 3–5 events were observed in the peptide-vaccine trial. Conclusion: Both DNA- and peptide-based HER2 vaccines were well tolerated and capable of inducing HER2-specific T-cell responses in patients with advanced HER2-positive breast cancer. However, the DNA vaccine elicited broader and more consistent immune activation, including stronger epitope spreading to HER2 ECD and greater responses among patients without preexisting immunity. Notably, DNA-vaccine recipients demonstrated enhanced immunity even among non-responders, suggesting a more robust platform for overcoming baseline immune tolerance.
Presentation numberPS5-01-18
Dalpiciclib Plus Pyrotinib Plus Letrozole for Patients with Estrogen Receptor-Positive and HER2-Positive Metastatic Breast Cancer (DAPLET)
Zhanhong Chen, Zhejiang Cancer Hospital, Hangzhou, China
Z. Chen, P. Huang, H. Zhou, J. Chen; Breast Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, CHINA.
Title: Dalpiciclib Plus Pyrotinib Plus Letrozole for Patients with Estrogen Receptor-Positive and HER2-Positive Metastatic Breast Cancer (DAPLET) Abstract:Background: Estrogen receptor-positive and HER2-positive breast cancer accounts for approximately 10-15% of breast cancers. Standard metastatic regimens combining trastuzumab-based HER2-targeted therapy with chemotherapy show limited efficacy in this subtype. In SYSUCC-002 trial, trastuzumab plus endocrine therapy was not inferior to trastuzumab plus chemotherapy therapy in first-line HER2+HR+ metastatic breast cancer, suggesting the possibility of omission of chemotherapy. The CDK4/6 pathway may be involved in HER2-targeted therapy resistance due to the HER2-ER signaling cross-talk and thus CDK4/6 inhibitor may become a strategy for patients with HER2+ER+ breast cancer. Pyrotinib, an irreversible pan-HER tyrosine kinase inhibitor, shows synergistic activity with CDK4/6 blockade in preclinical models. Building on this rationale, we evaluated a novel, chemotherapy-free regimen of pyrotinib, dalpiciclib, and letrozole in patients with HER2+ER+ metastatic breast cancer.Methods: Eligible patients had ER+HER2+ metastatic breast cancer, defined as ER≥1%, and HER2 positivity (immunohistochemistry 3+ or in situ hybridization positive). Patients should previously receive at most one systemic treatment containing trastuzumab for metastatic breast cancer. Patients with stable brain metastases and no clinical symptoms could be enrolled. Eligible patients received pyrotinib (320 mg orally once daily, every 4 weeks), dalpiciclib (125 mg orally once daily on days 1-21 of a 4-week cycle), and letrozole (2.5 mg orally once daily, every 4 weeks) until disease progression or intolerable toxicity. Pre/peri-menopausal patients received GnRH agonist. Radiographic evaluation was conducted every two cycles. The primary endpoint was objective response rate (ORR). In SYSUCC-002 trial, the ORR of trastuzumab plus endocrine arm was 37.2%, thus we hypothesized ORR of 55% in this trial. Using Simon’s optimal two-stage design, at the significance level of type I error rate at 0.05 and power of 0.8, a total of 22 patients was accrued in stage I. If there were more than 9 responses among these evaluable 22 patients, the study would enter stage II. The registration number of this trial was NCT07014410.Results: Between February 2023 and March 2025, 18 patients were enrolled. The median age was 54.5 years (range, 34-71). Ki67 expression at baseline included 12 patients (66.7%) with ≥30%. Of these 18 patients, 12 (66.7%) previously received trastuzumab, of whom 6 patients (33.3%) injecting with trastuzumab at metastatic stage. As of June 20, 2025, 16 of 18 patients had evaluable radiographic data (two patients withdrew from the study within two cycles because of intolerable toxicity). In these evaluable 16 patients, 11 patients achieved confirmed responses (1 complete response, 10 partial responses), yielding an ORR of 68.9% (95% confidence interval [CI], 41-88%). Despite inadequate patients in stage I, outstanding efficacy result indicated the study could enter stage II. The most common grade 3 adverse events were white blood cell count decreased (61.1%) and neutropenia (50.0%). No grade≥4 events were reported.Conclusions: This chemotherapy-free, trigeminy-targeted regimen demonstrated promising antitumor activity and manageable toxicity in first or second line HER2+ER+ metastatic breast cancer. The data supported this regimen as a potential alternative to standard chemotherapy-based approaches in this subtype. Efficacy of this regimen needs to be further evaluated in Simon stage II.
Presentation numberPS5-01-19
Mechanisms of resistance to anti-HER2 therapies in brain metastatic derivatives of inflammatory HER2-positive breast cancer models
Caroline M. Sabotta, Baylor College of Medicine, Houston, TX
C. M. Sabotta1, F. Liao2, M. J. Shea2, S. Nanda2, L. Qin2, P. S. Steeg3, M. F. Rimawi4, C. Gutierrez2, C. Osborne4, S. G. Hilsenbeck4, J. Veeraraghavan4, R. Schiff4; 1Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 2Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 3Women’s Malignancies Branch, Center for Cancer Research National Cancer Institute, Bethesda, MD, 4Lester and Sue Smith Breast Center, Dan L. Duncan Comprehensive Cancer Center, and Dept of Medicine, Baylor College of Medicine, Houston, TX.
Background: HER2-positive (HER2+) breast cancer (BC) is an aggressive subtype with 30-50% incidence of brain metastases in metastatic patients. The HER2-selective tyrosine kinase inhibitor (TKI) tucatinib (Tuca) and the pan-HER TKI neratinib (Nrb), mostly used in the late line setting, are effective, including in treating brain metastases. The new antibody drug conjugate T-DXd is also highly effective in the brain metastatic setting. It is crucial to understand the mechanisms of resistance to these brain permeable anti-HER2 therapies. Materials and Methods: GFP/Luc tagged SUM190-BR3 (SUM190Br), a brain-tropic derivative of the HER2+/ER- inflammatory SUM190 cell model (harboring the PIK3CA H1047R mutation), was used (PMID: 27245829). A long-term highly Tuca resistant (TucaR) derivative of this line was developed through exposure to increasing doses of Tuca up to 1uM. Characterization of these resistant and naïve cells was performed by Western blot (WB), DNA-seq, and RNA-seq. Drug efficacy studies involved methylene blue-based cell growth and IC50 assays, and included the Akt inhibitor (i), capivasertib (Capi, 1uM), T-DXd (5ug/mL), and the EGFR-specific TKI gefitinib (Gef, 1uM) or monoclonal antibody cetuximab (Cetux, 10ug/mL). To characterize the in vivo tumorigenicity and metastatic capacity of our resistant model, immunocompromised mice were injected with cells via subcutaneous or intracardiac routes and monitored for primary tumor growth and metastasis by bioluminescence imaging. Results: Interestingly, the naïve SUM190Br cells are intrinsically resistant to Tuca (IC50>600nM), but not Nrb (IC50<60nM). Capi is effective as a single agent in the TucaR model (p<.0001). WB analysis of the TucaR model showed substantially elevated levels of total EGFR and modestly elevated HER2, as well as increased phosphorylation of EGFR and HER2, suggesting pathway reactivation at time of resistance vs. short-term Tuca treated naïve cells. Further, DNA and RNA-seq analysis showed high EGFR at the mRNA level with no change in copy number. As we previously showed in other HER2+ TucaR models such as BT474 (with acquired EGFR amplification), the SUM190Br TucaR cells remain sensitive to Nrb. Likewise, EGFRi together with Tuca was effective in overcoming resistance in this TucaR model. T-DXd is highly effective in metastatic BC, but high EGFR expression has been shown to reduce its efficacy (PMID: 39437778). Both Gef and Cetux improve the efficacy of T-DXd in our models, but Cetux is selectively effective against the TucaR model. Moreover, our preliminary in vivo results show that the highly aggressive TucaR model grows as xenografts, keeps brain tropism via intracardiac injection, and metastasizes via subcutaneous injection. Conclusions: Our findings suggest the role of high EGFR and PIK3CA mutations in resistance to Tuca, which warrants additional preclinical and clinical investigation. This underscores the importance of understanding if PIK3CA mutations are associated with reduced Tuca sensitivity and the testing of new mutant specific PIK3CAi or other PI3K/Akt pathway inhibitors. Our brain tropic TucaR cell and mouse models will be useful to understanding resistance in the brain metastatic setting, and future work will test the most promising treatments in vivo. The recent DESTINY-Breast09 trial points to the promise of T-DXd in combination with the monoclonal antibody pertuzumab as the new first line for metastatic BC. Since high-EGFR is suggested to play a potential role in reducing T-DXd efficacy, our ongoing and future work seeks to understand if the new T-DXd plus pertuzumab combination will be effective compared to T-DXd plus EGFRi in treating HER2+ BC with high EGFR.
Presentation numberPS5-01-20
Taxane induction before trastuzumab-endocrine therapy improves 10-year survival in ER-positive HER2-positive metastatic breast cancer: a propensity-matched real-world analysis of 646 patients
Ahmed Elkhanany, Baylor College of Medicine, Houston, TX
M. Eysha1, M. Elchouemi2, W. Zhang 3, R. del Toro-Mijares4, A. Asad5, A. Elkhanany6; 1Department of Medicine, Texas tech University health science center, El Paso, TX, 2Department of Medicine, Texas Tech University Health Science Center El Paso,, Houston, TX, 3Department of Medicine, Duke University Medical Center, Durham, NC, 4Department of Medicine, Texas Tech Health El Paso, El Paso, TX, 5Department of Medicine, UTMB John Sealy School of Medicine, Galveston, TX, 6Department of Medicine, Baylor College of Medicine, Houston, TX.
Background: CLEOPATRA trial, adding Pertuzumab to Docetaxel and Trastuzumab improved median overall survival (OS) from 40.8 to 57.1 months and remains guideline-defining for first-line HER2-positive metastatic breast cancer (MBC), albeit with risks of myelosuppression and long-term neurotoxicity. In HR+ HER2+ MBC, few studies presented good outcomes with addition of endocrine therapy (ET) and CDK4/6 inhibitors without chemotherapy (PERTAIN, SYSUCC-002, PERNETTA, BR 18-2 MINI). Robust real-world survival data comparing chemotherapy-free versus taxane-induction strategies in this subgroup are lacking. Methods: TriNetX Global Collaborative Network (152 sites, 2008-2024) was queried for adults (≥18 y) with incident HR+ HER2+ MBC treated with Trastuzumab (± Pertuzumab) plus ET. Patients receiving ≥1 dose of paclitaxel or docetaxel within 6 months of diagnosis before the first HER2-targeted dose constituted the induction cohort (IND, n = 365 pre-match); others served as controls (CTRL, n = 498). One-to-one propensity-score matching (caliper 0.10) on 13 demographics/comorbidities yielded 323 well-balanced pairs (N = 646); after matching all standardized differences were < 0.10 except male sex (3 % vs 0 %). Ten-year OS was the primary endpoint. Secondary outcomes were grade-coded neutropenia, sepsis, heart failure, emergency-department (ED) visits and intensive-care-unit (ICU) admission. Survival analysis done with univariate Kaplan Meier and Cox models. Results: Of 7184 HR+ HER2+ MBC cases, 2114 received first-line Trastuzumab (± Pertuzumab) + ET; 863 met prespecified criteria (365 IND, 498 CTRL); 646 remained after 1:1 matching. Matched cohorts were well balanced, with median age 52 years, 96 % female, 59 % White, 22 % Black. All baseline standardized differences < 0.10 (except male sex 3 % vs 0 %). Median follow-up was 1578 vs 1151 days (IND vs CTRL). Ten-year OS was 64.3 % in IND vs 53.1 % in CTRL (absolute gain 11.2 %; HR 0.64, 95 % CI 0.47-0.85, P = 0.002). Ten-year mortality fell from 31.9 % to 24.1 % (absolute risk reduction 7.8 %; NNT = 13). Survival curves diverged by 12 months and remained parallel. Induction increased neutropenia (25.7 % vs 13.9 %; HR 1.88, P < 0.001) and ED visits (63.5 % vs 51.7 %; HR 1.24, P = 0.009); with median time to first ED visit shortened from 1084 to 678 days in the IND group. Serious events were comparable: sepsis 17.6 % vs 14.9 % (P = 0.34), heart failure 18.0 % vs 16.4 % (P = 0.60), ICU admission 16.1 % vs 16.4 % (P = 0.92). Conclusions: In contrast to some signals from chemotherapy-free regimens in PERTAIN, SYSUCC-002 and PERNETTA, this real-world study shows that a short taxane induction in HR+ HER2+ MBC before trastuzumab-ET confers a 36 % relative and 11 % absolute 10-year OS advantage; one extra life saved per thirteen treated; at the cost of predictable, manageable myelosuppression and higher acute-care use without excess cardiotoxicity, sepsis or ICU need. This data strengthens guideline endorsements of chemotherapy-containing first-line regimens and asks for careful discussion if upfront de-escalation is considered, as well as provides a benchmark for ongoing chemo-sparing strategies utilized in other trials (DESTINY-Breast09, DEMETHER, etc.). Selection and lead-time bias are possible but were mitigated by robust propensity matching.
Presentation numberPS5-01-21
Prognostic Significance of Circulating Tumor Cell Subtypes in HER2+ Metastatic Breast Cancer with Central Nervous System Metastases
Diana Jaber, McGaw Medical Center of Northwestern University, Chicago, IL
D. Jaber1, S. Marini1, N. Raptis1, N. K. Heater2, Y. Ma2, S. Warrior2, L. Flaum2, R. Stein2, P. Robinson2, H. Liu2, Y. Zhang2, R. Chen2, Q. Zhang2, L. C. Platanias2, W. Gradishar2, P. U. Kumthekar2, J. Lu2; 1Internal Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, 2Hematology-Oncology, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL.
Background: Patients with HER2-positive (HER2+) metastatic breast cancer (MBC) are at increased risk for developing central nervous system (CNS) metastases, presenting a significant clinical challenge and a major cause of morbidity and mortality in this population. Circulating tumor cells (CTCs) offer a noninvasive tool for disease monitoring; however, the prognostic significance of CTC subtypes in the context of CNS involvement remains unclear. This study examines the association between CTC subtypes and CNS progression and outcomes in patients with HER2+ MBC and detectable HER2+ CTCs. Methods: This retrospective cohort study examined patients with MBC who had CTCs collected from blood samples between 2016-2024 under an IRB-approved protocol (NU16B06) at Northwestern University Robert H. Lurie Comprehensive Cancer Center. CTC enumeration was performed via CELLTRACKS (Menarini). Patients with CTC samples collected within 6 months prior to radiographic detection of CNS metastases, defined as parenchymal brain metastases (PBM) or leptomeningeal disease (LMD), were included. Early CNS disease was defined as detection of CNS metastases within 12 months of metastatic diagnosis. Patients with at least one HER2+ CTC were analyzed. The relative proportion of CTCs was assessed by including both HER2+ and non-HER2+ CTC counts in multivariable models to capture the distinct contributions of each subtype. Results: 26 patients with HER2+ CTCs who developed CNS metastases were identified. The median age at diagnosis of CNS metastases was 54 years (interquartile range [IQR]: 46-64); 18 (75%) were White, 4 (17%) African American, and 2 (8.3%) Asian. At radiographic detection, 21 (80.8%) had PBM, 3 (11.5%) had LMD, and 2 (7.7%) had both. Median total CTC count was 15 (IQR: 5-108), and median HER2+ CTC count was 6 (IQR: 2-59). Following CNS detection, 13 (50%) received HER2-targeted therapy, 8 (31%) received non-HER2 therapy, and 5 (19%) received no therapy. Median overall survival (OS) was 13.0 months with HER2-targeted therapy versus 7.0 months with non-HER2 therapy (p=0.32), while median progression-free survival (PFS) was 2.6 vs. 2.0 months, respectively (p=0.98). After adjusting for extracranial disease burden, receipt of radiation, and CTC counts, HER2-targeted therapy was not associated with differences in OS (HR 0.50, 95% CI 0.16-1.50, p=0.2) or PFS (HR 1.89, 95% CI 0.55-6.47, p=0.3). Similarly, after multivariable adjustment, HER2+ CTC count was not associated with OS (HR 1.006 per CTC, 95% CI 0.99-1.02, p=0.5) or PFS (HR 0.997 per CTC, 95% CI 0.98-1.01, p=0.7), and non-HER2+ CTC count was not associated with OS (1.007 per CTC, 95% CI 0.10-1.02, p=0.2) or PFS (HR 1.004 per CTC, 95% CI 0.99-1.02, p=0.4). After multivariable adjustment, HER2+ CTC count was not associated with early (12 months) CNS progression (OR 1.00, 95% CI 0.97-1.03, p=0.9), whereas higher non-HER2+ CTC count was significantly associated with early CNS progression (OR 1.02 per CTC, 95% CI 1.00-1.07, p=0.041). Conclusion: In this real-world cohort of patients with HER2+ CTCs and CNS metastases, higher non-HER2+ CTC burden, included to capture total CTC burden while isolating the effects of HER2+ CTCs, was associated with early CNS progression. HER2+ CTC counts and HER2-targeted therapy were not associated with survival or progression outcomes. These findings suggest that CTC heterogeneity, particularly the presence of non-HER2+ subpopulations, may reflect a biologically aggressive or treatment-resistant phenotype in patients with HER2+ MBC and CNS involvement. The lack of association between HER2+ CTCs and outcomes may reflect the limited cohort size and treatment heterogeneity, highlighting the need for larger studies to clarify the prognostic value of HER2+ CTC burden in this population.
Presentation numberPS5-01-22
Efficacy of second- or third-line Tucatinib, Trastuzumab, and Capecitabine (TTC) following trastuzumab deruxtecan (T-DXd) in HER2-positive metastatic breast cancer: A multicenter French cohort study.
Jean Sebastien Frenel, Institut de Cancerologie de L’Ouest, Saint-Herblain, France
J. S. Frenel1, A. de Nonnevile2, C. Guerin-Charbonnel3, J. Zeghondy4, L. Mathiot1, A. Mailliez5, F. Poumeaud6, N. Isambert7, M. Arnedos8, F. Le Du9, L. Galland10, R. Kabirian11, S. Guiu12, L. Poestch13, E. Deluche14, F. Cherifi15, F. Dalenc6, E. Volant1, B. Pistilli4, T. San16, T. Bachelot17, A. Patsouris18, F. Bocquet19, L. Larrouquere17, D. Loirat20; 1Medical Oncology, Institut de Cancerologie de L’Ouest, Saint-Herblain, FRANCE, 2Medical Oncology, Institut Paoli Calmette, Marseille, FRANCE, 3Statistics, Institut de Cancerologie de L’ouest, Saint-Herblain, FRANCE, 4Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 5Medical Oncology, Centre Oscar Lambert, Lille, FRANCE, 6Medical Oncology, Oncopole, Toulouse, FRANCE, 7Medical Oncology, CHU Poitiers, Poitiers, FRANCE, 8Medical Oncology, Centre Bergonie, Bordeaux, FRANCE, 9Medical Oncology, Centre Eugène Marquis, Rennes, FRANCE, 10Medical Oncology, Centre Georges-Francois Leclerc, Saint-Herblain, FRANCE, 11Medical Oncology, Institut Curie, Paris, FRANCE, 12Medical Oncology, Institut de Cancer Montpellier, Monptellier, FRANCE, 13Medical Oncology, Polyclinique Bordeaux Nord Aquitaine, Bordeaux, FRANCE, 14Medical Oncology, CHU de Limoges, Limoges, FRANCE, 15Medical Oncology, Centre Francois Baclesse, Caen, FRANCE, 16Medical Oncology, CORT37, Chambray les tours, FRANCE, 17Medical Oncology, Centre Leon Berard, Lyon, FRANCE, 18Medical Oncology, Institut de Cancerologie de L’ouest, Angers, FRANCE, 19Data Factory and analytics, Institut de Cancerologie de L’ouest, Saint-Herblain, FRANCE, 20Medical Oncology, Institut Curie, PARIS, FRANCE.
Background: Trastuzumab deruxtecan (T-DXd) is currently the standard second-line therapy for HER2-positive metastatic breast cancer (MBC). Recent results from the DESTINY-Breast09 (DB-09) trial suggest it may soon be considered for first line setting. However, optimal treatment sequences after T-DXd remain unclear. Currently, data regarding the efficacy of the combination of tucatinib, trastuzumab, and capecitabine (TTC) administered as second- or third-line therapy after T-DXd is limited. Materials and Methods: We conducted a cohort study across 17 French cancer centers. Eligible patients had HER2-positive MBC and received TTC as second- or third-line therapy in the metastatic setting after prior treatment with T-DXd. The primary endpoint was progression-free survival (PFS). Secondary endpoints included time to next treatment (TTNT), overall survival (OS), objective response rate (ORR). Results: Between July 2021 and June 2025, 103 patients were enrolled. The median age at TTC initiation was 54.6 years (range: 21.0-86.0), 42.7% had de novo metastatic disease and 55.3% had brain metastases. Prior (neo)adjuvant chemotherapy and anti-HER2 therapy were administered to 55.3% of patients, while 88.3% had received pertuzumab as treatment for metastatic disease. Most patients (91.3%) progressed on T-DXd, while 8.7% discontinued due to toxicity or other reasons. The median duration of prior T-DXd therapy was 8.0 months (range: 0.7-37.2). TTC was administered as second-line therapy in 8.7% and third line in 91.3%, immediately following T-DXd in all cases. After a median follow-up of 12.7 months (95% CI: 11.0-17.0), 74 patients (71.8%) had discontinued TTC due to disease progression, 26 patients (25.2%) remained on treatment, and 3 patients (3.0%) discontinued TTC because of toxicity. Median PFS with TTC was 4.7 months (95% CI: 3.5-5.7), median TTNT was 6.6 months (95% CI: 4.8-9.6), and median OS was 12.3 months (95% CI: 10.5-19.5). Among the 96 RECIST-evaluable patients, 9.4% achieved a complete response, 20.8% had a partial response, 27.1% showed stable disease, and 42.7% experienced progressive disease as best response. Patients treated with T-DXd for >18 months (n=15) had a median PFS of 6.9 months (95% CI: 4.8-NR) and a median TTNT of 8.9 months (95% CI: 5.9-NR) compared to to a median PFS of 3.9 months (95% CI: 3.2-5.3) and a median TTNT of 5.5 months (95% CI: 4.2-9.6) in those treated with T-DXd for ≤ 18 months (n=88). Patients with brain metastases had a median PFS of 5.0 months (95% CI: 3.8-7.4) and a median TTNT of 8.5 months (95% CI: 5.5-11.7). For those without brain metastases, median PFS was 3.5 months (95% CI: 2.7-6.8) and median TTNT was 4.8 months (95% CI: 3.4-9.6). Following TTC progression, 60 patients received further therapy, with a median PFS of 2.7 months (95% CI: 2.5-3.9). Conclusion: In this multicenter French cohort, TTC showed clinically meaningful activity as a second- or third-line therapy in HER2-positive MBC patients previously treated with T-DXd, especially among long-responders (PFS>18 months) to T-DXd. Final updated results will be presented at the meeting.
Presentation numberPS5-01-23
Validation of the HER2DX Genomic Test in First-Line Advanced HER2-Positive Breast Cancer: Identifying Long-Term Responders to THP
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
M. Kubeczko1, S. Cobo2, R. Sanchez-Bayona3, B. Pycinski4, J. Soberino5, E. Chmielik6, E. Sanfeliu7, M. Rey8, F. Pardo9, A. Aguirre8, O. Castillo8, A. Lesniak1, M. Oczko-Wojciechowska10, E. Carcelero11, B. Adamo12, M. Vidal12, M. Bergamino13, J. Maues14, G. Villacampa15, L. Pare9, E. Ciruelos16, A. Prat17, M. Jarzab1, F. Braso-Maritany18; 1Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 2Breast Cancer Center, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, SPAIN, 3Unidad de Cáncer de Mama y Ginecológico, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 4Faculty of Biomedical Engineering, Silesian University of Technology, Zabrze, POLAND, 5Breast Cancer Unit, IOB-QuirónSalud, Barcelona, SPAIN, 6Tumor Pathology Department, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 7Department of Medicine, Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS); Hospital Clinic Barcelona; University of Barcelona, Barcelona, SPAIN, 8Translational Genomics and Targeted Therapies in Solid Tumors, Cancer Institute and Blood Disorders, Hospital Clinic; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN, 9Reveal Genomics, Reveal Genomics, Barcelona, SPAIN, 10Department of Clinical and Molecular Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 11Pharmacy Department, Hospital Clinic Barcelona, Barcelona, SPAIN, 12Department of Medical Oncology, Hospital Clinic of Barcelona; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS); SOLTI Breast Cancer Research Group, Barcelona, SPAIN, 13Medical Oncology Department, Hospital Clinic Barcelona, Barcelona, SPAIN, 14GRASP, GRASP, Baltimore, MD, 15Statistics Unit, Vall d’Hebron Institute of Oncology, Cellex Centre; SOLTI Breast Cancer Research Group, Barcelona, SPAIN, 16Medical Oncology Department Breast Cancer Unit, Hospital Universitario 12 de Octubre; HM CIOCC; SOLTI Breast Cancer Research Cooperative Group, Madrid, SPAIN, 17Cancer Institute and Blood Diseases, Hospital Clínic Barcelona; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute; University of Barcelona; Reveal Genomics; SOLTI Cancer Research Group, Barcelona, SPAIN, 18Translational Genomics and Targeted Therapies in Solid Tumors, Hospital Clinic Barcelona; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN.
Background: The HER2DX genomic test is a standardized, quantitative assay with demonstrated prognostic value in first-line advanced HER2+ breast cancer treated with trastuzumab, pertuzumab, and chemotherapy (THP). Here, we validated the test in an independent cohort (n=122) and explored its association with long-term outcomes in a combined real-world population (n=215). Methods: HER2DX testing was performed on baseline tumor tissue from 122 consecutive patients with advanced HER2+ breast cancer treated with first-line THP at the Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice (Poland). In addition, we evaluated a 93-patient real-world cohort from Spain (NPJ Breast Cancer 2025) with updated survival data in order to create a combined cohort. The assay was conducted at a central laboratory in Barcelona, blinded to clinical outcomes. Outcomes were assessed using pre-established ERBB2 mRNA score cutoffs. Multivariable Cox models were adjusted for clinicopathological variables. Prognostic value was also assessed in the subgroup of patients achieving a response (partial or complete) to first-line THP. An improved version of the test, called HER2DX metastatic prognostic score (HER2DX-mets-score), which combines the HER2DX ERBB2 score with additional molecular signatures, was trained in the Spanish cohort and validated in the Polish cohort. Results: In the Polish cohort (n=122), the mean age was 59.2 years (range: 36-85.7), 72.1% of patients presented with de novo metastatic disease, and 59% had hormone receptor-positive tumors. The overall response rate was 68.9%, the median progression-free survival (PFS) was 28.4 months (95% CI 17.9-42.9), and the median overall survival (OS) was 51.1 months (95% CI 43.8-87.6). Patients in the high ERBB2 mRNA group had significantly improved outcomes compared to those in the combined medium/low groups: PFS (hazard ratio [HR] 0.57; 95% CI 0.35-0.92; p=0.02) and OS (HR 0.48; 95% CI 0.28-0.83; p=0.009). In the combined cohort (n=215), the prognostic value of the ERBB2 score was confirmed. Patients in the high ERBB2 group had significantly longer PFS (median 33.8 months, 95% CI 25.6-52.5) and OS (median not reached) compared to those in the medium/low group (median PFS 12.5 months, 95% CI 9.79-25; median OS 37.1 months, 95% CI 25.4-47.9), with HRs of 0.50 (95% CI 0.35-0.71; p<0.001) for PFS and 0.36 (95% CI 0.23-0.54; p<0.001) for OS. The objective response rate (ORR) was significantly higher in the High HER2DX ERBB2 group (84.4%) compared to the Low group (52.0%; p <0.001). In multivariable models, the ERBB2 score remained independently prognostic. Among patients with a response to THP, those with high ERBB2 scores had a median PFS of 33.9 months (95% CI 26.9-61) and median OS not reached, compared to 17.6 months (95% CI 10.3-42.9) and 37.1 months (95% CI 25.4-51.1) in the medium/low group, respectively. Notably, in the subgroup of patients with fewer than three metastatic sites, those with high ERBB2 scores experienced a median PFS of 51.7 months (95% CI 28.6-NA) and a median OS that was not reached, compared to 20.3 months (95% CI 10.4-41.5) and 42.4 months (95% CI 27.6-55.8) in the medium/low group. HER2DX-mets-score outperformed the ERBB2 score alone in both cohorts; detailed results will be presented at the conference. Conclusions: The HER2DX ERBB2 mRNA score provides robust, independent prognostic information in patients treated with first-line THP for advanced HER2+ breast cancer. The test may help identify individuals who could be managed with chemotherapy followed by maintenance HP, while others may require treatment intensification. These findings suggest a potential role for the HER2DX ERBB2 score, and its improved version, HER2DX metastatic prognostic score, in informing more personalized treatment strategies.
Presentation numberPS5-01-24
Evorpacept (ALX-148) combined with trastuzumab deruxtecan in patients with HER2 positive/HER2low metastatic breast cancer (mBC): results from the PRE-ISPY phase Ib trial
PAULA R POHLMANN, University of Texas MD Anderson Cancer Center, Houston, TX
P. R. POHLMANN1, N. Chen2, J. A. Mouabbi3, N. Jahan4, M. Rampurwala5, A. Discacciati6, M. Eklund6, A. Chapple7, G. L. Hirst8, S. Venters9, L. Brown-Swigart9, E. Petricoin III10, A. D. Borowsky11, L. J. van ‘t Veer9, A. L. Delson12, S. M. Asare13, S. Jaffari14, P. Henderson15, D. Tripathy3, L. J. Esserman8, H. S. Rugo16, R. Nanda2, E. Stringer-Reasor4, D. Yee17, F. Meric-Bernstam18, D. A. Potter19; 1Breast Medical Oncology and Investigational Cancer Therapeutics (joint appointment), University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Medicine, University of Chicago, Chicago, IL, 3Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 4Division of Hematology and Oncology, University of Alabama at Birmingham O’Neal Comprehensive Cancer Center, Birmingham, AL, 5Hematology and Medical Oncology, UChicago Medicine Center for Advanced Care Orland Park, Orland Park, IL, 6Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SWEDEN, 7Statistics, Quantum Leap Healthcare Collaborative, San Francisco, CA, 8Department of Surgery, University of California San Francisco, San Francisco, CA, 9Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 10Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 11Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, 12Breast Science Advocacy Core, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 13Program Management, Quantum Leap Healthcare Collaborative, San Francisco, CA, 14QLHC, QLHC, San Francisco, CA, 15Collaborations, Quantum Leap Healthcare Collaborative, San Francisco, CA, 16Breast Medical Oncology, City of Hope Cancer Center, Duarte, CA, 17Departments of Medicine and Pharmacology (joint appointment), Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 18Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 19Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN.
BackgroundDespite advances in therapies such as trastuzumab deruxtecan (T-DXd) for HER2-expressing metastatic breast cancer, the disease remains incurable, underscoring a critical unmet need. Evorpacept (evo) is a high-affinity CD47-blocker with an inactive IgG Fc region designed to enhance antibody-dependent cellular phagocytosis. Here, we report results from a phase Ib trial of evo + T-DXd in patients (pts) with advanced HER2-expressing (HER2+/HER2low) breast cancer [NCT05868226].MethodsPRE-ISPY Phase I/Ib trial is an open-label platform study evaluating therapies in the advanced disease setting to identify promising regimens for rapid transition into I-SPY2, K-SPY or other trials. This multicenter study arm included 30 pts with previously treated, T-DXd naïve, advanced/metastatic HER2+/HER2low breast cancer. Part 1 dose finding contained 2 cohorts (A and B) of 10 patients each evaluating 2 doses of evo 30 mg/kg and 45 mg/kg combined with standard doses of T-DXd 5.4 mg/kg IV Q3W. Part 2 expansion (cohort C) enrolled additional 10 patients at RP2D. The primary objectives were to establish safety, Recommended Phase 2 Dose (RP2D) and antitumor activity (Overall Response Rate, ORR) in HER2+/HER2low mBC pts. PFS, Disease Control Rate (DCR), evo PK and ADA, CD47 levels, were secondary endpoints. ResultsAs of May 30th 2025, enrollment was complete with 30 pts (n=10 in each cohort A, B and C). Median follow-up was 6.6 months (4.6 in HER2+ vs 7.4 in HER2low), with 11 pts remaining on treatment at data cutoff. Median age (range) was 54 (30-76) yrs; 14 (47%), 8 (27%) and 2 pts (7%) had a history of liver, lung and brain metastasis, respectively; and 28 patients had measurable disease at enrollment. By local HER2 assessment, 7/10 (70%) pts in cohort A and 13/20 (65%) pts in cohorts B/C had HER2low mBC diagnosis, respectively. 13/30 (43%) of pts had 3 or more prior treatment lines in the advanced setting. There were no DLTs. The RP2D was evo 45 mg/kg and T-DXd 5.4 mg/kg IV Q3W. Among all 30 pts, treatment-emergent adverse events (TEAE) of any grade occurring in the most participants were nausea (73%), fatigue (67%), diarrhea (53%) and anemia (50%). Most of these events were Grade 1 or 2 in severity. Overall, 14 participants (47%) experienced Grade 3 events (anemia, hemolytic anemia, decreased counts of neutrophils, lymphocytes and platelets, dental caries, portal hypertension, fatigue, mastitis, back pain, hypokalemia, syncope, embolism, dyspnea, and pneumonia). Four pts experienced 8 total SAEs: grade 3 hemolytic anemia, grade 4 thrombocytopenia¸ grade 3 pneumonia, portal hypertension, embolism, mastitis, and grade 2 pyrexia, each event occurring once. All SAEs were resolved except for portal hypertension. There were no grade 5 TEAEs. One AE of special interest was described (grade 2 decreased left ventricular ejection fraction). There was one grade 1 ILD/pneumonitis. Dose reductions were made for 4 participants; one included a temporary dose hold. The confirmed ORR (cORR) was 33.3% (95% CI 17.9%-52.9%) and the DCR was 73.3% (95% CI 53.8%-87.0%). In HER2+ mBC pts, cORR was 40% (95% CI 13.7%-72.6%) and the DCR was 90% (95% CI 54.1%-99.4%). In the HER2low pts (n=20), cORR was 30% (95% CI 12.8%-54.3%) and DCR was 65% (95% CI: 40.9%-83.7%). Median progression free survival (mPFS) was 8.6 months, with a mPFS of 5.6 months in HER2low (13/20 events) and not yet reached in HER2+ (2/10 events). Updated data and correlatives will be provided at the Symposium. Conclusions In this phase I study, Evo + T-DXd demonstrated a manageable safety profile comparable with T-DXd alone and evidence of antitumor activity in pretreated HER2+/HER2low mBC. There were no new safety signals.
Presentation numberPS5-01-25
Long-term response Patterns in HER2-positive advanced breast cancer within the geicam/2014-03 (registem) study
Isabel Alvarez, Unidad de Cáncer de Guipúzcoa – Osakidetza; GEICAM Spanish Breast Cancer Group, San Sebastián, Spain
I. Alvarez1, Á. Guerrero-Zotano2, J. Cruz-Jurado3, S. Antolín4, E. Galve5, C. Rodríguez6, M. Hernández7, A. Tibau8, C. Falo9, J. Chacón10, A. Miguel11, Á. Rodríguez-Lescure12, E. Adrover13, M. Margelí-Vila14, R. Andrés15, S. Servitja16, M. Merino17, I. González18, J. Alonso-Romero19, R. Villanueva20, M. Echarri21, A. Antón-Torres22, S. Varela23, M. Ruíz-Borrego24, J. Guerra25, M. Corbellas26, M. Escudero27, S. Bezares27, F. Rojo28, S. López-Tarruella29; 1Medical Oncology, Unidad de Cáncer de Guipúzcoa – Osakidetza; GEICAM Spanish Breast Cancer Group, San Sebastián, SPAIN, 2Medical Oncology, Fundación Instituto Valenciano de Oncología (FIVO); GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 3Medical Oncology, Hospital Universitario de Canarias; GEICAM Spanish Breast Cancer Group, Santa Cruz de Tenerife, SPAIN, 4Medical Oncology, Complejo Hospitalario Universitario A Coruña (CHUAC); GEICAM Spanish Breast Cancer Group, A Coruña, SPAIN, 5Medical Oncology, Hospital Universitario de Basurto; GEICAM Spanish Breast Cancer Group, Bilbao, SPAIN, 6Medical Oncology, Hospital Universitario de Salamanca-IBSAL; GEICAM Spanish Breast Cancer Group, Salamanca, SPAIN, 7Medical Oncology, Hospitalario Universitario de Gran Canaria Dr. Negrín; GEICAM Spanish Breast Cancer Group, Las Palmas, SPAIN, 8Medical Oncology, Hospital de la Santa Creu i Sant Pau; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 9Medical Oncology, ICO Hospitalet; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 10Medical Oncology, Hospital Universitario de Toledo; GEICAM Spanish Breast Cancer Group, Toledo, SPAIN, 11Medical Oncology, ALTHAIA Xarxa Asistencial de Manresa; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 12Medical Oncology, Hospital General Universitario de Elche; GEICAM Spanish Breast Cancer Group, Elche, SPAIN, 13Medical Oncology, Hospital General Universitario de Albacete; GEICAM Spanish Breast Cancer Group, Albacete, SPAIN, 14Medical Oncology, ICO-Badalona. B-ARGO (Badalona Applied Reasearch Group in Oncology); GEICAM Spanish Breast Cancer Group, Badalona, SPAIN, 15Medical Oncology, Hospital Clinico Universitario Lozano Blesa; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 16Medical Oncology, Hospital del Mar-Hospital del Mar Research Institute ; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 17Medical Oncology, Hospital Universitario Infanta Sofía; GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 18Medical Oncology, Hospital Son Llátzer; GEICAM Spanish Breast Cancer Group, CR/ De Manacor, Km 4, SPAIN, 19Medical Oncology, Hospital Clínico-Universitario Virgen de la Arrixaca-IMIB; GEICAM Spanish Breast Cancer Group, Murcia, SPAIN, 20Medical Oncology, Hospital de Sant Joan Despi Moises Broggi; GEICAM Spanish Breast Cancer Group, Sant Joan Despí, SPAIN, 21Medical Oncology, Hospital Universitario Severo Ochoa; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 22Medical Oncology, Hospital Universitario Miguel Servet; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 23Medical Oncology, Hospital Universitario Lucus Augusti; GEICAM Spanish Breast Cancer Group, Lugo, SPAIN, 24Medical Oncology, Hospital Universitario Virgen del Rocío; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 25Medical Oncology, Hospital Universitario Fuenlabrada; GEICAM Spanish Breast Cancer Group, Fuenlabrada, SPAIN, 26Medical Oncology, Hospital Universitario Dr. Peset; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 27Medical Oncology, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 28Medical Oncology, Fundación Jiménez Díaz; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 29Medical Oncology, Hospital General Universitario Gregorio Marañón; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: Breast cancer (BC) is a heterogeneous disease with distinct subtypes, each having different prognosis and treatment responses. RegistEM aims to understand the distribution of subtypes in advanced BC (ABC) and their clinical implications. Significant advances have led to better median overall survival (OS) in patients (pts) with HER2+ BC. Targeted anti-HER2 therapies have changed the management and prognosis of HER2+ ABC, although there is a lack of data from long-term responders (LTRs).Methods: ABC pts, either recurrent early BC (EBC) or de novo metastatic (dnMBC), diagnosed from Jan-16 to Dec-19, were enrolled in this ambispective non-interventional cohort study. Biological samples from primary tumors, metastatic lesions, and blood were collected. This subanalysis aims to better characterize LTRs among pts with HER2+ ABC receiving 1st-line (1L) or 2nd-line (2L) anti-HER2 targeted therapies. We defined LTR as pts who did not progress to 1L therapy for more than 35 months (mo.), or for more than 16 mo. in 2L therapy. BC subtype was assessed by immunohistochemistry (IHC) with or without in situ hybridization (ISH) in the most recent tumor sample before 1L therapy. In the current analysis, 1,900 pts from 38 GEICAM sites were included (database ongoing, cut-off date Mar 17, 2025).Results: 341/1,900 (18%) pts had HER2+ ABC, 133(39%) were considered as LTR and 197 (58%) as non-LTR (NLTR), 11 pts were excluded due to lack of information or not receiving systemic treatment for ABC. At ABC diagnosis, median age was 56 years (interquartile range 48-69), all pts were female (70% postmenopausal), 68 (51%) LTR and 77 (39%) NLTR had dnMBC (p=0.0881). Hormone receptors positive (HR+) were observed in 92 (69%) LTR and 122 (62%) NLTR. HER2 status was confirmed by IHC 3+ in 89 (67%) LTR and 140 (71%) NLTR, and ISH amplified in 55 (41%) LTR and 81 (41%) NLTR. A different pattern of metastatic spread was observed between LTR and NLTR (p<0.01; χ2 test): visceral (58% vs. 76%), bone without visceral (27% vs. 16%), and only soft tissue (15% vs. 8%), respectively. Tumor burden with ≤2 locations was present in 57 (50%) LTR and 96 (49%) NLTR. Most frequent metastatic locations were lymph nodes (53%) and bone (51%), similar in both groups; more NLTR had liver (30% vs. 40%), lung (28% vs. 35%), pleura (2% vs. 11%), and brain (4% vs. 9%). Neo-/adjuvant therapy was given to 61 (95%) LTR and 112 (94%) NLTR with EBC, mainly chemotherapy (CT)/anti-HER2 (34% LTR vs. 38% NLTR), CT/endocrine therapy (ET)/anti-HER2 in 30% LTR, and CT in 22% NLTR, with 55 and 48 mo. as median time to ABC in LTR and NLTR, respectively. The most common 1L therapies in both groups were CT/biological therapy (BT) (33% LTR vs. 48% NLTR) and CT/ET/BT (50% LTR vs. 28% NLTR); BT was mainly anti-HER2 and antiangiogenics, and ET, aromatase inhibitors (AI). 2L-therapy was given to 57% LTR and 72% NLTR. Most common 2L therapies were BT (50% LTR and 59% NLTR), ET/BT (mainly AI plus anti-HER2) in LTR (29%), and CT/BT (anti-HER2 in 89%) in NLTR (20%). At database cut-off date and after a median follow-up of 46 mo, 1L median PFS (mPFS) was significantly longer in LTR patients: 50 mo. (95% confidence interval [CI]: 42-64) LTR compared to 12 mo. (95% CI: 10-13) in NLTR pts. No PFS event on 1L was seen in 52 (39%) LTR and 13 (7%) NLTR. 2L mPFS (95% CI) was 23 mo. (19-30) LTR vs. 5 (4-7) NLTR. Death was reported in 31/133 (23%) LTR and 149/197 (76%) NLTR, mainly because of PD (65% LTR vs. 77% NLTR). Median (95% CI) OS from ABC diagnosis was not reached in LTR and 34 mo. (31-38) NLTR.Conclusions: In our cohort, pts with HER2+ ABC classified as LTR more frequently had bone or soft tissue-only metastases compared to visceral disease. No significant differences were observed between LTR vs NLTR in terms of HR, HER2-IHC score (3+ vs. 2+), oligo- vs. non-oligometastatic presentation, or dnMBC vs recurrent EBC disease.
Presentation numberPS5-01-26
Long-term outcomes of first-line (1L), guideline-recommended treatment versus alternative regimens in HER2+ metastatic breast cancer (mBC): a retrospective observational study of US electronic health records
Maki Inoue-Choi, AstraZeneca, Gaithersburg, MD
M. Inoue-Choi1, B. Adeyemi2, J. Schmidt3, L. Luo4, A. Ali5; 1Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD, 2US Medical Affairs, Oncology Business Unit, AstraZeneca, Gaithersburg, MD, 3GMA/Payer Biometrics, Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 4Oncology Data & Analytics, AstraZeneca, Gaithersburg, MD, 5Department of Hematology and Oncology, Cleveland Clinic Foundation, Case Western Reserve University Cleveland, Cleveland, OH.
Background: The current guideline-recommended 1L standard of care (SOC) for HER2+ mBC is taxane, trastuzumab, and pertuzumab (THP) followed by maintenance therapy after initial response; however, many patients may not receive SOC in routine clinical practice. This study describes demographics, disease characteristics, and long-term treatment outcomes in US patients treated with SOC or other treatments for 1L HER2+ mBC. Methods: This retrospective observational study used the nationwide Flatiron Health database of derived and de-identified electronic health records. Patients aged ≥18 years with HER2+ mBC who received 1L treatment for mBC from January 2015 to February 2024 (excluding clinical trial drugs), with confirmatory HER2+ biomarker test results before initiation of 1L treatment, were included. Demographic and clinical characteristics were captured on the index date (start of 1L treatment). Patients were followed until death or last activity date on or before end of study (September 2024). SOC was defined as THP followed by maintenance trastuzumab/pertuzumab after response (with concurrent endocrine therapy for hormone receptor-positive [HR+] disease). Study outcomes included real-world time from initiation of 1L treatment to initiation of 3L treatment or death (rwTTNT2) and real-world time from initiation of 1L treatment to disease progression after initiation of 2L treatment or death (rwPFS2). Results: Overall, 3277 patients were included; 1L SOC was received by 35.5% (n=1164) of patients, with mean (standard deviation) age at index date of 58.7 (12.4) years, compared with 61.4 (14.1) years for other regimens (see Table for further characteristics). Of the 64.5% (n=2113) of patients who received other regimens, 17.1% (n=362) received HER2-targeted monotherapy and 36.8% (n=778) HER2-targeted combination therapy. Recurrent disease and HR+ status were observed in a greater proportion of patients treated with other regimens than with SOC. During follow up, 32.9% of patients treated with 1L SOC were observed to receive 3L treatment, 26.5% died, and 40.6% were censored before 3L. In patients receiving other regimens at 1L, these proportions were 37.1%, 33.3%, and 29.6%, respectively. Median rwTTNT2 (95% CI) was prolonged in patients treated with 1L SOC versus other regimens (30.1 [27.4, 33.6] vs 17.4 [16.1, 19.0] months). Median rwPFS2 (95% CI) was also prolonged in patients treated with 1L SOC (26.1 [24.2, 28.0] months) versus other regimens (18.9 [17.7, 19.9] months). Conclusion: Among patients with HER2+ mBC, receipt of guideline-recommended SOC (vs not) in the 1L setting was associated with substantially prolonged long-term outcomes. However, disease characteristics at treatment initiation may have influenced treatment regimen choice and impacted long-term outcomes.
| SOC (n=1164) | Other regimens (n=2113) | ||||
| Stage at initial breast cancer diagnosis, n (%) | |||||
| I-III (recurrent at index date) | 401 (34.5) | 1209 (57.2) | |||
| IV (de novo) | 714 (61.3) | 677 (32.0) | |||
| Unknown | 49 (4.2) | 227 (10.7) | |||
| Eastern Cooperative Oncology Group performance status ≤60 days before index date, n (%) | |||||
| 0-1 | 688 (59.1) | 968 (45.8) | |||
| 2 | 109 (9.4) | 172 (8.1) | |||
| 3-4 | 18 (1.5) | 63 (3.0) | |||
| Unknown | 349 (30.0) | 910 (43.1) | |||
| HR status, n (%) | |||||
| HR+ | 711 (61.1) | 1582 (74.9) | |||
| HR− | 433 (37.2) | 512 (24.2) | |||
| Unknown | 20 (1.7) | 19 (0.9) | |||
| Number of metastatic sites, n (%) | |||||
| 1 | 460 (39.5) | 727 (34.4) | |||
| 2-3 | 484 (41.6) | 651 (30.8) | |||
| ≥4 | 153 (13.1) | 158 (7.5) | |||
| Missing | 67 (5.8) | 577 (27.3) | |||
| Sites of metastasis, n (%) | |||||
| Bone | 664 (57.0) | 868 (41.1) | |||
| Brain | 88 (7.6) | 235 (11.1) | |||
| Liver | 472 (40.5) | 468 (22.1) | |||
| Lung | 386 (33.2) | 524 (24.8) |
Presentation numberPS5-01-27
Outcomes by hormone receptor (HR) status in patients (pts) with HER2+ advanced/metastatic breast cancer (mBC) with brain metastases (BM) treated with trastuzumab deruxtecan (T-DXd): a post-hoc subgroup analysis of DESTINY-Breast12
Hans Wildiers, University Hospitals Leuven, KU Leuven, Leuven, Belgium
H. Wildiers1, N. Harbeck2, E. Ciruelos3, G. Jerusalem4, V. Müller5, N. Niikura6, G. Viale7, R. Bartsch8, C. Kurzeder9, M. Higgins10, R. Connolly11, S. Baron-Hay12, M. Gión13, V. Guarneri14, G. Bianchini15, S. Escrivá-de-Romaní16, M. Prahladan17, S. Anand18, N. Toms17, R. Antony19, N. Lin20; 1Department of General Medical Oncology, University Hospitals Leuven, KU Leuven, Leuven, BELGIUM, 2Breast Center, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Munich, LMU University Hospital, Munich, GERMANY, 3Breast Cancer Unit, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 4Department of Medical Oncology, CHU Liège and Liège University, Liège, BELGIUM, 5Department of Gynecology and University Breast Center, University Medical Center Hamburg-Eppendorf, Hamburg, GERMANY, 6Department of Breast Oncology, Tokai University School of Medicine, Isehara City, Kanagawa Prefecture, JAPAN, 7Department of Pathology and Laboratory Medicine, IEO European Institute of Oncology IRCCS, Milan, ITALY, 8Division of Oncology, Department of Medicine 1, Medical University of Vienna, Vienna, AUSTRIA, 9Breast Center, University Hospital Basel, Basel, SWITZERLAND, 10UCD Cancer Trials Cluster, St. Vincent’s University Hospital, Dublin, IRELAND, 11Cancer Research @ UCC, University College Cork, Cork, IRELAND, 12Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, AUSTRALIA, 13IOB-Madrid, Beata María Ana Hospital, Madrid, SPAIN, 14Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, ITALY, 15Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, ITALY, 16Medical Oncology Department, Vall d’Hebron Institute of Oncology and Vall d’Hebron University Hospital, Barcelona, SPAIN, 17Global Medical Affairs, Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 18Biostatistics, Oncology R&D, AstraZeneca, Gaithersburg, MD, 19Global Medical Affairs, Oncology R&D, AstraZeneca, Gaithersburg, MD, 20Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: T-DXd demonstrated substantial and durable overall and intracranial clinical activity in DESTINY-Breast12. In pts with BM, 12-month progression-free survival (PFS) and central nervous system (CNS) PFS rates were 61.6% and 58.9%, respectively, and CNS objective response rate (ORR) was 71.7% (79.2% in pts with stable and 62.3% in pts with active BM). In the cohort of pts without BM, efficacy of T-DXd was in line with previous data, and there were no new safety signals. To date, T-DXd has shown clinical benefit irrespective of HR status, most recently in first-line HER2+ mBC in DESTINY-Breast09. In a post-hoc analysis of DESTINY-Breast12, outcomes by HR status in pts with BM were explored. Methods: Pts ≥18 years old with HR-positive (HR+) or HR-negative (HR−) HER2+ mBC whose disease had progressed after ≤2 previous lines of therapy were eligible; BM were required to be stable or active (untreated/treated progressing) and not in need of immediate local therapy. All pts received intravenous T-DXd 5.4 mg/kg every 3 weeks. Efficacy outcomes were analyzed by baseline HR status using the Kaplan-Meier technique for PFS. Results: In total, 262 pts with baseline BM were included in this analysis, of whom 165 (63.0%) had HR+ disease. Among pts with BM, the median age (range) was 52.0 (30-80) years in the HR+ subgroup and 54.0 (28-86) years in the HR− subgroup (n=97). Median follow up for PFS at data cutoff (February 8, 2024) was 13.1 months in both subgroups. Median PFS was not calculable for either HR subgroup. The 12-month PFS and CNS PFS, as well as CNS ORR, were indicative of consistent responses with T-DXd regardless of HR subgroup; confirmed ORRs, including in pts with measurable disease at baseline, indicated activity of T-DXd in pts with HR+ and HR− tumors (Table). The median total treatment duration (range) was 12.0 (0.7-26.9) months in the HR+ subgroup and 11.1 (0.1-26.8) months in the HR− subgroup. Adverse events of any grade in pts occurred with a similar overall incidence in both HR subgroups; the most common Grade ≥3 events were neutropenia (12.1%), anemia (7.3%), and fatigue (6.7%) in the HR+ subgroup, and neutropenia (11.3%), decreased neutrophil count (8.2%), and anemia (7.2%) in the HR− subgroup. Interstitial lung disease / pneumonitis occurred in 26 (Grade 5, n=3) and 16 (Grade 5, n=3) pts in the HR+ and HR− subgroups, respectively. Results by HR subgroup in the cohort without BM (data not presented) were consistent with those from the cohort with BM. Conclusion: T-DXd demonstrated consistent intracranial activity and safety in pts with BM, regardless of HR status. These data reinforce the utility of T-DXd as a second-line treatment regardless of HR status in pts with HER2+ mBC.
| HR+ (n=165) | HR− (n=97) | ||||
| PFS at 12 months, % (95% CI) | 64.2 (55.8, 71.5) | 57.7 (46.3, 67.6) | |||
| CNS PFS at 12 months, % (95% CI) | 60.1 (51.4, 67.8) | 56.8 (44.5, 67.4) | |||
| Confirmed ORR, % (95% CI) | 49.1 (41.5, 56.7) | 55.7 (45.8, 65.6) | |||
| In pts with measurable disease at baseline* | 60.8 (52.2, 69.4) | 69.4 (58.8, 80.1) | |||
| Confirmed CNS ORR, % (95% CI)† | 66.7 (56.6, 76.7) | 79.2 (68.3, 90.2) |
*n=125 (HR+) and n=72 (HR−); †based on n=84 (HR+) and n=53 (HR−) with measurable disease at baseline
Presentation numberPS5-01-28
Us real-world clinical outcomes of tucatinib, trastuzumab, and capecitabine following trastuzumab deruxtecan for the treatment of her2+ metastatic breast cancer
Jo Chien, UCSF, San Francisco, CA
J. Chien1, E. Neuberger2, S. Simon3, K. Watkins2, G. Curigliano4, B. Li2, S. Stergiopoulos2, M. Czachorowski5, H. Itakura6; 1Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA, 2Pfizer Inc., Pfizer Inc., New York, NY, 3Pfizer S.L.U., Pfizer S.L.U., Madrid, SPAIN, 4European Institute of Oncology, IRCCS, and Department of Oncology and Hemato-Oncology, University of Milano La Statale, Milano, ITALY, 5Pfizer Ltd., Pfizer Ltd., Surrey, UNITED KINGDOM, 6Medicine – Med/Oncology, Stanford University School of Medicine, Stanford, CA.
Background: US and EU treatment guidelines currently recommend tucatinib in combination with trastuzumab and capecitabine (TTC) in third-line (3L) for patients with human epidermal growth factor receptor 2-positive (HER2+) metastatic breast cancer (MBC), and in second-line (2L) for those patients with brain metastases (BM). More recently, trastuzumab deruxtecan (T-DXd) has become the recommended 2L treatment option and may be considered as first-line treatment for patients with rapid progression after (neo)adjuvant therapy. Previous real-world studies in patients receiving TTC after T-DXd have reported median time to next treatment (TTNT) of 6 months and median overall survival (OS) of 13 months; however, these study populations included later line treatment, a small sample size, and did not solely include TTC immediately following T-DXd, limiting applicability to current clinical practice. This study aimed to describe real-world outcomes in patients with HER2+ MBC in the US who were treated with TTC in 2L, 3L and fourth-line (4L) immediately following T-DXd. Methods: We conducted a retrospective cohort analysis of patients with HER2+ MBC receiving TTC therapy using the Flatiron Health electronic health record (EHR)-derived deidentified longitudinal database. This US nationwide database, containing patient-level structured and unstructured data curated using natural language processing with machine learning and technology-enabled abstraction, includes >720,000 patients with breast cancer. Patients included were diagnosed with HER2+ MBC (January 2017 to July 2024), treated with TTC in 2L, 3L or 4L immediately following T-DXd, and were not enrolled in clinical trials. Patients were assessed from the start of TTC (index date) to death, last medical activity, or end of available study follow-up (January 2025), whichever came first. Primary outcomes included, but were not limited to, TTNT and OS in all patients. Exploratory analyses will assess TTNT and OS in patients stratified by BM status, duration of T-DXd treatment, and hormone receptor status. Descriptive statistics were used for patient characteristics, and outcomes were evaluated using Kaplan-Meier analyses. Results: In total, 92 patients with HER2+ MBC received TTC immediately following T-DXd. Overall, median (95% CI) TTNT was 8.6 (5.7-11.6) months and median (95% CI) OS was 19.5 (12.0-26.1) months (Table). Exploratory outcomes will be presented in future analyses. Conclusion: Patients with HER2+ MBC benefit from TTC immediately after treatment with T-DXd, demonstrating its clinically meaningful activity in a contemporaneous post-T-DXd setting. These results reinforce the effectiveness of tucatinib, with longer TTNT and OS than in previous real-world studies, in a population more applicable to current clinical practice.
| Overall (N=92) |
2L (n=17) |
3L (n=46) |
4L (n=29) |
|
| Age at TTC initiation (years), median (IQR) | 58.0 (15.0) | 53.0 (14.0) | 58.0 (12.0) | 57.0 (16.0) |
| De novo at initial diagnosis, n (%) | 46 (50.0) | 2 (11.8) | 28 (60.9) | 16 (55.2) |
| Follow-up (months), median (IQR) | 7.7 (10.6) | 7.9 (8.9) | 7.6 (10.9) | 8.5 (11.2) |
| Median (95% CI) TTNT (months) | 8.6 (5.7-11.6) | 5.3 (3.3-12.2) | 8.6 (5.1-11.9) | 11.3 (4.5-22.6) |
| Median (95% CI) OS (months) | 19.5 (12.0-26.1) | 19.9 (10.0-not estimable) | 21.3 (14.7-31.6) | 11.9 (6.7-28.0) |
Presentation numberPS5-01-30
Trastuzumab deruxtecan (T-DXd) versus trastuzumab emtansine (T‑DM1) in patients (pts) with human epidermal growth factor receptor 2-positive (HER2+) metastatic breast cancer (mBC): Final analysis from DESTINY-Breast03
Seock-Ah Im, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea, Republic of
S. Im1, J. Cortes2, S. Kim3, G. Curigliano4, G. Buscacio de Sousa5, S. E. Nagai6, T. Sun7, J. Ignacio Delgado Mingorance8, P. Stephens9, V. Kaklamani10, F. Puglisi11, C. Levy12, H. Iwata13, S. A. Hurvitz14, S. Nakatani15, Z. Liang16, S. Ashfaque17, A. Egorov18, E. P. Hamilton19; 1Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Department of Oncology, International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Barcelona, SPAIN, 3Department of Oncology, Asan Medical Center, University of Ulsan, Seoul, KOREA, REPUBLIC OF, 4Department of Oncology and Hemato-Oncology, European Institute of Oncology, IRCCS, University of Milano, Milano, ITALY, 5Department of Breast Cancer, Instituto Americas, Rio de Janeiro, BRAZIL, 6Department of Breast Oncology, Saitama Cancer Center, Saitama, JAPAN, 7Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, CHINA, 8Servicio de Oncología Médica, Hospital Universitario de Badajoz, Badajoz, SPAIN, 9Exeter Oncology Centre, Royal Devon and Exeter Hospital, Exeter, UNITED KINGDOM, 10Department of Medicine, Breast Oncology Program, UT Health San Antonio, MD Anderson Cancer Center, San Antonio, TX, 11Department of Medicine (DMED), University of Udine, Department of Medical Oncology, National Cancer Institute, Centro di Riferimento Oncologico (CRO), Udine, Aviano, ITALY, 12Department of Medical Oncology, Centre François Baclesse, Caen, FRANCE, 13Department of Advanced Clinical Research and Development, Nagoya City University, Graduate School of Medical Sciences, Nagoya, JAPAN, 14Clinical Research Division (FHCC) and Division of Hematology/Oncology, Department of Medicine (UW), Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, 15Department of Clinical Science, Daiichi Sankyo Co., Ltd., Tokyo, JAPAN, 16Department of Biostatistics & Data Management, Daiichi Sankyo, Basking Ridge, NJ, 17Department of Clinical Safety, Daiichi Sankyo, Basking Ridge, NJ, 18Department of Global Oncology Clinical Development, Daiichi Sankyo Oncology France, Rueil-Malmaison, FRANCE, 19Department of Oncology, Sarah Cannon Research Institute, Nashville, TN.
Background In DESTINY-Breast03 (NCT03529110), T-DXd demonstrated a statistically significant and clinically meaningful improvement in progression-free survival (PFS) and overall survival (OS) over T-DM1 in pts with HER2+ mBC previously treated with trastuzumab and a taxane. Here, we report the 5-year follow-up analysis of efficacy and safety. Methods DESTINY-Breast03 is a randomized, multicenter, open-label, phase 3 study. Pts were randomly assigned (1:1) to receive either T-DXd 5.4 mg/kg or T-DM1 3.6 mg/kg intravenously every 3 weeks. Exploratory analyses (investigator assessed) at 5 years included OS, PFS, PFS2 (time from randomization to disease progression on the next line of therapy or death), confirmed objective response rate (cORR), duration of response (DoR), and safety. Results In all, 524 pts were randomly assigned (261 to T-DXd and 263 to T-DM1), with the last pt enrolled on June 23, 2020. As of the data cutoff (DCO), June 27, 2025, median (m; range) duration of follow-up was XX.X mo (XX.X-XX.X) for T-DXd and XX.X mo (XX.X-XX.X) for T-DM1; XX pts (XX.X%) in the T-DXd group and XX pts (XX.X%) in the T-DM1 group remained on treatment. The most common reasons pts discontinued study treatment were progressive disease (PD; [XX.X%]) and adverse events (AEs; [XX.X%]) with T-DXd, and PD (XX.X%) and AEs (XX.X%) with T-DM1. XX pts (XX.X%) in the T-DXd group and XX pts (XX.X%) in the T-DM1 group received subsequent therapy of physician’s choice. Median treatment duration (range) was XX.X mo (XX.X-XX.X) with T-DXd and XX.X mo (XX.X-XX.X) with T-DM1. mOS (95% CI) was XX.X mo (XX.X-XX.X) with T-DXd and XX.X mo (XX.X-XX.X) with T-DM1 (hazard ratio [HR], X.XX [95% CI, X.XX-X.XX], using a Cox regression model with treatment as the only covariate). The OS (95% CI) rate at 5 years was XX.X% (XX.X-XX.X%) with T-DXd vs XX.X% (XX.X-XX.X%) with T-DM1. mPFS (95% CI) was XX.X mo (XX.X-XX.X) with T-DXd vs XX.X mo (XX.X-XX.X) with T-DM1 (HR, X.XX [95% CI, X.XX-X.XX]). The PFS (95% CI) rate at 5 years was XX.X% (XX.X-XX.X%) with T-DXd vs XX.X% (XX.X-XX.X%) with T-DM1. mPFS2 (95% CI) was XX.X mo (XX.X-XX.X) with T-DXd vs XX.X mo (XX.X-XX.X) with T-DM1. Rates of treatment-emergent adverse events (TEAEs) were consistent with the prior DCO. Adjudicated drug-related interstitial lung disease (ILD)/pneumonitis occurred in XX.X% of pts treated with T-DXd (X new events [grade X] since last DCO) and XX.X% of pts with T-DM1 (X new events [grade X]), with no grade 4/5 events in either group. Updated data are shown in the Table. Conclusions The final analysis of DESTINY-Breast03 confirms the superior efficacy of T-DXd over T-DM1 in pts with HER2+ mBC whose disease had progressed following prior treatment with trastuzumab and a taxane. The safety profile of T-DXd remains manageable, with no new safety signals observed with longer follow-up.
| Efficacy |
T-DXd n = 261 |
T-DM1 n = 263 |
HR (95% CI) |
| mOS (95% CI), mo | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) |
| 5-year OS rate (95% CI), % | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | |
| mPFS (95% CI), mo | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) |
| 5-year PFS rate (95% CI), % | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | |
| mPFS2 (95% CI), mo | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) |
| cORR (95% CI), % | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | |
| mDoR (95% CI), mo | XX.X (XX.X-XX.X) | XX.X (XX.X-XX.X) | |
| Safety, n (%) |
T-DXd n = 257 |
T-DM1 n = 261 |
|
| Any-grade drug-related TEAEs | XXX (XX.X) | XXX (XX.X) | |
| Grade ≥3 drug-related TEAEs | XXX (XX.X) | XXX (XX.X) | |
| Drug-related serious TEAEs | XXX (XX.X) | XXX (XX.X) | |
| Drug-related TEAEs leading to permanent discontinuation | XXX (XX.X) | XXX (XX.X) | |
| Drug-related TEAEs leading to dose reduction | XXX (XX.X) | XXX (XX.X) | |
| Drug-related TEAEs leading to dose interruption | XXX (XX.X) | XXX (XX.X) | |
| Drug-related fatal TEAEs | XXX (XX.X) | XXX (XX.X) | |
| Adjudicated drug-related ILD/pneumonitis | XX (XX.X) | XX (XX.X) | |
| Grade 3 adjudicated drug-related ILD/pneumonitis | XX (XX.X) | XX (XX.X) |
Presentation numberPS5-02-01
Efficacy of cross-line therapy with anti-HER2 TKI in HER2-positive MBC: a multicenter real-world study
Ruixia Song, Chinese Academy of Medical Sciences and Peking Union Medical College – National Cancer Center, Cancer Hospital, Beijing, China
R. Song, Y. Fan, B. Xu, B. Lan, F. Ma; Medical Oncology, Chinese Academy of Medical Sciences and Peking Union Medical College – National Cancer Center, Cancer Hospital, Beijing, CHINA.
Objective: Human epidermal growth factor receptor (HER2)-targeted tyrosine kinase inhibitors (TKIs) have transformed the treatment landscape for HER2-positive metastatic breast cancer (MBC). Previous studies demonstrated the robust antitumor activity of TKIs in both first-line and later-line settings. Nevertheless, the benefits of maintaining TKI-based regimens after resistance remain uncertain. Based on real-world data, this study comprehensively assessed the clinical value of cross-line TKI application and investigated key factors influencing efficacy, aiming to provide insights for TKI administration in the advanced stage. Methods: This real-world study consecutively enrolled HER2-positive MBC patients receiving TKI-based regimens at 3 institutions (including the National Cancer Center, China) from October 2014 to April 2024. Inclusion criteria included patients who received at least two lines of TKI-based therapy in the advanced settings. Patients receiving cross-line TKI therapy not due to disease progression and with incomplete clinical records were excluded. The primary endpoint was progression-free survival (PFS). Results: A total of 132 patients with HER2-positive metastatic breast cancer who received two or more lines of TKI-based regimens were enrolled, with 49 receiving three lines and 19 receiving four lines of TKI therapy. The median age of all patients was 52 years. In patients receiving two lines of TKI therapy (N = 132), second-line PFS (PFS2) was significantly shorter than first-line (median PFS (mPFS): 5.5 vs. 9.0 months; hazard ratio (HR) 2.048; 95% confidence interval (CI) 1.588 – 2.640; P < 0.001). Multivariate Cox regression analysis identified PFS1 ≥ 6 months as an independent predictor of prolonged PFS2 (HR 0.492; 95% CI 0.333 – 0.728; P < 0.001). Among patients receiving TKI as the sole HER2-targeted agent (TKI with/without chemotherapy or endocrine therapy) (N = 70), the addition of chemotherapy in the second-line setting was associated with a significantly longer PFS2 compared to chemotherapy-free regimens (mPFS: 6.4 vs. 2.2 months; HR 0.404; 95% CI 0.208 – 0.787; P = 0.005). Similarly, patients who switched to a different TKI agent experienced a longer PFS2 than those who continued the original TKI regimen (mPFS: 6.8 vs. 4.5 months; HR 0.581; 95% CI 0.356 – 0.948; P = 0.025). Importantly, patients who switched from lapatinib to pyrotinib had a significantly longer PFS2 compared to those who continued pyrotinib in the second-line setting (mPFS: 7.0 vs. 4.1 months, HR 0.523, 95%CI 0.298 – 0.917, P < 0.001). Further analysis of patients receiving three lines of TKI therapy (N = 49) showed that PFS3 was shorter than PFS2 and PFS1 (mPFS: 4.1, 6.2, and 9.1 months, respectively; P < 0.001, P for trend < 0.0001). Notably, patients with high HER2 expression (immunohistochemistry 3+ vs. 2+ and fluorescence in situ hybridization positive, HR 0.282; 95% CI 0.117 – 0.680; P = 0.005) and PFS1 > 9.1 months (HR 0.549; 95% CI 0.306 – 0.988; P = 0.045) were associated with markedly longer PFS3. In the second- and third-line TKI settings, PFS did not significantly differ between regimens using TKI as the sole anti-HER2 agent and those combining TKI with another anti-HER2 agent (antibody or antibody-drug conjugate) (P > 0.05). Conclusions: This study indicated that cross-line anti-HER2 TKI therapy confers sustained PFS benefits in HER2-positive MBC. Notably, patients with longer frontline PFS and those who switched TKI agents exhibited superior survival outcomes in subsequent treatment lines. Further prospective studies are warranted to confirm these findings.
Presentation numberPS5-02-02
Early use of CDK4/6 inhibitor combined with endocrine therapy plus anti-HER2 antibody may get more survival benefit in HR+/HER2+ advanced breast cancer: result from a phase II single-arm mini cohort study (CABC016)
Yaxin Liu, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
Y. Liu, S. Guohong, R. Ran, J. Hanfang, S. Bin, X. Liang, Z. Jiayang, Z. Ruyan, L. Huiping; Department of Breast Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, CHINA.
Background: Based on anti-HER2 therapy, the combination of CDK4/6 inhibitors proved to be a potential regimen, which can simultaneously inhibit the crosstalk between the ER and HER2 pathways. PATINA study showed a meaningful prolonged PFS after a chemo-based induction therapy when added Palbociclib to the maintaining treatment, same kind of de-chemo treatment which also achieve better efficacy is still expected to be the initial regimen during metastatic setting.Methods: CABC016 is a phase II, single-arm, prospective cohort study (NCT05969184), assessing palbociclib combine with anti-HER2 antibody and endocrine therapy (ET) to treat HR+/HER2+ metastatic breast cancer (MBC). Patients are treated with palbociclib at a dose of 125 mg daily on days 1 – 21 of a 28 – day cycle, plus trastuzumab (H, trastuzumab or inetetamab) or trastuzumab plus pertuzumab (HP) intravenously on day 1 of each 21-day cycle. ET options included aromatase inhibitor (AI), tamoxifen or fulvestrant depend on endocrine sensitivety, with ovarian suppression required for premenopausal patients. The primary endpoint is investigator-assessed progression-free survival (PFS); key secondary endpoints were safety, response rate (ORR), disease control rate (DCR), and survival (OS).Result Analysis cutoff date is June 21, 2025. Enrolled 47 eligible MBC patients with a median age of 56 (32 – 74). 72.3% and 68.1% have >50% ER and >30% Ki – 67 expression. 72.3% patients had visceral and 17.0% had stable brain disease. At least one prior chemotherapy regimen was administered in 70.2% of patients, of whom 46.8% have been treated with taxane. The mPFS of ITT population is 16.5 m(95%CI, 11.3 – 21.9), with 19.3 m (95%CI, 15.5 – 22.5) of median follow – up. The mOS is 34.9 m(95%CI, 29.1 – 40.6), which is immature. Further analysis found that patients who were ET-naïve during metastatic setting had better mPFS (22.0 m[95%CI, 16.3 – 27.6], n = 26) than who had been treated by ET (8.3 m[95%CI, 3.2 – 13.5], n = 21) (P < 0.001). mPFS is longer in the front lines than late lines (mPFS of 1, 2 and 3 or more prior ET is 11.1 m[95%CI, 3.2 – 19.0; n = 12], 4.8 m[95%CI, 3.7 – 6.1; n = 7] and 1.5 m[95%CI, 0.3 – 2.7; n = 2], respectively (P < 0.001). At a median of 3 lines of this cohort, mPFS is better in 1st- line treatment (18.9 m[95%CI, 12.5 – 25.5; n = 17]) than in second/third – line (15.8 m[95%CI, 9.9 – 21.7; n = 18]), than fourth or later(up to 12 lines)treatment (7.7 m[95%CI, 1.5 – 13.9; n = 12]) (P = 0.008). In front line (line 1-3) patients, mPFS is even longer in ET-naïve patients (24.1 m[95%CI, 18.6 – 29.6], n = 23]) than those under ET treated (7.0 m[95%CI, 3.3 – 10.8], n = 12). No significant difference in mPFS was observed between the inetetamab (13.9 m[95%CI, 9.3 – 18.5; n = 26]) and trastuzumab (14.5 m[95%CI, 7.4 – 21.6; n = 21]) (P = 0.28), or H and HP (11.3month[95%CI, 6.0 – 16.5] vs. 18.1m[95%CI, 11.0 – 25.1; P = 0.418). The ORR of ITT population is 34.1%, included 1 CR, and DCR is 87.8%. While in ET-naïve setting, the ORR rise to 45.5%, and DCR rise to 95.5%. For patients with stable brain disease, the longest survival reached 34.6 months, no significant difference in mPFS was observed between patients with or without brain disease (P= 0.293), with a 50% ORR and 62.5% DCR, respectively. The overall safety profile was manageable. The most common grade 3-4 AEs were neutropenia (44.2%) and thrombocytopenia (20.9%). Dose reduction of palbociclib (from 125mg to 100mg) had occurred in 18 patients (38.3%) because of myelotoxicity. No patient fell out because of AEs.Conclusion: This mini cohort study showed palbociclib plus ET plus anti-HER2 antibody could be one of the optimal initial treatment options for metastatic setting of HR+/HER2+ breast cancer, especially for early use during ET-based regimen, even for stable brain disease.
Presentation numberPS5-02-03
Clinical Outcomes of Trastuzumab Emtansine (T-DM1) Following Trastuzumab Deruxtecan (T-DXd) in Metastatic Breast Cancer: A Single-Center Experience
Vicente Valero, MD ANDERSON CANCER CENTER, HOUSTON, TX
V. Valero, Z. WANG, R. BASSETT, R. K. MURTHY, P. POHLMANN, D. TRIPATHY, A. RAGHAVENDRA; BREAST MEDICAL ONCOLOGY, MD ANDERSON CANCER CENTER, HOUSTON, TX.
Clinical Outcomes of Trastuzumab Emtansine (T-DM1) Following Trastuzumab Deruxtecan (T-DXd) inMetastatic Breast Cancer: A Single-Center Experience Background: HER2-positive metastatic breast cancer (MBC) remains an oncological challenge afterdisease progression to pertuzumab-trastuzumab, and fam-trastuzumab deruxtecan. Data on treatmentoutcomes with T-DM1 following progression on trastuzumab deruxtecan (T-DXd) remain limited. This study evaluates the clinical outcomes of patients with HER2-positive MBC who received T-DM1 post-T-DXd. Methods: Demographics and clinical characteristics were summarized for the whole cohort who received T-DM1 after T-DXd. Continuous variables were summarized using medians and ranges, while categorical variables were summarized using frequencies and percentages. Overall survival (OS), defined as the time from the date of receiving T-DM1 to the date of death or last follow-up, progression-free survival (PFS),defined as the time from the date of receiving T-DM1 to the date of first documented disease progression or death, whichever occurred first, and time to progression (TTP), defined as the time from the date of receiving T-DM1 to the date of disease progression, were evaluated using the Kaplan-Meier method, and differences in survival curves were compared using the log-rank test. Cox proportional hazards regression models assessed associations between survival outcomes and clinical variables. The overall response rate was calculated based on the proportion of patients experiencing progressive or stable disease. A p-value <0.05 was considered significant. Analyses were performed using R version 4.2.2.Results: All 22 patients were female with a median age of 55.5 years. Median OS was 13.54 months (95% CI:8.74-NE), median PFS was 2.14 months (95% CI: 1.35-2.69), and median TTP was 2.20 months (95% CI: 1.38-2.69). HER2-low expression was associated with shorter OS (HR: 3.05), PFS (HR:4.30), and TTP (HR: 5.30). Overall response rate was 91%, with 68% showing progressive disease and 23% stable disease. No significant associations were identified between best response and other clinical variables. Conclusions: This study provides real-world evidence of the limited clinical efficacy of T-DM1 following resistant to T-DXd in HER2-positive MBC. Further research is warranted to develop novel therapeutic agents in this subgroup of patients and also to develop strategies to avoid resistant to T-DXd.Keywords: Fam-trastuzumab deruxtecan, Trastuzumab emtansine, metastatic breast cancer, outcomes, Antibody-drug conjugates, metastatic breast cancer
Presentation numberPS5-02-04
Incidence of the development of brain metastases in patients with advanced stage HER2+ breast cancer who have previously received a HER2 directed cancer vaccine.
Ying Liu, University of Washington, Seattle, WA
Y. Liu1, J. Bahia2, C. Haghighi1, S. Vinayak1, J. S. Childs1, M. L. Disis1; 1Cancer Vaccine Institute, University of Washington, Seattle, WA, 2Division of General Internal Medicine, University of Washington, Seattle, WA.
Incidence of the development of brain metastases in patients with advanced stage HER2+ breast cancer who have previously received a HER2 directed cancer vaccine. Ying Liu, Jaspreet Bahia, Candace B Haghighi, Shaveta Vinayak, Jennifer S. Childs, and Mary L. Disis. UW Cancer Vaccine Institute, University of Washington, Seattle WA Background: HER2-positive breast cancer, characterized by the overexpression of the HER2 protein, has been associated with an aggressive disease course, higher rates of recurrence, and poorer prognosis compared to HER2-negative breast cancer and accounts for approximately 15-20% of all breast cancer cases. The introduction of HER2-targeted therapies has transformed the treatment of HER2+ breast cancer and significantly improved survival rates. A major challenge for patients is the development of brain metastases, which can occur in up to 30–50% of patients with breast cancer. Patients with HER2+ breast cancer have a higher risk of developing brain metastases. Immune system cells, specifically T-cells, are one of the few modalities that are capable of crossing the blood brain barrier and may offer an immunologic approach to the prevention of brain metastases in high risk patients. We have been immunizing HER2+ breast cancer patients with HER2 directed vaccines for over 2 decades and sought to determine the impact of HER2-targeted cancer vaccines on the development of brain metastases compared to historical controls. In addition, we will explore the link between the clinical outcomes of patients and the level of immunity they achieved after HER2 specific immunization. Methods: We conducted a retrospective review of 146 patient medical records of advanced stage (III or IV) breast cancer patients, with a HER2+ breast cancer diagnosis confirmed by either IHC 3+ or FISH amplification, who had received a HER2 directed vaccine from one of four clinical trials conducted at the University of Washington, Cancer Vaccine Institute. Patients were followed for the development of brain metastases from the time of initiation of vaccination for at least 5 years. The diagnosis of brain metastases was based on surgical and pathological reports or radiology records and clinical notes from the medical record. IRB approval was obtained prior to initiation of the study. Results: Of the 146 patients with retrospective data, 12 patients had documented brain metastases prior to trial enrollment and were excluded from further evaluation. Of the remaining 134 patients who had no brain metastases at enrollment and subsequently received HER2-targeted vaccines, 21 developed brain metastases, yielding a cumulative incidence (CI) of 15.7%. Stratified by hormone receptor (HR) status, the incidence was 16.9% in HR+/HER2+ patients (n = 13) and 14.5% in HR-/HER2+ patients (n = 8). When compared to a recent historical control cohort of 18,075 real-world patients from the U.S. Flatiron Health database, which reported a 60-month CI of brain metastases of 22.7% in HR+/HER2+ and 33.6% in HR-/HER2+ subgroups, our findings suggest a 26% relative reduction of brain metastases in HR+/HER2+ patients and a 57% reduction in HR-/HER2+ patients. Conclusion: HER2 directed vaccination could have a protective effect on the development of brain metastases in patients with advanced stage HER2+ breast cancer. Phase II trials are planned to prospectively evaluate the vaccine in this setting.
Presentation numberPS5-02-06
Is genetic testing of Her2-negative metastatic breast cancer patients implemented into clinical practice?
Georg Pfeiler, Medical University of Vienna, Vienna, Austria
G. Pfeiler, F. Heinzl, C. Leser, D. Gschwantler-Kaulich, C. Singer, S. Kostic, A. Golescu, C. Deutschmann; Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, AUSTRIA.
BackgroundTimely genetic testing in HER2-negative metastatic breast cancer (mBC) patients is important to identify BRCA mutations and allow optimal treatment options including Poly(ADP-ribose)polymerase inhibitors (PARPi) and platinum salts. Yet, testing rates have been reported unsatisfactory. Evaluation of the implementation of genetic testing in a real-world setting is important to reveal and subsequently target potential inadequacies and risk factors that are associated with a low testing rate. MethodsWe performed a retrospective analysis including all HER2- mBC patients treated at the Department of Obstetrics and Gynecology of the Medical University of Vienna, Austria, starting from 10.4.2019 (date of EMA approval of Olaparib for gBRCAmut HER2- mBC). The primary objective of the study was to evaluate the rate of HR+/HER2- mBC patients treated at a single academic center in Austria that were recommended to undergo genetic testing by the treating physicians. The secondary objective of the study was to identify factors that were associated with a higher likelihood of having undergone genetic testing. We performed descriptive statistics as well as logistic regression analysis using R version 4.3.2. Results47.6% (109 of 229) of HER2- mBC patients and therefore with an indication for genetic testing had been recommended to undergo genetic testing by the treating physicians. Of these informed patients, 89.0% (97 of 109) underwent genetic testing, of which 11.6% (11 of 95) had a BRCA mutation and were eligible for targeted treatment. In multivariate analysis younger age (p-value: 0.0007), HR+/HER2- subtype (p-value: 0.0000) and positive family history for breast and ovarian cancer (p-value: 0.0001) were significantly associated with the conduct of genetic counseling.ConclusionThe present study showed low genetic counseling rates of HER2- mBC patients in a real-world academic setting especially in patients without specific risk factors for hereditary breast cancer. Informed patients showed a high willingness to undergo genetic testing. Genetic testing revealed targetable mutations in over 10% of tested patients and allowed additional treatment options.
Presentation numberPS5-02-07
Her2-overexpressing circulating tumor cells in patients treated by trastuzumab deruxtecan for metastatic her2<sup>+</sup> breast cancer: clinical and prognostic implications
William Jacot, Institut du Cancer de Montpellier, Montpellier, France
W. Jacot1, F. Bidard2, R. Colin-Chevalier3, S. Guiu1, S. Renault4, G. Lossaint5, J. Pierga2, L. Cayrefourcq6, F. Fiteni7, C. Alix-Panabières6; 1Department of Medical Oncology, Institut du Cancer de Montpellier, Montpellier, FRANCE, 2Department of Medical Oncology, Institut Curie, Paris and Saint Cloud, Paris, FRANCE, 3Department of Biometry, Institut du Cancer de Montpellier, Montpellier, FRANCE, 4Circulating Tumor Biomarkers Laboratory, Inserm CIC-BT 1428, Department of Translational Research, Institut Curie, Paris, FRANCE, 5Department of Clinical Research and Innovation, Institut du Cancer de Montpellier, Montpellier, FRANCE, 6Laboratoire Cellules Circulantes Rares Humaines, University Medical Centre of Montpellier, Montpellier, FRANCE, 7Department of Medical Oncology, University Medical Centre of Nimes, Nimes, FRANCE.
Background: Trastuzumab deruxtecan (T-DXd) has become the standard second-line treatment of HER2-overexpressing metastatic breast cancer (HER2+ MBC). However, to date, no reliable circulating biomarker has been identified to anticipate the resistance or sensitivity to this compound, and to better understand the underlying mechanisms of resistance / sensitivity. We evaluated in a prospective cohort the respective value of circulating tumor cell (CTC) levels, as well as CTC HER2 immunostaining and its heterogeneity, before and early during therapy as a tool to evaluate the prognosis of HER2+ MBC patients. Methods: We prospectively enrolled patients with HER2+ MBC receiving T-DXd across three French clinical centers (NCT04025541). Peripheral blood samples were collected at three timepoints: prior to treatment initiation (T1), before the second treatment cycle (T2), and at the time of confirmed disease progression (T3). CTCs were detected using the CellSearch® system. For each detected CTC, both enumeration and quantitative assessment of HER2 expression by immunofluorescence were performed. HER2 staining intensity and intra-patient heterogeneity were evaluated using the ACCEPT image analysis algorithm, enabling characterization of HER2-positive CTCs (HER2+ CTCs). Progression-free survival (PFS) was modeled based on longitudinal trajectories of HER2+ CTC levels using shared random effects joint models. Results: A total of 60 evaluable patients were enrolled between June 2021 and February 2025. As of the data cutoff (July 1, 2025), with a median follow-up of 17.2 months (95% CI: 11.6-21.7 months), 28 patients had experienced disease progression. CTCs were detectable at baseline (T1) in 20 patients (33.3%) and at the second timepoint (T2) in 4 patients (6.6%). No baseline clinical or pathological variables were found to be significantly associated with CTC positivity at T1. Neither the baseline presence of CTCs (with a median PFS of 13.6 months [7.5 – 25.4] vs. 17.1 [8.9 – NA], p=0.4, for the CTC+ (≥1 detected cell) and CTC- populations, respectively) nor the longitudinal evolution model were associated with PFS. HER2+ CTCs were detected in 7 patients (11.6%). Among CTC-positive cases, no statistically significant correlation was observed between the number of CTCs and the presence of HER2+ CTCs. Notably, HER2+ CTCs were exclusively identified in patients whose tumor tissue exhibited HER2 3+ expression. While not significant (p=0.2), the HER2+ CTC population presented a PFS twice longer compared to the HER2+ CTC-negative population (25.3 [8.1 – NA] vs. 13.6 months [8.7 – 21.7], respectively, HR=0.48), even if the HER2+ CTC population was more heavily pretreated (median of 3 previous cytotoxic treatments in the MBC setting compared to 1 for the HER2+ CTC-negative group, p=0.01). The longitudinal evolution model was not associated with PFS. Conclusions: Detection of HER2+ CTCs appears to be a rare event in HER2+ MBC. However, their presence may help identify a subset of patients who derive greater benefit from T-DXd. Larger studies and evaluation of additional circulating biomarkers, such as ctDNA, are warranted.
Presentation numberPS5-02-08
Impact of Prior T-DM1 Exposure on the Efficacy of Trastuzumab Deruxtecan in HER2-Positive Metastatic Breast Cancer
Seyda Gunduz, Koc University School of Medicine, Istanbul, Turkey
F. Kemik1, T. K. Sahin2, H. Ozcelik3, A. Sezer4, G. Basaran5, S. Biter6, D. Erdem7, S. Tunbekici8, A. Oruc9, A. K. Guren10, K. Kaban11, M. B. Aykan12, O. B. Ekinci13, I. Deliktas Onur14, A. Kalem15, O. Altunok16, M. Seyyar17, B. Guney18, O. Akdogan19, M. Yazici20, N. Majidova21, B. Koylu1, C. I. Kikili1, N. Demir1, D. Tunali1, S. Lacin1, D. Tural1, A. Bilici3, E. Bayram6, E. Goker8, M. Araz9, I. V. Bayoglu10, N. Molinas Mandel11, N. Karadurmus12, E. Celik13, O. Ates14, H. Yesil Cinkir15, M. Yimaz16, R. U. Gursu18, O. Yazici19, D. Cabuk20, D. C. Guven2, F. Selcukbiricik1, S. Aksoy2, S. Gunduz1; 1Department of Medical Oncology, Koc University School of Medicine, Istanbul, TURKEY, 2Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, TURKEY, 3Department of Medical Oncology, Istanbul Medipol University, Istanbul, TURKEY, 4Department of Medical Oncology, Başkent University, Adana, TURKEY, 5Medical Oncology Department, Faculty of Medicine, Acıbadem University, Istanbul, TURKEY, 6Department of Medical Oncology, Faculty of Medicine, Çukurova University, Adana, TURKEY, 7Department of Medical Oncology, Samsun Medical Park Hospital, Samsun, TURKEY, 8Department of Medical Oncology, Ege University, Izmir, TURKEY, 9Department of Medical Oncology, Necmettin Erbakan University School of Medicine, Konya, TURKEY, 10Department of Medical Oncology, Marmara University Faculty of Medicine, Istanbul, TURKEY, 11Department of Medical Oncology, American Hospital, Istanbul, TURKEY, 12Department of Medical Oncology, Health Science University, Gulhane School of Medicine, Ankara, TURKEY, 13Department of Medical Oncology, Prof. Dr. Cemil Taşcıoğlu City Hospital, University of Health Sciences, Istanbul, TURKEY, 14Department of Medical Oncology, Health Sciences University Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Ankara, TURKEY, 15Department of Medical Oncology, Gaziantep University, Gaziantep, TURKEY, 16Department of Medical Oncology, Bakirkoy Dr Sadi Konuk Training and Research Hospital, Istanbul, TURKEY, 17Department of Medical Oncology, Gaziantep City Hospital, Gaziantep, TURKEY, 18Department of Medical Oncology, Istanbul Training and Research Hospital, Istanbul, Istanbul, TURKEY, 19Department of Medical Oncology, Gazi University School of Medicine, Ankara, TURKEY, 20Department of Medical Oncology, Kocaeli University, Kocaeli, TURKEY, 21Department of Medical Oncology, VM Medical Park Maltepe Hospital, Istanbul, TURKEY.
Introduction: Trastuzumab deruxtecan (T-DXd) has emerged as a highly effective antibody-drug conjugate for patients with HER2-positive metastatic breast cancer, particularly following progression on prior HER2-directed therapies. However, the clinical relevance of prior exposure to another HER2-targeted ADC, trastuzumab emtansine (T-DM1), on the efficacy of subsequent T-DXd remains unclear. Clarifying this sequencing effect is essential for optimizing treatment strategies in the advanced disease setting. Methods: This multicenter, retrospective cohort study evaluated patients with HER2-positive metastatic breast cancer who received T-DXd therapy. The primary objective was to assess the impact of prior exposure to another antibody-drug conjugate, T-DM1, on progression-free survival (PFS) following T-DXd treatment. Eligible patients were identified from institutional records across 21 oncology centers in Turkey, and clinical, pathological, and treatment data were collected. PFS was defined as the time from initiation of T-DXd to radiologic or clinical disease progression or last follow-up. Cox proportional hazards regression was used to estimate hazard ratios for progression. Results: A total of 199 patients were included, with a mean age of 45.8 ± 12.0 years (range: 21-83). T-DXd was administered at a median line of 3 (range: 1st-9th line). The majority of patients received T-DXd as a third (29.2%) or fourth-line therapy (25.6%). The median follow-up was 8.3 months, and the mean follow-up was 10.5 months (95% CI: approximately 9.5-11.5). Comorbidities were present in 31.2% of patients, and 57.3% were premenopausal. ER and PR positivity rates were 65.3% and 47.7%, respectively. HER2 status was IHC 3+ in 75.9% and IHC 2+/ISH+ in 24.1%. Prior to T-DXd, 67.3% of patients received T-DM1. Disease progression after T-DXd occurred more frequently in patients previously treated with T-DM1 (47.0% vs. 13.8%, p<0.001). In the univariate analysis, prior exposure to T-DM1 was significantly associated with a higher risk of progression under T-DXd treatment (HR = 3.03, 95% CI: 1.51-6.10, p = 0.002). Consistent with this finding, progression-free survival (PFS) was significantly shorter in the T-DM1-pretreated group compared to the T-DM1-naive group (p = 0.001, log-rank test). The median PFS was 12.1 months (95% CI: 7.7-16.5) in the T-DM1 group, whereas the median was not reached in the T-DM1-naive group during the study period. Conclusion: Prior exposure to T-DM1 was associated with significantly shorter progression-free survival and a higher risk of disease progression following T-DXd treatment in patients with HER2-positive metastatic breast cancer. These findings highlight the potential impact of treatment sequencing on T-DXd efficacy and underscore the need for personalized therapeutic strategies in the advanced setting.
|
Variable |
Category | n (%) |
| Age Group | <65 / ≥65 | 182 (91.5) / 17 (8.5) |
| Menopausal Status | Premenopausal / Postmenopausal | 114 (57.3) / 85 (42.7) |
| ECOG Performance Status | 0 / 1 / ≥2 | 136 (68.3) / 61 (30.7) / 2 (1) |
| Comorbidities | Yes / No | 62 (31.2) / 137 (68.8) |
| Histological Subtype | IDC / ILC / Other | 177 (88.9) / 9 (4.5) / 10 (5) |
| ER Status | Positive / Negative | 130 (65.3) / 69 (34.7) |
| PR Status | Positive / Negative | 95(47.7) / 104 (52.3) |
| HER2 IHC Score | IHC 3+ / IHC 2+ ISH+ | 151 (75.9) / 48 (24.1) |
| Number of Metastatic Sites | 1 / ≥2 | 104 (52.3) / 95 (47.7) |
| Visceral Metastasis | Yes / No | 159 (79.9) / 40 (20.1) |
| Prior T-DM1 exposure | Yes / No | 134 (67.3) / 65 (32.7) |
| Line of T-DXd therapy | 1st–2nd / 3rd–4th / 5th or later | 51 (26.2) / 107 (54.8) / 37 (19.0) |
Presentation numberPS5-02-09
Unconjugated MMAE Pharmacokinetics in Combination with Tucatinib in Patients with HER2 Expressing Locally Advanced or Metastatic Breast and Gastric Cancer
Andrew SyBing, Pfizer, San Diego, CA
A. SyBing1, E. Garcia2, J. Meyer3, M. Miller4, X. Li4, V. Kumar5, A. Topletz-Erickson1; 1Clinical Pharmacology & Translational Sciences, Pfizer, San Diego, CA, 2Pharmacometrics & Systems Pharmacology, Pfizer, San Diego, CA, 3Clinical Bioanalytics, Pfizer, San Diego, CA, 4Late Stage Clinical Development – Breast, Oncology, Pfizer, San Diego, CA, 5Pharmacokinetics, Dynamics, and Metabolism, Pfizer, San Diego, CA.
Background: Disitamab vedotin (DV) is a human epidermal growth factor receptor 2 (HER2) targeting antibody drug conjugate (ADC) with a monomethyl auristatin E (MMAE) payload via a maleimidocaproyl-valine-citrulline (MC-VC) linker. MMAE is primarily metabolized in vitro by cytochrome P450 3A4 (CYP3A4) and is a substrate of P-glycoprotein (P-gp). A clinical drug-drug interaction (DDI) study previously showed that ketoconazole, a strong CYP3A4 and P-gp inhibitor, increased unconjugated MMAE exposure 1.34-fold following brentuximab vedotin (BV) treatment in patients with CD30+ hematologic malignancies1. Thus, current recommendations for concomitant use of MMAE-conjugated ADCs and a strong CYP3A4 inhibitor are limited to close monitoring for adverse events. However, it’s unclear whether a similar trend of increased exposure holds true for other MMAE-conjugated ADCs. In this study, unconjugated MMAE PK following DV administration was characterized with and without tucatinib, a strong CYP3A4 inhibitor and a known P-gp inhibitor currently approved for previously treated HER2 expressing metastatic breast cancer, to assess the impact of tucatinib-mediated CYP3A4 and P-gp inhibition on unconjugated MMAE PK. Methods: C5731004/SGNDV-004 (NCT06157892) is a phase 1b/2 open-label, multicenter study designed to identify the maximum tolerated dose (MTD) and/or optimal dose of DV when administered in combination with tucatinib in participants with HER2+ (IHC 3+ or IHC 2+/ISH+) and HER2-low (IHC 1+ or IHC 2+/ISH-) expressing locally advanced or metastatic breast cancer (LA/mBC) and locally advanced or metastatic gastric cancer and gastroesophageal junction adenocarcinoma (LA/mGC/GEJC). Participants in the dose escalation phase of SGNDV-004 received DV (1.25 mg/kg or 1.50 mg/kg IV Q2W) starting Cycle 1 Day 1 and tucatinib (TUC, 300 mg PO BID) starting Cycle 1 Day 8. Unconjugated MMAE PK blood samples were collected on Days 1, 2, 3, and 8 in Cycle 1 (DV alone) and Cycle 2 (DV+TUC). Unconjugated MMAE plasma concentrations were analyzed using validated high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). The DDI assessment was made using unconjugated MMAE PK parameters following DV administration including plasma area under the concentration-time curve up to Day 8 (AUC0-168h) and maximal concentrations (Cmax) in Cycle 2 (test) relative to Cycle 1 (reference) and were calculated using PKNCA (R v4.3.1, PKNCA v0.10.2). Results: PK data were available for 6 patients at 1.25 mg/kg and 3 patients at 1.50 mg/kg. The geometric mean (geometric CV%) of Cycle 1 and Cycle 2 dose-normalized MMAE AUC0-168h values were 232.2 (83%) and 275.7 (101%) ng*hr/mL/(mg/kg), respectively. The geometric mean (geometric CV%) of Cycle 1 and Cycle 2 dose-normalized MMAE Cmax values were 1.87 (88%) and 2.40 (94%) ng/mL/(mg/kg), respectively. Geometric mean ratios (CI90%) for Cycle 2/Cycle 1 MMAE AUC0-168h and Cmax were 1.18 (1.05-1.35) and 1.28 (1.17-1.40), respectively. Conclusions: Unconjugated MMAE exposure following DV administration of 1.25 mg/kg or 1.5 mg/kg Q2W was increased <1.3-fold in the presence of tucatinib in participants with HER2 expressing LA/mBC or LA/mGC/GEJC, translating to an overall weak DDI impact. References1.Han TH, Gopal AK, Ramchandren R, Goy A, Chen R, Matous JV, Cooper M, Grove LE, Alley SC, Lynch CM, O’Connor OA. CYP3A-mediated drug-drug interaction potential and excretion of brentuximab vedotin, an antibody-drug conjugate, in patients with CD30-positive hematologic malignancies. J Clin Pharmacol. 2013 Aug;53(8):866-77. doi: 10.1002/jcph.116. Epub 2013 Jun 10. PMID: 23754575; PMCID: PMC3777854.
Presentation numberPS5-02-10
Pyrotinib plus capecitabine with or withoutradiotherapyin patients with HER2-positive metastatic breast cancer and brain metastases: Amulticenter, retrospective-prospective, real-world study in China (Post-PERMEATE)
Min Yan, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
L. Niu1, X. Liang2, S. Wang3, S. Zhang4, Z. Lv5, H. Li6, Y. Zhao7, Y. Yin8, Y. Song9, G. Sun10, B. Zhao11, L. Cai12, Q. Ouyang13, C. Geng14, H. Yao15, X. Wu16, Z. Chen17, Z. Jiang18, H. Li2, M. Yan1; 1Department of Breast Disease, Henan Breast Cancer Centre, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, CHINA, 2Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, CHINA, 3Department of Medical Oncology, State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 4Department of Breast Cancer, Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA, 5Cancer Center, The First Hospital of Jilin University, Changchun, CHINA, 6Department of Breast Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, CHINA, 7Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, CHINA, 8Department of Oncolog, Jiangsu Province Hospital, Nanjing, CHINA, 9Breast Center B Ward, The Affiliated Hospital of Qingdao University, Qingdao, CHINA, 10Department of Breast and Thyroid Surgery, Affiliated Tumor Hospital of Xinjiang Medical University, Xinjiang, CHINA, 11Department of Breast Cancer, The Clinical Research Center of Breast Tumor and Thyroid Tumor in Xinjiang Uygur Autonomous Region, Affiliated Tumor Hospital of Xinjiang Medical University, Xinjiang, CHINA, 12Department of Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, CHINA, 13Department of Breast Medicine, Hunan Cancer Hospital,The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, CHINA, 14Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, CHINA, 15Department of Medical Oncology, Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA, 16Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, Wuhan, CHINA, 17Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, CHINA, 18Department of Breast Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA.
Background The combination of pyrotinib and capecitabine has shown promising efficacy in patients with HER2-positive metastatic breast cancer and brain metastases (BMs). However, it remains uncertain whether the timing of local radiotherapy affect overall survival (OS) on the basis of this systemic regimen. The purpose of our study is to investigate the impact on OS of the addition of brain radiotherapy to the treatment with pyrotinib and capecitabine in patients with HER2-positive breast cancer BMs in a real-world clinical practice setting. MethodsThis is a multicenter, retrospective-prospective, real-world study conducted across 41 centers in China (NCT05359120). Patients diagnosed with HER2-positive breast cancer and BMs, with complete diagnostic and treatment records available in hospital electronic medical record systems were eligible for this study. Enrolled patients were categorized into three cohorts based on the radiotherapy status while undergoing systemic treatment: those who were radiotherapy-naïve (Cohort A), those who received whole-brain radiotherapy (WBRT) and/or stereotactic radiosurgery (SRS) within three months of initiating systemic treatment (Cohort B), and those who received systemic treatment more than three months after radiotherapy (Cohort C). Systemic treatment was defined as pyrotinib (no less than 240 mg once daily) combined with capecitabine (no less than 50% of 1000 mg/m² twice daily on days 1-14 of each 21-day cycle) until disease progression or unacceptable toxicity. The endpoints included OS and progression-free survival(PFS). Here we present the results of Cohort A and B. ResultsFrom September 2018 to January 2024, a total of 287 female patients were enrolled in the study. In Cohort A, 27.4% (37/135) of patients presented with symptomatic BMs, while 52.5% (64/122) in Cohort B. Additionally, a greater percentage of patients in Cohort A (88.9%) had active BMs compared to those in Cohort B (61.5%).As of May 6, 2025, the median follow-up duration was 35.9 months (95% CI: 34.3-37.5) in Cohort A and 45.3 months (95% CI: 41.9-48.6) in Cohort B. The median OS was 35.4 months (95% CI: 31.4-39.4) in Cohort A and 37.3 months (95% CI: 27.4-47.2) in Cohort B, with no statistically significant difference between the two groups (p = 0.793). Similarly, the median progression-free survival (PFS) was comparable between Cohort A and Cohort B, at 15.0 months (95% CI: 11.7-18.3) and 15.5 months (95% CI: 12.1-18.9), respectively (p = 0.073). ConclusionTo our knowledge, this is the largest study of pyrotinib plus capecitabine in HER2-positive breast cancer patients with BMs. No significant difference was observed in OS or PFS between the two groups: priority systemic therapy with pyrotinib plus capecitabine until disease progression or unacceptable toxicity, or systemic therapy concurrently with radiotherapy. Therefore, future studies should further elucidate the characteristics of patients most likely to benefit from specific systemic-local treatment combinations and enabling personalized therapeutic strategies for HER2-positive breast cancer patients with BMs. These findings warrant prospective clinical investigation. Research Sponsor: None.
Presentation numberPS5-02-11
Staying Ahead of Resistance: ctDNA-Guided Camizestrant Switching in HR+ Breast Cancer (SERENA-6)
Chiugo Okoye, Northeast Georgia Medical Center, Gainesville, GA
C. Okoye1, C. Emeasoba2, O. Ntukidem3, L. Odion-Omonhimin4; 1Internal Medicine, Northeast Georgia Medical Center, Gainesville, GA, 2Internal Medicine, UAMS Northwest, Fayeteville, AR, 3Internal Medicine, Trinity Health Ann Arbor, Ypsilanti, MI, 4Internal Medicine, Southern Regional Medical Center, Riverdale, GA.
BackgroundESR1 (estrogen receptor) mutations are a common mechanism of acquired resistance to first-line aromatase inhibitor (AI) therapy in HR-positive, HER2-negative advanced breast cancer. These mutations often emerge during combined AI plus CDK4/6 inhibitor treatment, making tumors insensitive to estrogen deprivation. The SERENA-6 trial is built on the rationale that serial plasma ctDNA monitoring can detect emergent ESR1 mutations before radiographic progression. Early identification of an ESR1 mutation allows preemptive switching from the AI to the novel oral SERD camizestrant (while continuing the same CDK4/6 inhibitor) to target resistant clones. The goal is to extend control of ER-driven tumor growth and delay the need for chemotherapy.MethodsSERENA-6 (NCT04964934) is a global, double-blind, randomized phase III trial in postmenopausal women with HR+/HER2- advanced breast cancer on first-line AI+CDK4/6i therapy. Patients underwent serial plasma ctDNA testing (every 2-3 months) during stable disease. When a new ESR1 mutation was detected (before any clinical progression), 315 patients were randomized 1:1 to either continue the AI (plus placebo) or switch to camizestrant 75 mg once daily (plus placebo AI), with all patients remaining on their original CDK4/6 inhibitor. The primary endpoint was investigator-assessed progression-free survival (PFS). Key secondary endpoints included overall survival, time to second progression event (PFS2), chemotherapy-free survival, patient-reported outcomes (including quality of life), and safety.ResultsAt the planned analysis, median PFS was significantly longer in the camizestrant arm than in the control arm (16.0 vs. 9.2 months; HR = 0.44; 95% CI 0.31-0.60; P < 0.0001). This corresponds to a 56% relative reduction in risk of progression or death. The 1-year and 2-year PFS rates were markedly higher with camizestrant (60.7% and 29.7%) versus continued AI (33.4% and 5.4%). Patients who switched to camizestrant also experienced a clinically meaningful delay in symptom deterioration: the median time to clinically significant worsening of quality-of-life was 23.0 months vs 6.4 months for those who remained on AI. Camizestrant was well tolerated; adverse events were consistent with its known profile. Very few patients required treatment discontinuation, and no new safety signals emerged. A mild ocular effect, photopsia, was noted in some patients, but it did not affect vision or daily function.ConclusionThe SERENA-6 findings demonstrate that real-time molecular profiling with ctDNA to guide early therapeutic switching can substantially improve outcomes in endocrine-resistant HR+ breast cancer. Preemptive replacement of the AI with camizestrant upon molecular progression more than doubled median PFS, extended quality-adjusted survival, and deferred progression by targeting ESR1-mutant clones. These results support a new paradigm of personalized endocrine therapy: intervening at the first molecular sign of resistance to prolong endocrine benefit and delay chemotherapy.
Presentation numberPS5-02-12
Intracranial Activity and Systemic Efficacy of Trastuzumab Deruxtecan in Breast Cancer Patients with Brain Metastases
Zouina Sarfraz, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
Z. Sarfraz1, V. Podder2, K. Vazquez3, F. Akkoc Mustafayev1, M. Jaramillo1, K. A. Qidwai1, M. Ganiyani1, V. Andion Camargo1, R. Mahtani4, M. Ahluwalia1; 1Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 2Gynecologic Oncology, Mount Sinai Medical Center, Miami, FL, 3Research, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 4Breast Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Trastuzumab deruxtecan (T-DXd), a novel antibody-drug conjugate (ADC), has shown promising intracranial efficacy in breast cancer (BC) patients with brain metastases (BM). This meta-analysis evaluates the intracranial activity and systemic efficacy of T-DXd in patients with metastatic BC and BM. Methods: A systematic search was conducted from inception until May 6, 2025, across PubMed, Cochrane Library, and oncology conference abstracts (ASCO, ESMO, SABCS, and EANO). Eligible studies included clinical trials and cohort studies reporting outcomes for BC patients with BM receiving T-DXd. Random-effects meta-analysis models with Freeman-Tukey transformations were used for pooling proportions (intracranial clinical benefit rate [IC-CBR], intracranial objective response rate [IC-ORR], and 12-month systemic overall survival [OS]). Systemic progression-free survival (PFS) was pooled using a random-effects inverse-variance model. For PFS, means and standard deviations (SDs) were extracted when available or estimated from medians and IQRs using the Wan et al. (2014) method: mean = (Q1 + median + Q3)/3 and SD = (Q3 − Q1)/1.35, adjusted for sample size. Heterogeneity was assessed using the I² statistic, with values ≤25% considered low, 26-75% moderate, and >75% high. Registration: osf.io/swghm. Results: Of the 489 screened records, 14 studies were included; 1 was a randomized controlled trial, 6 were prospective single-arm trials, and 7 were retrospective cohort studies, including real-world data from early access programs. Median patient age was 56.3 years; 97.3% had HER2-positive BC, and 2.7% HER2-low. Prior cranial radiotherapy was administered in 61%, with concurrent radiotherapy at T-DXd initiation in 1.4%. Median follow-up duration was 12.9 months (IQR: 11-15). T-DXd was administered uniformly at 5.4 mg/kg IV every three weeks, with protocol-specified dose reductions allowed. Pooled IC-CBR was 81% (95% CI: 69-91%, p<0.001), indicating strong intracranial clinical efficacy, with low heterogeneity (I²=18.6%). IC-ORR was 62% (95% CI: 51-74%, p<0.001), suggestive of substantial intracranial tumor responses. Moderate heterogeneity was noted (I²=71.8%). The pooled mean systemic PFS was 12.6 months (95% CI: 7.1-18.1, I²=99.8%, p<0.001). Pooled 12-month OS was 81% (95% CI: 66-93%, p<0.001), highlighting favorable survival outcomes despite high heterogeneity (I²=86.5%). Conclusion: T-DXd demonstrates robust intracranial activity and 12-month survival in breast cancer patients with brain metastases. The pooled IC-CBR of 81% notably exceeds historical benchmarks reported with T-DM1 or chemotherapy, which typically range from 30-50% in this population. Prospective studies are needed to refine patient selection and optimize therapeutic sequencing.
Presentation numberPS5-02-13
Journeying Through the Spectrum of HER2 Expression Breast Cancer Clinic: Education Outcomes on Guiding Treatment Decisions
Tariqa Ackbarali, Medlive, Lake Worth, MA
T. Ackbarali1, K. Jones2, A. Bardia3, S. Hurvitz4; 1Hematology/Oncology Team, Medlive, Lake Worth, MA, 2Hematology/Oncology Team, Medlive, Needham, MA, 3Division of Hematology and Oncology, University of California, Los Angeles, Santa Monica, CA, 4Division of Hematology and Oncology, Fred Hutchinson Cancer Center, Bellevue, WA.
BACKGROUND: A significant change in practice has occurred in breast cancer based on an improved understanding of HER2 expression existing on a broader spectrum than previously known. Despite the availability of personalized treatments for patients with metastatic breast cancer (mBC) based on their HER2-expression status, many patients who express HER2 are not receiving the optimal, tailored treatment for their disease. This is largely due to a lack of clarity surrounding HER2 testing, integration of HER2-targeted therapies across all subtypes of breast cancer, and management of unique adverse events (AEs) associated with novel HER2-targeting therapies. An interactive, case clinic was designed for oncology clinicians to engage in applying the latest guideline-recommended treatment approaches for mBC to close these gaps and ensure patients receive optimal therapy. METHODS: The branched-learning curriculum was comprised of two 30-minute tracks evaluating best practices in the management of HER2-positive and HER2-low breast cancer. The education launched on Medlive.com in September 2024 and will remain available for 12 months. Following a pre-assessment, learners selected their initial preferred track, which included several concise and easy-to-digest modules framed by clinical cases. Faculty experts provided rationales for each decision-point option before moving on to the next module. Discussions ensued on factors influencing treatment selection, guidelines and clinical evidence, treatment-sequencing strategies, and strategies for adverse-event management. Pre-post assessment data through case decisions for HER2+ (track 1) vs HER2-low MBC (track 2)will be presented. RESULTS: To date, more than 1,300 clinicians have engaged with the education program, 74% of whom were MD/DO and 81% designating their specialty as oncology. Given a choice of selecting tracks, 91% initiated the education in track 1 and 89% of all learners completed both tracks. An approximate 16% improvement was seen across all knowledge and competence questions for intended learners (pathology and oncology speciaties) with higher gains – upwards of 50% – demonstrated for confidence treating HER2-low MBC vs HER2+ MBC. The reported intended changes in clinical practice included: • Greater personalized treatment sequencing based on patient-specific characteristics (36%) • Higher frequency of use of novel HER2-targeted agents in 2nd-line therapy (29%) • Implementing stricter monitoring for AEs associated with HER2-targeted therapies (27%) • Rigorous testing for HER2-low expression in patients with MBC (25%) • Increasing referrals for clinical trials of novel HER2-targeted agents (20%) Pre-test variability in responses indicated that some learners were unclear on when additional testing is warranted, likely due to inconsistent application of updated HER2 testing protocols in clinical practice. CONCLUSIONS: The post-test gains indicate an improved ability to differentiate between available ADCs, with greater recognition of novel therapies as the preferred next-line therapy in HER2-low disease due to its targeted mechanism against HER2, even at low levels. However, the persistent gap suggests an opportunity to reinforce how HER2 expression levels influence ADC selection and where other ADCs may fit into later-line treatment. Improvements were seen in testing protocols, treatment application, and AE management, for relevant populations likely to benefit. These outcomes demonstrated the impact of this education initiative focusing on point-of-care decisions. Support was provided by an independent educational grant from Daiichi Sankyo, Inc. and AstraZeneca Pharmaceuticals.
Presentation numberPS5-02-14
“longterm” – long-term response (>18 months) in metastatic her2-positive breast cancer: a retrospective analysis of patients diagnosed between 2010 and 2020 in the german bngo network
Georg Erich Wilhelm Heinrich, Practice for gynecological oncology, Fürstenwalde, Germany
G. E. Heinrich; Gynocological Oncology, Practice for gynecological oncology, Fürstenwalde, GERMANY.
“LONGTERM” – Long-Term Response (>18 Months) in Metastatic HER2-Positive Breast Cancer: A Retrospective Analysis of Patients Diagnosed Between 2010 and 2020 in the German BNGO NetworkHeinrich G., Nuti P., Busch S., Olbermann A., Ruhmland B., Urban H., Lenk I., Kreiss-Sender J., Lorenz R., Baerens D.-T., Förster F., Wierick E., Großmann C., Wolf C., Altmann F., Uleer C., Müller A., Massinger-Biebl H., Apel K., Schubert R., Oskay-Özcelik G., Fiege J., Hielscher C., Steinfeld-Birg D.BackgroundProgression-free survival (PFS) and overall survival (OS) have significantly improved in patients with HER2-positive metastatic breast cancer (mBC) due to advances in targeted therapies. The current standard first-line treatment includes dual antibody therapy with trastuzumab and pertuzumab combined with chemotherapy, as established in the CLEOPATRA trial. This real-world data (RWD) analysis, conducted by the German Association of Private Practice-Based and Outpatient Gynecologic Oncologists (BNGO), retrospectively examined patients with a PFS >18 months.MethodsThis multicenter, retrospective, non-interventional RWD analysis included patients newly diagnosed with HER2-positive metastatic or inoperable breast cancer between January 1, 2010, and July 31, 2019. Data collection occurred between March 1, 2022, and December 31, 2023, using existing site documentation. Data were pseudonymized and entered into standardized electronic case report forms (eCRFs) via a common electronic data capture (EDC) system. Patients were included if they had HER2 IHC 3+ and/or ISH positivity and a PFS of at least 18 months following first-line therapy.Inclusion Criteria– First diagnosis of metastatic or inoperable disease between 01.01.2010 and 31.07.2020- HER2 IHC 3+ and/or ISH-positive at first metastatic diagnosis or in the primary tumor if unknown in metastases- PFS >18 months, assessed clinically or radiologically (RECIST 1.1)- Receipt of anti-HER2 therapy as first-line treatmentExclusion Criteria– No documented HER2+ status in metastases or primary tumor- Disease progression within 18 months of diagnosisResultsOf 111 patients from 24 sites, 95 were eligible. Median age at diagnosis was 61.1 years. Of these, 58.9% had prior early-stage HER2+ disease, 33.7% were de novo metastatic, and 7.4% showed HER2 receptor conversion. ER and PR positivity were recorded in 72.6% and 58.9% of patients, respectively. Table 1 summarizes the distribution of metastatic sites at diagnosis.Chemotherapy combined with anti-HER2 therapy was administered in 74.7% of cases, with trastuzumab used in 98.9% and pertuzumab in 66.3%. Endocrine therapy was given in 63.2% of patients, often sequentially. At the end of follow-up, 69.5% of patients remained under therapy, 14.7% had died, and 2.1% were lost to follow-up.ConclusionThis RWD analysis provides insight into the evolving treatment patterns and outcomes of long-term responders with HER2-positive metastatic breast cancer. All patients received anti-HER2 therapy, most commonly the trastuzumab-pertuzumab combination. Hormone receptor-positive patients often received sequential endocrine therapy. The findings reflect adherence to international guidelines (ASCO, NCCN, AGO) and highlight the role of national networks such as BNGO in generating meaningful real-world oncology data.
Presentation numberPS5-02-15
Concurrent Trastuzumab Deruxtecan and Radiation Therapy in HER2- positive and HER2-low Metastatic Breast Cancer: Assessing the efficacy
Jihane BOUZIANE, CHU HASSAN II , Fez, Morocco, Fez, Morocco
J. BOUZIANE1, P. Loap2, S. Allali2, L. Escalup3, J. Pierga4, Y. Kirova2; 1Radiation Oncology, CHU HASSAN II , Fez, Morocco, Fez, MOROCCO, 2Radiation Oncology, Institut Curie, Paris, Paris, FRANCE, 3Pharmacy, Institut Curie, Paris, Paris, FRANCE, 4Medical Oncology, Institut Curie, Paris, Paris, FRANCE.
Background: Metastatic breast cancer, particularly HER2-positive disease, remains a therapeutic challenge and is often associated with poor prognosis. While anti-HER2 therapies such as trastuzumab have improved outcomes, trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate, has shown promising efficacy even in heavily pretreated patients. Clinical trials, including DESTINY-Breast, have demonstrated significant improvements in progression-free survival (PFS) and overall survival (OS). However, the safety and efficacy of combining T-DXd with radiotherapy (RT) have yet to be fully evaluated. This study aims to assess the outcomes of this combination in patients with HER2-positive and HER2-low metastatic breast cancer.Methods: We conducted a retrospective study including patients treated between November 2020 and January 2024. Patients with HER2-positive and HER2-low metastatic breast cancer who received concurrent trastuzumab der uxtecan and radiotherapy were identified. Data on patient demographics, treatment regimens, radiation doses, toxicity profiles, efficacy and treatment discontinuations were collected. Data on tumor response were collected through imaging examinations, and follow-up was conducted from the last day of radiotherapy until death or the last examination, and toxicities were graded using CTCAE V5.0. Results: The studied population includes 33 patients with HER2-positive and HER2-low metastatic breast cancer who underwent concurrent treatment with trastuzumab deruxtecan and radiotherapy. The median follow-up was 14 months. Treatment details indicated that trastuzumab deruxtecan was administered at the recommended dose across various treatment modalities. Of the patients evaluated, 39.4% achieved partial remis sion, while 9.1% attained complete remission. Additionally, 39.4% experienced stable disease, and 12.1% faced disease progression necessitating a change in therapy. Safety assessment revealed that acute toxicities were mainly associated with systemic treatment. Survival analysis showed 11 deaths (33.3%) during the follow-up period, with a median overall survival of 26 months and median progression-free survival of 12 months.Conclusion: The combination of trastuzumab deruxtecan with radiotherapy in HER2-positive and HER2-low metastatic breast cancer demonstrates promising efficacy with a manageable safety profile. Further studies are warranted to fully elucidate the potential synergistic effects of this treatment regimen and its impact on patient outcomes.
Presentation numberPS5-02-16
Clinical outcomes and treatment attrition rates of HER2 positive metastatic breast cancer (MBC) patients at a tertiary referral cancer centre in London: the Guy’s Cancer experience.
Haixi Yan, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
H. Yan, M. Toki, H. Kristeleit; Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, UNITED KINGDOM.
Background: Treatment options for HER2 positive MBC are increasing and trastuzumab deruxtecan (T-DXd) is now used in earlier lines of treatment, having shown superiority to trastuzumab emtansine (T-DM1) in DESTINY-Breast03 and to THP in DESTINY-Breast09. Uncertainty remains on the optimal sequencing of these drugs and their impact on OS with concerns raised regarding the generalisability of reported progression free survival (PFS) and treatment attrition rates in these trials. In this series, we report treatment attrition rates and PFS for patients starting 1st line chemotherapy between 2019 and 2024. Methods: Electronic health records were searched for patients with HER2 positive advanced breast cancer starting 1st line treatment between 01/01/19 and 31/12/24. Dates to calculate PFS and OS as well as attrition rates were extracted for each line of therapy. Results: 143 women commenced treatment during the study period. Median age was 51.6 years at time of first breast cancer diagnosis and 55.2 years at time of metastatic disease. 82 patients had de-novo metastatic disease, invasive disease-free survival in the remainder was 46.6 months. Observed median PFS for THP (23.0 months) appeared comparable to the THP arms of CLEOPATRA (18.7 months)1 and DESTINY-Breast09 (26.9 months)2. T-DM1 (9.0 months) performed similarly to the EMILIA (9.6 months) study3 and better than the control arm of DESTINY-Breast03 (6.8, CI 5.6-8.2 months)4. T-DXd was used in 11 patients in the 2nd line setting and 16 patients in the 3rd line setting and in this small series performed considerably worse (11.7 months) than in DESTINY-Breast03 (28.8 months)4 and DESTINY-Breast02 (17.8 months)5, but was comparable to other real-world experience (12.3 months)6. With median follow-up of 27.6 months, 44 patients have died, and median OS has not been reached. 15 (34%) events were due to CNS progression and two (7%) patients treated with T-DXd experienced grade 5 pulmonary toxicity. Attrition rates mirrored published findings.7 In subgroup analysis, ER+ patients had a longer median PFS than ER- although this was not statistically significant (26.1 vs 19.9 months, p = 0.26). Notably, patients with HER2 2+ (ISH+) had shorter median PFS compared to those with HER2 3+ (10.5 vs 27.9 months, p = 0.0089). Conclusion: Guy’s Cancer serves one of the most ethnically diverse populations in the world. In this retrospective real-world study, 1st and 2nd line PFS was similar to published phase 3 studies. Although T-DXd appeared to perform better than T-DM1 in our population, it fell short of the outcomes seen in the DESTINY studies. However, due to the small number of patients treated with T-DXd and the short follow-up our confidence intervals are wide. High attrition rates and death from CNS disease in 1/3 of patients supports the earlier use of T-DXd in this population.
Presentation numberPS5-02-18
Soluble E-cadherin-STAT3/NFκB axis in inflammatory breast cancer progression
Kiros Haddish Tesfamariam, MD Anderson Cancer Center, Houston, TX
K. H. Tesfamariam, X. Hu, I. Longa Rizzo, E. S Villodre, L. TH Phi, J. Song, S. Krishnamurthy, Y. Gong, W. Priebe, W. A. Woodward, B. G. Debeb; Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX.
sEcad-STAT3/NFκB axis in inflammatory breast cancer progression Kiros H. Tesfamariam1,6,*, Xiaoding Hu1,6,*, Isabella Longa Rizzo1,6, Emilly S Villodre1,6, Lan TH Phi1,6, Juhee Song2, Savitri Krishnamurthy3,6, Yun Gong3, Waldemar Priebe4, Wendy A Woodward5,6, Bisrat G. Debeb1,6 Departments of 1Breast Medical Oncology, 2Biostatistics, 3Pathology, 4Experimental Therapeutics, 5Breast Radiation Oncology, and 6MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research Program, The University of Texas MD Anderson Cancer Center, Houston TX. (* Equal contributors) Abstract Background: Inflammatory breast cancer (IBC) is a rare and aggressive subtype of breast cancer, accounting for 2-5% of all cases. It is characterized by rapid progression and a high propensity for distant metastasis, including to the brain. We showed that elevated serum levels of soluble E-cadherin (sEcad), an 80 kDa ectodomain fragment, are independently correlated with poor survival and increased incidence of brain metastasis in IBC patients. sEcad overexpression also promoted brain metastasis growth in IBC xenograft models. RNA-seq analyses revealed activation of STAT3 and NFκB signaling in sEcad-overexpressing cells. Given the interaction between sEcad and Protein Disulfide Isomerase A4 (PDIA4), we hypothesize that sEcad promotes metastasis in IBC through PDIA4-dependent activation of STAT3 and NF-κB signaling pathways. Methods: Using lentiviral transduction, we stably overexpressed sEcad in the IBC cell lines SUM149 and MDA-IBC3. Protein expression and phosphorylation states of STAT3 and NFκB were validated by immunoblotting. Functional assays, including colony formation, migration, and invasion, were conducted to assess the impact of sEcad overexpression and PDIA4 knockdown using shRNA constructs. Pharmacological inhibition studies with STAT3 and NFκB pathway inhibitors (WP1066 and ATL) were conducted to evaluate pathway dependencies. To test the in vivo efficacy of these inhibitors on brain metastasis, SUM149-sEcad cells were injected via the intracardiac route into SCID/Beige mice. Molecular docking simulations using the Glowworm Swarm Optimization algorithm for protein-protein interactions, as well as a deep learning model based on diffusion algorithms for protein-ligand interactions, were used to assess binding interactions among sEcad, PDIA4, STAT3, and the inhibitors. Results: Overexpression of sEcad in SUM149 and MDA-IBC3 cells significantly increased phosphorylation of STAT3 and NFκB, and enhanced proliferation, colony formation, migration, and invasion. Knockdown of PDIA4 in sEcad-overexpressing cells reversed these effects and reduced pSTAT3 and pNFκB levels, highlighting PDIA4’s critical role in mediating sEcad-driven signaling. Both STAT3 and NFκB pathway inhibitors, either alone or in combination, reduced cell proliferation and colony formation in sEcad-high cells in a dose- and time-dependent manner. Moreover, treatment with brain-permeable inhibitors of STAT3 and NFκB reduced metastatic burden in an IBC brain metastasis model (p=0.03). Finally, molecular docking studies confirmed energetically favorable interactions between sEcad and PDIA4 (swarm 110; score: 38.6), as well as effective binding of PDIA4 with ATL (RMSD: 1.8 Å) and WP1066 (RMSD: 1.3 Å). Conclusion: Our findings demonstrate that sEcad is a key driver of aggressive oncogenic features in IBC through PDIA4-dependent activation of STAT3 and NFκB pathways. Targeting the sEcad-STAT3/NFκB signaling axis represents a promising therapeutic strategy for IBC.
Presentation numberPS5-02-19
Impact of HER2 kinase domain mutations on trastuzumab deruxtecan efficacy in HER2 low metastatic breast cancer
Yoshiya Horimoto, Tokyo Medical University, Tokyo, Japan
Y. Horimoto1, M. Oshi2, A. Yamada2, Y. Ueki3, R. Semba3, R. Wu1, H. Kusama1, T. Kawate1, F. Murakami3, T. Ishikawa1, G. Kutomi3, J. Watanabe3; 1Breast Surgery and Oncology, Tokyo Medical University, Tokyo, JAPAN, 2Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 3Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, JAPAN.
PURPOSE: In metastatic breast cancer (MBC) treatment, HER2 expression is a key factor in determining the indication for trastuzumab deruxtecan (T-DXd). While HER2 mutations have been identified as therapeutic biomarkers for T-DXd in several solid tumors, their clinical relevance in breast cancer has received little attention. Consequently, the impact of HER2 mutations on the efficacy of T-DXd in MBC remains unclear. METHODS: We retrospectively examined 25 Japanese women with HER2-low MBC treated with T-DXd from January 2022 to October 2024. Tumor tissue from primary or metastatic sites was analyzed for four HER2 kinase domain mutations (L755S, D769Y, V777L, and V842I) using droplet digital PCR. These mutations were selected for their potential functional relevance in solid tumors based on recent studies. Treatment efficacy was assessed by time to treatment discontinuation (TTD), and its association with each mutation was examined. Subgroup analysis was performed based on the number of mutations (single vs. multiple). RESULTS: Clinicopathological characteristics are summarized in the Table. The frequencies of the four HER2 kinase domain mutations were as follows: L755S (64%), D769Y (32%), V777L (8%), and V842I (92%), with notable variation by mutation site. Tumors harboring D769Y tended to have a higher Ki67 labeling index (p = 0.027), suggesting increased proliferation activity. Patients with the L755S mutation had a shorter median TTD than those without the mutation (32 vs. 41 weeks), suggesting a potential association with reduced sensitivity to T-DXd. No apparent differences in TTD were observed for D769Y, V777L, or V842I mutations. Patients with multiple (≥ 2) mutations had a significantly shorter median TTD than those with a single mutation (30 vs. 52 weeks). CONCLUSION: Specific HER2 kinase domain mutations may influence the response to T-DXd in HER2-low MBC. This is the first study to report the frequencies of these four mutations and to assess mutation-specific differences in treatment duration using real-world data.
| Variables | [range] / (%) | |
| Age (mean, year) | 59.0 [46-77] | |
| Hormone receptor status | Positive | 22 (88%) |
| Negative | 3 (12%) | |
| Ki67 labeling index (mean, %) | > 20% | 12 (48%) |
| ≤ 20% | 12 (48%) | |
| Unknown | 1 (4%) | |
| Visceral metastases | Yes | 20 (80%) |
| No | 5 (20%) | |
| Prior chemotherapy regimens (mean) | 2.8 [0-8] |
Presentation numberPS5-02-20
Treatment patterns and outcomes of sacituzumab govitecan in older versus younger patients with mTNBC: multinational real-world data
Milos Holanek, Masaryk Memorial Cancer Institute, and Faculty of Medicine, Masaryk University, Brno, Czech Republic
A. Konieczna1, M. Holanek2, M. Pieniazek3, J. Zubrowska4, A. Polakiewicz-Gilowska5, R. Soumarova6, H. Studentova7, A. Mlodzinska1, K. Winsko-Szczesnowicz8, M. Lisik-Habib9, A. Pekala9, D. Krejci10, J. Sustr11, I. Kolarova12, I. Kolouskova2, I. Danielewicz13, M. Szymanik-Resko13, T. Ciszewski14, L. Rusinova15, B. Czartoryska-Arlukowicz8, A. Lacko3, M. Jarzab5, R. Pacholczak-Madej16, Z. Bielcikova17, M. Malejcikova18, M. Puskulluoglu19; 1Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology-National Research Institute, Warszawa, POLAND, 2Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, and Faculty of Medicine, Masaryk University, Brno, CZECH REPUBLIC, 3Department of Oncology, Breast Unit Clinical Oncology Day Care Department, Wroclaw Medical University, Lower Silesian Comprehensive Cancer Center, Wroclaw, POLAND, 4Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, POLAND, 5Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 6Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 7Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 8Department of Clinical Oncology, M. Sklodowska-Curie Bialystok Oncology Center, Bialystok, POLAND, 9Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 10Department of Oncology, First Faculty of Medicine, Charles University in Prague, and Bulovka University Hospital, Prague, CZECH REPUBLIC, 11Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University, and University Hospital Pilsen, Plzen, CZECH REPUBLIC, 12Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Kralove, CZECH REPUBLIC, 13Oddzial Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 14Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 15Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 16Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, POLAND, 17Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 18II. Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 19Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, POLAND.
Background: Patients over 65 years constitute a substantial subset of those diagnosed with metastatic triple-negative breast cancer (mTNBC), yet are frequently underrepresented in clinical trials due to comorbidities, frailty, and polypharmacy. Sacituzumab govitecan (SG), approved for pretreated mTNBC, demonstrated comparable efficacy and tolerability in this age group in the ASCENT trial. However, real-world data remain limited. This multinational study investigates the safety and effectiveness of SG in patients aged >65 years treated in routine clinical practice. Materials and Methods: We retrospectively analyzed clinical data from female patients receiving SG across 18 oncology centers in Slovakia, Poland, and the Czech Republic between August 2021 and May 2025. Patients were stratified by age at SG initiation (≤65 vs. >65 years). Baseline characteristics, treatment patterns, and adverse events (graded according to CTCAE v5.0) were compared. Clinical information was extracted retrospectively from routine medical records. The analysis included progression-free survival (PFS; time from SG initiation to progression or death), overall survival (OS; time from SG initiation to death from any cause), objective response rate (ORR), and disease control rate (DCR) as clinical endpoints. Tumor responses were evaluated according to RECIST 1.1. Univariate Cox regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). P < 0.05 was considered statistically significant. Results: Among 303 patients included, 76 (25.1%) were >65 years. Only 10 of them (13.2%) underwent geriatric assessment as part of routine clinical practice. Older patients had received a higher number of prior palliative systemic therapies (median 2 [IQR 1-2] vs. 1 [IQR 1-2], p=0.039). Visceral metastases (77.5% vs. 60.5%, p=0.006) and central nervous system (CNS) metastases (11.5% vs. 2.6%, p=0.038) were more frequent in the younger group. A reduced starting dose of SG (≤8 mg/kg at cycle 1 day 1) was used in 13.2% of older and 7.5% of younger patients (p=0.204). Adverse event-related dose reductions occurred in 46.1% of individuals >65 and 34.8% ≤65 (p=0.106). Delays in SG administration were observed in 71.1% and 60.4%, respectively (p=0.125). Grade ≥3 neutropenia occurred in 42.1% vs. 45.8% (p=0.733). No other significant differences in toxicity were detected. Median number of full SG cycles was similar across age groups (7 [IQR 3-12] vs. 6 [IQR 3-10], p=0.240). ORR was 30.3% vs. 30.8% (p>0.999), and DCR was 72.4% vs. 61.7% (p=0.122). Survival analyses demonstrated that patients >65 years achieved significantly longer PFS and OS compared to younger individuals (median PFS: 5.42 vs. 4.07 months; HR=0.716; 95% CI: 0.534-0.960; p=0.026; and median OS: 12.81 vs. 10.91 months; HR=0.691; 95% CI: 0.485-0.985; p=0.041, respectively). Conclusions: Older women with mTNBC treated with SG in routine clinical practice achieved significantly longer PFS and OS than younger patients, despite similar treatment exposure, toxicity, and response rates. These findings challenge the assumption that age alone predicts poorer outcomes and support SG use in selected older individuals. However, only 13.2% of patients >65 years underwent formal geriatric assessment, suggesting a possible selection bias toward clinically fit individuals. The observed survival advantage may also reflect lower rates of CNS and visceral metastases or less aggressive tumor biology. Our findings emphasize the need for individualized treatment decisions based on clinical fitness rather than age and support broader integration of geriatric assessment into routine oncology practice. Prospective studies are warranted to clarify the biological and clinical drivers of age-related outcome differences.
Presentation numberPS5-02-21
Chronic stress promotes brain metastasis via serotonin-HTR2B axis in triple-negative breast cancer: insights from spatial transcriptomic and functional analysis
Ju Young Ahn, Houston Methodist, Houston, TX
J. Ahn, J. Zambelas, D. Chen, M. Vasquez, W. Dong, H. Zhao, S. Wong; Systems Medicine and Bioengineering (SMAB), Houston Methodist, Houston, TX.
Background: Brain metastasis from triple-negative breast cancer (TNBC) is highly aggressive and remain difficult to treat, with a disproportionate burden among African American patients, especially in Texas. Chronic psychological stress, anxiety, or depression affecting more than 50% of cancer patients, disrupts neurotransmitter homeostasis and has been increasingly implicated in promoting cancer progression and metastasis. Among stress-associated neurotransmitters, serotonin (5-HT) levels are markedly elevated both under chronic stress and with selective serotonin reuptake inhibitor (SSRI) use, and can exacerbate brain metastasis. In this study, we investigate the mechanistic contribution of serotonin-HTR2B signaling to stress-induced brain metastatic progression in TNBC, using spatial transcriptomics and functional analysis. Methods: We conducted a comprehensive analysis of neurotransmitter-related gene expression using both single-cell RNA-seq (scRNA-seq) and bulk transcriptomic data from TNBC brain metastases and brain-tropic cell lines. Chronic restraint stress models were used to study metastasis development following inoculation with 4T1-BR or MDA-MB231-BR cells. Visium spatial transcriptomics was performed on brain tissue sections to spatially resolve transcriptional alterations within the metastatic niche under stress conditions. Functional validation included CRISPR -Cas9-mediated knockout (KO) of HTR2B in TNBC cell lines and in vivo bioluminescence imaging to monitor metastatic burden. Clinical datasets (GSE12276, n=204; TNBC cohort n=422) were analyzed to examine HTR2B expression levels and their association with patient survival outcomes. Results: TNBC brain metastases and brain-seeking cell lines showed widespread neurotransmitter-related gene upregulation. In vivo, chronic restraint stress significantly accelerated brain metastasis formation (p=0.022) and increased anxiogenic behaviors in mice. Spatial transcriptomic analysis revealed serotonin-HTR2B signaling enriched at the tumor-microenvironment interface in stressed mice, with activation of ERK/MAPK, PI3K/AKT, and STAT signaling pathways. CRISPR-mediated knockout of HTR2B resulted in a ~60% reduction in brain metastatic burden under stress conditions (p < 0.01), without affecting in vitro proliferation. Exogenous serotonin modestly increased in vitro growth but likely promotes metastasis via microenvironmental interactions. High tumor HTR2B expression was associated with shorter brain metastasis-free survival (p=0.012) and distant metastasis-free survival (p=0.013). Conclusions: Chronic stress promotes TNBC brain metastasis through activation of serotonin-HTR2B signaling, creating a pro-metastatic niche. Ongoing studies aim to identify specific stromal and immune cell populations responding to this signaling axis, using spatial transcriptomics-guided multiplex imaging. This work addresses a critical knowledge gap in understanding how stress reshapes the brain metastatic microenvironment. Collectively, these findings highlight serotonin-HTR2B as a promising therapeutic target to mitigate brain metastasis risk in stressed cancer patients.
Presentation numberPS5-02-22
Impact of overall survival on the use of prophylactic granulocyte colony-stimulating factor with sacituzumab govitecan-hziy in the treatment of triple negative metastatic breast cancer patients
Fred Kudrik, South Carolina Oncology Associates, Columbia, SC
F. Kudrik1, R. Choksi2, V. Gorantla2, S. Reddy3, S. Reganti3, S. Rosenfeld4, G. Cioffi5, E. Alwon5, D. Parris5, A. Rui5, M. Gart5, C. Wall5, B. Wang5, P. Varughese5, J. Donegan5, L. Morere5, R. Geller5, J. Scott5, D. Patt6; 1Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 2Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3Medical Oncology, Cancer Care Specialists, Reno, NV, 4Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 5PrecisionQ, IntegraConnect, West Palm Beach, FL, 6Medical Oncology, Texas Oncology, Austin, TX.
Background: Treatment options for metastatic triple-negative breast cancer (mTNBC) are limited, but antibody-drug conjugates (ADCs) such as sacituzumab govitecan (SG) have shown improved survival outcomes compared to single-agent chemotherapies. However, individuals undergoing this treatment are at an increased risk of neutropenia. Furthermore, updates to the package insert recommend primary prophylaxis with granulocyte-colony stimulating factor (G-CSF) for at-risk patients (pts). This study evaluated the impact of prophylactic G-CSF use on time to treatment discontinuation (TTD) and real-world overall survival (rwOS) among individuals with mTNBC who received SG. Methods: The IntegraConnect PrecisionQ de-identified electronic health record database, consisting of >3 million cancer pts across >500 care sites, was used to identify individuals with mTNBC that initiated SG between 4/22/2020 and 4/9/2025. Eligibility criteria included age ≥18 years at diagnosis and ≥2 documented visits. Prophylactic G-CSF was defined as any G-CSF use within 8 days following initiation of SG. Pts were excluded from the analysis if there was documentation of neutropenia (neutrophil count <1500/µL), discontinuation of SG, death, or censoring within 8 days of SG initiation. Demographic and clinical characteristics, including age at SG initiation, race, and Eastern Cooperative Oncology Group (ECOG) performance status, were assessed overall and stratified by prophylactic G-CSF use. Gray’s test was performed to assess cumulative incidence of neutropenia, adjusted for the competing risk of death, and 4-month estimates are reported. Kaplan-Meier survival curves were evaluated to assess TTD and rwOS by prophylactic G-CSF. To assess for the time-varying effect of prophylactic G-CSF use on TTD and rwOS, time-stratified multivariable Cox proportional hazards regression was performed with time-interval-specific effects (0-4 months, 4+ months) for prophylactic G-CSF use, and adjusted for additional demographic and clinical characteristics (age, race, ECOG status). Hazard ratios (HR) and 95% confidence intervals (CI) are reported. Results: Overall, 685 pts with mTNBC treated with SG were identified in the PrecisionQ database that met the study criteria. Most identified as White (67%), 41% had an ECOG status of 1, and 88% did not receive prophylactic G-CSF. Median age at SG initiation was 60 years (interquartile range [IQR]: 53, 69). Age, race, and ECOG status did not significantly differ by prophylactic G-CSF use. At 4 months, cumulative incidence of neutropenia was significantly higher among those who did not receive prophylactic G-CSF (42% vs. 30%, Gray’s test p=0.002). Median TTD and rwOS did not significantly differ by prophylactic G-CSF use. From 0-4 months, pts who did not receive prophylactic G-CSF were >2 times more likely to die compared to those who received prophylactic G-CSF within the first 8 days of SG initiation (HR: 2.37; 95% CI: 1.22-4.59 p=0.011). This impact was attenuated after 4 months (HR: 1.02, 0.79-1.50, p=0.4), indicating a time-varying effect of prophylactic G-CSF use on rwOS after SG initiation. There was no significant difference observed in TTD in either time-interval. Conclusions: Prophylactic use of G-CSF had a significant initial impact on rwOS among pts with mTNBC receiving SG. While prophylactic use of G-CSF did not confer a significant survival benefit after 4 months, the upfront effect warrants clinical consideration and integration into early treatment practices. These findings also indicate the need for further research assessing the impact of continued G-CSF use on overall survival.
Presentation numberPS5-02-23
Aretha trial: Safety run-in results of eribulin + evexomostat in metastatic triple-negative breast cancer with metabolic dysfunction
Sherry Shen, Memorial Sloan Kettering Cancer Center, New York, NY
S. Shen1, V. Solomon1, D. Williams1, C. White1, Y. Chen1, M. Meem1, I. Warren1, P. Drullinsky1, S. Schweber1, R. Wang1, M. Gorsky1, A. Gucalp1, J. Shen1, R. Sanford1, A. Widman1, M. Robson1, L. Norton1, T. Traina1, D. Graham2, R. Mahtani3, N. M. Iyengar4; 1Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 2Medicine, Hackensack Meridian Health, Hackensack, NJ, 3Medicine, Baptist Health, Miami, FL, 4Medicine, Winship Cancer Institute at Emory University, Atlanta, GA.
Background: The prognosis for metastatic triple negative breast cancer (metTNBC) remains poor and novel therapeutic strategies are urgently needed. Insulin resistance & obesity are independent predictors of worse survival in metTNBC. Moreover, Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and related metabolic markers often worsen significantly during chemotherapy. Methionine aminopeptidase 2 (MetAP2/p67) is an enzyme involved in angiogenesis and regulation of metabolic hormones that is overexpressed in breast cancer. In a phase I study of evexomostat, a MetAP2 inhibitor, 80% of patients with advanced solid tumors had stable disease and improvements in fasting insulin, adiponectin, and VEGF-C. The primary objective of this phase 2 study is to assess the efficacy of evexomostat on insulin resistance in combination with standard of care eribulin in metTNBC. Here, we report results from the safety run-in phase and exploratory biomarker changes. Methods: This is a multicenter phase 2 randomized controlled trial of evexomostat + eribulin (NCT05570253). Key eligibility include metTNBC, baseline HbA1c >5.5% and/or BMI ≥30, and ≤2 prior treatment lines in the metastatic setting. In the safety run-in period, 15 patients received evexomostat 49 mg/m2 Q2 weeks + eribulin 1.4 mg/m2 on D1&8 of a 21-day cycle. The safety threshold was defined as ≥4/15 patients experiencing serious adverse events (SAEs) not resolving within 14 days. The primary endpoint of the randomized phase is change in HOMA-IR at 12 weeks. Secondary endpoints include objective response rate (ORR) and safety and tolerability. Patients were considered evaluable if they received ≥1 dose of evexomostat. Change in HOMA-IR and other markers was analyzed using Wilcoxon signed rank test. Results: Among the 15 patients in the safety run-in phase, 2/15 experienced SAEs, both of which were grade 3 anemia. The most common any-grade AE was anemia (9/15, 60%). AEs of grade ≥3 were reported in 11 patients; the most frequent grade ≥3 AE was decreased neutrophil count. Accordingly, the safety stopping rule was not triggered and the trial advanced to the randomization phase. Exploratory analyses from baseline to week 12 demonstrated significant changes in LDL cholesterol, adiponectin, leptin, resistin, VEGF-D, and sFRP-5 (p=0.016, 0.009, 0.021, 0.016, 0.027, and 0.012 respectively; see Table). There was no increase in HOMA-IR (p>0.9). Among 12 patients evaluable for response; ORR was 17% and clinical benefit rate was 67%. Conclusions: Evexomostat + eribulin safety was confirmed and the trial advanced to the randomization phase including a placebo-controlled arm with eribulin. Preliminary biomarker results indicate that insulin resistance was unchanged, and favorable changes were seen in other metabolic markers.
| Characteristic | Baseline (N=12), mean (range) | Week 12 (N=12), mean (range) | Difference2 | p-value1 |
| HOMA-IR3 | 5.0 (0.4-15.1) | 4.1 (1.3-10.6) | -0.9 | |
| Glucose3, mg/dL | 101 (74-168) | 104 (89-137) | 3.4 | 0.4 |
| Insulin3, mcU/mL | 19 (2-56) | 16 (5-40) | -2.9 | 0.6 |
| HDL3, mg/dL | 47 (23-70) | 43 (30-71) | -4.1 | 0.4 |
| LDL3, mg/dL | 116 (62-153) | 99 (44-137) | -16.6 | 0.016 |
| Triglycerides3, mg/dL | 108 (76-159) | 110 (68-160) | 2.6 | 0.4 |
| Adiponectin, ng/mL | 8428 (20623-16989) | 10882 (3474-18431) | 2453 | 0.009 |
| Leptin, pg/mL | 35382 (1283-85050) | 21614 (1112-102339) | -13768 | 0.021 |
| Apelin, pg/mL | 1042 (378-2072) | 1348 (410-3910) | 305 | 0.5 |
| Resistin, ng/mL | 69 (23-134) | 173 (19-501) | 104 | 0.016 |
| FGF-21, pg/mL | 510 (100-1299) | 1527 (90-9808) | 1016 | 0.2 |
| C-reactive protein, ng/mL | 61679 (6003-316052) | 52459 (3483-224261) | -9220 | 0.7 |
| IL-6, pg/mL | 21 (4-64) | 36 (5-151) | 15 | 0.2 |
| VEGF, pg/mL | 190 (43-715) | 188 (26-608) | -1.9 | 0.3 |
| VEGF-C, pg/mL | 1374 (556-4294) | 880 (325-1956) | -495 | 0.06 |
| VEGF-D, pg/mL | 279 (136-556) | 363 (182-501) | 84 | 0.027 |
| Cyclophilin A, ng/mL | 536 (201-1095) | 589 (271-1158) | 53 | 0.6 |
| bFGF, pg/mL | 39 (9-136) | 36 (6-79) | -3.1 | 0.6 |
| sFRP-14, pg/mL | 1.46 (0.91-2.34) | 1.37 (0.78-1.90) | -0.1 | 0.6 |
| sFRP-5, pg/mL | 39205 (10666-82615) | 54983 (12986-97705) | 15779 | 0.012 |
|
1Wilcoxon signed rank test 2Absolute difference in means 3Complete data available in 7 patients 4Complete data available in 4 patients |
Presentation numberPS5-02-25
Homologous Recombination Deficiency is the major determinant of response of Triple-Negative Breast Cancer to Cisplatin+Radiotherapy
Simon N Powell, memorial sloan kettering cancer center, New York, NY
S. N. Powell1, A. J. Khan1, R. Delsite1, N. Riaz1, J. S. Reis-Filho2; 1radiation oncology, memorial sloan kettering cancer center, New York, NY, 2pathology, memorial sloan kettering cancer center, New York, NY.
Purpose/objectives: Women with recurrent triple-negative breast cancer (TNBC) who have recurrent disease have a poor prognosis. We know that recurrent TNBC has a high rate of homologous recombination deficiency (HRD). HRD cells are sensitive to cisplatin and radiation therapy (RT) independently, but their combination may enhance the sensitivity of HRD tumors. Materials/Methods: From April 2015 to September 2022, we enrolled 50 patients with locally recurrent or metastatic TNBC to a phase II trial (NCT02422498) of weekly cisplatin (25mg/m2) x12 and concurrent RT (50-60 Gy in 25 fractions or 37.5-48 Gy in 15 fractions). The primary end-point was the imaging response at 4-8 weeks after completion of therapy. Imaging response was a >50 reduction in the cross-sectional area of the index lesion. Fresh biopsies of all recurrent tumor masses were obtained in order to assess RAD51 function in viable cells, using ex-vivo irradiation for RAD51 foci in single cell suspensions from the tumor specimens. Tumors were categorized as HRD or HR-proficient (HRP) using the criteria established (PMID 28299801). All tumors with available material were subjected to whole genome sequencing (WGS) and categorized as HRD by criteria published recently (PMID 37587346).Results: The median age of enrolled patients was 50 (range 31-70). All patients had received at least two prior systemic therapies. 39 had local recurrence on the chest wall or regional nodes, with 25 having additional metastatic disease. 11 patients had only metastatic disease, with radiotherapy given to the dominant and painful site of disease. 1 patient was later determined to not have TNBC and was excluded from analysis. 42 of 49 patients had successful evaluation of HRD status, with 7 patients failing due to an inadequate cellular specimen on a limited core biopsy and the lack of any prior DNA for WGS. Of the 42 currently evaluated patients, 23 were HRD and 19 were HRP. For the patients with HRD tumors, the imaging response rate to cisplatin and RT was 78% (n=18, of which 10 showed a complete imaging response). In contrast, for HRP tumors the response rate was 21% (n=4, of which none showed a complete imaging response). The response of HRD tumors without radiotherapy, but to cisplatin alone was 24% (n=5) and only 1/15 HRP tumor showed a response to cisplatin alone. For the tumors where we had both RAD51 foci and WGS (n=29) the correlation of HRD calling was 27/29, with the two discordant tumors having evidence of BRCA2 reversion. HRD tumors were both sporadic biallelic and unknown germline at the time of recruitment to the study. Only 2 tumors were HRD without any detectable mutation in HR genes, suggesting that target gene mutation is the predominant cause of HRD. Further details of the whole genome signatures will be presented, with a mixture of BRCA1-like and BRCA2-like tumors observed (PMID 37587346). Although the study did not extend beyond the intitial treatment and response evaluation, follow-up durability of response was observed in the complete imaging responders.Conclusions: HRD status as determined by RAD51 focus formation or by WGS for genomic scars is the major determinant of imaging response of the index lesion (receiving cisplatin and RT). Even though these patients were heavily pre-treated, the imaging response and clinical benefit rate was reassuringly high for HRD patients, many of whom were non-germline cases. This trial supports the use of modern HRD testing as a method for selecting patients for HRD-directed therapies in future clinical trials. The incidence of HRD in recurrent TNBC is well above 50%, suggesting that many patients may benefit from newer agents for exploiting HRD in the future.
Presentation numberPS5-02-26
A phase I/II, single arm, non-randomized study of ribociclib, a CDK 4/6 inhibitor, in combination with bicalutamide, an androgen receptor inhibitor, in advanced AR-positive triple-negative breast cancer: A Big Ten Cancer Research Consortium Study
Julia Knight-Shefner, University of Wisconsin Carbone Cancer Center, Madison, WI
J. Knight-Shefner1, R. M. O’Regan2, E. Sampene3, O. Danciu4, K. Hoskins4, M. E. Burkard5, M. T. West1, V. Carreno1, I. Fernandez1, K. B. Wisinski1, M. N. Sharifi1; 1Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, 2Hematology and Oncology, University of Rochester, Rochester, NY, 3Biostatistics, University of Wisconsin, Madison, WI, 4Hematology and Oncology, University of Illinois Cancer Center, Chicago, IL, 5Medicine, University of Iowa Holden Cancer Center, Iowa City, IA.
Background: Triple-negative breast cancer (TNBC) is a heterogenous disease encompassing different intrinsic molecular subtypes. Outcomes remain poor for those with advanced disease despite recent advancements in immunotherapy and antibody-drug conjugates. The androgen receptor (AR) is a luminal nuclear hormone steroid receptor in the same family as the estrogen receptor and progesterone receptor. Although dependent on assay and cutoff used, about 25-35% of TNBC have AR protein expression detectable by immunohistochemistry (IHC). Preclinical data suggest the luminal androgen receptor (LAR) molecular subtype of TNBC is dependent on AR signaling and is particularly susceptible to CDK 4/6 inhibition. Thus, AR has emerged as a therapeutic target via androgen blockade in AR+ TNBC. We hypothesized that the combination of ribociclib, a CDK 4/6 inhibitor, and bicalutamide, an androgen receptor inhibitor, will demonstrate clinical benefit for patients with advanced AR+ TNBC. Methods: In this single arm, non-randomized phase I/II study, patients with AR+ TNBC were treated with combination ribociclib and bicalutamide. AR+ was defined as >0% in phase I and ≥10% in phase II. Up to 3 prior lines of systemic therapy for metastatic disease were allowed. Dose escalation was by 3+3 design with fixed bicalutamide at 150 mg daily and ribociclib (1) 400 mg QD on days 1-21, (2) 400 mg on days 1-28, and (3) 600 mg on days 1-21. A Simon two stage design was utilized with a planned total of 25 patients, including phase I patients treated at the recommended phase II dose (RP2D). The primary efficacy endpoint was clinical benefit rate (CBR) at 16 weeks. Secondary objectives were progression free survival (PFS), objective response rate (ORR), overall survival (OS), duration of response (DOR), and safety/tolerability. Additionally, a circulating tumor cell (CTC) AR gene signature was explored. The study was concluded early due to low enrollment. Results: 21 patients were enrolled. Median age was 56 (range 37-75 yrs) and all patients were female. 76% of patients were non-Hispanic white, 19% Black, and 5% unknown. AR positivity was between 2-95%. In the phase I dose escalation, the RP2D was determined to be the standard doses of each agent: ribociclib 600 mg daily on days 1-21 of the 28-day cycle and bicalutamide 150 mg daily. Of the 19 patients with available adverse event data, the most common treatment-emergent adverse events included: leukopenia 79% (47% grade 3), neutropenia 74% (47% G3, 16% G4), lymphopenia 47% (11% G3), fatigue 47%, thrombocytopenia 37% (11% G3), anemia 37% (5% G3), nausea 37%, AST elevation 26% (5% G3), ALT elevation 21%, constipation 21%, hot flashes 21%, creatinine elevation 16%. Two patients experienced a grade 3 infection: one with urinary tract infection and one with skin infection. 18/21 patients were evaluable for efficacy, which included both the phase I and phase II cohorts. The CBR at 16 weeks was 27% (5/18 patients, 95% CI [0.097, 0.535]; p-value 0.0481). There was a PR in 1 patient. OS, PFS, ORR, and DOR will be presented, as well as efficacy in patients treated at RP2D. AR-V7 was detected in baseline CTC samples in 2 out of the first 11 patients in the cohort, neither of whom achieved disease control at 16 weeks. CTC analysis for the remaining patients is ongoing and will be reported. Conclusions: Although the study did not complete accrual, there were 5 patients with AR+ TNBC treated with ribociclib and bicalutamide combination therapy who achieved disease control at 16 weeks. Further, the AR V7 CTC gene signature may be an emerging biomarker of treatment response to AR and CDK 4/6 inhibition. Future investigation of biomarkers beyond tissue AR IHC staining could be investigated to improve patient selection.
Presentation numberPS5-02-27
Variations in lines of treatment and clinical outcomes in US health system patients with locally advanced, inoperable or metastatic triple negative breast cancer (la/m TNBC)
Yuanhui Zhang, Gilead Sciences, Inc, Foster City, CA
X. Wang1, Y. Zhang1, A. Petrilla2, S. McElwee3, B. Momani3, J. Dinoso4, B. Stwalley5, C. Lai6, A. Brufsky7; 1Global Value and Access, HEOR, Gilead Sciences, Inc, Foster City, CA, 2HEOR, Norstella, Boston, MA, 3RWD, Norstella, Boston, MA, 4Global Medical Affairs, Gilead Sciences, Inc, Foster City, CA, 5US Medical Affairs, Gilead Sciences, Inc, Foster City, CA, 6Clinical Development, Gilead Sciences, Inc, Foster City, CA, 7Division of Hematology/Oncology Breast Cancer Center, Clinical Investigation, UPMC Hillman Cancer Center, Pittsburgh, PA.
Introduction: Patients with la/m TNBC have poor prognosis and limited effective treatment options. Clinical guidelines recommend testing for biomarkers, including PD-L1 expression, to inform clinical decision making and eligible treatments. This study describes treatment patterns and clinical outcomes in patients with la/m TNBC treated in US community and academic health systems. Methods: This observational, cohort study used deidentified person-level NorstellaLinQ EMR/lab and supplemental claims data. Patients were required to have diagnosis of la/m TNBC followed by initiation of 1L mTNBC anti-cancer treatment between 1/1/ 2020 and 12/31/2023. Patients were followed from initiation of 1L treatment (index date) until earliest of death, end of EMR, or end of study period (12/31/2024). Patients were stratified by PD-L1 status closest to the index date. Tumor PD-L1 status were assessed using the PD-L1 assays in EMR laboratory results or evidence of PD-L1 tumor status in unstructured clinical notes. Clinical outcomes included real-world time on treatment (rwTOT) and real-world time to next treatment (rwTTNT). Results: A total of 724 patients met criteria for analysis (PD-L1 Negative: 163, PD-L1 Positive: 398, Unknown/did not receive IO in la/m setting: 163). Mean(sd) age at index: 59(13) years. Demographics were comparable across PD-L1 Negative/Positive subgroups. Less than 5% of study patients were exposed to IO therapy in (neo)adjuvant setting. Among all patients, there were over 30 distinct 1L treatment agent-based regimens observed. Among patients with PD-L1 negative tumors, most common 1L regimens were chemotherapy monotherapy (“chemo mono”) (73.6%) and chemotherapy + immunotherapy (chemo + IO) (17.2%)). Among patients with PD-L1 positive tumors, most common 1L regimens were chemo + IO (60.6%) and chemo mono (31.7%). Approximately 34% of study patients cycled through at least 3 LOTs during the study period. rwTOT and rwTTNT were described in Table 1. Conclusions: In this real-world study of US patients with la/m TNBC and confirmed PD-L1 status, there is variability in 1L, 2L and 3L regimens. The majority of the PD-L1 positive cohort received 1L chemo (+/- IO) followed by chemo + IO or ADCs in 2L/3L. Negative PD-L1 cohort was largely prescribed 1L chemo followed by ADCs or chemo in 2L/3L, while there is a proportion of patients who used chemo+ IO combination therapy that is higher than previously published literature. The proportion of patients receiving subsequent LOTs was low, highlighting the need for new and efficacious frontline treatment options.
| Measure | Overall Study Population with la/m TNBC Initiating 1LOT (n=724) | With Negative PD-L1 Expression (n=163) | With Positive PD-L1 Expression (n=398) | ||||
| Most common 1 L regimens | |||||||
| Chemotherapy monotherapy | 378 (52.2%) | 120 (73.6%) | 126 (31.7%) | ||||
| Chemotherapy + IO | 283 (39.1%) | 28 (17.2%) | 241 (60.6%) | ||||
| rwTOT: Duration of 1LOT (in months) (median, 95% CI) | 2.67 (2.47, 2.83) | 2.80 (2.33, 3.23) | 2.77 (2.57, 3.03) | ||||
| rwTTNT: Time from 1LOT Start to 2LOT Start (in months) (median, 95% CI) | 6.47 (5.47, 7.70) | 5.60 (4.70, 7.03) | 5.20 (4.20, 7.30) | ||||
| Advanced to 2L during study period (n, % of cohort) | 427 (59.0%) | 107 (65.6%) | 248 (62.3%) | ||||
| Most common 2L Regimens (n, % with 2L treatment) | |||||||
| Chemo + IO | 138 (32.3%) | 10 (9.3%) | 127 (51.2%) | ||||
| Chemo monotherapy | 176 (41.2%) | 54 (50.5%) | 71 (28.6%) | ||||
| ADCs | 79 (18.5%) | 33 (30.8%) | 32 (12.9%) | ||||
| Advanced to 3L during study period (n, % of cohort) | 247 (34.1%) | 71 (43.6%) | 143 (35.9%) | ||||
| Most common 3L regimens (n, % of patients with 3L treatment) | |||||||
| Chemo monotherapy | 113 (45.7%) | 36 (50.7%) | 55 (38.5%) | ||||
| ADCs | 66 (26.7%) | 22 (31.0%) | 41 (28.7%) |
Presentation numberPS5-02-28
Safety analysis of phase 3 ASCENT-04 study of sacituzumab govitecan (SG) + pembrolizumab (pembro) vs chemotherapy (chemo) + pembro for previously untreated PD-L1+ metastatic triple-negative breast cancer (mTNBC)
Kevin Kalinsky, Winship Cancer Institute, Emory University, Atlanta, GA
K. Kalinsky1, P. Schmid2, E. de Azambuja3, S. Loi4, S. Kim5, C. Yam6, B. Rapoport7, S. Im8, B. Pistilli9, W. Mchayleh10, D. W. Cescon11, J. Watanabe12, M. Bañuelas13, R. Freitas-Junior14, A. Lortholary15, D. Gary16, K. Caldwell16, E. Harting17, S. M. Tolaney18; 1Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 2Medical Oncology, Centre for Experimental Cancer Medicine, Bart’s Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 3Medical Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (U.L.B.), Bruxelles, BELGIUM, 4Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 5Medical Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, KOREA, REPUBLIC OF, 6Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 7Medical Oncology, The Medical Oncology Centre of Rosebank, Personalised Cancer Care, Saxonworld, SOUTH AFRICA, 8Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, KOREA, REPUBLIC OF, 9Cancer Medicine, Gustave Roussy; IHU-National PRecISion Medicine Center in Oncology, Villejuif, FRANCE, 10Hematology and Oncology, AdventHealth Cancer Institute, Orlando, FL, 11Medical Oncology, Princess Margaret Cancer Centre/UHN, Toronto, ON, CANADA, 12Breast Oncology, Juntendo University Graduate School of Medicine, Tokyo, JAPAN, 13Medical Oncology, SCIENTIA Investigación Clínica S.C., Chihuahua, MEXICO, 14Medical Oncology, Advanced Center for Diagnosis of Breast Diseases Federal University of Goiás, Goiás, BRAZIL, 15Medical Oncology, Groupe d’Investigateurs National des Etudes des Cancers Ovariens et du sein (GINECO) and Hôpital Privé du Confluent, Nantes, FRANCE, 16Patient Safety, Gilead Sciences, Inc, Foster City, CA, 17Biostatistics, Gilead Sciences, Inc, Foster City, CA, 18Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.
Background: SG is a Trop-2-directed antibody-drug conjugate approved for patients (pts) with previously treated mTNBC. In PD-L1+ mTNBC, SG + pembro demonstrated statistically significant and clinically meaningful progression-free survival improvement vs chemo + pembro in the phase 3 ASCENT-04/KEYNOTE-D19 study; we report the first in-depth safety analysis. Methods: Pts were randomized 1:1 to SG (10 mg/kg IV, days 1 and 8) + pembro (200 mg, day 1, max 35 cycles) in 21-day cycles or chemo (gem + carbo, pac, or nab-pac) + pembro until disease progression or unacceptable toxicity. Exploratory analysis of exposure-adjusted incidence rates (EAIRs) was performed, defined as number of pts with ≥1 specified treatment-emergent adverse event (TEAE) per pt-year of exposure. Incidence, severity, time to onset, duration, and impact of AE management were assessed for select TEAEs for SG and/or pembro. Results: 441 pts (SG + pembro: 221; chemo + pembro: 220) were included in the analysis. EAIRs, time to onset and duration of TEAEs of interest, and immune-mediated (im)AE rates are summarized in the Table. G-CSF as primary prophylaxis was used in 43 (19%) and 20 (9%) pts in the SG + pembro and chemo + pembro arms, respectively; as secondary prophylaxis, G-CSF was used in 75/122 (61%) and 37/118 (31%) eligible pts. Incidence of grade ≥3 neutropenia was 35% with SG + pembro and 50% with chemo + pembro in pts who received G-CSF as primary prophylaxis; 50% and 45% in those who did not. Diarrhea was experienced by 155 (70%) and 63 (29%) pts in the SG + pembro and chemo + pembro arms, respectively; most cases were grade 1 (37% vs 17%) or 2 (24% vs 10%) diarrhea (60% vs 26% grade 1/2). The proportion of pts with diarrhea leading to dose reduction was similar in both arms (5% vs 1%) and there was no discontinuation due to diarrhea in the SG + pembro arm. Among pts with diarrhea, antidiarrheal treatment (tx) was used for 101 (65%) pts who received SG + pembro including loperamide (93%) and atropine (12%). The imAE of colitis occurred in 13 (6%) and 3 (1%) pts in the SG + pembro and chemo + pembro arms, respectively; one led to discontinuation in the SG + pembro arm. Colitis was managed according to pembro labeling guidance. Conclusions: In pts with previously untreated PD-L1+ mTNBC, EAIRs were generally similar between tx arms. With SG + pembro, there was a lower rate of TEAEs leading to dose reduction or discontinuation and a higher rate of any TEAEs; the rate of imAEs was lower vs chemo + pembro, although diarrhea and colitis were higher. Neutropenia, diarrhea, and colitis were manageable with supportive care. The overall safety profile was consistent with that of each agent, with no exacerbation of imAEs. Taken with previously reported efficacy benefit, these data support a favorable benefit/risk profile for SG + pembro vs chemo + pembro in this population.
Presentation numberPS5-02-29
Clinical data of DB-1305/BNT325 (TROP2 antibody-drug conjugate [ADC]) in patients (pts) with metastatic triple negative breast cancer (mTNBC): Efficacy and safety data from a phase 1/2 trial
Erika Hamilton, Sarah Cannon Research Institute, Nashville, TN
E. Hamilton1, D. A. Aysel1, M. Yan2, C. Wang3, H. Yang4, J. Shi5, W. Xie6, H. Wang7, Y. Sun8, X. Sun9, B. Zhang9, Q. Yang10, Z. Jia9, H. Mu9, S. Tillmanns11, Y. Barbachano12, J. Furlanetto13, T. Sun14; 1SCRI Oncology Partners, Sarah Cannon Research Institute, Nashville, TN, 2Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, CHINA, 3Cancer Institute and Hospital, Tianjin Medical University, Tianjin, CHINA, 4Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, CHINA, 5Department of Internal Medicine, Linyi Cancer Hospital, Linyi, CHINA, 6Department of Breast & Bone and Soft Tissue Tumor, Guangxi Medical University Cancer Hospital, Nanning, CHINA, 7Department of Breast & Bone and Soft Tissue Tumor, Guangxi Medical University Cancer Hospital,, Nanning, CHINA, 8Cancer Hospital of Shandong First Medical University, Cancer Hospital of Shandong First Medical University, Jinan, CHINA, 9Clinical Development, Duality Biologics, Shanghai, CHINA, 10Research and Development, Duality Biologics, Shanghei, CHINA, 11Clinical Development, BioNTech SE, Mainz, GERMANY, 12Biostatistics, BioNTech UK Ltd, London, UNITED KINGDOM, 13Medical Safety, BioNTech SE, Mainz, GERMANY, 14First Ward of Breast Medicine, Liaoning Cancer Hospital and Institute, Shenyang, CHINA.
Background DB-1305/BNT325 is a novel investigational ADC with a humanized TROP2 IgG1 monoclonal antibody linked to a DNA topoisomerase I inhibitor via a cleavable linker. Early clinical data showed a manageable safety profile and encouraging activity across tumor types, particularly in ovarian cancer (Marathe, ESMO 2023 & Rubinstein, SGO 2025). This study evaluated DB-1305/BNT325 in pts with previously treated mTNBC. Methods In the dose expansion part of this Phase 1/2 trial, pts with mTNBC (w/o prior sacituzumab govitecan) received DB-1305/BNT325. The primary endpoints were objective response rate (ORR), based on investigator assessment per RECIST 1.1, and safety. Secondary endpoints included disease control rate (DCR), duration of response (DOR) and progression-free survival (PFS). Results As of March 31, 2025, 26 pts with TNBC had received ≥1 dose of DB-1305/BNT325, with 6 pts still on treatment. The cohort is fully enrolled. Median age was 49 years (range: 29-77); most pts were Asian (92.3%) and had ECOG PS 1 (65.4%). Most common metastases sites were bone (50%), lung (38.5%), and liver (34.6%). The median number of prior lines of treatment was 2 (range: 2-6), including immunotherapy in 8 (30.8%), platinum-based chemotherapy in 19 (73.1%), and bevacizumab in 5 (19.2%) pts. Efficacy data (summarized in the table) is available for all 26 pts after a median follow-up of 9.3 months. Treatment-emergent and treatment-related adverse events (TRAEs) occurred in all 26 pts, with Grade ≥3 in 9 (34.6%) pts. TRAEs led to dose reduction in 5 pts (19.2%) and discontinuation in 1 pt (3.8%, due to anemia). No TRAEs resulted in death. The most common TRAEs were stomatitis (any grade: 69.2%, Grade ≥3: 7.7%), anemia (any grade: 53.8%, Grade ≥3: 11.5%), nausea (any grade 46.2%, Grade ≥3: 0%), decreased neutrophil (any grade: 46.2%, Grade ≥3: 0%) and white blood (any grade: 46.2%, Grade ≥3: 3.8%) cell count. Updated analyses from the cohort will be presented. Conclusions The data suggest an encouraging efficacy and manageable safety profile of DB-1305/BNT325 in pts with pretreated TNBC, with only 1 patient discontinuing treatment due to TRAE. DB1305/BNT325 is now being evaluated in combination with BNT327, an investigational anti-PD-L1 x VEGF-A bispecific antibody in pts with TNBC.
| Best overall response, n (%) | n=26 | ||
| PR | 9 (34.6) | ||
| SD | 12 (46.2) | ||
| PD | 4 (15.4) | ||
| Confirmed Objective response rate, n (%) | 9 (34.6) | ||
| Disease control rate, n (%) | 21 (80.8) | ||
| Median duration of response, months | Not reached | ||
| Median progression-free survival (95% CI), months | 5.5 (2.76, NE) |
Presentation numberPS5-02-30
Prolonged survival following PD-L1/PD-1 immune checkpoint inhibitor therapy after leronlimab induced PD-L1 upregulation on cancer-associated macrophage-like cells and circulating tumor cells in patients with metastatic or locally advanced triple-negative breast cancer
Milana V Dolezal, Stanford University School of Medicine, Stanford, CA
M. V. Dolezal1, V. G. Abramson2, N. Chittoria3, S. Ehsani4, R. G. Pestell5, H. S. Rugo6, H. Rui7, D. L. Adams8, J. Meidling9, M. Lataillade9, J. P. Lalezari9; 1Department of Medicine, Stanford University School of Medicine, Stanford, CA, 2Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN, 3Department of Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, 4Department of Medicine, University of Arizona Cancer Center, Tucson, AZ, 5Department of Medicine, Pennsylvania Cancer and Regenerative Medicine Research Center, Wynnewood, PA, 6Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, 7Cancer Biology, Thomas Jefferson University, Philadelphia, PA, 8Department of Clinical R&D, Creatv MicroTech, Inc., Monmouth Junction, NJ, 9Department of R&D, CytoDyn Inc., Vancouver, WA.
Background. Pretreated patients with metastatic triple-negative breast cancer (mTNBC) have a poor prognosis. In clinical trials among previously treated mTNBC patients the median overall survival (mOS) has been reported as 6.6 months for ≥3rd line chemotherapy, 11.8 months for ≥2nd line sacituzumab govitecan, and 9.9 months for ≥1st line pembrolizumab. It has also been reported that in the real-world, after first line therapy around a third of patients die before receiving 2nd line treatment. Furthermore, there remains an unmet need in the PD-L1 negative mTNBC patient population. It has been shown that >95% of TNBCs are positive for C-C chemokine receptor 5 (CCR5). Leronlimab (LRM) is a humanized monoclonal antibody that blocks CCR5 and reduces TNBC metastasis by >98% in preclinical models. Methods. In a retrospective post hoc analysis, data were pooled from 28 patients with mTNBC from 3 LRM clinical trials (NCT03838367 [N=10]; NCT04313075 [N=16]; NCT04504942 [N=2]). LRM was given subcutaneously weekly at a dose of 350 mg (N=10), 525 mg (N=15), or 700 mg (N=3) in combination with various chemotherapies ± immune checkpoint inhibitors (ICI). PD-L1 staining was measured on cancer-associated macrophage-like cells (CAMLs) and circulating tumor cells (CTCs) prior to and after (≈40 days) LRM treatment. Cox proportional univariate analyses were run to determine Hazard Ratios (HRs) for progression-free survival (PFS) and overall survival (OS) at 48 months. Results. Median age was 48.5 years (range 32-83) with a median of 2 prior metastatic therapies (range 0 to ≥3). Overall, 18% (5/28) of patients had PD-L1 positive tumor staining (CPS≥10%). LRM was well tolerated with no patients withdrawing due to LRM-related adverse events and no dose-limiting toxicities. Overall, mPFS was 3.8 months and mOS was 6.8 months. Survival at 1, 2, 3, and 4 years was 35.7%, 21.4%, 17.9%, and 17.9%, respectively. An upregulation from baseline of PD-L1 was observed in CAMLs/CTCs in 76% (16/21) of patients (7 patients had no post-baseline samples) after any dose of LRM, and in 88% (15/17) of patients receiving the 525 mg or 700 mg doses. Sixteen patients showed a drop in CAMLs/CTCs after initiating LRM while 12 showed an increase. The mOS for patients who showed a drop in CAMLs/CTCs was 17.2 months (95% CI, 9.4–N/A) compared to 3.7 months (95% CI, 1.7–5.6) for those patients who showed an increase in CAMLs/CTCs after LRM treatment (HR: 7.11 [95% CI, 2.5–20.2; p=0.0007]). Further, of the seven patients treated with an ICI with, or after LRM, 5/5 (100%) patients with PD-L1 upregulation remained alive at 4 years, compared to none of the 21 patients (0%) who did not receive an ICI with or after LRM (HR: 4.14 [95% CI, 1.7–10.2; p=0.0041]). Conclusions. In this retrospective pooled analysis of 28 mTNBC patients, LRM was well tolerated. A 4-year OS rate of 17.9% (5/28) in a population with a median of 2 prior metastatic therapies is encouraging. Among a key subgroup, all 5 patients with PD-L1 upregulation who subsequently received ICI remained alive at 4 years possibly indicating a correlation with durable responses. A reduction in CAMLs/CTCs after LRM treatment may relate to improved survival, suggesting its potential as a prognostic biomarker. These findings support the hypothesis that LRM may enhance PD-L1 expression on CAMLs/CTCs, potentially priming tumors for improved responses to ICIs. In an evolving treatment landscape these data warrant prospective evaluation of LRM, particularly in combination with ICIs in mTNBC.
Presentation numberPS5-03-01
Phase I Study of Stereotactic Radiation and Sacituzumab Govitecan with Zimberelimab in the Management of Metastatic Triple Negative Breast Cancer with Brain Metastases
Kamran A Ahmed, Moffitt Cancer Center, Tampa, FL
K. A. Ahmed1, Y. Kim2, R. Dowell1, D. Oliver1, M. Mills1, R. Nair3, A. Armaghani4, R. Costa4, T. O’Connor4, G. Rajasekaran Rathnakumar4, H. Soliman4, A. Soyano4, E. Tan-Chiu4, P. Forsyth5, J. Arrington6, H. Yu1, H. Han4; 1Radiation Oncology, Moffitt Cancer Center, Tampa, FL, 2Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, 3Pharmacy, Moffitt Cancer Center, Tampa, FL, 4Breast Oncology, Moffitt Cancer Center, Tampa, FL, 5Neuro Oncology, Moffitt Cancer Center, Tampa, FL, 6Radiology, Moffitt Cancer Center, Tampa, FL.
Background: The prognosis and quality of life for triple negative breast cancer (TNBC) brain metastases remains poor. Sacituzumab govitecan (SG) has shown potential blood brain barrier (BBB) penetration in preclinical and clinical settings with anti-tumor activity in the phase III ASCENT Trial (NCT02574455). Radiotherapy can open the BBB and has been shown to be safe in combination with anti-PD-1 therapy in our previous study (NCT03807765). Combining radiation with the anti-PD-1 monoclonal antibody, zimberelimab, and SG may provide a synergistic anti-tumor approach. Methods: The study is designed as a single-arm, nonrandomized, open-label, phase I/II trial of SG and zimberelimab with stereotactic radiosurgery (SRS) among patients with metastatic TNBC with brain metastases. The primary endpoint is neurologic toxicity defined by CTCAE v5 criteria (phase I) assessed via a 3+3 design. Eligibility includes TNBC patients ≥ 18 years old with brain metastases eligible for SRS with at least one measurable lesion ≥ 0.5 cm per RANO-BM criteria. Treatment is initiated with SRS followed 1 week later by SG on days 1 and 8 (10 mg/kg) with zimberelimab on day 1 (360 mg IV) repeated every 3 weeks. Phase I results are reported, clinical trial information: NCT06238921. Results: Three patients were enrolled between December 2024 through April 2025. Median age was 56 (range: 49-57) with a baseline KPS of 90 in 2 patients and 70 in 1 patient. Median lines of prior systemic therapy in the stage IV setting was 1 (range 0-2). No intracranial dose limiting toxicities were noted. The most common grade ≥ 2 adverse events were neutropenia in 3 patients (100%) (grade 4 in 1 and grade 2 in 2 patients) and grade 2 fatigue in 1 patient. The best RANO-BM intracranial response has been a partial response (PR) in 3 patients (100%). The best irRECIST systemic response has been a PR (n=1), stable disease (n=1), and progressive disease (n=1). Two patients continue study therapy currently. No deaths have occurred. Conclusions: Results from the phase I portion reveals safety of a combined approach. Efficacy continues to be assessed in the phase II portion.
Presentation numberPS5-03-02
Specific site of metastasis in stage IV triple-negative breast cancer: a national registry analysis of survival outcomes and treatment patterns
Shawn M Doss, Medical College of Georgia, Augusta, GA
S. M. Doss, P. Raval; Department of Medicine, Medical College of Georgia, Augusta, GA.
Background: Prior studies of metastatic triple-negative breast cancer (TNBC) have generally categorized multiple metastatic sites broadly as “multiple” or have focused on single-organ involvement, limiting the ability to counsel patients on prognosis for specific metastatic groups. To assist oncologists in these discussions, we analyzed how site-specific patterns of distant metastasis affected survival and treatments received for de-novo stage IV TNBC using a large, real-world national registry. Methods: Using the National Cancer Database (NCDB), we identified women over age 40 who were diagnosed with de-novo stage IV TNBC between 2010 and 2020. Metastatic groups were classified as brain only, lung only, liver only, or combinations of these sites at diagnosis. We focused only on brain, lung, and liver involvement because bone metastases are known to have a more favorable prognosis. Median overall survival (OS) and six-month and one-year survival rates were calculated using Kaplan-Meier analysis. Multivariate Cox regression analyzed associations between metastatic group and two-year OS after adjustment for age, comorbidity index, race/ethnicity, facility type, and diagnosis year. Treatment patterns were summarized for each metastatic group. Results: We identified 119,373 women with stage IV breast cancer, of whom 13,345 (11.2%) had TNBC. After excluding patients with missing variable data, 7,448 women comprised the final cohort for analysis. Median OS by metastatic group ranged from 3.6 months (brain + liver) to 12.7 months (lung only). In ascending order, one-year OS rates were 13.1% for brain + lung + liver, 17.3% for brain + liver, 29.2% for brain + lung, 30.8% for brain only, 34.0% for lung + liver, 47.6% for liver only, and 51.8% for lung only. In multivariate Cox regression, compared to brain only, patients with metastases to lung only (adjusted hazard ratio [aHR] 0.55, 95% confidence interval [CI] 0.49-0.61, p<0.001) and liver only (aHR 0.69, 95% CI 0.62-0.77, p<0.001) had increased two-year OS, while patients with metastases to brain + liver (aHR 1.67, 95% CI 1.37-2.03, p<0.001) and brain + lung + liver (aHR 1.72, 95% CI 1.47-2.01, p<0.001) had worse OS. Chemotherapy receipt ranged from 57.0% (brain + liver) to 75.7% (liver only). Radiation was most common in patients with CNS involvement, ranging from 60.6% (brain + liver) to 69.9% (brain + lung). Surgery to the primary breast site was rare, highest in lung only (27.1%) and lowest in brain + liver (6.3%). Conclusions: Sites of distant metastasis at diagnosis strongly influenced survival and treatment choice in stage IV TNBC. Patients with lung-only metastases showed the highest OS, while those with brain and/or liver involvement had markedly worse OS. These results can help guide clinicians and patients in counseling, expectations, and treatment planning for stage IV TNBC.
| Metastatic Group | n | Median OS (months) | 6-month OS (%) | 1-year OS (%) | % Received Chemotherapy | % Received Radiation | % Received Primary Breast Site Surgery |
| Brain + Liver | 142 | 3.6 | 30.3 | 17.3 | 57.0 | 60.6 | 6.3 |
| Brain + Lung + Liver | 276 | 3.8 | 34.9 | 13.1 | 62.0 | 68.8 | 6.5 |
| Brain + Lung | 501 | 6.0 | 50.0 | 29.2 | 64.5 | 69.9 | 9.0 |
| Brain only | 544 | 6.7 | 52.5 | 30.8 | 61.8 | 64.5 | 16.2 |
| Lung + Liver | 1,127 | 7.3 | 54.3 | 34.0 | 67.4 | 19.3 | 14.8 |
| Liver only | 1,800 | 11.2 | 66.6 | 47.6 | 75.7 | 18.9 | 21.7 |
| Lung only | 3,058 | 12.7 | 71.7 | 51.8 | 73.9 | 21.3 | 27.1 |
Presentation numberPS5-03-03
Trop2-directed antibody-drug conjugate shr-a1921 combined with pd-l1 inhibitor adebrelimab for patients with advanced triple negative breast cancer: results from a phase 2, multi-cohort, open-label, non-controlled trial
Tao Wang, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
T. Wang1, W. Zhao2, J. Xiao1, J. Zhou1, X. Shi2, Q. Li2, Z. Jiang1; 1Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA, 2Department of Oncology, Beijing Gobroad Hospital, Beijing, CHINA.
Background: The advent of novel antibody-drug conjugates (ADCs) has greatly changed the treatment landscape of advanced HER2-negative breast cancer. Preclinical studies have showed potential synergistic effect between ADCs and immunotherapies, while the clinical evidence remains limited. Our phase 2, multi-cohort, open-label, non-controlled trial (NCT06433609), was designed to evaluate the efficacy and safety of ADCs with distinct targets combined with PD-L1 inhibitor adebrelimab in pts with pretreated advanced HER2-negative breast cancer. Here, we present the preliminary results of TROP2-ADC SHR-A1921 combined with adebrelimab in pts with advanced triple negative breast cancer (TNBC). Methods: Pts with HER2-negative advanced breast cancer who had experienced disease recurrence or progression on or after at least one line of systematic therapy were eligible for this trial. Prior PD-1/L1 inhibitors were allowed. Eligible pts with advanced TNBC enrolled in Cohort 2 received SHR-A1921 (3 mg/kg IV on day 1, Q3W) and adebrelimab (1200mg IV on day 1, Q3W). The primary endpoint was objective response rate (ORR) per investigator based on RECIST v1.1. Secondary endpoints included disease control rate (DCR), clinical benefit rate (CBR), duration of response (DoR), progression-free survival (PFS), overall survival (OS) and safety. Results: Between Sept. 6th, 2024 and Dec. 31st, 2024, 15 pts were enrolled in Cohort 2. The median age was 52 years (range, 29-70), and pts had the median one (range, 0-8) prior treatment at the advanced setting. Eleven pts (73.3%) had ECOG PS of 1, eight (53.3%) were PD-L1-positive (CPS≥1) and 13 (86.7%) had visceral metastases, with 8 (53.3%) having ≥3 metastatic sites. With a median follow-up of 5.5 mo (range, 1.2-8.9), all of pts had at least one efficacy assessment. The confirmed ORR was 46.7% (95% confidence interval [CI], 21.2-73.4) with a median DoR of 4.6 mo. CBR was 46.7% (95%CI, 21.2-73.4), and DCR was 86.7% (95%CI, 59.5-98.3). Of note, among 6 pts with PD-L1-negative diseases, 2 pts achieved PR and 1 pt achieved PR to be confirmed. Nine PFS events were observed, and the median PFS was 5.1 mo (95%CI 3.1-NA). Two pts died after disease progression, and the median OS was unmatured. Treatment-related adverse events (TRAEs) occurred in all pts, and mostly were of grade 1-2. Grade≥3 TRAEs occurred in 2 pts (13.3%), including one (6.7%) grade 3 stomatitis and one (6.7%) grade 3 lymphocyte decreased. Eight pts experienced dose reduction of SHR-A1921 due to TRAEs. No dose discontinuation and no death due to AE was reported. Conclusion: The combination of SHR-A1921 and adebrelimab demonstrated promising anti-tumor efficacy and manageable safety in pretreated TNBC pts regardless of PD-L1 status, warranting further investigations in this population. Our exploration of ADCs combined with adebrelimab in HER2-negative advanced breast cancer is still ongoing.
Presentation numberPS5-03-04
Timing of final oncologist visit and systemic treatment use near the end of life in advanced triple-negative breast cancer: a multinational cohort study
Milos Holanek, Masaryk Memorial Cancer Institute, and Faculty of Medicine, Masaryk University, Brno, Czech Republic
J. Zubrowska1, M. Holanek2, M. Pieniazek3, A. Polakiewicz-Gilowska4, R. Soumarova5, H. Studentova6, A. Konieczna7, A. Mlodzinska7, K. Winsko-Szczesnowicz8, M. Lisik-Habib9, A. Pekala9, D. Krejci10, J. Sustr11, I. Kolarova12, I. Danielewicz13, M. Szymanik-Resko13, T. Ciszewski14, L. Rusinova15, B. Czartoryska-Arlukowicz8, A. Lacko3, M. Jarzab4, R. Pacholczak-Madej16, I. Lugowska17, Z. Bielcikova18, M. Malejcikova19, M. Puskulluoglu20; 1Department of Clinical Oncology, Holy Cross Cancer Center, Kielce, POLAND, 2Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, and Faculty of Medicine, Masaryk University, Brno, CZECH REPUBLIC, 3Department of Oncology, Breast Unit Clinical Oncology Day Care Department, Wroclaw Medical University and Lower Silesian Comprehensive Cancer Center, Wroclaw, POLAND, 4Breast Cancer Center, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 5Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 6Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 7Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology-National Research Institute, Warszawa, POLAND, 8Department of Clinical Oncology, M. Sklodowska-Curie Bialystok Oncology Center, Bialystok, POLAND, 9Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 10Department of Oncology, First Faculty of Medicine, Charles University in Prague, and Bulovka University Hospital, Prague, CZECH REPUBLIC, 11Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University, and University Hospital Pilsen, Plzen, CZECH REPUBLIC, 12Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Kralove, CZECH REPUBLIC, 13Oddzial Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 14Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 15Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 16Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, POLAND, 17Centre of Excellence for Precision Oncology, Maria Sklodowska-Curie National Research Institute of Oncology (NIO-PIB), Warsaw, POLAND, 18Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 19II. Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 20Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, POLAND.
Background: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer (BC) characterized by rapid clinical progression. Further lines of palliative systemic therapy typically offer diminishing benefit and increasing toxicity. Unlike hormone receptor-positive BC, TNBC lacks low-toxicity, long-term treatment options. In heavily pretreated patients, the decision to continue systemic therapy is particularly challenging. In the absence of robust evidence and amid growing expectations for individualized, patient-centered care, clinicians must rely on judgment to determine when to shift focus to palliative care. Timely recognition of the end-of-life (EOL) phase is essential to avoid overtreatment, reduce harm, and ensure care aligns with patient goals. The aim of this study was to assess the timing of the final oncologist visit relative to death in heavily pretreated women with advanced TNBC. We also analyzed systemic treatment status and palliative care involvement at that time and explored clinical factors associated with this interval. Materials and Methods: This multinational ambispective cohort study included women with advanced TNBC who received ≥2 lines of palliative systemic therapy and died between June 20, 2022, and June 20, 2025. Data from 17 centers in Poland, the Czech Republic, and Slovakia were extracted from medical records. The primary endpoint was the interval (in days) between the final oncologist visit and death. Collected variables included age at last visit, number of prior chemotherapy lines, performance status (PS), systemic treatment continuation, and palliative care involvement prior to last visit. Analyses were performed in R (v4.3.3), with significance set at p < 0.05. Results: A total of 183 women were included. The median age at death was 53.6 years (range: 28.6-86.5). The median number of previous palliative chemotherapy lines was 4 (range: 2-12), and the median interval between the last oncologist consultation and death was 17 days (range: 0-410). At the final visit, systemic treatment was ongoing in 53 patients (29.5%), and palliative care involvement prior to the last visit was documented in 89 patients (49%). The median interval between the final oncologist consultation and death among patients continuing systemic therapy was 27 days (range: 2-194). PS at the last visit was 0-1 in 36 patients (20%), 2 in 38 (21%), 3 in 65 (36%), and 4 in 35 (19%). A significantly longer interval between the last visit and death was observed in patients with better performance status 0-1 vs ≥2 (p<0.001). No significant difference was observed with respect to palliative care involvement (p=0.849). Conclusions: These findings highlight the need for earlier recognition of clinical decline and more proactive integration of palliative care in patients with advanced TNBC. Avoiding non-beneficial systemic therapy near the EOL may support care focused on symptom relief, patient autonomy, and quality of life. Given the particularly poor prognosis of metastatic TNBC, ensuring timely access to appropriate EOL care has become a recognized priority at the European level, including within the Network of Expertise on Complex and Poor Prognosis Cancers (NoE PPC), as part of broader efforts to address inequalities and improve outcomes for this high-risk population. Structured EOL planning remains a critical gap in international real-world practice and requires urgent attention in this context.
Presentation numberPS5-03-06
Real-world progression-free survival from first-line treatment initiation in patients with PD-L1-negative locally recurrent inoperable or metastatic triple-negative breast cancer in the United States
Tiffany A. Traina, Memorial Sloan Kettering Cancer Center, New York, NY
T. A. Traina1, E. Serra2, J. Bedard2, M. Levesque-Leroux2, P. Gagnon-Sanschagrin2, A. Guérin2, S. Guenther3, G. Joseph4, V. Guan4, J. Salcedo4; 1Department of Medicine, Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY, 2-, Analysis Group ULC, Montréal, QC, CANADA, 3Global Medical Affairs, BioNTech SE, Mainz, GERMANY, 4Health Economics & Outcomes Research, BioNTech US Inc, Cambridge, MA.
Background: Patients with PD-L1-negative locally recurrent inoperable or metastatic triple-negative breast cancer (lr/m TNBC) have limited treatment options and typically receive first-line (1L) chemotherapy in clinical practice. Real-world evidence on patient outcomes, particularly real-world progression-free survival (rwPFS) from 1L treatment initiation in PD-L1-negative lr/m TNBC, remains scarce. Additional data are needed to better understand clinical outcomes in this population and support evidence-based treatment decision-making. Methods: A retrospective cohort of adults (≥18 years old) receiving 1L treatment for PD-L1-negative lr/m TNBC was identified in Komodo Research Data (01/2016-12/2023), a large claims database that covers approximately 30% of the United States (US) population. Patients with lr/m TNBC were identified using an algorithm developed with medical expert guidance that required systemic treatment initiation within 3 months of a biopsy and no observed surgery in the 12 months following the biopsy. Triple-negative and PD-L1 statuses were inferred from treatments received in the lr/m setting. Demographics were described on the date of 1L treatment initiation, and comorbidities were assessed during the preceding 12 months. Real-world proxies of progression were also defined with medical expert guidance and included death, hospice admission, initiation of a second line of systemic treatment, or initiation of radiotherapy. Among patients who initiated 1L treatment ≥6 months before the end of data availability, the Kaplan-Meier method assessed rwPFS from 1L treatment initiation until the first real-world proxy of progression or end of follow-up, defined as the earliest of end of continuous health plan enrollment or end of data availability. Results: A total of 929 patients with lr/m TNBC were identified in this study. The median (interquartile range [IQR]) age at 1L initiation was 59 (51-66) years. Most patients were female (98.6%). Among those with a recorded race (69.3%), 60.1% were White, 26.4% were Black or African American, and 9.2% were Hispanic or Latino. Most patients had commercial insurance (64.9%), followed by Medicare (26.7%) and Medicaid (8.4%). The most frequent comorbidities prior to 1L treatment initiation were liver disease (26.8%), chronic obstructive pulmonary disease (22.7%), diabetes (22.0%), and peripheral vascular disease (13.6%). Median (IQR) follow-up from 1L initiation was 10.3 (5.7-20.1) months. The most common 1L treatments in the lr/m TNBC setting included cyclophosphamide+doxorubicin-based regimens (28.1%), followed by capecitabine (12.7%), paclitaxel (10.4%), carboplatin+paclitaxel (9.4%), and carboplatin+gemcitabine (8.6%), with frequent use of non-NCCN-guideline-preferred regimens. Among the 910 patients with sufficient follow-up to assess rwPFS, 703 (77.3%) had an indicator of progression following 1L initiation. Median (95% confidence interval [CI]) rwPFS was 4.7 (4.5-5.2) months. rwPFS rates (95% CI) were 69.0% (65.9%-71.9%) at 3 months, 41.7% (38.4%-45.0%) at 6 months, 22.6% (19.7%-25.6%) at 12 months, and 18.3% (15.5%-21.2%) at 18 months. The observed indicators of progression were, in order of frequency, initiation of a second line of systemic treatment (43.8%), death (25.5%), initiation of radiotherapy (18.1%), and hospice admission (12.7%). Conclusions: In US clinical practice, patients with PD-L1-negative lr/m TNBC experience rapid disease progression following initiation of 1L treatment, with a median time to progression of less than 5 months. These findings underscore a critical unmet need for effective 1L treatment options with tolerable safety profiles to improve outcomes in this patient population.
Presentation numberPS5-03-07
A Real-World Study on the Current Status of First-Line Treatment and Clinical Outcomes in Patients with Advanced Triple-Negative Breast Cancer (aTNBC) in Western China
Ting Luo, West China Hospital, Sichuan University, China, Chengdu, China
T. Luo1, C. Zhuang1, Y. Song1, P. He1, D. Zheng1, X. Yan1, X. Zhong1, T. Tian1, B. Wei2, Y. Xie1, J. Chen1, Q. Lv1; 1Institute of Breast Health Medicine, Breast Center, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, China, Chengdu, CHINA, 2Department of pathology, West China Hospital, Sichuan University, China, Chengdu, CHINA.
Background: Triple-negative breast cancer (TNBC) is characterized by limited treatment options and poor prognosis, with chemotherapy serving as the primary treatment approach. Immunotherapy and targeted therapy have become critical strategies for improving prognosis in patients with TNBC, but real-world adoption remains unclear. This study aimed to characterize first-line (1L) treatment and clinical outcomes in patients with advanced TNBC (aTNBC) in Western China.Methods: This retrospective study enrolled aTNBC patients initiating first-line therapy (2018-2023) in West China Hospital. Descriptive statistics summarized treatment adoption and patient characteristics.The primary endpoint was overall survival (OS), progression-free survival (PFS), and time to next treatment or death (TTNTD).Results: A total of 282 patients with aTNBC were included, with a median age of 51 years (range, 22-80). The mOS was 22.0 months(95% CI, 19.6-25.1), mPFS was 7.8 months(95% CI, 7.1-9.0) and mTTNTD was 8.3 months(95% CI, 7.2-9.4) among patients. Patients aged 40-65 years had significantly longer OS (p=0.04). Patients with recurrent metastatic TNBC demonstrated significantly prolonged PFS (p=0.04) and TTNTD (p=0.04) compared to those with de novo metastatic TNBC; however, no statistically significant difference in OS was observed. Patients with a treatment-free interval (TFI) exceeding 12 months demonstrated significantly longer OS (p<0.001). Ipsilateral chest metastasis was associated with shorter OS(p=0.005), while primary breast recurrence correlated with reduced PFS (p=0.02) and TTNTD (p=0.02) in locoregional recurrence patients. Patients who developed distant metastasis exhibited significantly shorter OS (p=0.04), PFS (p=0.002), and TTNTD (p=0.002); notably, those with liver or lung metastasis as the first metastatic site showed particularly poor prognosis (liver PFS (p<0.001) and TTNTD (p=0.002); lung OS (p=0.02), PFS (p=0.003) and TTNTD (p<0.001)). PD-L1 testing (22C3 assay) was performed in 20.6% (58/282) of patients, yielding a 50.0% positivity rate (29/58, CPS ≥1), among whom 8 received immune checkpoint inhibitors (ICIs) in 1L treatment. 17 patients received ICIs without established PD-L1 biomarker eligibility (either PD-L1-negative or untested status). Pathogenic germline BRCA1/2 mutations were identified in 19 of 83 tested patients (22.9%), among whom only 5 received PARP inhibitor in 1L treatment. Additionally, ADC-based therapy was administered to 9 patients in the 1L treatment. Conclusions: In Western China, 1L treatment for aTNBC predominantly relies on chemotherapy with limited ICI and targeted therapy due to economic and logistical barriers. Future expanding access to immunotherapy and targeted therapy in Western China is clinically imperative to address current care disparities.
|
|
OS |
PFS |
TTNTD |
|||||||
|
Characteristics |
group(n=282) |
mOS (m) |
p |
HR(95%CI) |
mPFS (m) |
p |
HR(95%CI) |
mTTNTD (m) |
p |
HR(95%CI) |
|
Metastatic breast cancer |
||||||||||
|
De novo |
53(18.79%) |
20.8 |
7.3 |
7.5 |
||||||
|
Recurrent |
229(81.21%) |
22.5 |
0.54 |
0.86(0.53-1.40) |
8.4 |
0.04* |
0.71(0.51-0.98) |
8.5 |
0.04* |
0.71(0.51-0.98) |
|
Sites of distant metastasis in 1L |
||||||||||
|
Bone |
50(17.73%) |
21.0 |
0.50 |
1.19(0.72-1.95) |
7.4 |
0.87 |
1.03(0.72-1.47) |
7.5 |
0.97 |
0.99(0.69-1.42) |
|
Brain |
13(4.61%) |
14.1 |
0.72 |
1.21(0.44-3.29) |
7.3 |
0.79 |
0.91(0.45-1.85) |
7.2 |
0.93 |
1.03(0.51-2.10) |
|
Liver |
43(15.25%) |
19.5 |
0.19 |
1.45(0.83-2.52) |
4.8 |
<0.001*** |
2.06(1.42-3.00) |
4.9 |
0.002** |
1.84(1.26-2.70) |
|
Lung |
93(32.98%) |
20.7 |
0.02* |
1.63(1.08-2.44) |
7.4 |
0.003** |
1.57(1.16-2.11) |
7.4 |
<0.001*** |
1.67(1.24-2.26) |
|
Lymph node |
48(17.02%) |
18.6 |
0.07 |
1.57(0.96-2.57) |
6.6 |
0.18 |
1.28(0.90-1.82) |
7.1 |
0.42 |
1.16(0.81-1.67) |
|
Other sites |
24(8.51%) |
19.8 |
0.20 |
1.49(0.81-2.73) |
7.4 |
0.41 |
1.22(0.77-1.93) |
7.2 |
0.26 |
1.30(0.82-2.07) |
|
TFI (month) |
||||||||||
|
TFI = 0 |
45(19.57%) |
13.8 |
|
5.9 |
5.9 |
|||||
|
0 < TFI ≤ 6 |
28(12.17%) |
14.7 |
0.63 |
0.83(0.39-1.77) |
5.4 |
0.25 |
1.41(0.79-2.52) |
6.5 |
0.31 |
0.79(0.51-1.23) |
|
6 < TFI ≤ 12 |
43(18.70%) |
17.4 |
0.29 |
0.70(0.36-1.37) |
6.4 |
0.60 |
1.15( 0.68-1.94) |
7.1 |
0.76 |
1.09( 0.63-1.87) |
|
TFI > 12 |
114(49.57%) |
31.4 |
<0.001*** |
0.33(0.19-0.57) |
11.9 |
0.22 |
0.76(0.49-1.18) |
11.9 |
0.25 |
1.41(0.79-2.53) |
|
1L treatment |
||||||||||
|
Platinum-free chemotherapy |
126(44.68%) |
23.6 |
6.9 |
7.2 |
||||||
|
Platinum-containing chemotherapy |
108(38.30%) |
20.3 |
0.97 |
0.99(0.64-1.55) |
8.6 |
0.21 |
0.83(0.61-1.12) |
8.6 |
0.41 |
0.88(0.65-1.19) |
|
ICI+chemotherapy |
21(7.45%) |
19.3 |
0.46 |
1.33(0.63-2.83) |
7.7 |
0.77 |
1.08(0.64-1.84) |
7.8 |
0.47 |
1.22(0.71-2.07) |
|
ADC+chemotherapy |
5(1.77%) |
9.1 |
0.10 |
3.34(0.80-13.95) |
4.8 |
0.46 |
1.55(0.49-4.91) |
7.9 |
0.59 |
1.38(0.43-4.37) |
|
ICI+ADC (clinical trial) |
4(1.42%) |
23.6 |
0.58 |
1.50(0.36-6.23) |
11.6 |
0.84 |
0.89(0.28-2.82) |
11.9 |
0.91 |
0.94(0.30-2.97) |
|
Other |
18(6.38%) |
32.1 |
0.31 |
0.62(0.25-1.57) |
13.8 |
0.04* |
0.53(0.29-0.97) |
15.9 |
0.05 |
0.55(0.30-1.00) |
Presentation numberPS5-03-08
Effectiveness of sacituzumab govitecan in later-line treatment of mTNBC: real-world evidence from Poland, Czech Republic, and Slovakia according to prior first-line pembrolizumab use
Marcin Kubeczko, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
J. Żubrowska1, M. Kubeczko2, M. Pieniążek3, A. Polakiewicz-Gilowska2, M. Malejčíková4, A. Konieczna5, M. Holánek6, I. Kolářová7, R. Soumarová8, H. Študentová9, A. Młodzińska5, K. Winsko-Szczęsnowicz10, M. Lisik-Habib11, A. Pękala11, D. Krejčí12, J. Šustr13, I. Danielewicz14, M. Szymanik-Resko14, T. Ciszewski15, L. Rusinova,16, A. Rudzińska17, B. Czartoryska-Arłukowicz10, A. Łacko18, M. Jarząb2, R. Pacholczak-Madej19, Z. Bielčiková20, M. Püsküllüoğlu21; 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, 4II. Oncology Clinic of LFUK, National Cancer Institute, Bratislava, SLOVAKIA, 5Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, POLAND, 6Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, and Masaryk Memorial Cancer Institute, Brno, CZECH REPUBLIC, 7Department of Oncology and Radiotherapy, Faculty of Medicine in Hradec Kralove and University Hospital in Hradec Kralove, Charles University, Hradec Králové, CZECH REPUBLIC, 8Department of Oncology, Third Faculty of Medicine, Charles University, University Hospital Kralovske Vinohrady, Prague, CZECH REPUBLIC, 9Department of Oncology, Faculty of Medicine and Dentistry, Palacky University and University Hospital, Olomouc, CZECH REPUBLIC, 10Department of Clinical Oncology, M. Skłodowska-Curie Bialystok Oncology Center, Białystok, POLAND, 11Department of Proliferative Diseases, Nicolaus Copernicus Multidisciplinary Centre for Oncology and Traumatology, Lodz, POLAND, 12Department of Oncology, First Faculty of Medicine, Charles University in Prague, and Bulovka University Hospital, Prague, CZECH REPUBLIC, 13Department of Oncology and Radiotherapy, Faculty of Medicine in Pilsen, Charles University, and University Hospital Pilsen, Plzen, CZECH REPUBLIC, 14Oddział Onkologii i Radioterapii, Szpitale Pomorskie sp. z o.o., Gdynia, POLAND, 15Department of Metabolic Diseases and Immuno-oncology, Medical University of Lublin, Lublin, POLAND, 16Department of Oncology, Stefan Kukura Hospital Michalovce, Michalovce, SLOVAKIA, 17Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND, 181. Department of Oncology 2. Breast Unit Clinical Oncology Day Care Department, Maria Sklodowska-Curie National Research Institute of Oncology, Wrocław, POLAND, 19Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow, POLAND, 20Department of Oncology, First Faculty of Medicine, Charles University, and General University Hospital, Prague, CZECH REPUBLIC, 21Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków, POLAND.
Background Patients with metastatic triple-negative breast cancer (mTNBC) have limited treatment options and poor outcomes in later lines. Sacituzumab govitecan (SG) has shown benefit in this setting, including among those previously treated with programmed death-1 (PD-1) inhibitors. In the ASCENT trial, outcomes were numerically worse in patients with prior immunotherapy. As SG is used after progression on first-line chemoimmunotherapy with the PD-1 inhibitor pembrolizumab, concerns arise about its efficacy in this subgroup. Several biological and clinical mechanisms could support the hypothesis that prior anti-PD-1 therapy might influence SG efficacy, including selection of resistant clones, immune remodeling, functional decline, absence of mechanistic synergy, and acquired resistance to SN-38. This study evaluates SG outcomes according to prior use of pembrolizumab-based first-line treatment in Poland, the Czech Republic, and Slovakia. Materials and Methods We conducted a retrospective analysis of patients with mTNBC treated with SG across 18 oncology centers between August 2021 and May 2025. Patients were stratified by prior exposure to first-line palliative chemoimmunotherapy with pembrolizumab, as defined by KEYNOTE-355 criteria. Baseline data included prior treatment history, metastatic burden, and starting dose of SG. Outcomes assessed were progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR), with tumor response evaluated per RECIST 1.1. Safety was analyzed based on adverse events (AEs) graded by CTCAE v5.0. Comparisons were performed using non-parametric and exact tests, and survival was analyzed using univariate Cox models with p<0.05 considered significant. Results Of the 302 evaluable women, 38 (12.6%) had previously received first-line anti-PD-1-based therapy. Baseline characteristics were similar between the groups. The median number of prior lines of palliative systemic therapy was 1 (IQR 1-2; range 1-7 in the anti-PD-1 group vs. 1-10 in the chemotherapy group; p=0.158). Brain metastases were present in 5/38 (13.2%) vs. 23/264 (8.7%) (p=0.372), and visceral metastases in 33/38 (86.8%) vs. 188/264 (71.2%) (p=0.066). A reduced starting dose of SG (≤8 mg/kg) was administered in 2/38 patients (5.3%) previously treated with first-line anti-PD-1 therapy and in 25/264 (9.5%) of others (p=0.550). Median OS was 10.74 months in the anti-PD-1 group and 11.43 months in the chemotherapy group (HR=1.30; 95% CI: 0.836-2.021; p=0.244). Median PFS was 3.22 vs. 4.44 respectively (HR=1.14; 95% CI: 0.782-1.662; p=0.497). ORR was 28.9% (11/38) in the anti-PD-1 group vs. 31.1% (82/264) in the chemotherapy group (p=0.939), while DCR was 60.5% (23/38) vs. 64.8% (171/264) (p=0.742). The median number of full SG cycles was 4 (IQR 2.25-9) in the anti-PD-1 group vs. 6 (IQR 3-11) (p=0.124). Toxicity profiles were similar. Grade ≥3 neutropenia occurred in 12/38 (31.6%) in the anti-PD-1 group and 124/264 (47.0%) in the chemotherapy group (p=0.137). Dose reductions due to AEs were required in 15/38 (39.5%) vs. 99/264 (37.5%) (p=0.956), and treatment delays occurred in 24/38 (63.2%) vs. 167/264 (63.3%) (p=1.000). Conclusions This real-world analysis provides insight into SG use after first-line pembrolizumab-based chemoimmunotherapy in mTNBC. While no clear reduction in efficacy or safety was observed, the impact of prior anti-PD-1 exposure remains clinically relevant. In light of the growing adoption of perioperative pembrolizumab and the anticipated shift of SG into earlier treatment lines, the impact of prior immunotherapy on SG performance should be further explored across diverse mTNBC settings and disease trajectories.
Presentation numberPS5-03-09
Trial in progress: A phase 2/3 trial of iza-bren (BMS986507/BL-B01D1), an EGFRxHER3 antibody-drug conjugate, vs standard-of-care chemotherapy in patients with previously untreated, locally advanced, recurrent inoperable, or metastatic triple negative breast cancer ineligible for anti-PD-(L)1 treatment (IZABRIGHT-Breast01)
Gustavo Werutsky, Latin American Cooperative Oncology Group, Porto Alegre, Brazil
S. Loi1, G. Werutsky2, Y. Park3, S. Loibl4, G. Curigliano5, D. Eiger6, J. Spiridigliozzi7, A. K. Dubey8, J. Cheng9, S. M. Tolaney10; 1Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 2Medical Oncology, Breast Cancer Program, Latin American Cooperative Oncology Group, Porto Alegre, BRAZIL, 3Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF, 4N/A, German Breast Group (GBG) and Centre for Haematology and Oncology, Neu-Isenburg, GERMANY, 5Department of Oncology and Hemato-Oncology, University of Milano and European Institute of Oncology IRCCS, Milan, ITALY, 6Clinical Development | Late Oncology Team 2, Bristol-Myers Squibb, Princeton, NJ, 7Department of Oncology Clinical Development, Bristol-Myers Squibb, Princeton, NJ, 8Global Biometrics and Data Sciences, Bristol-Myers Squibb, Princeton, NJ, 9Research and Development, SystImmune , Inc., Redmond, WA, 10Division of Breast Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.
Background: The epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor (HER) 3 are well-established oncogenic drivers frequently overexpressed in advanced cancers, including triple-negative breast cancer (TNBC) and HER2-negative BC. Their prevalence and critical role in tumor progression make these receptors suitable targets for iza-bren. Iza-bren is a potentially first-in-class antibody-drug conjugate (ADC) composed of an EGFR×HER3 bispecific antibody, conjugated to a novel topoisomerase 1 inhibitor payload (Ed-04) via a cleavable stable linker. Iza-bren has shown promising clinical activity and a manageable safety profile in early-phase clinical trials across various solid tumors, including pre-treated metastatic BC. Methods: IZABRIGHT-Breast01 (NCT06926868) is an open-label, randomized phase 2/3 trial evaluating the efficacy and safety of iza-bren vs chemotherapy of investigator’s choice (paclitaxel, nab-paclitaxel, capecitabine, or carboplatin plus gemcitabine) in patients with previously untreated, locally advanced, recurrent inoperable or metastatic TNBC or estrogen receptor (ER)-low, HER2-negative BC who are ineligible for approved anti-programmed death-1/programmed death ligand 1 (PD-[L]1)-based treatment combinations or endocrine therapy-based treatments, respectively. Key inclusion criteria include ≥ 18 years of age, ECOG performance status 0-1, measurable disease by RECIST v1.1, and either histologically or cytologically confirmed locally advanced, recurrent inoperable, or metastatic TNBC or ER-low, HER2-negative BC. Additional eligibility criteria include recent tumor biopsy collected (≤ 6 months), brain MRI or CT scan ≤ 28 days pre-randomization, and eligibility for ≥ 1 chemotherapy option. Key exclusion criteria include known germline BRCA 1/2 mutation with PARP inhibitor as best treatment option, untreated symptomatic CNS metastases, known bleeding disorders or conditions affecting bone marrow reserve, history of interstitial lung disease or pneumonitis, and prior therapy with iza-bren or any other ADC targeting EGFR and/or HER3 or containing a topoisomerase 1 inhibitor payload. In phase 2, patients will be randomized 1:1:1 to receive iza-bren at dose 1 (arm A1) or dose 2 (arm A2) on days 1 and 8 of each 3-week cycle, or chemotherapy of investigator’s choice (arm B). After a dose is selected, the study will proceed to phase 3 to confirm efficacy and safety with the selected dose of iza-bren. In phase 3, patients will continue to be randomized 1:1 into arms A and B. Randomization will be stratified by early recurrence (6-12 months after last treatment with curative intent) vs late recurrence (> 12 months) vs de novo disease presentation (no prior early-stage breast cancer), hormone receptor status (ER-low [1%-10%] and/or progesterone receptor [PgR]-low [1%-10%] vs negative [ER and PgR < 1%]), and intended chemotherapy (taxanes [nab-paclitaxel or paclitaxel] vs capecitabine vs carboplatin plus gemcitabine). Treatment will continue until disease progression or unacceptable toxicity. The primary endpoint is progression-free survival (PFS) per blinded independent central review using RECIST v1.1. The key secondary endpoint is overall survival. Other secondary endpoints include investigator-assessed PFS, objective response, disease control rate, duration of response, time to response, time to subsequent treatment, PFS after next line of treatment, safety, and health-related quality of life.
Presentation numberPS5-03-10
Btg2 suppresses the invasion and metastasis of triple-negative breast cancer by inhibiting the ubiquitination-mediated degradation of becn1
Hui Hui Li, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, China
H. H. Li, J. L. Zhang, D. D. Zhou, S. J. Sun, Y. S. Gao; Department of Breast Medical Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, Jinan, CHINA.
Objective: Triple-negative breast cancer (TNBC) is the most challenging subtype of breast cancer due to its high heterogeneity, aggressive metastatic potential, and lack of effective targeted therapies and predictive biomarkers. Although btg2 has been implicated as a tumor suppressor in tumorigenesis and cancer progression, its specific role in TNBC invasion and metastasis remains unclear. This study aims to elucidate the mechanism through which btg2 regulates the invasion and metastasis of TNBC by modulating the degradation of becn1. Methods: Using the TCGA database, we analyzed btg2 expression levels and their prognostic significance in TNBC, which we further validated through IHC staining of clinical TNBC samples. Functional assays, including cell viability, colony formation, transwell assays, as well as mouse xenograft experiments, were performed to assess the impact of btg2 on TNBC cell proliferation, migration, and invasion. To elucidate the underlying mechanism, we employed mass spectrometry (MS), co-immunoprecipitation (Co-IP), and immunofluorescence confocal microscopy to examine the interaction between btg2 and becn1. Additionally, Western blotting, qPCR, cycloheximide chase, and ubiquitination assays were conducted to determine how btg2 regulates becn1 protein stability and ubiquitination, including the potential involvement of rnf123 in btg2-mediated becn1 degradation. Results: btg2 was significantly downregulated in TNBC tissues, and this downregulation correlated with poor prognosis. Functional studies demonstrated that btg2 suppressed TNBC cell proliferation, colony formation, migration, and invasion, findings further corroborated by xenograft tumor models. Mechanistically, btg2 increased becn1 levels by inhibiting its proteasomal degradation. Specifically, btg2 expression reduced becn1 ubiquitination, prolonged its half-life, and activated autophagy. Furthermore, we discovered that btg2 stabilizes becn1 by binding to rnf123, thereby blocking rnf123-mediated ubiquitination of becn1. Conclusions: Our findings demonstrate that btg2 interacts with the E3 ligase rnf123 to suppress becn1 ubiquitination and proteasomal degradation, leading to autophagy activation and inhibition of TNBC invasion and metastasis. These results highlight btg2 as a potential diagnostic marker and therapeutic target for TNBC.
Presentation numberPS5-03-11
Combinatorial EZH2 Inhibition and Dopamine D1 Agonism Suppresses Pro-Inflammatory Monocytes and TNBC Tumor Progression
Eswar Shankar, The Ohio State University, columbus, OH
R. Shukla1, K. Ormiston1, G. Sarathy1, S. Dhekne1, D. Quiroga1, S. Gupta2, D. Stover1, P. Giglio3, C. Rolfo1, E. Shankar1; 1Internal Medicine, The Ohio State University, columbus, OH, 2Urology, The Case Western Reserve University, Cleveland, OH, 3Neurology, The Ohio State University, columbus, OH.
Background: Triple-negative breast cancer (TNBC) is an aggressive disease and lacks effective therapies due to tumor heterogeneity, resistance, and poor immune infiltration. There is a critical need to develop novel and less toxic therapies to improve TNBC survival outcomes. Treatment with Enhancer of Zeste Homolog 2 (EZH2) inhibitors have shown improved efficacy suggesting that these inhibitors play a pivotal role in TNBC progression. We have previously demonstrated that combined treatment with GSK126, an EZH2 inhibitor and dopamine D1 receptor agonist (A77636) exhibit higher efficacy in inhibiting tumor growth and metastasis of TNBC cells both in vitro and in vivo. Here, we examined the immune phenotype/function by which EZH2 and A77636 combination reprogram the TNBC microenvironment. Methods: Female NSG mice (4-6 weeks old) were orthotopically injected with MDA-MB-231 cells into the mammary fat pads. Once tumors became palpable, mice were randomized into four treatment groups (n = 8 per group): Vehicle control, GSK126 (2 mg/kg body weight), A77637 (50 mg/kg body weight), and the combination of GSK126 and A77637. Treatments were administered intraperitoneally once per week for four weeks. Tumor size was measured weekly using calipers, and tumor volume was calculated using the formula: (length × width²)/2. At the end of the treatment period, mice were euthanized. Tumors were excised and weighed. Tumor tissue, bone, blood, and spleen were harvested for flow cytometry analysis. Results: The combination of GSK126 and A77636 demonstrated a synergistic antitumor effect, significantly reducing both tumor weight and volume compared to vehicle or single-agent treatments. Tumor weight was significantly lower in the combination group compared to vehicle (mean difference = 0.278 g, 95% CI: 0.109-0.446, p = 0.0018). Similarly, tumor volume was significantly reduced in the combination group relative to vehicle (mean difference = 101 mm³, 95% CI: 51.7-151, p < 0.0001). This combinatorial treatment also led to a marked reduction in monocyte populations within both the peripheral blood and tumor microenvironment. EZH2 expression was significantly decreased in tumor-associated monocytes and neutrophils in the combination group. Kinetic analysis of tumor-infiltrating monocytes revealed a biphasic response: initial recruitment of pro-inflammatory Ly6Chi monocytes followed by a transition to anti-inflammatory Ly6Clo monocytes. Notably, the combination therapy significantly reduced Ly6Chi monocyte infiltration compared to the vehicle control (0.32 vs. 0.92, representing a 65% decrease; p = 0.0138). In contrast, it increased Ly6Clo monocyte levels relative to vehicle control (2.5 vs. 1.5), showing a 1.67-fold increase, (p = 0.5126) compared to single-agent and single-dose treatments. Furthermore, the combination group showed a trend toward increased production of the anti-inflammatory cytokine IL-10, while reduction of pro-inflammatory cytokine IL-1β production in both Ly6Chi and Ly6Clo monocyte. Conclusion: These findings demonstrate that dual targeting of EZH2 and A77636 not only suppresses TNBC tumor growth but also reprograms the tumor immune repertoire by shifting monocyte dynamics toward an anti-inflammatory phenotype, offering a promising strategy to overcome resistance and improve therapeutic outcomes. (This work is supported by DOD: W81XWH2010065, for Eswar Shankar)
Presentation numberPS5-03-12
Real-world BRCA and PD-L1 testing among first-line triple negative metastatic breast cancer patients
Fred Kudrik, South Carolina Oncology Associates, Columbia, SC
F. Kudrik1, R. Choksi2, D. Patt3, S. Reddy4, S. Reganti4, S. Rosenfeld5, S. W. Champaloux6, A. Shrivastava6, D. Parris6, A. Rui6, M. Gart6, C. Wall6, B. Wang6, P. Varughese6, J. Donegan6, L. Morere6, R. Geller6, J. Scott6, V. Gorantla2; 1Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 2Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 3Medical Oncology, Texas Oncology, Austin, TX, 4Medical Oncology, Cancer Care Specialists, Reno, NV, 5Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 6PrecisionQ, IntegraConnect, West Palm Beach, FL.
Background: Triple negative breast cancer is a less common, aggressive subtype that lacks the expression of estrogen and progesterone receptors as well as HER2, making standard endocrine and HER2-directed therapies ineffective. Current NCCN guidelines recommend germline BRCA testing for all patients (pts) with metastatic breast cancer to identify candidates for poly (ADP-ribose) polymerase (PARP) inhibitors. In addition, PD-L1 testing, defined as a combined positive score (CPS) ≥10, guides utilization of pembrolizumab in combination with chemotherapy. Taken together, biomarker testing is an important component in guiding care. The study describes patterns and timing of BRCA and PD-L1 testing among pts with mTNBC who initiated therapy. Methods: Pts with metastatic triple-negative breast cancer (mTNBC) who initiated therapy on or after January 1, 2021, were identified from the IntegraConnect PrecisionQ database, which includes electronic health records from over 3 million de-identified pts in the U.S. Metastatic status and biomarker information were verified through manual chart review, including extraction from unstructured clinical notes, genomic reports and pathology reports. The review captured hormone receptor and HER2 status, as well as documentation of BRCA and PD-L1 testing. Descriptive analyses compared testing rates by demographics, ECOG status, recurrent vs de novo presentation, and treatment patterns. Results: Overall, 720 curated pts were identified as having mTNBC and initiated a line of therapy on or after 1/1/2021 (date initiated minimum 1/1/2021; maximum 3/5/2025). Of the 720 pts, 651 (90%) received a BRCA genetic test, yet just 61% (n=437) were tested prior to the mTNBC therapy initiation date and among those 71% (N =311) received Germline testing. Out of the 651 pts who received a BRCA test, germline testing was recorded in 463 pts (71%), somatic testing in 536 pts (82%) and both types of tests were identified in 361 pts (55%). Prior to mTNBC therapy initiation, 42% (n=303) were tested for PD-L1 and 83% (N=600) were tested for PD-L1 at any time. The mean age for pts at treatment initiation date differed for those who received a BRCA test at any time compared to those who did not receive a BRCA test (mean age at treatment initiation date: BRCA tested 60 years old versus BRCA Not tested: 63 years old). Distribution of ECOG scores did not differ by PD-L1 or BRCA testing status at time of treatment of mTNBC. BRCA testing was similar across racial groups, but PD-L1 testing was lower in the African American population (68% vs. 76%). Out of all the PD-L1 CPS tested pts (N=384), a positive result, defined as a CPS that was ≥10, occurred in 49% (N=188) of cases and a negative result occurred in 51% (N=196) of cases. Among those who received a PD-L1 test after initiation of therapy, the median time to test was 29 days (interquartile range 10-151 days). Pts with recurrent disease had similar testing rates compared to those with de novo disease for both BRCA (89% vs. 91%) and PD-L1 (74% vs. 73%) testing. In the pts who tested positive for BRCA and negative for PD-L1 (n=36), PARP inhibitors were used in a subsequent line of therapy for 19 pts (53%). In the pts with a PD-L1 CPS ≥10 prior to mTNBC treatment initiation and did not receive pembrolizumab in the neo-adjuvant or adjuvant setting (n=166), pembrolizumab was utilized in 92 pts (55%). Conclusions: Out of all pts who were tested, only 42% of pts received PD-L1 testing and 61% received BRCA testing prior to initiating first-line treatment for mTNBC. Further opportunities for timely testing should be explored to optimize treatment choice.
Presentation numberPS5-03-13
Combinatory inhibition of Aurora Kinases and EZH2 leads to a synergistic antitumor effect in Triple Negative Breast Cancer
Jia Xu, University of Alabama at Birmingham, Birmingham, AL
K. Graciano1, C. Liu1, B. Ning1, H. Guo2, J. Xu1; 1Genetics, University of Alabama at Birmingham, Birmingham, AL, 2Pathology, University of Alabama at Birmingham, Birmingham, AL.
Triple-negative breast Cancer (TNBC) accounts for around 15-20% of all breast cancers and is associated with poor long-term outcomes. It remains the most challenging subtype to treat because of its aggressive phenotype and limited treatment options. Aurora Kinases are overexpressed in multiple types of solid tumors, including breast cancer. Our preliminary studies discovered that AURKB expression disproportionally increased in African American TNBC patients more than in European American TNBC patients. Aurora kinase inhibitors, such as alisertib (MLN8237), have demonstrated early signals of efficacy with a manageable safety profile for the treatment of patients with locally advanced or metastatic breast cancer (NCT02860000). However, they did not show significant efficacy in TNBC unless combined with chemotherapy. EZH2, a histone methyltransferase and catalytic subunit of Polycomb Repressive Complex 2 (PRC2), has a broad role in cell stemness, and its overexpression promotes the development of many cancers, including TNBC. Multiple independent studies have identified EZH2 as an attractive drug target in TNBC. Several EZH2 inhibitors, which inhibit the methyltransferase activity of EZH2, have been approved for the treatment of sarcoma and follicular lymphoma in clinical settings. However, EZH2 inhibitors are ineffective at inhibiting TNBC cell growth. We unexpectedly discovered that Aurora Kinases interact with and phosphorylate EZH2, and inhibition of Aurora Kinases upregulates the chromatin silencing marker H3K27me3. Moreover, combined inhibition of EZH2 and Aurora kinases shows a synergistic antitumor effect in TNBC. Phosphorylation of EZH2 by Aurora Kinase could retain EZH2 in the cytosol, leading to a functional shift from a PRC2 complex-dependent activity to a potential non-canonical EZH2 pro-metastatic function. Furthermore, RNA-seq analysis of the TNBC cells treated with Aurora Kinases inhibitors (AKi) Barasertib (AZD1152) and/or EZH2 PROTAC degrader MS177 reveals that AKi inhibits the expression of a panel of genes, which EZH2 degrader also regulates. Combinatory treatment leads to a synergistic effect on this panel of gene expression, which is critical for cell apoptosis, proliferation, and metabolism in TNBC cells. To summarize, we discovered Aurora Kinases have a less-studied, non-canonical oncogenic function, which acts in parallel with the canonical cell division activity to produce more aggressive tumor metastasis phenotypes seen in TNBC. This non-canonical function highly depends on EZH2-mediated cancer cell apoptosis, proliferation, invasion, and metastasis, which differs from the well-known cell division function of Aurora Kinases, regulating chromosomal alignment, segregation, and cytokinesis during mitosis.
Presentation numberPS5-03-14
Clinical features and drug-drug interactions of patients treated with Sacituzumab Govitecan for metastatic triple-negative breast cancer: a multicenter correlative real-world updated analysis
Claudia Martinelli, Istituto Nazionale Tumori-IRCCS “Fondazione G. Pascale”, Naples, Italy
R. Caputo1, C. Martinelli1, M. Piezzo1, G. Buono1, V. Di Lauro1, R. Buonaiuto1, A. Verrazzo1, D. Cianniello1, M. V. Bonomo2, F. Pantano3, S. Scagnoli2, S. Cocco1, G. R. Ricciardi4, F. Giotta5, L. S. Stucci6, N. Staropoli7, S. Turano8, M. V. Sanò9, P. Trasacco10, L. Orlando11, L. Del Mastro12, A. Fabi13, M. De Laurentiis1, A. Botticelli2; 1Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori-IRCCS “Fondazione G. Pascale”, Naples, ITALY, 2Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I-Sapienza University of Rome, Rome, ITALY, 3Department of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, ITALY, 4Department of Onco-hematology, Papardo Hospital, Messina, ITALY, 5Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, ITALY, 6Medical Oncology, Policlinico Hospital of Bari, Bari, ITALY, 7Department of Experimental and Clinical Medicine, Magna Graecia University, Azienda Ospedaliera Universitaria R. Dulbecco Catanzaro, Catanzaro, ITALY, 8Department of Oncohematology, Azienda Ospedaliera di Cosenza, Cosenza, ITALY, 9Medical Oncology, Istituto Clinico Humanitas, Misterbianco, Catania, ITALY, 10Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, ITALY, 11Medical Oncology, “Antonio Perrino” Hospital, Brindisi, ITALY, 12Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genoa, ITALY, 13Precision Medicine in Senology, Scientific Directorate – Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, ITALY.
Background: Sacituzumab Govitecan (SG), an antibody-drug conjugate (ADC) targeting TROP2, has demonstrated significant improvements in progression-free survival (PFS) and overall survival (OS) compared to standard chemotherapy in previously treated patients with metastatic triple-negative breast cancer (mTNBC). However, drug-drug interactions (DDIs), particularly in real-world settings, may influence the pharmacological activity of anticancer agents, potentially affecting both treatment efficacy and toxicity. Drug-PIN® (Personalized Interactions Network) is a tool designed to detect DDIs and integrate them with demographic, clinical, and biochemical patient data. In this multicenter retrospective analysis, we aim to evaluate the impact of clinical toxicites and DDIs on treatment outcomes in patients receving SG in a real world context.Methods: Toxicities, drug-drug interactions (DDIs), and clinical outcomes—including PFS and OS— in patients with mTNBC treated with SG in routine clinical practice were retrospectively collected from 11 Italian centers, as part of the NCT02284581 multicenter study. DDIs were assessed using the Drug-PIN® platform, which generated a Drug-PIN® score and assigned each patient to a Drug-PIN® tier: green (no clinically significant DDIs), and yellow to red (increasing DDI severity). Patients with relevant DDIs (tiers yellow to red) were compared to those without significant interactions (tier green). PFS and OS were estimated using the Kaplan-Meier method. The association between Drug-PIN® classification and clinical outcomes was assessed through multivariate logistic regression.Results: A total of 123 pts were included. Patient’s characteristics are reported in table 1. Median time from diagnosis of mBC to SG start was 17 months (range 0- 117). Among pts taking any medications, 20 pts (16.3%) had a relevant DDIs. Median rwPFS was 5.0 months in pts without any DDIs and 2.8 months in patients with DDIs [HR 1.89 (1.14-3.15) p 0.0138]. Using a multivariate model, both Drug-PIN tier [green vs other; HR 2.18 (1.3-3.5); p 0.0015] and presence of brain metastases at baseline [HR 1,62 (1,07-2,47) p 0,022] resulted as independent predictor of shorter PFS. Median OS was numerically higher but not statistically significant (p 0.098) in pts without DDIs [7.57 months vs 5.3 months [HR 1.69 (0.91-3.15)]. No significant association among DDIs and the report of common treatment related toxicity or dose reduction was detected.Conclusion: In our real-world cohort, a low rate of clinically relevant DDIs was observed. While DDIs were associated with shorter PFS, no differences in toxicity or dose modifications emerged. Further investigations are needed to assess the potential impact of DDIs in mBC pts treated with ADCs.
| Characteristic | N = 123 |
| Median age at SG start (range) | 53 (26-78) |
| > 60 years, no. (%) | 36 (29.3) |
| ≤ 60 years, no. (%) | 87 (70.7) |
| Median number of previous anticancer regimens for metastatic disease (range) | 2 (0-7) |
| Major tumor locations | N (%) |
| Lung | 61 (49.6) |
| Liver | 40 (32.5) |
| Lymph nodes | 77 (62.6) |
| Brain | 33 (26.9) |
| Bone | 61 (49.6) |
| Comorbidities | N (%) |
| 1-2 | 56 (45.5) |
| ≥3 | 11 (9.0) |
| Concomitant drugs | N (%) |
| 1-2 | 46 (37.4) |
| 3-5 | 30 (24.4) |
| ≥6 | 11 (8.9) |
| Median drug pin score (range) | 8.85 (0-155) |
| Drug pin tier | N (%) |
| No concomitant drugs/Green | 101 (82.1) |
| Yellow | 14 (11.4) |
| Dark Yellow | 6 (4.9) |
| Red | 2 (1.6) |
Presentation numberPS5-03-15
Real-world outcomes with sacituzumab govitecan in patients with triple-negative breast cancer and central nervous system metastases
Roberta Caputo, Istituto Nazionale Tumori-IRCCS “Fondazione G. Pascale”, Naples, Italy
R. Caputo1, M. Piezzo1, C. Martinelli1, G. Di Mauro2, V. Granata3, M. Pagliuca4, A. Botticelli5, F. Pantano6, F. Giotta7, A. Diana8, L. S. Stucci9, N. Staropoli10, S. Turano11, E. Bajardi12, G. Pernice13, M. V. Sanò14, A. Caltavituro15, L. Orlando16, S. Scagnoli5, C. De Angelis4, A. Fabi17, G. Buono1, M. De Laurentiis1; 1Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori-IRCCS “Fondazione G. Pascale”, Naples, ITALY, 2School of Specialization in Medical Oncology, University of Messina, Naples, ITALY, 3Divisions of Radiology, Istituto Nazionale Tumori-IRCCS “Fondazione G. Pascale”, Naples, ITALY, 4Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, ITALY, 5Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I-Sapienza University of Rome, Rome, ITALY, 6Department of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, ITALY, 7Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, ITALY, 8Medical Oncology Unit, Ospedale del Mare, Naples, ITALY, 9Medical Oncology, Policlinico Hospital of Bari, Bari, ITALY, 10Department of Experimental and Clinical Medicine, Magna Graecia University, Azienda Ospedaliera Universitaria R. Dulbecco Catanzaro, Catanzaro, ITALY, 11Department of Oncohematology, Azienda Ospedaliera di Cosenza, Cosenza, ITALY, 12Medical Oncology, Casa di Cura La Maddalena, University of Palermo, Palermo, ITALY, 13Medical Oncology, Fondazione Istituto Giglio, Cefalù, ITALY, 14Medical Oncology, Istituto Clinico Humanitas, Misterbianco, Catania, ITALY, 15Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, ITALY, 16Medical Oncology, “Antonio Perrino” Hospital, Brindisi, ITALY, 17Precision Medicine in Senology, Scientific Directorate – Department of Women and Child Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, ITALY.
Background: Central nervous system (CNS) metastases, including brain metastases (BMs) and leptomeningeal disease (LMD), occur in approximately 20%-40% of patients (pts) with breast cancer during the course of their disease. Triple-negative breast cancer (TNBC) is associated with a high incidence of BMs (up to 34%) and poor survival rates. Sacituzumab Govitecan (SG) has demonstrated significant improvements in progression-free survival (PFS) and overall survival (OS) in pre-treated pts with metastatic TNBC (mTNBC) in the pivotal ASCENT trial. However, data on SG efficacy in pts with TNBC and CNS metastases are limited, as only 32 pts with stable/treated BMs were enrolled in the study, and patients with active BMs or LMD were excluded. This study aimed to provide further insight into the clinical activity of SG in pts with mTNBC and CNS involvement. Methods: Clinical and treatment data of pts with TNBC and BMs who were treated in a real-life setting were retrospectively collected from 14 Italian centers as part of a multicenter, observational study (NCT02284581). CNS treatment response was retrospectively categorized as objective response, stable disease, or progressive disease. Real-world (rw)PFS, rwOS, and CNS-specific progression-free survival (CNS-PFS) were described using the Kaplan-Meier method and reported with a 95% confidence interval. Survival outcomes between groups were compared by log-rank tests. Analysis was carried out with R software (version 4.5.1). Results: A total of 54 pts were included in the analysis. Of these, 46 (85.2%) had treated/stable CNS metastases, 4 (7.4%) had active CNS metastases, and 4 (7.4%) had LMD. The median age at SG initiation was 54 years (range: 31-75). The median time from mTNBC diagnosis to CNS metastasis was 12 months, and the median time from CNS metastasis diagnosis to SG initiation was 5 months. Pts had received a median of 2 prior lines of therapy before CNS involvement. At a median follow-up of 18.2 months, the median rwPFS was 3.75 months (95% CI, 3.06-7.89 months) and the median rwOS was 9.49 months (95% CI, 6.21-15.3). The CNS objective response rate (ORR) was 19.6% among pts with treated/stable BMs, and 25% in both pts with active BMs and those with LMD. The median CNS-PFS in the overall population was 9.26 months (range: 4.8-16.2), with subgroup medians of 12.1 months for stable/treated BMs, 4.8 months for active BMs, and 4.1 months for LMD. The table provides a summary of the efficacy results.Conclusion: To our knowledge, this study represents the largest real-world analysis to date assessing the activity of SG in pts with TNBC and CNS metastases. Our findings are consistent with the efficacy profile observed in the ASCENT trial. These data support the potential role of SG in this difficult-to-treat population, although further studies are warranted to better characterize its activity in pts with mTNBC and CNS involvement.
| Response to SG | Overall population N = 54 | Treated/Stable BM N = 46 | Active BM N = 4 | LMD N = 4 |
| CNS response* – n (%) | ||||
| Response (complete or partial) | 11 (20.4%) | 9 (19.6%) | 1 (25%) | 1 (25%) |
| Stable disease | 9 (16.6%) | 7 (15.2%) | 0 | 2 (50%) |
| Progressive disease | 20 (37.0%) | 16 (34.8%) | 3 (75%) | 1 (25%) |
| Not evaluable | 14 (26.0%) | 14 (30.4%) | 0 | 0 |
| Disease Control Rate | 20 (48.8%) | 16 (34.8%) | 1 (25%) | 3 (75%) |
| Clinical Benefit Rate at 6 months | 8 (14.8%) | 8 (17.4%) | 0 | 0 |
| Median CNS-PFS | ||||
| Months (95% CI) | 9.26 (4.8-16.2) | 12.1 (4.07-16.2) | 4.8 (2.0-NR) | 4.1 (2.8 – NR) |
Presentation numberPS5-03-16
Clinical Outcomes of ER-Positive to Negative Conversion Versus TNBC: Evidence from an<sup>18</sup>F-FES PET/CT Guided Propensity-Score-Matched Study
Biyun Wang, Fudan University Shanghai Cancer Center, Shanghai, China
B. Wang1, M. Li1, Y. Xie1, Z. Yang2; 1Department of Breast and Urological Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 2Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Background:Patients with estrogen receptor (ER)-positive breast cancer may experience ER-negative conversion (ER loss) during disease progression. 18F-fluoroestradiol positron emission tomography/computed tomography (18F-FES PET/CT) offers a practical approach for comprehensive ER evaluation across all metastatic sites, allowing early identification of ER conversion—especially valuable for cases where biopsy is technically challenging. This study aimed to compare survival outcomes and clinical characteristics between ER loss and metastatic triple-negative breast cancer (mTNBC) patients, exploring the prognostic implications of dynamic ER changes. Methods:This retrospective study screened 808 patients with ER-positive, HER2-negative breast cancer at initial diagnosis who underwent 18F-FES PET/CT after diagnosis of recurrent or metastatic disease from 2017 to 2023 at Fudan University Shanghai Cancer Center. Among them, 82 patients with functional ER loss detected by FES-PET at metastatic sites before first-line treatment were identified. 115 contemporaneous standard mTNBC patients with complete medical records were enrolled in control group. Propensity score matching (PSM) was used to balance baseline characteristics (number of metastatic sites, visceral/liver/lung/bone metastasis, taxane therapy history; caliper = 0.1). Results:PSM matched 59 patients per group and baseline clinical characteristics are well-balanced. FES-PET showed 71.7% concordance with IHC. Median progression-free survival (PFS) was significantly shorter in ER loss group compared with mTNBC (6.7 vs 12.0 months, HR=1.55, P=0.042). Objective response rate (ORR) (15.3% vs 37.3%) and disease control rate (DCR) (72.9% vs 84.7%, P=0.044) favored mTNBC, as well. Androgen receptor (AR) expression is available in 25 and 32 patients in ER loss and mTNBC group, respectively, and AR pathway positivity (≥10%) was significantly higher in ER loss group (88.0% vs 25.0%, P<0.0001). 28 of 59 (47.5%) ER loss patients received chemotherapy, and 18 of them received capecitabine based first-line therapy, while 34 of 59 (57.6%) mTNBC patients received platinum-based chemotherapy and 8 of 59 mTNBC patients had PD-1/PD-L1 exposure in first-line therapy. Conclusions:Our study reveals that patients with functional ER loss defined by FES-PET exhibit worse outcomes than mTNBC, potentially attributed to lighter treatment intensity and AR pathway activation. These findings suggest that functional ER-negative conversion, detectable via whole-body FES PET/CT, may identify a distinct high risk subgroup requiring TNBC-level treatment intensification. Future studies should investigate underlying mechanisms and evaluate AR-targeted therapies in this population.
Presentation numberPS5-03-17
Ai-derived Morphometric And Transcriptomic Biomarkers From H&E-stained Images Predict Response To Durvalumab And Olaparib In Metastatic Triple Negative Breast Cancer
Hassan Muhammad, PATHOMIQ, Inc., Cupertino, CA
H. Muhammad1, S. S. Chavan1, C. Feng1, SMMART Clinical Trials Program, H. S. Basu1, W. Huang1, R. Rajat1, G. Wilding1, S. Kummar2, G. B. Mills3; 1AI, PATHOMIQ, Inc., Cupertino, CA, 2Molecular Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 3Precision Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR.
Introduction: PARP inhibitors (PARPi) have demonstrated the potential to enhance tumor immunogenicity and sensitize cancer cells to immune checkpoint blockade, as shown in the AMTEC clinical trial. However, identifying PARPi responders and understanding the tumor’s adaptive response remain significant clinical challenges. Advances in multi-omic profiling offer unprecedented resolution into these dynamics, yet the complexity and volume of such data demand sophisticated analytical approaches. By applying AI to histopathology images, we aim to uncover predictive morphogenomic biomarkers of PARPi response, potentially enabling more precise treatment selection and to reveal novel mechanisms of therapeutic adaptation in the tumor and its immune microenvironment. Method: We applied two complementary approaches to identify predictive morphometric and morphogenomic biomarkers in biopsy samples collected during the AMTEC trial. Using the PATHOMIQ Phenotype Atlas, we quantified fold changes in individual morphological phenotypes between matched pre-treatment and on-treatment biopsies to identify morphology-based predictors of treatment response. Using the PATHOMIQ Genoscope, a deep learning model that infers RNA expression from H&E-stained slides, we estimated gene expression profiles directly from histology images. Genes that were significant in univariate Cox modeling were evaluated in multivariate models to identify transcriptomic predictors. Result: To assess phenotypic changes across serial biopsies, we computed fold changes in the prevalence of individual morphological phenotypes between pre-treatment and on-treatment samples. Specific morphometric changes related to desmoplasty and necrosis were significantly associated with progression-free survival (PFS). Their emergence in on-treatment biopsies enabled clear patient stratification, as confirmed by a log-rank test (p < 0.001). Separately, univariate Cox proportional hazards modeling identified 15 genes, predicted directly from H&E images via Genoscope, as significantly associated with PFS. A multivariate model incorporating the top-ranked genes along with FOXC1 demonstrated robust stratification of patient outcomes. Kaplan-Meier estimation using a median-expression cutoff revealed a significant survival difference (p < 0.01, log-rank test). Discussion: These results show that AI-based analysis of H&E slides can identify both morphological and gene expression markers predictive of PARP inhibitor response. The identification of these desmoplastic changes during treatment may signal early resistance and these necrotic changes signal early response, while inferred expression of genes such as FOXC1, a known biomarker, was linked to poor progression-free survival. This morphogenomic approach enables non-destructive, scalable patient stratification and may support real-time treatment adaptation in future studies.
Presentation numberPS5-03-18
Clinicopathological features and outcomes of triple-negative breast cancer relapsing after pembrolizumab-based neoadjuvant therapy.
Delphine Loirat, Institut Curie – Institute of Women’s Cancers, Paris, France
R. Kabirian1, M. Gendreau1, F. C. Bidard1, F. Lerebours2, A. Vincent Salomon3, L. Djerroudi3, P. H. Cottu1, F. Coussy1, J. Y. Pierga1, L. Cabel1, D. Loirat1; 1Medical Oncology department, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 2Medical Oncology department, Institut Curie – Institute of Women’s Cancers, Saint-Cloud, FRANCE, 3Department of Diagnostic and Theranostic Medicine, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE.
Background: Neoadjuvant pembrolizumab combined with paclitaxel and carboplatin followed by anthracycline and cyclophosphamide has become the standard of care for stage II-III early triple-negative breast cancer (TNBC). However, clinicopathological features and outcomes of metastatic relapse after neoadjuvant chemo-immunotherapy remain poorly documented. Methods: We conducted a retrospective analysis at Institut Curie Hospitals (France) including patients diagnosed with a metastatic relapse between Nomvember 2022 and May 2025, after having received pembrolizumab-based neoadjuvant regimen for early TNBC. Patients’ characteristics at early and metastatic settings were collected, as well as Distant Metastasis-Free Interval (DMFI, from primary diagnosis to metastatic relapse), Progression-Free Survival on 1st line therapy (PFS1, from the date of metastatic relapse) and Overall Survival (OS, from the date of metastatic relapse). Results: We identified N=32 patients out of 386 patients treated with neoadjuvant pembrolizumab who experienced metastatic relapse following a pembrolizumab-based neoadjuvant regimen. At primary diagnosis, all 32 tumors were TNBC, including N=3 tumors (9%) with low (1-9%) expression of hormone receptors and N=13 tumors (41%) with HER2low status. Strikingly, androgen receptor (AR) expression was apparently enriched, with N=8/22 (36%) primary tumors showing high (≥10%) AR expression. Most primary tumors were of grade 3 (N=25/32, 78%) and stage II (N=20, 63%). Only N=2/29 (7%) patients displayed a germline BRCA1/2 mutation. Following neoadjuvant chemo-immunotherapy, N=30 patients underwent surgery, of whom only N=5 patients (17%) had a pCR (RCB=0). Post-operative treatments were heterogeneous: N=11 patients (37%) received adjuvant pembrolizumab monotherapy, N=9 (30%) received capecitabine monotherapy, N=5 (17%) received a combination of both, N=3 (10%) were included in a clinical trial while N=2 (7%) relapsed before any adjuvant therapy. The median DMFI from initial diagnosis was 16 months, IQR[9-19], and most relapses (N=23, 72%) occurred during or within 6 months after the completion of adjuvant therapy. At relapse, median age was 43 years (IQR[37-50]). Visceral metastases were present in N=22 patients (69%), while brain and bone-only metastases were identified in N=6 (19%) and N=3 (9.4%) patients, respectively. A prior or concurrent local recurrence was observed in N=12 of patients (38%). Although N=13/24 tumors (54%) displayed a high PD-L1 CPS score, only 3 patients (9%) were rechallenged with pembrolizumab and chemotherapy as first line therapy. N=11 patients (34%) received standard chemotherapy, N=16 patients (50%) received sacitumab govitecan (SG) when olaparib and experimental therapy were each administered to N=1 patient (3%). After a median follow-up of 11.2 months, median PFS1 was 3.9 months [95%CI:2.4-5.5]. We found no prognostic impact of disease sites and no survival difference between standard chemotherapy (median PFS1: 3.7mo) and SG (4.1mo). N=15 deaths have been observed, with a median OS of 8.5 months [95%CI: 7.7-NA]. Updated results with longer follow-up will be presented. Conclusions: This real-world cohort of patients with TNBC relapsing after pembrolizumab-based neoadjuvant therapy shows an extremely poor prognosis upon metastatic recurrence, regardless of metastatic site and first line therapy. There remains a critical need to improve treatment strategies in this population.
Presentation numberPS5-03-19
Label-free multimodal multiphoton microscopy reveals spatial lipid alterations associated with inflammation in triple-negative breast cancer following NOS2 suppression and COX-2 inhibition
Wesley Poon, Texas A&M Health Science Center, Bryan, TX
W. Poon1, R. Uzma2, R. Master3, L. Wang4, H. Zhao4, A. Veeraraghavan2, L. Ridnour5, S. J. Lockett6, R. Raghunathan4, D. A. Wink5, S. T. Wong4; 1Department of Medical Sciences, Texas A&M Health Science Center, Bryan, TX, 2Department of Electrical and Computer Engineering, Rice Univeristy, Houston, TX, 3School of Engineering Medicine, Texas A&M University, Houston, TX, 4Department of Systems Medicine and Bioengineering, Houston Methodist, Houston, TX, 5Center for Cancer Research, National Cancer Institute, Frederick, MD, 6Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD.
Triple-negative breast cancer (TNBC, 15–20% of breast cancers) has poor prognosis, high metastatic potential, and limited treatment options due to the absence of estrogen receptor (ER), progesterone receptor (PR), and HER2 amplification. The tumor microenvironment (TME) plays a key role in driving progression, with inflammatory enzymes such as inducible nitric oxide synthase (NOS2) and cyclooxygenase-2 (COX-2) upregulated in aggressive subtypes. High NOS2 expression in TNBC has been linked to poor overall survival, possibly due to its role in shaping an immunosuppressive microenvironment and activating pro-metastatic signaling cascades. Elevated COX-2 expression is also a hallmark of TNBC, where it facilitates M2 macrophage polarization, regulatory T cell recruitment, and resistance to immune checkpoint blockade. However, their influence on tumor metabolism, lipid accumulation, and collagen dynamics remains unclear. The TME is also spatially heterogeneous, made up of diverse niches that lead to therapy resistance. Microscopy offers insight into these regions, but exogenous dyes hinder translation to live tissues. To overcome this, we employed a label-free, multimodal multiphoton microscopy platform combining second harmonic generation (SHG) for collagen, two-photon autofluorescence (TPAF) for redox metabolism, and coherent anti-Stokes Raman scattering (CARS) for lipid imaging. We examined how NOS2-derived nitric oxide depletion and COX-2 inhibition via indomethacin affect TNBC metabolism, lipid profiles, and collagen architecture. Orthotopic 4T1 TNBC tumors were established in 4 cohorts of BALB/c mice: Group 1 (n=5, wild type (WT) untreated), Group 2 (n=7, WT + indomethacin), Group 3 (n=5, NOS2 knockout (NOS2-/-)), and Group 4 (n=7, NOS2-/- + indomethacin). 12-15 mm diameter tumors were excised, sectioned, and imaged ex vivo. A dual-output ultrafast tunable laser (Insight DeepSee, Santa Clara, USA) was used as the optical source of the label-free multimodal platform. For CARS, TPAF of NADH, TPAF of FAD, and SHG, the following excitation wavelengths were used: 803 nm combined with 1040 nm, 755 nm, 860 nm, and 803 nm. Samples were imaged at 25x magnification (0.94 µm/pixel). Multimodal images of whole slices were acquired and processed using MATLAB. SHG images were quantified by collagen area fraction (CAF), anisotropy parameter, Hough transform-based standard deviation, and Fourier transform-based metrics. Redox ratio was used to assess TPAF images. Size distribution of lipid droplets through CARS was also analyzed. CARS imaging revealed distinct lipid remodeling across both NOS2-NO depletion and indomethacin treatment. NOS2- tumors show a significant increase in large lipid droplets (1,000–10,000 pixels) compared to wild-type, suggesting enhanced lipid accumulation suggesting altered storage or β-oxidation. Indomethacin-treated tumors, independent of NOS2, had a greater share of small lipid droplets (1–10 pixels), reflecting altered lipolysis or turnover. The combined treatment in NOS2- mice further augmented the accumulation of small lipid droplets (p<0.0075), supporting a synergistic effect. Global SHG and TPAF analyses did not reveal significant differences, but initial regional analyses are ongoing to assess collagen structure and metabolic redox states in peritumoral and immune-rich zones. Early inspection suggests disorganized collagen in immune dense regions of NOS2- tumors, potentially facilitating immune cell infiltration.
Presentation numberPS5-03-20
Impact of granulocyte-colony stimulating factor use on clinical outcomes in metastatic breast cancer patients receiving chemotherapy<b></b>
Mara Hofherr, Washington University, St. Louis, MO
M. Hofherr; Medical Oncology, Washington University, St. Louis, MO.
Background: The mainstay of triple negative breast cancer (TNBC) continues to be cytotoxic treatment, specifically carboplatin/gemcitabine, carboplatin/paclitaxel or sacituzumab govitecan. The most common adverse effect of these regimens is dose-dependent and dose-limiting neutropenia. Recent meta-analyses of randomized controlled trials (RCTs) have shown a reduction in all-cause mortality in patients receiving prophylactic G-CSF with cytotoxic chemotherapy as well as an association with improved overall survival (OS). However, these meta-analyses did not exclusively look at metastatic breast cancer patients. To the authors knowledge, this is the first real-world evidence study looking at G-CSF use in metastatic triple negative breast cancer. This study investigated the use of prophylactic G-CSF agents to determine if metastatic breast cancer patients receiving intermediate or high-risk chemotherapy regimens remained on treatment longer, resulting in less febrile neutropenia and better outcomes. Methods: In this retrospective, single center study we examined pts with early-stage breast cancer who received and discontinued denosumab or zoledronic acid. IRB approval was obtained. Number and length of treatments, fractures of any kind, and delay in BMA were collected from the electronic medical record using diagnosis codes. The objectives of this study were to describe the rate and severity of neutropenia, febrile neutropenia, progression-free survival (PFS) metastatic TNBC who received carboplatin/gemcitabine, carboplatin/paclitaxel or nab-paclitaxel, and sacituzumab govitecan. Results: A total of 197 patients were included with 79 receiving carboplatin plus gemcitabine, 20 receiving carboplatin plus paclitaxel or nab-paclitaxel, and 98 receiving Sacituzumab govitecan. The primary reason for exclusion was having not received the chemotherapy regimen despite having the treatment plan ordered. The median PFS was 4.9 months (95 % confidence interval [CI], 3.5 – 6.3) in the G-CSF group compared to 2.5 months in the no G-CSF group (95% CI, 2.2 – 3.2). PFS was significantly longer in the G-CSF group than in the no-G-CSF group (hazard ratio for disease progression or death, 0.66; 95% CI, 0.453 – 0.951; p = 0.026). The percentage of patients who were alive and free from progression at 12 months was 26.8% and 20.5% at 24 months. The incidence of febrile neutropenia was higher in the G-CSF group than in the no-G-CSF group (18 and 4 incidences, respectively; p = 0.012). However, when comparing patients who received primary prophylaxis and secondary prophylaxis, those receiving primary prophylaxis had a lower incidence of febrile neutropenia (4 and 14, respectively; p = 0.009). Conclusions: In this study, we found that PFS significantly improved in patients who received G-CSF at any point during their course of the treatment, with carboplatin plus gemcitabine, carboplatin plus paclitaxel or nab-paclitaxel, or Sacituzumab govitecan and patients receiving primary prophylaxis had a lower incidence of febrile neutropenia, treatment plan adjustment, and treatment discontinuation.
Presentation numberPS5-03-21
Rewiring fibroblast function in breast cancer
Prabhat Suman, USA Health Mitchell Cancer Institute, University of South Alabama, Mobile, AL
P. Suman1, S. Kakkat1, B. Kola1, E. T. Herrera1, A. Richter2, A. Mahadevan2, S. Atterberry2, K. Sawrie2, R. Gupta2, D. T. Tambe3, C. Sarkar1, D. Chakroborty1; 1Department of Pathology, USA Health Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 2USA Health Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 3Pharmacology and Mechanical Engineering, University of South Alabama, Mobile, AL.
In the United States, breast cancer (BCa) is the second most common cause of cancer-related death for women and the most commonly diagnosed malignancy. Cancer-associated fibroblasts (CAFs) are a crucial component of the tumor microenvironment (TME) that influences breast cancer progression. By interacting with cancer cells and altering the extracellular matrix (ECM), CAFs actively promote tumor growth and metastasis. In contrast, normal fibroblasts restrict tumor progression. Despite the significant role of CAFs, little is known about the mechanisms underlying the regulation and conversion of normal fibroblasts into CAFs within the TME. Lysine Deficient Protein Kinase 1 (WNK1) is a serine/threonine kinase that is important for maintaining electrolyte balance and tissue homeostasis in the body. It is overexpressed in the BCa microenvironment, particularly in CAFs. This study investigated the role of WNK1 in regulating CAFs in BCa. Studies were conducted using BCa patient tissues, available public data, orthotopic mouse 4T1 tumor models, and in vitro assays to identify the effect of WNK1 on regulating fibroblast functions and BCa progression. WNK1 expression levels were determined in BCa tissues, orthotopic 4T1 BCa mouse embryonic fibroblasts (MEFs), and 4T1 BCa cells using Western blot analysis, RT-PCR, immunofluorescence, and confocal microscopy. Quantitative RT-PCR (qRT-PCR) and flow cytometry were used to identify the molecular changes associated with WNK1 manipulation in MEFs in vitro. Our results indicated that WNK1 is overexpressed in BCa and is associated with a poor prognosis and worse disease outcomes. WNK1 is expressed by both cancer cells and CAFs in tumors; however, WNK1 expression in CAFs was significantly higher. Our in vitro data revealed molecular changes in MEFs, accompanied by the upregulation of WNK1, when these cells were exposed to 4T1 cell-conditioned medium. Furthermore, significant upregulation in the expression of CAF-specific markers, α-SMA, FAP, and FSP1, along with ECM crosslinking genes and fibronectin, was observed in 4T1 medium-stimulated MEFs, which was significantly repressed in the presence of WNK1 inhibitors. Interestingly, upon overexpression of WNK1 in MEFs through expression plasmid vector, CAF markers and ECM crosslinking genes were found to be expressed significantly higher in WNK-overexpressed MEFs compared to normal MEFs, as indicated by qRT-PCR and flow cytometry analysis. The findings suggest a critical role for WNK1 in fibroblast-to-CAF transition. Analyzed publicly available datasets also indicated a strong correlation between WNK1 expression and the expression of ECM crosslinking genes in BCa. Taken together, our findings indicate that WNK1 contributes to BCa progression by regulating CAF conversion and the production of ECM proteins. Our results suggest that WNK1 may be a potential therapeutic target in BCa.
Presentation numberPS5-03-22
Lactate driven upregulation of BACH1 promotes metastatic dissemination in triple negative breast cancer
Joselyn Padilla, The George Washington University, Washington, DC
J. Padilla1, Y. Park1, A. Bansal2, A. Banduru3, J. Lee1; 1Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, 2School of Medicine and Health Sciences, The George Washington University, Washington, DC, 3College of Arts and Sciences, Case Western Reserve University, Cleveland, OH.
Patients with triple negative breast cancer (TNBC) experience the highest mortality and lowest metastasis-free survival rate amongst all other breast cancer subtypes. TNBC is characterized by absence of hormone receptors; estrogen receptor and progesterone receptor, and HER2 receptor, which limits the effectiveness of targeted therapies, highlighting the need for further study. One key factor, BTB and CNC homology 1 (BACH1), a heme-binding transcription factor, is upregulated in TNBC compared to other breast cancer subtypes. Previous studies have identified BACH1 as a regulator of metastasis in cancers, including TNBC. While much of the focus has been on the intrinsic regulation of BACH1, the impact of the tumor microenvironment (TME), specifically nutrients like lactate, on BACH1 regulation and TNBC metastasis is unknown. Cancer cells rely on heightened glycolysis, leading to excessive lactate production, even in the presence of oxygen. This metabolic shift supports both tumor growth and metastasis. Given the emerging role of lactate as a key signaling molecule, we explored how lactate in the TME directly regulates BACH1 expression and contributes to metastasis of TNBC. In vitro simulation of a lactate rich microenvironment using human and murine TNBC cell lines resulted in increased BACH1 protein expression while mRNA expression remained unregulated. Additionally, lactate accumulation in the microenvironment led to BACH1-dependent increase in invasion and migration of TNBC, while proliferation remained unregulated. To understand the mechanism of lactate mediated upregulation of BACH1, a recently identified posttranslational modification, lysine lactylation (Kla) was investigated utilizing global lactylation analysis and BACH1 specific interactome under lactate rich conditions. These findings represent a novel regulatory mechanism by which lactate in tumor microenvironment contributes to metastasis of TNBC through lactylating BACH1 or its interacting partners. These results suggest that targeting BACH1 stability through lactylation inhibition could serve as a promising approach to limit TNBC metastatic progression, especially in aggressive, therapy-resistant cases.
Presentation numberPS5-03-23
Outcomes with first-line treatments for patients with locally advanced or metastatic triple negative breast cancer eligible for anti-PD-(L)1 therapy: A systematic literature review of randomized trials
Maryam Afshari, Gilead Sciences, Inc, Foster City, CA
X. Wang1, J. Frampton2, S. Pathak3, M. Afshari4, J. Dinoso5, A. Brufsky6; 1Global Value and Access, HEOR, Gilead Sciences, Inc, Foster City, CA, 2Evidence Synthesis, Thermo Fisher Scientific, London, UNITED KINGDOM, 3Evidence Synthesis, Thermo Fisher Scientific, Bengaluru, INDIA, 4Clinical Development, Gilead Sciences, Inc, Foster City, CA, 5Global Medical Affairs, Gilead Sciences, Inc, Foster City, CA, 6Division of Hematology/Oncology, Magee-Womens Hospital and the Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburg, PA.
Objective: To evaluate current treatment outcomes for patients with inoperable locally advanced or metastatic triple negative breast cancer (LA/mTNBC) whose tumors express programmed death-ligand 1 (PD-L1+) treated in the first line. Methods: Literature databases were searched to June 2024. Randomized trials involving adults with PD-L1+, (combined positive score [CPS] ≥1/immune cells ≥1% or receiving anti-PD-[L]1 therapy) inoperable LA/mTNBC treated in the first line were included. Results: From 4140 records, 50 reporting on eight phase 3, six phase 2 or 1/2 trials were included. The primary endpoint was progression-free survival (PFS) in 9 trials and overall survival (OS) in 5. Median PFS ranged from 3.6-12.2 months. Adding atezolizumab to taxane-based chemotherapies (7.2-7.5 vs. 5.3-6.4 months)—but not gemcitabine-carboplatin or capecitabine (4.2 vs. 3.6 months)—resulted in a significant PFS benefit. Adding pembrolizumab or toripalimab to standard chemotherapy also significantly improved PFS (9.7-10.8 vs. 5.6-6.7 months). There were also statistically significant improvements in PFS with molecular subtyping-based treatments and a numerical improvement in PFS when replacing nab-paclitaxel with sacituzumab govitecan (SG) combined with immunotherapy. Similarly, replacing chemotherapy with SG combined with pembrolizumab significantly increased PFS. Conversely, trials evaluating the addition of oleclumab, nivolumab, or ipatasertib to chemotherapy and/or immunotherapy did not significantly improve PFS. Median OS ranged from 10.7-32.8 months across trials; most showed no significant differences between treatment arms. However, adding atezolizumab (25.4 vs. 17.9 months) or toripalimab (32.8 vs. 19.5 months) to nab-paclitaxel significantly improved OS. Adding pembrolizumab to chemotherapy significantly improved OS only in patients with CPS ≥10 (23.0 vs. 16.1 months). Further trials investigating other immunotherapies added to standard chemotherapy did not significantly improve OS. Overall response rates (ORR) ranged from 23.1%-72.7%. All treatment arms, except nivolumab and toripalimab, resulted in a numerically higher ORR vs. comparator arms; however, statistical significance was rarely assessed and demonstrated only for the addition of atezolizumab to nab-paclitaxel. Overall, patients experiencing a serious adverse event ranged from 16%-44.4%. Conclusions: This review indicates an evolving treatment landscape, with the adoption of regimens with atezolizumab, toripalimab, pembrolizumab, and SG being associated with improved efficacy in patients with PD-(L)1-eligible LA/mTNBC. Specifically, the most recent publication revealed that SG in combination with pembrolizumab being a potential new frontline standard of care.
| Study Name, Phase (PD-L1 Group) | Treatment vs Comparator |
PFS HR
(95% CI)
|
OS HR
(95% CI)
|
ORR (%) | SAEs (%) |
| ASCENT-04, Ph3 (CPS≥10)* | SG + PEM vs. Chemo + PEM |
0.65
(0.51, 0.84)
|
NR | 60 vs. 53 | NR |
| CBCSG006, Ph3 (≥1% IC) | GEM + PTX vs. GEM + Cisplatin | NR | NR | 73 vs. 63 | NR |
| FUTURE-SUPER, Ph2 (CPS≥10) | Subtyping-based vs. NP |
0.26
(0.09, 0.77)
|
NR | NR | NR |
|
Garrido-Castro, 2022, Ph2
(≥1% IC)
|
CARBO + NIVO vs. CARBO |
0.54
(0.18, 1.62)
|
0.75
(0.26, 2.16)
|
23 vs. 27 | NR |
| IMpassion130, Ph3 (≥1% IC) | ATZ + NP vs. PBO + NP |
0.63
(0.50, 0.79)
|
0.67
(0.53, 0.86)
|
59 vs. 43 | NR |
| IMpassion130, Ph3 (≥5% IC) | ATZ + NP vs. PBO + NP |
0.71
(0.48, 1.05)
|
0.76
(0.46, 1.26)
|
NR | NR |
| IMpassion131, Ph3 (≥1% IC) | ATZ + PTX vs. PBO + NP |
0.73
(0.56, 0.96)
|
1.11
(0.76, 1.64)
|
52 vs. 44 | NR |
| IMpassion132, Ph3 (≥1% IC) | ATZ + Chemo vs. PBO + Chemo |
0.84
(0.67, 1.06)
|
0.93
(0.73, 1.20)
|
40 vs. 28 | 22 vs. 20 |
| IPATunity170, Ph3 (≥1% IC) | ATZ + Ipatasertib + PTX vs. ATZ + PTX |
0.99
(0.63, 1.58)
|
1.04
(0.52, 2.06)
|
47 vs. 39 | 26 vs.16 |
| KEYLYNK-009, Ph2 (CPS≥10) | PEM + CARBO + GEM → PEM + OLA vs. PEM + CARBO + GEM → PEM + CARBO + GEM |
0.92
(0.59, 1.43)
|
0.97
(0.53, 1.76)
|
NR | NR |
| KEYNOTE-355, Ph3 (CPS≥1) | PEM + Chemo vs. PBO + Chemo |
0.75
(0.62, 0.91)
|
0.86
(0.72, 1.04)
|
45 vs. 39 | NR |
| KEYNOTE-355, Ph3 (CPS≥10) | PEM + Chemo vs. PBO + Chemo |
0.66
(0.50, 0.88)
|
0.73
(0.55, 0.95)
|
53 vs. 41 | NR |
| KEYNOTE-355, Ph 3 (CPS≥20) | PEM + Chemo vs. PBO + Chemo | NR |
0.72
(0.51, 1.01)
|
NR | NR |
| MORPHEUS-pan BC, Ph1/2 (NR) | ATZ + SG vs. ATZ + NP |
0.27
(0.11, 0.70)
|
NR | NR | 23 vs. 44 |
| TORCHLIGHT, Ph3 (CPS≥1) | TORI + NP vs. PBO + NP |
0.64
(0.47, 0.88)
|
0.62
(0.41, 0.91)
|
66 vs. 68 | NR |
| TORCHLIGHT, Ph3 (CPS≥10) | TORI + NP vs. PBO + NP |
0.74
(0.45, 1.24)
|
0.55
(0.30, 0.98)
|
NR | NR |
| * The ASCENT-04 trial was identified after the SLR cut-off date, and added to reflect recent literature. Note: Two studies not shown, due to reporting median survival instead of HRs, and reporting no ORR or SAE data. Abbreviations: ATZ = atezolizumab; CARBO = carboplatin; Chemo = chemotherapy; CPS = combined positive score; GEM = gemcitabine; HR = hazard ratio; IC = immune cells; NIVO = nivolumab; NP = nab-paclitaxel; NR = not reported; OLA = olaparib; ORR = overall response rate; OS = overall survival; PBO = placebo; PEM = pembrolizumab; PFS = progression-free survival; Ph = phase; PTX = paclitaxel; SAE = serious adverse event; SG = sacituzumab govitecan; TORI = toripalimab |
Presentation numberPS5-03-24
Thromboxane A2-Prostanoid Receptor (TPR) Blockade with Ifetroban Disrupts Platelet-Tumor Cell Interactions to Reduce Metastasis in Triple-Negative Breast Cancer
Veeresh Toragall, University of Mississippi, Oxford, MS
V. Toragall1, A. Rester1, S. Begum2, O. Shofolawe-Bakare1, K. Hulugalla1, J. Monroe2, Y. Gibert3, T. Werfel1; 1Biomedical Engineering, University of Mississippi, Oxford, MS, 2Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi, Jakson,, MS, 3Department of Cell and Molecular Biology,, University of Mississippi Medical Center, Jackson, MS, USA, Jakson,, MS.
Platelets play a central role in tumor metastasis by promoting circulating tumor cell (CTC) survival, vascular arrest, and extravasation. Thromboxane A2 (TXA2), a key platelet activation mediator, drives these processes via its receptor (TPr). Here, we investigate the therapeutic potential of the TPr antagonist ifetroban to disrupt platelet-tumor cell crosstalk and suppress metastasis in triple-negative breast cancer (TNBC). In vitro, ifetroban significantly reduced platelet activation and binding to MDA-MB-231 cells. Co-culture assays showed U46619 (TPr agonist) increased platelet-tumor cell interactions by 160%, while ifetroban pretreatment reduced binding by 65% (p<0.05). Flow cytometry confirmed decreased CD62P expression (activation marker) in ifetroban-treated platelets, supporting the established antiplatelet effect of TPr blockade. The in vivo zebrafish model results revealed that ifetroban reduced MDA-MB-231 xenograft metastasis without attenuating platelet (CD41+) recruitment. Murine studies demonstrated ifetroban’s efficacy across TNBC models: 4T1 subcutaneous tumor model (1×106 cells) showed higher lung and liver metastatic foci in control animals, while ifetroban treatment (50 mg/kg BW, oral dosing, pretreatment for 2 weeks before tumor induction and continued until the end) resulted in significantly lower (p<0.05) numbers of lung and liver metastases and lower circulating levels of TXA2 metabolite, TXB2. Additionally, a decrease in tumor volume and weight was observed in ifetroban-treated animals, although ifetroban (up to 500 µM) did not show any direct toxic effects on tumor cell lines (MDA-MB-231 and 4T1), suggesting an indirect anticancer effect. Further confirmation came from a xenograft model using Athymic nude mice induced with MDA-MB-231 RFP cells (1×106 cells, subcutaneous injection). The treatment regime was similar (50 mg/kg BW, oral dosing, pretreatment for 2 weeks before tumor induction and continued until the end), and results from in vivo imaging system (IVIS) studies showed a significant (p<0.05) reduction in MDA-MB-231 cell metastasis to the liver and lungs compared to the control. Tumor weight and volume reductions were consistent with previous animal studies, demonstrating ifetroban’s therapeutic efficacy in different TNBC models. Histopathological analysis of lungs and liver confirmed findings in both mouse models. Mechanistically, ifetroban suppressed circulating TXB2 (TXA2 metabolite) and disrupted platelet-mediated CTC protection. Notably, tumor volume/weight reductions occurred without direct tumor cell toxicity, implicating immune-modulatory effects. These findings highlight ifetroban’s ability to target platelet-driven metastatic niches by blocking TXA2-TPr signaling. Our data provide preclinical evidence for repurposing ifetroban as an adjuvant therapy to reduce metastasis in TNBC, offering a novel strategy to counteract platelet-supported metastatic progression.
Presentation numberPS5-03-25
Bexmet trial for metastatix tnbc patients
YING ZHANG, Genome institute of Singapore, A*STAR, singapore, Singapore
Y. ZHANG1, E. Pathak1, C. Koh1, J. Lidster1, J. Yeong2, E. Lim3, W. Tam1; 1GIS, Genome institute of Singapore, A*STAR, singapore, SINGAPORE, 2IMCB, Institute for Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), singapore, SINGAPORE, 3Division of Medical Oncology, Division of Medical Oncology, National Cancer Centre Singapore, singapore, SINGAPORE.
The ability to precisely manipulate cancer cell states represents a promising approach to enhance therapeutic response. Our previous work demonstrated that retinoids can reduce tumorigenicity in triple-negative breast cancer (TNBC) models by shifting cells from a mesenchymal to an epithelial state, thereby sensitizing resistant TNBC cells to standard-of-care treatments. Here, we designed a Phase I trial, BEXMET, to investigate whether bexarotene, a retinoid X receptor agonist, can induce epithelial cell state changes and increase sensitivity to capecitabine in patients with metastatic TNBC. This cohort included 12 patients who received bexarotene during a two-week lead-in period, followed by repeated treatment cycles consisting of two weeks of oral capecitabine and one week of bexarotene. Tumor biopsies were collected at four time points: pre-treatment, post-bexarotene lead-in, post-capecitabine (cycle 1), and at disease progression (if applicable). RNA-Seq and multiplex immunohistochemistry were performed on all biopsies. Our results show that bexarotene induced a mesenchymal-to-epithelial transition (MET) in 3 of 9 patients, as indicated by transcriptional and histological markers. These bexarotene responders demonstrated increased sensitivity to capecitabine, evidenced by enrichment of cell death-related gene expression following treatment. Notably, the MET responders exhibited reduced immune cell infiltration and a more epithelial-like phenotype after two weeks of bexarotene treatment. At baseline, these responders tended to have a more mesenchymal and immune-infiltrated tumor profile, suggesting potential predictive biomarkers for response. In conclusion, this study provides early clinical evidence that pharmacologic induction of cell state changes can enhance the efficacy of existing chemotherapies in metastatic TNBC. Targeting cell plasticity may offer a cost-effective and accessible therapeutic strategy for this aggressive breast cancer subtype.
Presentation numberPS5-03-26
A Prospective Observational Study of Atezolizumab plus Nab-Paclitaxel in Patients with Triple-Negative Breast Cancer: First Analysis of JBCRG-C08/ATTRIBUTE
Tatsunori Shimoi, National Cancer Center Hospital, Tokyo, Japan
T. Shimoi1, N. Shibata2, M. Kitada3, M. Tsuneizumi4, M. Yamamoto5, Y. Ozaki6, Y. Fujimoto7, S. Akiyoshi8, N. Hashimoto9, M. Yamaguchi10, Y. Yamamoto11, N. Masuda12, H. Tada13, T. Yamanaka14, Y. Kikawa15, T. Taira16, S. Nakagawa17, K. Kiyota18, M. Oba19, N. Niikura20; 1Department of Medical Oncology, National Cancer Center Hospital, Tokyo, JAPAN, 2Department of Clinical Oncology, Kansai Medical University Hospital, Osaka, JAPAN, 3Department of Breast Disease Center, Asahikawa Medical University Hospital, Hokkaido, JAPAN, 4Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, JAPAN, 5Department of Breast Oncology, National Hospital Organization Hokkaido Cancer Center, Hokkaido, JAPAN, 6Department of Breast Medical Oncology, Cancer Institute Hospital of JFCR, Tokyo, JAPAN, 7Division of Breast Oncology, Saitama Cancer Center, Saitama, JAPAN, 8Department of Breast Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, JAPAN, 9Department of Breast Surgery, Aomori Prefectural Central Hospital, Aomori, JAPAN, 10Department of Breast Surgery, JCHO Kurume General Hospital, Fukuoka, JAPAN, 11Department of Breast and Endocrine Surgery, Kumamoto University Hospital, Kumamoto, JAPAN, 12Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, JAPAN, 13Department of Surgery, Division of Breast and Endocrine Surgery, Tohoku University Hospital, Miyagi, JAPAN, 14Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Kanagawa, JAPAN, 15Department of Breast Surgery, Kansai Medical University Hospital, Osaka, JAPAN, 16Department of Medical Oncology, Social Medical Corporation Hakuaikai Sagara Hospital, Kagoshima, JAPAN, 17Biometrics Department, Clinical Development Division, Chugai Pharmaceutical Co., Ltd., Tokyo, JAPAN, 18Oncology Medical Science Department, Medical Affairs Division, Chugai Pharmaceutical Co., Ltd., Tokyo, JAPAN, 19Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and Psychiatry, National Center of Neurology and Psychiatry Hospital, Tokyo, JAPAN, 20Department of Breast and Oncology, Tokai University School of Medicine, Kanagawa, JAPAN.
Background: Atezolizumab plus nab-paclitaxel has been available as a standard treatment for Programmed Death-Ligand 1 (PD-L1)-positive metastatic triple-negative breast cancer (mTNBC) in Japan since 2019, based on the results of the IMpassion130 trial. However, real-world evidence remains insufficient. Moreover, while immune-related adverse events (irAEs) are known to occur not only during treatment but also after discontinuation, detailed characterization of these events is limited. The ATTRIBUTE study was designed to evaluate the safety and effectiveness of atezolizumab plus nab-paclitaxel in a real-world patient population, including those who would have been excluded from the pivotal clinical trial. Methods: This multicenter, prospective observational study was conducted in Japan. Key eligibility criteria included: 1) age ≥20 years, 2) PD-L1-positive mTNBC, 3) planned atezolizumab treatment, 4) no hypersensitivity to atezolizumab, 5) ≤2 prior systemic therapy lines for mTNBC. The primary outcome was the incidence of adverse events as assessed by investigators. Secondary outcomes included progression-free survival (PFS), objective response rate (ORR), clinical benefit rate (CBR), and others. The observation period was set at 2 years from the final patient enrollment, with this report presenting the first analysis data collected 6 months after the final patient enrollment. The protocol was approved by the institutional review board and informed consent was obtained from all patients. Results: A total of 151 patients were enrolled from 79 institutions between February 15, 2021, and April 30, 2024. The safety analysis population comprised 149 patients, and the full analysis set included 148 patients. Baseline characteristics included: median age: 59 years, ECOG PS 0: 75.2%, postoperative recurrence: 73.2%, and history of neoadjuvant/adjuvant therapy: 59.7%. This study included patients who would have been excluded from IMpassion130: ECOG PS ≥2 (4.1%) and history or presence of autoimmune disease (7.4%). The incidence of serious adverse events (SAE) was 16.8%. SAE occurring in ≥1% of patients included pneumonitis (3.4%, n=5), diarrhea (1.3%, n=2), fever (1.3%, n=2), adrenal insufficiency (1.3%, n=2) and heart failure (1.3%, n=2). Two cases of Grade 5 pneumonitis were observed. The incidence of late-onset irAEs (31 days to 6 months after treatment discontinuation) associated with atezolizumab was 2.7%, including reports of adrenal insufficiency, pneumonitis, and palmar-plantar erythrodysesthesia syndrome. The median progression-free survival details will be presented at the meeting. The ORR was 33.6% (95% CI: 25.3-42.7), and the CBR was 37.7% (95% CI: 29.1-46.9). Conclusion: In this first analysis of one of the largest real-world patient populations, no new safety signals were observed for atezolizumab combination therapy compared with previous reports. Further follow-up is ongoing, and findings will be reported at upcoming scientific conferences and publications. Clinical trial identification: UMIN000041747 Funding: Chugai Pharmaceutical Co., Ltd.
Presentation numberPS5-03-27
Multifaceted Therapeutic Approaches Targeting Oncogenic Pathways to Combat Triple-Negative Breast Cancer
Arisha Arora, Indian Institute of Technology Guwahati, Guwahati, India
A. Arora, S. S. Ghosh; Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, INDIA.
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype, characterized by the lack of targetable bioreceptors, high chemoresistance, metastasis and limited treatment options. Therefore, identifying novel biological targets and therapeutic avenues is crucial to improve TNBC outcomes. It is known that the plasticity of key signaling pathways like Wnt and MAPK, contributes to TNBC progression and metastasis. In the efforts to identify actionable targets, we performed a high-throughput analysis of TNBC microarray datasets and identified Maternal embryonic leucine zipper kinase (MELK), a component of MAPK pathway, to be significantly upregulated, with high MELK expression correlated with poor overall survival of TNBC patients. Additionally, our bioinformatic and RT-PCR analyses identified Pigment epithelium-derived factor (PEDF), a suppressor of the Wnt pathway through the inhibition of its co-receptor LRP6, to be significantly downregulated in TNBC cells. Building on these findings, our current study aims to target these dysregulated pathways by leveraging drug repurposing approach to target MELK in the MAPK pathway and PEDF recombinant protein therapy to restore Wnt suppression in TNBC. Methods: To inhibit MELK, an FDA-approved drug library was screened utilizing molecular docking, molecular dynamics simulations (MDS), and MMPBSA energy calculations. The top drug candidates were validated for their cellular activity using functional assays, flow cytometry, RT-PCR, and western blot analyses in TNBC cell lines (MDA-MB-231 and MDA-MB-468). The recombinant PEDF (rPEDF) protein was produced in E.coli with His-tag for purification using affinity chromatography and characterized by circular dichroism (CD) and MALDI-TOF biophysical analyses. The effect of rPEDF in EMT-induced TNBC cell lines and free-floating generated 3D spheroid cultures was evaluated by MTT assay, scratch assay, matrigel invasion assay and cellular signaling by RT-PCR, western blot and confocal microscopy. Results: Virtual Screening of a library of 1293 FDA-approved drugs and MDS parameters like RMSD, RMSF and pair distance revealed that the top 10 drugs (binding energy <-67.75 kJ/mol) were strongly stabilized in the kinase domain of MELK. The top two candidates: Netarsudil and Dutasteride, led to a dose-dependent reduction in cell proliferation, migration, G1-phase cell cycle arrest, significant reduction of MELK expression and its target genes, including cyclin B1, cyclin D1, p21 and p27 in TNBC cell lines. In protein therapy, MALDI-TOF confirmed the purified rPEDF had the legitimate 54 kDa molecular weight, and CD demonstrated the protein retained its secondary structure integrity. rPEDF treatment demonstrated anti-proliferative effects in EMT-induced TNBC cells and 3D tumor spheroids along with increased intracellular ROS and mitochondrial membrane depolarization. A significant decrease in colony and sphere formation, invasion and migration ability of TNBC cells was also observed following rPEDF treatment. The reduction in migration ability was corroborated by significant reduction in EMT markers and Wnt signaling proteins: N-cadherin, Vimentin, and β-catenin, underscoring the role of rPEDF in suppressing Wnt pathway-mediated TNBC survival and metastasis. Conclusion: Our findings accentuate the translational potential of repurposed drugs Netarsudil and Dutasteride and rPDEF protein as novel therapeutic agents, effectively targeting the MAPK and Wnt pathways, leading to reduced proliferation and metastasis in TNBC cell lines. By leveraging these approaches, including ongoing in vivo validation, our research aims to bridge the gap between identification of therapeutic targets and development of tailored treatment options, facilitating future clinical investigations in TNBC.
Presentation numberPS5-03-28
Reciprocal activation of breast cancer metastases and the lung epithelium during metastatic outgrowth
Jessica L. Christenson, University of Colorado Anschutz Medical Campus, Aurora, CO
J. L. Christenson, N. S. Spoelstra, K. I. O’Neill, M. M. Williams, J. K. Richer; Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO.
Introduction. During metastasis, tumor cells interact extensively with cells in distant organs. These interactions can interfere or modify the function of the normal cells at metastatic sites to induce a more pro-tumor microenvironment. The lung is one of the most common sites of breast cancer (BC) metastasis, and alveolar epithelial cells are the most common cell type in the lung. My data suggests that the lung alveolar epithelium contributes to metastatic outgrowth, and I hypothesize that BC lung micrometastases activate surrounding lung epithelial cells which, in turn, support the outgrowth of BC metastases within the lung. The overall purpose of these studies is to identify factors secreted by resident lung epithelial cells that could be used as metastasis-specific therapeutic targets and/or agents. Methods. To test this hypothesis, I utilized immunocompetent preclinical metastatic mouse mammary carcinoma models to study lung metastatic outgrowth, the stage most relevant to patients diagnosed with metastatic disease. To quantify lung wound repair, I developed a custom multispectral imaging panel. Single-cell RNA-sequencing (scRNAseq) was performed on mouse lungs with high or low metastatic burden to identify genes in the lung epithelium that produce potentially targetable pro-metastatic factors. No-contact co-culture experiments were used to study reciprocal paracrine interactions between lung type II alveolar epithelial (AT2) and BC cells. Results. A wound repair-related phenotype, characterized by chronic inflammation, developed within the lung microenvironment during metastatic outgrowth, including an increase in the number and activation of AT2 cells surrounding metastases as they grow. Single-cell RNA-sequencing of mouse lungs with a high vs. low metastatic burden showed that metastatic outgrowth significantly changed AT2 gene expression resulting in a modified secretome. No-contact co-culture experiments indicated that TNBC cells directly alter AT2 gene expression, and AT2 secreted factors promoted TNBC growth. I investigated the possible mechanism(s) responsible for these effects and discovered that AT2 pulmonary surfactant protein and lipid levels are altered by BC-derived secreted factors. Interestingly, treatment with the naturally derived calf surfactant Infasurf, which contains native surfactant proteins/lipids and is FDA-approved for use in premature infants, inhibited BC cell proliferation. Conclusion. Low levels of surfactant are commonly associated with pulmonary disease and lung cancer. My BC lung metastasis data suggests that something similar may be occurring in the lungs during metastatic outgrowth. Overall, my studies demonstrate that in addition to directly targeting malignant cells, there is potential to target lung epithelial cells in the metastatic BC microenvironment as well, in order to effectively treat BC lung metastasis.
Presentation numberPS5-03-29
A retrospectIve observatioNal multicenter study on SacItuzumab Govitecan in mTNBC post NHIA reimbursement in Taiwan (inSiGht)- An interim analysis
Hsu-Huan Chou, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan City, Taiwan
H. Chou1, M. Peng2, W. Kuo1, S. Shen1, C. Yu1, W. Shen3, C. Yang2, H. Kuo4, H. Lin5, D. F. Lin5, M. Wang6, N. Sadetsky7, Y. Lin2, S. Chen1; 1Department of General and Breast surgery, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan City, TAIWAN, 2Department of Oncology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan City, TAIWAN, 3Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan City, TAIWAN, 4Division of Hematology-Oncology, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, New Taipei City, TAIWAN, 5Medical affairs, Gilead, Taipei, TAIWAN, 6Real world evidence, Gilead, Stockley Park, UNITED KINGDOM, 7Real world evidence, Gilead, Foster City, CA.
Background Sacituzumab govitecan (SG), a Trop-2-directed antibody-drug conjugate, is approved globally for second-line (2L) or later treatment of unresectable locally advanced or metastatic triple-negative breast cancer (mTNBC) and HR+/HER2- mBC. However, pivotal trials largely included Western populations. Given known ethnic differences in mTNBC biology and treatment responses, evaluating SG’s real world effectiveness and safety in Asians is crucial. SG was approved in Taiwan in Nov 2022 and reimbursed by the National Health Insurance in Feb 2024. This interim analysis from the inSiGht study provides early real-world data in Taiwanese mTNBC patients post-reimbursement. Methods This retrospective analysis included mTNBC patients treated with SG in the 2L setting in Chang Gung Memorial Hospital Breast Cancer Registry (Taipei, Linkou and Tucheng) from Feb 2024 to Apr 2025. Clinical characteristics, treatment outcomes, and adverse events were assessed. Kaplan-Meier estimates on treatment outcomes and subgroup analyses by treatment line were performed. Results Among 42 patients (median age: 56 years); 15 received SG as 2L and 27 as third-line or later (3L+). ECOG 0-1 was reported in 90.5% (2L: 86.7%; 3L+: 92.6%). Visceral metastases were present in 90.5% (2L: 86.7%; 3L+: 92.6%) and brain metastases in 21.4% (2L: 26.7%; 3L+: 18.5%). HER2-low expression was seen in 47.6% (2L: 53.3%; 3L+: 44.4%), and 52.4% were HER2 IHC 0. Prior PD-L1 inhibitor use was reported in 28.6% (2L: 46.7%; 3L+: 18.5%). Median time from mTNBC diagnosis to SG initiation was 11 months (2L: 6.2 months; 3L+: 15 months). Median rwPFS was 5 (95%CI: 2.6, 8.8) months overall. By line of treatment, median rwPFS was 9.0 (95%CI: 2.6, NR) months in 2L and 3.8 (95%CI: 2.3, NR) months in 3L+. Median follow up was 9 months (IQR 6.4, 13.8). The 9-month OS rate (95% CI) was 73% (59- 89%) overall, with higher OS in 2L (92% [79-100%]) vs. 3L+(61% [43-85%]). Among 33 patients with response data, 36.4% achieved partial response (PR), 27.3% had stable disease (SD) and 36.4% showed progression. PR rate was higher in 2L (53.8%) vs. 3L+ (25.0%). Progression occurred more in 3L+ (45.0%) vs. 2L (23.1%). Among patients who discontinued SG, the primary reason was disease progression (79.3%), followed by death (13.8%) and adverse events (6.9%). Notably, none of the 2L patients discontinued due to adverse events. Grade 2 diarrhea occurred in 7.1%, grade 1 in 16.7%. Grade 3 and 4 neutropenia were observed in 4.8% each. Granulocyte colony-stimulating factors use was reported in 23.8% of patients. Conclusion This interim analysis demonstrates the real-world effectiveness and tolerability of SG in Asian patients with mTNBC. Effectiveness outcomes are particularly encouraging when SG is used earlier in the treatment course and are comparable to those observed in the ASCENT trial. These findings support SG’s clinical value in Asian populations, including those with aggressive disease features such as brain and visceral metastases. Further follow up will help confirm these observations as additional data mature.
Presentation numberPS5-03-30
Neoadjuvant Chemo-Immunotherapy Yields Survival Comparable to Upfront Surgery in Metaplastic Breast Cancer: A National Cancer Database Analysis
Margo Nelis, Cleveland Clinic Foundation, Cleveland, OH
M. Nelis1, A. Chichura2, L. Zhang3, V. Gadi4; 1Obstetrics and Gynecology Institute, Cleveland Clinic Foundation, Cleveland, OH, 2Surgical Oncology, University of Illinois, Chicago, Chicago, IL, 3Department of Surgery, University of Illinois, Chicago, Chicago, IL, 4Division of Hematology & Oncology, University of Illinois, Chicago, Chicago, IL.
Neoadjuvant Chemo-Immunotherapy Yields Survival Comparable to Upfront Surgery inMetaplastic Breast Cancer: A National Cancer Database AnalysisAuthors:Margo Nelis, MD1Lily Zhang, MS 2Vijayakrishna Gadi, MD, PhD3Anna M. Chichura, MD4,51. Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH 44195, USA.2. Biostatistician, Department of Surgery, University of Illinois Chicago, Chicago, IL60612, USA.3. Division of Hematology and Oncology, Department of Medicine, University of IllinoisChicago, Chicago, IL 60612, USA.4. Division of Surgical Oncology, Department of Surgery, University of Illinois Chicago,Chicago, IL 60612, USA.5. Division of Gynecologic Oncology, Department of Obstetrics and Gynecology,University of Illinois Chicago, Chicago, IL 60612, USA.Purpose:Metaplastic breast carcinoma (MpBC) is a rare and aggressive subtype of breast cancer, oftencharacterized by triple-negative (TN) receptor status and a limited response to standardchemotherapy. The optimal treatment strategy remains undefined. This study evaluates overallsurvival (OS) in patients with TN-MpBC treated with neoadjuvant chemotherapy (NAC) orneoadjuvant chemo-immunotherapy (NACIO) compared to upfront surgery using the NationalCancer Database (NCDB).Methods:A retrospective cohort analysis was conducted using the NCDB to identify patients diagnosedwith stage I-III TN-MpBC between 2010 and 2022 treated with NAC, NACIO, or upfront surgery. Primary endpoints included overall survival (OS), pathologic complete response (pCR),and breast conservation rates. Breast conservation and pathologic complete response wereanalyzed using logistic regression, and overall survival was analyzed using Cox proportionalhazard model.Results:Of 4,953 patients, 3429 (69.2%) underwent upfront surgery, 1232 (24.9%) received NAC, and292 (5.8%) received NACIO. No significant difference in OS was observed between NACIOand surgery-first approaches (HR = 0.80, 95% CI: 0.51-1.27; p = 0.3), but NAC was associatedwith a 37% increased risk of mortality compared to upfront surgery (HR = 1.37, 95% CI: 1.21-1.56; p < 0.001 NACIO was associated with a 62% higher likelihood of achieving pCRcompared to NAC (OR = 1.62, 95% CI: 1.11-2.38; p = 0.013). The rate of breast conservationwas not significantly different between NACIO and upfront surgery (OR = 0.8, 95% CI: 0.61-1.05; p = 0.11), but patients receiving NAC were 27% less likely to undergo breast-conservingsurgery compared to those undergoing surgery first (OR – 0.73, 95% CI: 0.63-0.84; p < 0.001).Conclusion:In patients with TN-MpBC, NACIO demonstrated comparable survival outcomes to upfrontsurgery and improved pCR rates compared to NAC. These findings suggest that integratingimmunotherapy into neoadjuvant treatment may offer clinical benefit, warranting furtherprospective investigation.
Presentation numberPS5-05-01
Treatment Intensity and Survival in Older Adults with Breast Cancer: Undertreatment or Overtreatment?
Soo-Young Lee, Inha University Hospital, Incheon, Korea, Republic of
S. Lee, S. Park, S. Lim; General Surgery, Inha University Hospital, Incheon, KOREA, REPUBLIC OF.
BackgroundOlder adults with breast cancer are under-represented in clinical trials despite the increasing aging population and extended life expectancy. Concerns about comorbidities and treatment-related toxicity lead to undertreatment. It remains unclear whether reduced treatment intensity leads to inferior survival outcomes, or whether intensive treatment in frail individuals contributes to harm. This study aimed to investigate real-world treatment patterns and their association with survival outcomes in older breast cancer patients. MethodsWe retrospectively reviewed patients aged ≥65 years who underwent curative surgery for breast cancer at Inha University Hospital between January 2007 and December 2018. Clinical characteristics, treatment modalities (chemotherapy, radiotherapy, endocrine therapy, anti-HER2 therapy), and treatment completion rates were recorded. Disease-free survival (DFS) and overall survival (OS) were compared based on age and treatment receipt and completion using Kaplan-Meier analysis and multivariate Cox regression. ResultsA total of 278 patients were included (median age 70.0 years). Total mastectomy was performed in 51.1%, and axillary lymph node dissection in 31.2%. For adjuvant therapies, 75.5% received endocrine therapy, 44.2% radiotherapy, 38.1% chemotherapy, and 8.4% anti-HER2 therapy. Treatment completion rates varied, with dropout rates of 31.8% for anti-HER2 therapy, 27.8% for endocrine therapy, and 15.9% for chemotherapy. OS significantly declined with increasing age groups (65-69 vs. 70-74 vs. ≥75), with 7-year OS rates of 96.4%, 94.0%, and 81.1%, respectively (p = 0.011), whereas DFS and breast cancer specific survival did not differ significantly by age. In univariate Cox models, OS was associated with age and treatment factors including chemotherapy and endocrine therapy completion, while DFS was more closely linked to pathologic features including lymphovascular invasion and Ki-67 proliferation index. In multivariate analysis adjusting for stage and endocrine therapy completion, older age (≥75 years) remained independently associated with worse overall survival (OR 1.79, 95% CI 1.01-3.18, p = 0.047). ConclusionsWhile older age was independently associated with worse OS, DFS did not differ by age, indicating that mortality in older patients is more attributable to non-cancer causes, and that appropriately treated older adults can achieve comparable recurrence outcomes. These findings highlight the need for individualized treatment decisions based on functional status rather than chronological age, particularly to avoid undertreatment in fit older adults.
Presentation numberPS5-05-03
Survival Outcomes of Chemotherapy Versus Endocrine Therapy in Symptomatic Medullary Infiltration from Metastatic Breast Cancer: A Retrospective Real-World Study
Raelson Rodrigues Miranda, Instituto do Câncer do Estado de São Paulo, Sao Paulo, Brazil
R. R. Miranda, L. d. Graziano, G. C. Trofa, W. A. De Lima, L. Testa; Clinical Oncology, Instituto do Câncer do Estado de São Paulo, Sao Paulo, BRAZIL.
Background: Symptomatic medullary infiltration (SMI) in metastatic breast cancer is rare and associated with significant hematologic compromise. No standard treatment exists, and data-guided management are limited. Retrospective studies suggest similar overall survival (OS)with chemotherapy (CT) and endocrine therapy combining aromatase inhibitors (AI) andCDK4/6 inhibitors. However, direct comparisons between CT and AI alone are scarce. This study evaluated OS in patients with SMI treated with CT or AI-based endocrine therapy in the first- or second-line setting. Methods: We retrospectively reviewed records of patients with metastatic breast cancer who underwent bone marrow biopsy at a single academic center from 2015 to 2023. Inclusion criteria were histologically confirmed SMI, symptomatic cytopenias, and receipt of either CT or hormone therapy (HT; AI or tamoxifen ± ovarian suppression, excluding CDK4/6 inhibitors) in the first or second line. Clinical, pathological, and survival data were analyzed. OS was estimated using Kaplan-Meier methods; independent prognostic factors were assessed via multivariable Cox regression. Results: Of 306 patients screened, 24 met inclusion criteria. Median age was 53 years; all tumors were hormone receptor-positive, and 5 were HER2-positive. Common manifestations included anemia (96%), thrombocytopenia (62%), dacrocytosis (58%), and leukopenia (33%). Most patients (n=23) had ≥3 metastatic sites; ECOG performance status was 0 (n=1), 1 (n=10), 2 (n=10), and 3 (n=3). In the first-line setting, median OS was 5.0 months with CT versus 2.0 months with HT (HR 2.4; 95% CI 0.9-6.4; p = 0.07). On multivariable analysis, CT was associated with improved OS (HR 0.06; p = 0.01), while HER2-positivity (HR 50; p = 0.01) and ≥3 metastatic sites (HR 4.3; p = 0.004) were associated with worse outcomes. Latin ethnicity emerged as a protective factor (HR 0.01; p = 0.01). In the second-line setting, CT conferred longer OS (5.0 vs.1.4 months; HR 0.2; p = 0.02), although not confirmed in the multivariable model. Age (HR 1.2;p = 0.02), ECOG 3 (HR 369; p = 0.04), and non-Latin race (HR 0.01; p = 0.03) were significant predictors of poorer survival. Conclusions: In this real-world cohort of patients with SMI from metastatic breast cancer, chemotherapy was associated with improved OS in the first-line setting compared to hormone therapy. No survival benefit was observed in second-line. HER2-positivity, advanced age, poor performance status, and extensive metastatic disease were associated with worse outcomes. Latin ethnicity was consistently protective across treatment lines.
Presentation numberPS5-05-04
Healthcare resource use among women treated with adjuvant endocrine therapy for hormone-receptor positive breast cancer: associations with induced vasomotor symptoms and treatment adherence
Aswanth Reddy, Mercy, Fort Smith, AR
A. Reddy1, N. Schoof2, M. Sophie3, C. Proenca4, C. Caetano5, C. Seitz6, A. Frenz7, K. Genga8, J. Milosavljevic9, S. Hagenaars10, C. Flach11, J. Nguyen12; 1Mercy Clinic Oncology, Mercy, Fort Smith, AR, 2Global Medical & Evidence Women’s Healthcare, Bayer AG, Berlin, GERMANY, 3Global Medical & Evidence Study Operations, Bayer Healthcare SAS, Bordeaux, FRANCE, 4HEOR, Bayer Consumer Health AG, Basel, SWITZERLAND, 5Global Medical & Evidence Women’s Healthcare, Bayer Consumer Health AG, Basel, SWITZERLAND, 6Clinical Development, Bayer AG, Berlin, GERMANY, 7Medical Affairs Statistics, Bayer AG, Berlin, GERMANY, 8Global Safety, Bayer SA, Sao Paulo, BRAZIL, 9Global regulatory, Bayer AG, Berlin, GERMANY, 10Epidemiology, IQVIA Solutions, Amsterdam, NETHERLANDS, 11Real-World Methods & Evidence Generation, IQVIA Ltd, London, UNITED KINGDOM, 12Global Medical & Evidence RWE Centre of Excellence, Bayer HealthCare Pharmaceuticals Inc, Whippany, NJ.
Background: Patients with breast cancer who are non-adherent to adjuvant endocrine therapy have worse clinical outcomes than those who are adherent.1 Side effects of treatment, including induced vasomotor symptoms (iVMS), are a common cause of non-adherence.2 There is a knowledge gap on how iVMS and non-adherence in this population are related to healthcare resource use (HCRU). Methods: In a retrospective cohort study using the Guardian Research Network electronic health record (EHR) US database, we identified women aged 18-70 years treated with adjuvant endocrine therapy following an initial diagnosis of stage 0-III hormone receptor-positive breast cancer from Jan 2016-Jun 2023. Women were stratified into 4 subgroups by adherence to adjuvant endocrine therapy (adherence; ≥80% days covered) and presence of iVMS at the start of follow-up (date of therapy initiation)/distinct follow-up periods. All-cause and breast cancer-related HCRU were identified from EHRs and manual curation of clinical notes. HCRU rates were calculated as events/patient-month. Quasi-Poisson regression models were used to calculate incident rate ratios (IRRs) with 95% CIs for all-cause and breast cancer-related HCRU between iVMS (vs. non-iVMS) and adherent (vs. non-adherent) subgroups, adjusted for confounders. Results: 4459 women were included; mean age at index date was 57 years. Most had ≥1 HCRU event during follow-up (99% for all-cause events, 94% for breast cancer-related events); the most common, respectively, were outpatient visits (98% & 93%), laboratory services (88% & 76%), and primary care visits (66% & 62%). As shown in the Table, non-adherent women with iVMS had the highest all-cause and breast cancer-related HCRU rates from 1 to <3 months’ follow-up (7.24 & 3.04 events/patient-month, respectively). Adherent women with iVMS had the highest HCRU rates during follow-up periods thereafter (3 to <12 months, 12 to <36 months, and 36 to 60 months; see the Table). In the regression models, patients with iVMS had higher rates of all-cause and breast cancer-related HCRU vs. those without iVMS over all follow-up periods (IRRs ranged from 1.16-1.30 and from 1.19-1.35, respectively). Irrespective of iVMS, adherent patients had a lower rate of all-cause and breast cancer-related HCRU vs. non-adherent patients in the first 3 months’ follow-up (IRR 0.84, 95% CI: 0.81-0.87 and IRR 0.77, 95% CI: 0.74-0.80, respectively) and higher rates in later follow-up periods (p<0.001). Conclusion: Our results suggest that women who experience iVMS after adjuvant endocrine therapy for hormone receptor-positive breast cancer have higher rates of HCRU than those who do not experience iVMS. Effective management of iVMS could translate into lower HCRU rates. Refs 1. Eliassen et al. BMC Cancer. 2023;23(1):625. 2. Franzoi et al. Lancet Oncol 2021; 22: e303-13
| Per patient month event rate (95% CI) | |||||
| Follow-up period* | Adherent & iVMS† | Adherent & no iVMS | Non-adherent & iVMS | Non-adherent & no iVMS | |
| 1 to <3 months | All-cause HCRU | 5.38 (5.18 – 5.59) | 4.43 (4.38 – 4.47) | 7.24 (6.54 – 8.00) | 5.55 (5.38 – 5.73) |
| Breast cancer-related HCRU | 1.53 (1.43 – 1.64) | 1.22 (1.19 – 1.24) | 3.04 (2.59 – 3.54) | 1.68 (1.59 – 1.78) | |
| 3 to <12 months | All-cause HCRU | 3.12 (3.08 – 3.16) | 2.51 (2.49 – 2.53) | 2.79 (2.68 – 2.90) | 1.99 (1.94 – 2.03) |
| Breast cancer-related HCRU | 1.08 (1.05 – 1.10) | 0.85 (0.84 – 0.87) | 0.92 (0.86 – 0.98) | 0.65 (0.63 – 0.68) | |
| 12 to <36 months | All-cause HCRU | 2.21 (2.19 – 2.23) | 1.90 (1.88 – 1.91) | 1.57 (1.53 – 1.60) | 1.00 (0.98 – 1.02) |
| Breast cancer-related HCRU | 0.69 (0.68 – 0.70) | 0.58 (0.57 – 0.59) | 0.41 (0.39 – 0.43) | 0.26 (0.25 – 0.27) | |
| 36 to 60 months | All-cause HCRU | 1.87 (1.85 – 1.89) | 1.62 (1.59 – 1.65) | 1.21 (1.17 – 1.24) | 0.82 (0.79 – 0.85) |
| Breast cancer-related HCRU | 0.55 (0.54 – 0.56) | 0.47 (0.45 – 0.48) | 0.31 (0.29 – 0.33) | 0.18 (0.17 – 0.20) | |
| *Number of women in the adherent & iVMS, adherent and no VMS, non-adherent & iVMS, and non-adherent & no iVMS subgroups were as follows: 251, 3801, 27, and 354 for 1 to <3 months; 986, 2821, 106, and 449 for 3 to <12 months; 1596, 1272, 325, and 502 for 12 to <36 months; 943, 467, 267, and 261 for 36 to 60 months. | †iVMS was determined at the start of each follow-up period. CI, confidence interval; HCRU, healthcare resource use; iVMS, induced vasomotor symptoms Values in bold show subgroup with the highest HCRU rate. |
Presentation numberPS5-05-05
Correlating time to treatment discontinuation with overall survival: real-world data on outcomes for first-line endocrine therapy + CDK4/6 inhibitor in metastatic breast cancer
Erica Mayer, Dana-Farber Cancer Institute, Boston, MA
E. Mayer1, K. A. Betts2, R. Ramasubramanian2, X. Nie2, T. Pham3, C. Chen3; 1-, Dana-Farber Cancer Institute, Boston, MA, 2-, Analysis Group, Los Angeles, CA, 3Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD.
Background Endocrine therapy (ET) + cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) is the standard-of-care first-line (1L) treatment for hormone receptor-positive (HR+)/HER2-negative (HER2−) metastatic breast cancer (mBC). Given that outcomes tend to worsen following progression to next-line therapy, understanding the relationship between the duration of 1L treatment and subsequent outcomes may be an important factor for clinical decision-making. This real-world, retrospective, observational cohort study investigated the relationship between time to discontinuation (TTD) of 1L therapy and overall survival (OS) in patients with HR+/HER2− mBC. Methods Adults with HR+/HER2− mBC were identified from electronic health records in the US Flatiron Health Database. Included patients had started 1L ET + CDK4/6i between March 1, 2018, and March 31, 2024, and had ≥2 clinical activities on different days after 1L initiation. Patients were followed until September 30, 2024. Descriptive statistics were used to summarize demographics, baseline characteristics, and treatment patterns. 1L TTD was defined as time from 1L initiation to final administration of 1L therapy (whichever component was administered last) or death. TTD and OS were assessed using Kaplan-Meier methodology. The relationship between 1L TTD and OS was assessed using rank correlation and a multivariate, covariate-adjusted, time-dependent Cox regression model. Results A total of 2910 patients were included, with a mean age of 63.2 years. Most were White (66.2%) and postmenopausal (71.4%), with Eastern Cooperative Oncology Group performance status (ECOG PS) ≤1 (63.2%), and estrogen receptor (ER) expression >70% (82.0%); 49% had recurrent disease (4.7% unknown), and few had liver metastases (16.1%). With a median (interquartile range) follow-up of 24.8 (14.0-41.0) months, median 1L TTD was 20.6 (95% CI: 19.4-22.0) months, and median OS was not reached. OS rate was 92.1% at 12 months and 83.8% at 24 months. There was a strong positive correlation between TTD and OS (r = 0.74 [95% CI: 0.70-0.78]). For each additional month on 1L treatment, the adjusted hazard ratio for OS was 0.95 (95% CI: 0.94-0.96; p<0.001). In real-world terms, this means that an additional 2, 4, 6, 8, and 10 months on 1L treatment would result in cumulative hazard ratios (calculated as 0.95^number of months) of 0.90, 0.81, 0.74, 0.66, and 0.60, respectively, and a 10%, 19%, 26%, 34%, and 40% lower risk of death, respectively. Lower ECOG PS, de novo disease, and ER expression >70% were also associated with better survival outcomes. Age, region, menopausal status, and practice type were not associated with survival outcomes. Conclusions In this real-world dataset, a longer duration of 1L treatment prior to clinical progression was highly positively correlated with longer OS, suggesting a clinical benefit to extending 1L treatment in HR+/HER2− mBC. It is therefore critical to identify ways to increase the duration of 1L therapy to improve patient outcomes.
Presentation numberPS5-05-06
Real-world Clinical Outcomes With Tratuzumab Deruxtecan in Metastatic HER2-low Breast Cancer
Zhonghua Wang, Fudan University Shanghai Cancer Center, Shanghai, China
K. Zhang, X. Zhu, X. Ying, Z. Wang; Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
BACKGROUND Trastuzumab deruxtecan (T-DXd) is a novel antibody-drug conjugate (ADC) that has demonstrated improved survival outcomes in patients with metastatic human epidermal growth factor receptor 2 (HER2)-low breast cancer, and is approved by Food and Drug Administration (FDA) for these patients. However, real-world evidence regarding the efficacy of T-DXd remains limited.METHODS We retrospectively collected clinical and demographic data of HER2-low mBC patients treated with T-DXd between November 2021 and December 2024 at Fudan University Shanghai Cancer Center. Real-world progression-free survival (rwPFS) was estimated using the Kaplan-Meier method. Treatment response was assessed according to RECIST 1.1 criteria. A T-DXd-prognostic score was established, according to regression coefficients found in the multivariate Cox regression analysis. RESULTS A total of 126 Chinese female patients with HER2-low mBC treated with T-DXd was enrolled. The median age was 56 years (IQR: 48 to 64), and 40 patients was TNM-IV stage. In this cohort, 72 (57.14%) had HER2-1+ mBC and 54 (42.86%) had HER2-2+ mBC. Hormone receptor (HR) status was positive in 82 (65.08%) patients and negative in 44 (34.92%). The median number of prior lines of advanced therapy was 3 (IQR: 2 to 5). Last follow-up was conducted on December 30, 2024, the median rwPFS was 9.10 months, which was significantly longer than the median rwPFS of 3.50 months observed with the last prior treatment chosen by physician. In patients with HR positive, those with low PR expression (≤ 10%) have significantly higher rwPFS than other patients (17.77 vs. 6.13 months; p < 0.01). Among 111 patients with measurable disease, the objective response rate (ORR) was 30.63%, and the disease control rate (DCR) was 72.07%. We developed a prognostic score using the regression coefficients from the multivariate Cox regression model, which included age, HER2 expression, and HR status as covariates. And then, patients were stratified into low- and high-risk groups by the prognostic score. According to risk stratification, the median rwPFS was 5.13 months in the high-risk group and 12.40 months in the low-risk group (hazard ratio [HR] 2.25, 95% confidence interval [CI] 1.31 – 3.88, p < 0.01). CONCLUSION In this real-world cohort of Chinese patients with HER2-low mBC, T-DXd demonstrated meaningful clinical benefit, with notably longer rwPFS than last prior treatment. Furthermore, we established a risk stratification model that effectively predict patient prognosis.
Presentation numberPS5-05-07
Real-world Patient Characteristics and Treatment Patterns with Pembrolizumab Among Patients with Early-Stage Triple-Negative Breast Cancer in the United States
Amin Haiderali, Merck, North Wales, PA
A. Haiderali1, Y. Sun2, L. Ai3, F. Beca3; 1Value & Implementation, Merck, North Wales, PA, 2Biostatistics and Research Decision Sciences (BARDS), Merck, Rahway, NJ, 3MRL, Merck, Rahway, NJ.
Background: Pembrolizumab (pembro) was approved by the Food and Drug Administration in July 2021 to treat high-risk, early-stage triple-negative breast cancer (TNBC) based on the KEYNOTE-522 trial. To support post-launch assessments, this real-world study aimed to outline patient characteristics and treatment patterns in patients with Stage II-III TNBC in the US community setting. Methods: This retrospective study used electronic medical records accessed via the Syapse data platform to examine previously untreated adults diagnosed with Stage II-III TNBC from August 2021 to July 2023. We analyzed patient characteristics, surgery rates and types, treatment patterns and duration, and reasons for treatment discontinuation in the neoadjuvant and adjuvant settings. Results: 322 patients were identified with Stage II (61.3%) or Stage III (38.7%) TNBC and who had undergone definitive surgery. Mean age was 58 years (standard deviation [SD]: 15). 47% had an ECOG performance status of 0, 14% had an ECOG of 1, while 37% had unknown status. In the neoadjuvant setting, most patients (75%) received pembro + chemotherapy, 12% received chemotherapy alone and 12% received no neoadjuvant therapy. The most common therapy in patients receiving neoadjuvant pembro + chemotherapy was pembro + carboplatin + taxanes + anthracyclines (A) + cyclosphosphamide (C) (83%) while the most common therapy in patients receiving chemotherapy alone was taxanes + AC (50%). Patients receiving pembro + chemotherapy were more likely to undergo mastectomy (56% vs 38%) and less likely to undergo lumpectomy (44% vs. 63%). Among patients receiving adjuvant therapy in the cohort previously treated with neoadjuvant pembro + chemotherapy, the most frequently administered adjuvant regimens were pembro monotherapy (70%) and pembro + capecitabine (10%). On the other hand, capecitabine was the most commonly administered treatment (75%) among patients receiving adjuvant therapy in the cohort that received neoadjuvant chemotherapy without pembro. The median (95% CI) time on pembro in the neoadjuvant and adjuvant settings was 5.3 (5.1, 5.6) and 5.6 (5.6, 5.7) months, respectively. Comparatively, median time on chemotherapy was 4.2 (3.4, 4.9) and 5.2 (4.4, 5.7) months, respectively. The primary reasons for discontinuation among patients who had received neoadjuvant chemotherapy + pembro (n=236) were completion of planned therapy (72%) and intolerance/toxicity (24%). In the adjuvant pembro group (n=97), primary reasons for discontinuation were completion of therapy (78%) and intolerance/toxicity (8.2%). Conclusion: Findings show a high utilization of pembro in neoadjuvant and adjuvant early-stage TNBC, highlighting its role as standard of care. Treatment duration for pembro was consistent with that reported in clinical trials and previous real-world studies, supporting use in routine clinical practice. High treatment completion rates suggest that pembro is generally well tolerated, supporting its use as a viable treatment option across perioperative TNBC setting.
Presentation numberPS5-05-08
Physician perspectives on treatment decision-making process in high-risk early-stage triple negative breast cancer (esTNBC) in a community oncology setting
Amin Haiderali, Merck, North Wales, PA
A. Haiderali1, L. Okoth2, S. Annavarapu2, J. K. Paulus2, W. Pan3, P. Conkling2, M. Danso4; 1Value & Implementation, Merck, North Wales, PA, 2Real World Research, Ontada, Boston, MA, 3MRL, Merck, Rahway, NJ, 4Virginia Oncology Associates, US Oncology, Norfolk, VA.
Background: Pembrolizumab, approved by the FDA in July 2021, is a standard of care (SOC) for neoadjuvant treatment of high-risk esTNBC, based on effectiveness established in the KEYNOTE-522 trial. Understanding factors affecting clinical integration are now critical to optimize patient outcomes. This study explored physician perspectives on pembrolizumab use in neoadjuvant and adjuvant settings and treatment decision-making process for high-risk esTNBC. Methods: In January 2025, physicians from The US Oncology Network completed a brief online survey and a virtual focus group discussion. Physicians were chosen for their TNBC management experience and geographical diversity. Survey data were analyzed quantitatively, while the discussion was transcribed and key themes identified qualitatively. Results: Six medical oncologists and two breast surgeons completed the survey and focus group discussion. SOC adoption The discussion revealed that the implementation of pembrolizumab is primarily driven by its status as the SOC, potential to enhance patient survival, and manageable side effect profile. On the survey, physicians estimated they use neoadjuvant pembrolizumab plus chemotherapy in 90% -100% of their esTNBC patients. Rare exceptions for non-use Focus group physicians reported rare exceptions to pembrolizumab as SOC. These include patients with immunotherapy contraindications, like severe autoimmune diseases or kidney transplants. Patients usually adhere to medical advice but occasionally, some patients decline neoadjuvant or adjuvant therapy due to side effect concerns or fear of progression and prefer immediate surgery. Additionally, focus group physicians reported that occasionally patients are referred post-surgery by general surgeons without having received neoadjuvant therapy. In other instances, focus group physicians indicated that patients with low disease burden, inoperable tumors, or urgent surgical needs may also not receive pembrolizumab. Treatment of ER-Low Breast Cancer All focus group physicians reported treating breast cancer patients with ER-low (1% – 9%) status similarly to TNBC patients. Extensive adjuvant use Seven surveyed physicians described that they use adjuvant pembrolizumab in >75% of their esTNBC patients who achieve pCR. However, patient-specific contraindications and clinician concerns for toxicity remain barriers to its recommendation as reported by five and four surveyed physicians, respectively. Conclusions: This qualitative study of breast cancer physicians reveals broad adoption of pembrolizumab as SOC for high-risk esTNBC in The US Oncology Network, due to survival benefits and tolerable side effects. Usage is limited by comorbidities, patient preferences, and post-surgery referrals. Future efforts are essential to guide improvements in patient communication, enhance surgeon awareness on current neoadjuvant guidelines, shared decision-making, and multidisciplinary care coordination, ultimately optimizing patient outcomes.
Presentation numberPS5-05-09
Treatment Persistence and Dose Modifications in US Patients with HR+, HER2-, Node-Positive, Early Breast Cancer Treated with Adjuvant Abemaciclib
Hatem Soliman, Moffitt Cancer Center and Research Institute, Tampa, FL
H. Soliman1, S. Dent2, A. Liepa3, V. Stefaniak3, M. Huang3, T. Sugihara4, K. Moreira3, K. Hudson5, M. Goetz6; 1Medical Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, 2Medicine, University of Rochester Medical Center, Rochester, NY, 3Oncology, Eli Lilly and Company, Indianapolis, IN, 4Research, Syneos Health, Morrisville, NC, 5Oncology, Texas Oncology, Austin, TX, 6Department of Oncology, Mayo Clinic, Rochester, MN.
Background: Two years of adjuvant abemaciclib plus endocrine therapy is approved and guideline-recommended in patients (pts) with HR+, HER2-, node-positive, early breast cancer (EBC) at high risk of recurrence. In initial real-world studies, the high rate (>85%) of treatment (tx) persistence beyond 3 months suggests that adjuvant abemaciclib is well-tolerated in routine clinical practice.1,2 Dose reductions were associated with higher persistence in prior studies and clinical trial data have shown that efficacy of abemaciclib was maintained with dose reductions. This study describes 6-month tx persistence and dosing patterns in pts with HR+, HER2-, node-positive EBC initiating abemaciclib 150 mg twice daily (BID). Methods: Retrospective data were accessed from the US de-identified Flatiron Health Research Database. Adults with HR+, HER2-, node-positive, EBC, who initiated abemaciclib 150 mg BID from Jan 2022-Jun 2024 were eligible and further characterized by ≥1 vs no dose reduction. Data cut-off was Dec 2024. Persistence rate was the proportion of pts on abemaciclib >6 months, allowing for ≤60-day medication gap within this period. Time to dose modifications and reasons for discontinuation (DC) were summarized. Subgroup analyses were conducted in pts confirmed as meeting the monarchE high-risk criteria (N2, N3, or N1 plus Grade 3 and/or tumor >5 cm). All analyses were descriptive. Results: Of 1063 eligible pts, median age was 56 years (IQR 47, 65). Most had N1 (48%) or N2 (34%) disease. Median follow-up was 17.5 months (IQR 11, 25). Tx persistence at 6 months was 75%. DC was mostly due to adverse events (AEs; 19%) and <1% due to recurrence. Approximately 50% of pts had ≥1 dose reduction. Persistence was 85% in pts with ≥1 dose reduction and 64% in pts with no dose reductions. 70% of pts who DC’d by 6 months did not have a dose reduction. Median time from start of abemaciclib to first dose change and/or hold was 49 days (IQR 23, 111). During the first 30 days of tx and Days 31-90, DCs due to AEs were lower in pts with dose reductions vs pts with no dose reductions (Table). Persistence and use of dose reductions were similar in pts confirmed as meeting the monarchE high-risk criteria. Conclusion: In US clinical practice, a majority (75%) who initiated adjuvant abemaciclib continued abemaciclib beyond 6 months. Tx persistence was higher among pts with dose reductions relative to those with no dose reductions, and rates of early DCs due to AEs were low in pts with dose reductions. Given that dose reductions in monarchE were not associated with reduced efficacy, these additional real-world data support the use of early dose modifications to improve tolerability and tx persistence for adjuvant abemaciclib in pts with high risk of recurrence.
| Time period for discontinuations | 150 mg BID(N=1063) | No Dose reductions(n=527) | >1 Dose reductions(n=536) |
| 0-30 days | 71 (7%) | 65 (12%) | 6 (1%) |
| 31-90 days | 77 (7%) | 53 (10%) | 24 (4%) |
| 91-182 days | 54 (5%) | 24 (5%) | 30 (6%) |
1Hudson K, et al. Clin Cancer Res (2025) 31 (12_Supplement): P1-11-29.2Gorantla V, et al. Clin Cancer Res (2025) 31 (12_Supplement): P1-11-21.
Presentation numberPS5-05-10
A Comparison of Participant-Reported and Medical Record Stage and Pathology Information in the Susan G. Komen ShareForCuresRegistry
James C Dickerson, Stanford University School of Medicine, Stanford, CA
J. C. Dickerson1, J. Epps2, A. Gyden2, A. Cleveland3, K. Sabelko2, A. W. Kurian1, J. Jourquin2, M. Bondy4; 1Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, 2Data Science & Scientific Programs, Susan G Komen Foundation, Dallas, TX, 3School of Medicine, Howard University College of Medicine, Washington DC, DC, 4Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA.
Background: ShareForCures is Komen’s crowd-sourced breast cancer research registry that collects and integrates data from and on behalf of its participants. Participants living in the U.S. with a history of breast cancer consent to share their medical records and complete surveys. In the U.S. primary care literature, patients and their medical records are concordant at rates ranging from 65% to 93%. However, there are limited data on how patients with breast cancer self-report details of their diagnoses. Here, we leveraged a cohort of ShareForCures participants to assess whether the recollection of their diagnosis information matches the information in their medical records. Not all researchers can access medical records to extract diagnosis information, and a high concordance could allow researchers to rely more heavily on patient-reported clinical details. Methods: We compared data for participants in ShareForCures who had completed the enrollment survey and had abstracted medical records (requested from the oncology and primary care practices) available by April 30, 2025. We had asked participants for the following variables in the intake survey: 1) stage at diagnosis, 2) estrogen-receptor (ER) and HER2 status (e.g., positive or negative), 3) receptor combination (e.g., triple-negative or triple positive), and 4) histology (e.g., ductal or lobular). These same variables are abstracted from the medical record. For staging, we only examined stages 1 – 4 and did not assess the accuracy of sub-stages (e.g., IB). We used the AJCC anatomic/clinical staging system in effect at the time of diagnosis. A board-certified medical oncologist specializing in breast cancer (JCD) adjudicated any unclear cases. We used descriptive statistics to summarize the data. We define concordance as the fraction of participant-reported details that match the abstracted medical records.Results: Data were obtained from 151 participants enrolled between 2022 and 2024. The majority (78%) had a diagnosis between 2010 – 2024. On median, 292 (IQR, 129 – 579) pages of records were available for abstraction per participant. The median age at diagnosis was 45 (IQR, 39 -52). The cohort was predominantly white (80%), and 82% of the participants had at least a college degree. The median time from diagnosis to enrollment in ShareForCures was 6 years (IQR, 2 – 11). 14% of the cohort reported as living with metastatic disease at enrollment. The diagnostic stage was concordant in 79% of the cases. ER status and HER2 status were concordant in 84% and 83% of the cases, respectively. When receptor statuses were combined (e.g., triple-negative, triple-positive), concordance was 75%. Histology was concordant in 73% of cases. We observed concordance for all these variables in 50% of the cases. Conclusions: Despite the inherent challenges of abstracting information from medical records and the complexities of breast cancer diagnoses, half of the responses from ShareForCures participants were concordant with all the diagnostic details abstracted from their medical records, with concordance rates ranging from 73% to 84% for individual variables. Although we may have anticipated higher concordance rates in a highly educated, young population, further research is needed to explore factors influencing concordance, such as the completeness of medical records and the time elapsed since diagnosis. This preliminary analysis highlights the importance of integrating both participant-reported and medical record-derived information to ensure the accurate reporting of breast cancer diagnosis information for research purposes.
Presentation numberPS5-05-11
Real-world evaluation of treatment trends in breast cancer: the PRISMA study
Maria Mafalda CPR Nogueira, MSD Portugal, Lisboa, Portugal
G. Sousa1, A. M. Ferreira2, I. Pereira3, A. Catarina4, D. Simão5, F. Machado6, G. Fernandes7, L. A. Ribeiro8, J. Fougo9, M. C. Nogueira10, P. H. Meireles11, J. Abreu Sousa12, P. Cortes13; 1Serviço de Oncologia, IPO Coimbra, Coimbra, PORTUGAL, 2Departamento de Oncologia, IPO Porto, Porto, PORTUGAL, 3Serviço de Oncologia, Hospital da Senhora da Oliveira, ULS Alto Ave, Guimarães, PORTUGAL, 4Serviço de Oncologia, Hospital de Santa Maria, ULS Santa Maria, Lisboa, PORTUGAL, 5Serviço de Oncologia, Hospital dos Capuchos, ULS São José, Lisboa, PORTUGAL, 6Direcção, Centro Académico Clínico Egas Moniz Health Alliance (EMHA), Aveiro, PORTUGAL, 7Serviço de Oncologia, Hospital da Luz Lisboa, Lisboa, PORTUGAL, 8Serviço de Oncologia, Hospital CUF Tejo, Lisboa, PORTUGAL, 9Unidade de Mama, Hospital de São João, ULS São João, Porto, PORTUGAL, 10Medical Departement, MSD Portugal, Lisboa, PORTUGAL, 11Serviço de Radioterapia, Hospital de São João, ULS São João, Porto, PORTUGAL, 12Departamento de Cirurgia, IPO Porto, Lisboa, PORTUGAL, 13Departamento de Oncologia, Hospital Lusíadas Lisboa, Lisboa, PORTUGAL.
Background: Breast cancer (BC) is the most common cancer and the leading cause of cancer-related death in women. Triple-negative BC (TNBC) and high-risk ER+ HER2- BC are aggressive BC subtypes with high recurrence rates. In Portugal, data on treatment of TNBC and high-risk ER+HER2- BC is limited. To address this, the PRISMA study collected quantitative data on treatment patterns and pathological complete response (pCR) from multiple Portuguese centers. Methods: Retrospective aggregated data from patients diagnosed between Jan-2019 and Jun-2023 were collected. The study included adult patients diagnosed with stage II – III TNBC or high-risk ER+ HER2- BC eligible for neoadjuvant (NAT) chemotherapy. Results: The study included 1585 patients, 642 (40%) with TNBC and 943 (60 %) with high-risk ER+HER2- BC. Most patients were ≥65 years with 1% being male. In both cohorts, most were diagnosed with stage II and were postmenopausal (62% and 73% in TNBC; 42% and 53% in ER+HER2-, respectively). Carcinoma of no special type was the predominant histology (88% TNBC and 83% ER+HER2- BC). In TNBC patients, the mean time between therapeutic decision and treatment initiation was five weeks (1 to 29 weeks), and from NAT completion to surgery was six weeks (3 to 8 weeks). Most TNBC patients underwent NAT chemotherapy (93%), primarily with carboplatin/paclitaxel and cyclophosphamide/doxorubicin or epirubicin (48%), resulting in a pCR rate of 47%. Surgery was performed in 99% of patients, with 59% undergoing conservative surgery. Adjuvant (AT) chemotherapy was given to 31% of patients, mostly with capecitabine (77%). Radiotherapy (RT) was performed in 72% of patients, primarily (66%) after conservative surgery, using conventional fractionation (50Gy/25fr, 48%). Among high-risk ER+HER2- BC patients, 81% cancers had Ki-67 ≥20%, and multi-gene assays were performed in 6%. 82% of high-risk ER+/HER2- patients underwent NAT and achieved a pCR of 9%. Dose dense AC or EC plus weekly paclitaxel was the most commonly used regimen (51%). The mean interval between NAT completion and surgery was 8 weeks (3 to 23 weeks). 59% of 939 patients undergoing surgery underwent breast conserving surgery. AT chemotherapy was administered to 18%, mostly with dose dense AC or EC and paclitaxel weekly (28%). RT was performed in 90% of patients, and 55% receiving it after conservative surgery. Regarding endocrine therapy (ET), 85% underwent adjuvant ET, with 68% using aromatase inhibitors. Ovarian suppression was performed in 26% of patients, frequently using gonadotropin-releasing hormone inhibitors (87%). Conclusions: The data provides a real-world characterization of TNBC and high-risk ER+HER2- BC treatment, demonstrating a high adherence to NAT chemotherapy and adjuvant ET, per guidelines, in this period. Observed delays in surgery suggest inter-hospital care variations that may inform healthcare policy, ultimately improving patient outcomes.
Presentation numberPS5-05-12
<sup>Real-world outcomes of neoadjuvant therapy for breast cancer: a regional australian cohort study.</sup>
Dianheng Bu, Goulburn Valley Health, Shepparton, Australia
D. Bu, J. Torres, A. Sahu; Department of Medical Oncology, Goulburn Valley Health, Shepparton, AUSTRALIA.
Background Neoadjuvant therapy is increasingly utilised in early and locally advanced breast cancer to improve operability and provide an early in vivo assessment of treatment response. Achieving pathological complete response (pCR) is associated with improved long-term outcomes, particularly in HER2-positive and triple-negative breast cancer (TNBC). While major trials have reported neoadjuvant therapy outcomes by tumour subtype, real-world data from regional centres remain limited. This study evaluates pCR rates, treatment completion, and treatment-related toxicities in a regional Australian cohort receiving neoadjuvant therapy for breast cancer. Methods A retrospective audit was conducted of all female patients with early or locally advanced breast cancer who received neoadjuvant systemic therapy followed by surgery at Goulburn Valley Health, Victoria, between January 2020 and May 2025. Data collected included demographics, tumour subtype, treatment details, hospitalisations during therapy, and rate of pathological complete response. Results 70 patients underwent neoadjuvant therapy at Goulburn Valley Health. Subtypes included HER2-positive (n=28), triple-negative breast cancer (TNBC; n=26), and hormone receptor-positive/HER2-negative (HR+/HER2−; n=16). 47 patients (67.1%) completed their planned neoadjuvant therapy. 21 patients (30%) required hospital admission during their neoadjuvant therapy. Surgical procedures after therapy included breast-conserving surgery in 36 patients, single mastectomy in 25, and bilateral mastectomy in 8 patients. 1 TNBC patient did not proceed to surgery due to disease progression. Pathologic complete response (pCR) rates were 42.9% (12/28) in HER2-positive, 38.5% (10/26) in TNBC, and 6.3% (1/16) in HR+/HER2− patients. Notable differences in pCR rates were observed between patients who completed neoadjuvant therapy and those who did not. In the HER2-positive group, 57.1% (12/21) of patients who completed therapy achieved pCR, compared to 0% (0/7) among non-completers. In the TNBC group, pCR was achieved in 50% (8/16) of patients who completed therapy versus 20% (2/10) of patients who did not. Conclusion This regional Australian cohort demonstrates pCR rates comparable to published literature, with a higher pCR rate seen in HER2+ and TNBC subtypes versus HR+/HER2−. Completion of the planned neoadjuvant therapy regimen was associated with increased pCR rates in both TNBC and HER2-positive subgroups. These findings highlight the importance of supportive care to minimise treatment interruptions, aiding in neoadjuvant therapy completion.
Presentation numberPS5-05-13
Pathologic complete response rates after neoadjuvant chemotherapy and pembrolizumab in patients with triple negative breast cancer treated at the Royal Marsden:A retrospective analysis
Nkemdilim Chukwuma, The Royal Marsden NHS foundation trust sutton, Sutton,Surrey, United Kingdom
C. A. Chigbo1, K. Udeogu2, F. Sheima3, N. Chukwuma3, A. Khan3, S. Redana3, S. Mcgrath3, A. Ring3, S. Johnston3, N. Battisti3; 1Radiation and clinical oncology, University of Nigeria teaching Hospital Ituku Ozalla Enugu and The Royal Marsden Foundation Trust, Enugu and Sutton Surrey, UNITED KINGDOM, 2Radiation and clinical oncology, University of Nigeria teaching Hospital Ituku Ozalla Enugu, Enugu, NIGERIA, 3medical oncology, The Royal Marsden NHS foundation trust sutton, Sutton,Surrey, UNITED KINGDOM.
Background: Triple-negative breast cancer (TNBC) is associated with high rates of early recurrence and poor prognosis. Chemotherapy remains the mainstay of treatment for early-stage TNBC in the preoperative setting. The KEYNOTE-522 recently demonstrated that adding pembrolizumab to chemotherapy significantly increases pathological complete response (pCR) rates in TNBC. However, real-world efficacy data remains limited. This study aims to evaluate the pCR rates among TNBC patients treated with neoadjuvant pembrolizumab at our Institution. Methods: We retrospectively retrieved data on patients diagnosed with stage I-III TNBC receiving neoadjuvant pembrolizumab at The Royal Marsden between June 2021 and June 2024. We collected data on patient demographics, tumour characteristics, treatment, and surgical pathology. We used simple statistics to evaluate the rate of pCR (ypT0/is, ypN0) after neoadjuvant systemic therapy. Results: Seventy-eight patients were included in the analysis. The median age at diagnosis was 49 years (interquartile range: 40-58). Most patients had clinical stage IIA at presentation (n=32, 41.0%) and invasive ductal carcinoma (n=74, 94.9%). Most tumours were grade 2 (n=65, 83.3%). Fifty-eight patients (74.3%) were able to complete the full regimen of chemotherapy. Among 20 patients discontinuing the treatment earlier, toxicity was the main reason for treatment discontinuation in 10 (50.0%). Forty-eight patients (61.5%) completed 8 cycles of pembrolizumab before surgery. Pembrolizumab was discontinued in 20 (25.2%), most commonly due to severe immune-related adverse effects in 15 patients (75.0%). Surgery involved breast-conservation in 43 patients (55.1%) and mastectomy in 35 patients (44.9 %). Nodal surgeries performed included sentinel lymph node biopsy in 43 patients (55.1%), axillary nodal clearance in 17 patients (21.8%), and targeted axillary dissection in 16 patients (20.5%). Overall, 41 patients achieved a pathologic complete response (pCR) rate (52.6%). pCR was more frequent in earlier-stage tumours: ( 100.0% [n=2] in stage I, 58.5% [n=31] in stage II, and 34.8% [n=8] in stage III) and in higher-grade tumours (36.4% [n=4] of grade 1 tumours, 55.4% [n=36] in grade 2 tumours, and 50% [n=1] in grade 3 tumours). pCR was achieved by 40 patients with ductal tumours (97.6%) versus 1 (2.4%) patient with other histology types pCR was seen in 29 patients who completed the planned course of chemotherapy (50%) versus 12 patients who did not (60%), and in 26 patients who completed the planned 8 cycles of pembrolizumab (54.2%) versus 15 of those who did not (50%). However, no statistically significant association was found between pCR and clinical stage, tumour grade /histology, number of chemotherapy or number of pembrolizumab cycles received (p = 0.05). Conclusion: In our analysis, the addition of pembrolizumab to chemotherapy resulted in similar pCR rates compared with published evidence. These findings reinforce the role of immunotherapy in early TNBC.. Further analyses with longer follow-up are warranted to evaluate the impact of pCR on survival in the real world.
Presentation numberPS5-05-14
Real-world outcomes of trastuzumab deruxtecan (T-DXd) in patients with HER2+ metastatic breast cancer: Turkish oncology group multicenter study
Taha Koray Sahin, Hacettepe University, Ankara, Turkey
T. Sahin1, F. Kemik2, H. Muglu3, S. Biter4, S. Tunbekici5, A. Oruc6, A. Guren7, K. Kaban8, M. Aykan9, B. Koylu2, O. Ekinci10, I. Deliktas Onur11, A. Kalem12, O. Altunok13, M. Seyyar14, B. Guney15, O. Akdogan16, M. Yazici17, N. Majidova18, E. Esin19, D. Cabuk17, O. Yazici16, R. Gursu15, M. Yilmaz13, H. Yesil Cinkir12, O. Ates11, E. Celik10, N. Karadurmus9, N. Mandel8, I. Bayoglu7, M. Araz6, E. Goker5, E. Bayram4, D. Erdem20, F. Selcukbiricik2, G. Basaran21, A. Sezer22, A. Bilici3, S. Gunduz2, D. Guven1, S. Aksoy1; 1Department of Medical Oncology, Hacettepe University, Ankara, TURKEY, 2Department of Medical Oncology, Koç University School of Medicine, Istanbul, TURKEY, 3Department of Medical Oncology, Istanbul Medipol University, Istanbul, TURKEY, 4Department of Medical Oncology, Cukurova University, Adana, TURKEY, 5Department of Medical Oncology, Ege University, Izmir, TURKEY, 6Department of Medical Oncology, Necmettin Erbakan University, Konya, TURKEY, 7Department of Medical Oncology, Marmara University, Istanbul, TURKEY, 8Department of Medical Oncology, American Hospital, Istanbul, TURKEY, 9Department of Medical Oncology, Health Science University, Gulhane School of Medicine, Ankara, TURKEY, 10Department of Medical Oncology, Prof. Dr. Cemil Taşcıoğlu City Hospital, University of Health Sciences, Istanbul, TURKEY, 11Department of Medical Oncology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research, Ankara, TURKEY, 12Department of Medical Oncology, Gaziantep University, Gaziantep, TURKEY, 13Department of Medical Oncology, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, TURKEY, 14Department of Medical Oncology, Gaziantep City Hospital, Gaziantep, TURKEY, 15Department of Medical Oncology, Istanbul Training and Research Hospital, Istanbul, TURKEY, 16Department of Medical Oncology, Gazi University, Ankara, TURKEY, 17Department of Medical Oncology, Kocaeli University, Kocaeli, TURKEY, 18Department of Medical Oncology, VM Medical Park Maltepe Hospital, Istanbul, TURKEY, 19Department of Medical Oncology, Bayındır Hospital, Ankara, TURKEY, 20Department of Medical Oncology, Samsun Medical Park Hospital, Samsun, TURKEY, 21Department of Medical Oncology, Acıbadem University, Istanbul, TURKEY, 22Department of Medical Oncology, Başkent University, Adana, TURKEY.
Background: Trastuzumab deruxtecan (T-DXd) has transformed the treatment landscape of HER2+ metastatic breast cancer (mBC), with significant improvements in survival reported in clinical trials. However, limited data exist regarding its performance in real-world settings, particularly in lower-middle-income countries (LMICs). We aimed to evaluate the real-world effectiveness and safety of T-DXd in patients with HER2+ mBC in Türkiye. Methods: This multicenter, retrospective cohort study, conducted by the Turkish Oncology Group, evaluated the real-world outcomes and tolerability of T-DXd in patients with HER2+ mBC across 22 oncology centers in Türkiye. The primary endpoints were real-world progression-free survival (rwPFS) and overall survival (OS). Secondary endpoints included objective response rate (ORR) and safety outcomes. Results: A total of 206 patients were included. The median age was 49 years (IQR: 42-59), and the median follow-up was 11.5 months. Among all patients, 72.3% had HER2 IHC 3+ disease, and 61.7% were HR-positive. T-DXd was administered in second line in 54 patients (26.2%), as third-line therapy in 63 patients (30.6%), and in the fourth line or beyond in 90 patients (43.7%). Patients had received a median of two lines of prior therapy (range: 0-8).The median rwPFS was 16.8 months (95%CI: 12.7-20.9), and the median OS was 29.2 months (95%CI: 22.1-36.3). Among the 192 patients evaluable for response, the ORR was 77.6% and the disease control rate (DCR) was 91.7%. Second line use of T-DXd in was associated with significantly longer rwPFS compared to later lines (p = 0.004). Treatment-related adverse events (AEs) of any grade occurred in 59% of patients. Interstitial lung disease (ILD) was reported in 20 patients (9.7%), with 4 cases being grade 3 or higher. Among the 12 patients with grade 1 ILD eligible for rechallenge, 83.3% (10/12 patients) were rechallenged. Conclusion: In this large real-world cohort from an LMIC, T-DXd demonstrated robust antitumor activity and a manageable safety profile in patients with HER2+ mBC. These findings are consistent with prior clinical trial data and support the applicability of T-DXd in broader clinical settings.
Presentation numberPS5-05-15
Sequencing Matters: Impact of EC-first versus Carboplatin-first Ordering on pCR in Triple-Negative Breast Cancer Real-World-Data
Jenny Katharina Wagner, Charité – Universitätsmedizin, Berlin, Germany
J. Wagner, I. Vasconcelos, J. Blohmer; Breast Center, Charité – Universitätsmedizin, Berlin, GERMANY.
Background Keynote-522 administers four cycles of Carboplatin + Paclitaxel (Carbo/P) together with pembrolizumab (Pembro), followed by Doxorubicin/Epirubicin + Cyclophosphamide (EC). In routine care, some centres invert the order, initiating therapy with EC. Whether sequencing influences delivery of the complete regimen or the likelihood of achieving a pathologic complete response (pCR) is uncertain.MethodsElectronic drug-administration logs from all patients with stage I-III TNBC treated with Pembro-EC-Carbo/P between January 2022 and July 2024 were analysed. Each EC or Carboplatin infusion was counted as a macro-cycle (maximum four each), and every Paclitaxel dose was grouped into twelve weekly micro-cycles, yielding a theoretical maximum of twenty cycles. Carboplatin administrations were classified as high-dose (area under the curve, AUC ≈ 5) or low-dose (AUC ≈ 1.5). pCR was defined as ypT0 or documented “pCR”. Pathogenic germline variants (pV) and clinical stage at diagnosis were retrieved from the hospital registry. Group comparisons used the Mann-Whitney U and Fisher’s exact tests (α = 0.05).ResultsSixty-five patients met inclusion criteria (median age 50 years; stage I 10%, II 46%, III 44%). Forty-four began with EC (EC-first) and twenty-one with Carbo/P (Carboplatin-first). EC-first patients completed a median of 18 cycles compared with 15 cycles in Carbo-first (p = 0.004); 73% versus 38% finished phase-2 Carboplatin, and 46% versus 24% received at least 18 cycles. High-AUC Carboplatin exposure (median four infusions) and cumulative Paclitaxel dose (2,300 mg versus 2,250 mg) were similar. The pCR rate was 48% (21/44) for EC-first and 38% (8/21) for Carboplatin-first (p = 0.46). Documented BRCA1/2 (pV) or other pVs were present in 8% of patients; their pCR rate was 57% compared to 43% in patients without a pV. Therapy discontinuation occurred in 14% of both cohorts, most commonly due to hematologic toxicity or disease progression.ConclusionsCommencing pembrolizumab-based neoadjuvant therapy with EC was associated with superior regimen completion and a numerically higher, though not statistically significant, pCR rate compared with starting with Carbo/P, without an increase in early termination. Of note, there were considerably more stage III patients than in the trial population—potentially harder to sterilise—which may account for the lower pCR rates seen. These real-world findings support prospective evaluation of an EC-first sequence and emphasise the importance of maintaining dose intensity in everyday practice.
Presentation numberPS5-05-16
From Keynote522 to real world evidence: The use of Pembrolizumab in early triple-negative breast cancer
Jonas Roth, University of Regensburg, Regensburg, Germany
J. Roth1, J. Albers1, M. Gerken2, O. Ortmann1, S. Seitz1; 1Gynecology and Obstetrics, University of Regensburg, Regensburg, GERMANY, 2Tumor Center Regensburg, University of Regensburg, Regensburg, GERMANY.
The KEYNOTE-522 trial demonstrated significantly higher rates of pathological complete response (pCR) with the addition of pembrolizumab to neoadjuvant chemotherapy in patients with non-metastatic triple-negative breast cancer (TNBC). In this analysis, we aimed to evaluate whether real-world data support the benefit of adding immunotherapy to neoadjuvant chemotherapy for patients with early-stage TNBC. Additionally, we analyzed several subgroups to identify whether specific patient populations derive greater benefit from immunotherapy.Data were collected at the Tumor Center Regensburg. We included 160 patients diagnosed with non-metastatic TNBC between January 2020 and May 2025 at the Comprehensive Cancer Center Regensburg. Of these, 88 patients received neoadjuvant chemotherapy plus pembrolizumab, while 72 patients received chemotherapy alone. We observed pCR rates of 61.4% in the chemotherapy plus pembrolizumab group compared to 51.4% in the chemotherapy-only group, demonstrating an absolute increase of 10% favoring the addition of pembrolizumab.Subgroup analyses revealed that patients with a high Ki-67 score, particularly above 35%, achieved significantly higher pCR rates. Patients with BRCA mutations also showed markedly higher pCR rates in the pembrolizumab arm. Interestingly, younger patients demonstrated significantly higher pCR rates in both treatment arms compared to older patients. Detailed subgroup analyses will be presented.This real-world study demonstrates that the addition of pembrolizumab in routine clinical practice is associated with improved pCR rates, showing an absolute difference of 10%. Moreover, it suggests that specific subgroups may particularly benefit from immunotherapy. However, larger real-world studies are needed to enable more robust subgroup analyses.
Presentation numberPS5-05-17
Real-world effectiveness of Trastuzumab Deruxtecan in HER2-low Metastatic breast cancer: a single-center retrospective study
Yifei Chen, Peking University Cancer Hospital, Beijing, China
Y. Chen, G. Song, H. Li; Department of Breast Oncology, Peking University Cancer Hospital, Beijing, CHINA.
Real-world effectiveness of Trastuzumab Deruxtecan in HER2-low Metastatic breast cancer: a single-center retrospective study Yifei Chen, Guohong Song, Huiping Li Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital &Institute, Beijing 100142, China Background: Clinical trials of trastuzumab deruxtecan (T-Dxd) have expanded options for HER2-low breast cancer, yet real-world patients often present with poorer prognostic features such as active brain metastases or heavy pretreatment. Robust effectiveness data in routine practice are therefore needed. Methods: This is a retrospective real-world monocentric study conducted by 81 patients with HER2-low advanced breast cancer, who were treated with T-Dxd. Database from the Department of Breast Oncology at Peking University Cancer Hospital was reviewed, spanning March 2023 to January 2025. All patients underwent at least one radiologic response assessment. Results: The median progression-free survival (PFS) was 6.0 months (95%CI, 3.8-8.1). There were 68 HR+(83.95%) and 13 HR- (16.05%) patients included. HR+ disease showed a median PFS of 6.7 months (95%CI, 5.2-8.2) versus 3.4 months (95%CI, 2.1-4.7) in triple-negative breast cancer (TNBC), no statistical significance between these two groups (P=0.062). TNBC patients had a significantly shorter median overall survival (OS) (8.7 months, (95%CI, 2.2-15.2) , P=0.002), while mOS was not reached in HR+ group.Presence of visceral, liver, lung, or CNS lesions did not materially alter PFS (6.0 vs 5.9 months, P=0.906). Among 13 patients (16.05%) with active brain metastases, disease-control rate was 84.62% and 5/6 evaluable patients achieved CNS partial response.Baseline IHC score (HER2 1+ vs 2+) did not affect PFS (5.9 vs 6.0 months, P=0.707). But patients whose metastatic sites showed up-regulated HER2 expression achieved longer PFS (7.0 vs 3.4 months, P=0.015).The cohort had received a median of 4 prior lines of systemic therapy, including a median of 2 chemotherapy regimens. Administering T-Dxd after more than 3 prior chemotherapy lines or after exposure to topoisomerase I/II inhibitors significantly shortened PFS (4.5 vs 7.0 months, P=0.018 and 3.2 vs 6.7 months, P=0.002, respectively).Of the 48 patients who underwent germline BRCA testing, 13 carried BRCA1/2 mutations and 35 were wild type. Although mPFS was numerically longer in the mutant subgroup (7.5 months) than in the wild-type subgroup (4.8 months), the difference was not statistically significant (P = 0.607). Among the 13 BRCA-mutated patients, 10 had previously received PARP inhibitors; their mPFS did not differ from that of PARP-naïve patients (7.5 vs 4.5 months, P = 0.989).PAM-pathway alterations (PIK3CA/AKT/PTEN) were evaluated in 41 patients, with 11 found to be mutant. Median PFS was similar between the PAM-mutated and wild-type groups (6.0 vs 4.2 months, P = 0.065). Conclusions: In routine clinical practice, T-Dxd delivers meaningful disease control for heavily pre-treated HER2-low metastatic breast cancer, including patients with active brain metastases—but its effectiveness is more modest than that seen in randomized trials. Greatest benefit is observed when: (i) metastatic lesions up-regulate HER2 expression, and (ii) T-Dxd is introduced earlier in the chemotherapy sequence. Outcomes are less favorable in triple-negative disease and after prior topoisomerase-targeted therapy, suggesting that biomarker- and timing-guided patient selection could optimize real-world use.
Presentation numberPS5-05-18
How Susan G. Komen’s ShareForCures® Research Hub will connect researchers with real-world data shared by breast cancer patients and survivors in the United States.
Kelsi O West, Susan G. Komen, Dallas, TX
K. O. West1, J. Epps1, E. G. Marks1, A. L. Gyden1, K. A. Sabelko1, M. L. Bondy2, J. Jourquin1; 1Data Science, Susan G. Komen, Dallas, TX, 2Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA.
Background: To optimize care and improve outcomes for all breast cancer patients, multi-modal data need to be integrated with the experiences of patients. However, access to these datasets that reflect the diversity of patients is often limited and siloed within single institutions. To address these challenges, Susan G. Komen® (Komen) launched ShareForCures® (SFC), a direct-to-consumer, online, IRB-approved breast cancer research registry, designed to integrate real-world, longitudinal data from adults who have been diagnosed with breast cancer and live in the United States and to provide this data for research. Methods: We analyzed data from participants who enrolled in SFC within the first two years and developed the SFC Research Hub, which facilitates data exploration, cohort creation and data access application. A taskforce of research experts (clinical oncologists, translational scientists, and research advocates) provided guidance and feedback throughout the development of the SFC Research Hub by completing surveys, meeting with Komen staff, and participating in user testing. Results: From July 2023 to June 2025, 1,214 participants enrolled in SFC (median age = 55 years [range: 27-93], 99% women, 75% non-Hispanic White). Only four participants withdrew after enrollment, resulting in a retention rate of 99.7%. Participants were geographically dispersed across the United States, including Puerto Rico. Most participants received oncology care outside of NCI-designated comprehensive cancer centers and were diagnosed within the past five years. The majority of participants reported their first breast cancer diagnosis as invasive ductal carcinoma (57%), with 39% reporting ER+/HER2- disease. Notably, 18% of participants reported living with metastatic breast cancer (18%) and 14% experienced a recurrence (14%). All participants reported demographic and diagnostic information, and 53% of participants completed additional questionnaires and/or shared saliva samples for germline whole genome sequencing. Medical records were requested for 52% of participants, 30% were retrieved and curated, and 13% have access to a summary of their records in the platform. The SFC Research Hub was designed and developed with end users in mind, offering secure access to integrated, deidentified data shared by SFC participants. To access SFC data, researchers must agree to a code of conduct, acknowledge their institution’s data use agreement with Komen, and receive approval of their data access application describing their research project from Komen. In return, researchers will share their findings with SFC participants. Conclusions: Through SFC, Komen is creating a real-world database that connects researchers with data contributed by participants with diverse experiences. The SFC Research Hub is expected to launch in 2026, enabling researchers to leverage SFC data to enhance our understanding of real-world breast cancer patient experiences, ultimately accelerating discoveries and improving outcomes for all breast cancer patients.
Presentation numberPS5-05-19
Real-world comparison of trastuzumab deruxtecan treatment outcomes in HER2-positive or HER2-low metastatic breast cancer patients with and without brain metastases
Vikram Gorantla, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA
V. Gorantla1, R. Choksi1, F. Kudrik2, S. Reddy3, S. Reganti3, S. Rosenfeld4, G. Cioffi5, D. Parris5, A. Rui5, M. Gart5, C. Wall5, B. Wang5, P. Varughese5, J. Donegan5, L. Morere5, R. Geller5, J. Scott5, D. Patt6; 1Medical Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 2Medical Oncology, South Carolina Oncology Associates, Columbia, SC, 3Medical Oncology, Cancer Care Specialists, Reno, NV, 4Medical Oncology, Highlands Oncology Group (HOG), Fayetteville, AR, 5PrecisionQ, IntegraConnect, West Palm Beach, FL, 6Medical Oncology, Texas Oncology, Austin, TX.
Background: In the DESTINY-Breast12 trial, individuals with HER2-positive (HER2+) metastatic breast cancer (mBC) who received trastuzumab deruxtecan (T-DXd) had similar progression-free survival (PFS) regardless of the presence of brain metastases (BrM). We sought to evaluate time to treatment discontinuation or death (TTD), time to next treatment (TTNT; a surrogate for PFS), and overall survival (OS) using real-world data to assess patients (pts) outside of a clinical trial setting for both HER2+and HER2-low mBC. Methods: We used the Integra Connect PrecisionQ real-world de-identified electronic health record database of >3 million cancer pts across >500 sites of care to identify 374 HER2+ mBC pts and 512 HER2-low mBC pts for the analysis. Eligible pts were adults with HER2+ or HER2-low mBC with ≥2 documented visits who initiated T-DXd between 1-1-2020 and 5-31-2024, with observation through 11-30-2024. The index date was the initiation of T-DXd. Documentation of BrM was determined at or prior to index date. Patient data included structured data supplemented with data obtained via chart abstraction. TTD, TTNT, and OS by documentation of BrM were analyzed using Kaplan-Meier analysis. Unadjusted hazard ratios (HR) were estimated using Cox proportional hazard models. Results: A total of 374 HER2+ pts were identified, including 94 with documented BrM and 280 without. The median age at index was 62.5 (interquartile range [IQR] 54, 71) years. A total of 512 HER2-low pts were identified, 80 of whom had documented BrM and 432 of whom did not. The median age at index was 65 (IQR 56, 71) years. Pts with HER2+ mBC did not have statistically significant differences in median TTD, TTNT, or OS regardless of the presence of BrM. However, pts with HER2-low mBC without BrM had significantly longer median TTD (5.5 months vs. 3 months, logrank p=0.002), TTNT (7.2 months vs. 5.8 months, logrank p=0.002), and OS (15.8 months vs. 10.2 months, logrank p<0.001) compared to those with BrM. Conclusions: This real-world comparison of T-DXd treatment reaffirmed findings from DESTINY-Breast12 in which PFS did not differ among HER2+ mBC pts based on the presence of BrM. While this study showed significantly longer median TTD, TTNT, and OS in HER2-low mBC pts without BrM compared to those with BrM, further research and larger samples sizes are needed to understand how the line of therapy for T-DXd impacts these findings.
| HER2-Positive | HER2-Low | |||||||||||||||||||||||||||||||||
| Median, months (95% CI) | HR (95% CI), P value | Median, months (95% CI) | HR (95% CI), P value | |||||||||||||||||||||||||||||||
| BrM (n=94) | No BrM (n=280) | BrM (n=80) | No BrM (n=432) | |||||||||||||||||||||||||||||||
| TTD | 9.2 (6.4, 14.7) | 12.4 (9.9, 14.7) | 1.07 (0.81, 1.42), 0.64 | 3.0 (2.1, 4.8) | 5.5 (4.6, 6.3) | 1.51 (1.16, 1.97), 0.0021 | ||||||||||||||||||||||||||||
| TTNT | 14.1 (7.9, 17.7) | 15.2 (13.4, 16.5) | 1.10 (0.81, 1.48), 0.55 | 5.8 (3.6, 6.5) | 7.2 (6.5, 8.2) | 1.52 (1.16, 1.97), 0.002 | ||||||||||||||||||||||||||||
| OS | 14.1 (7.9, 17.7) | 15.2 (13.4, 16.5) | 1.01 (0.68, 1.49), 0.97 | 10.2 (6.6, 11.9) | 15.8 (14.2, 18.9) | 1.98 (1.46, 2.69), <0.0001 |
Presentation numberPS5-05-20
Real-world treatment patterns and outcomes in patients with HER2+ metastatic breast cancer after treatment with trastuzumab deruxtecan (T-DXd) in the US
Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA
S. M. Tolaney1, S. Valliant2, S. Hunter3, O. Tymejczyk4, A. Yu5, J. Earla6, E. Pang7; 1Breast Oncology, Dana-Farber Cancer Institute, Boston, MA, 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, 7Global Oncology Medical Affairs, Daiichi Sankyo, Inc., Basking Ridge, NJ.
Background: Human epidermal growth factor receptor 2-positive breast cancer (HER2+ BC) accounts for 15% to 20% of approximately 300,000 new BC cases diagnosed annually in the US. The treatment landscape for HER2+ metastatic BC (mBC) has evolved significantly, and trastuzumab deruxtecan (T-DXd [a HER2 antibody-drug conjugate]) is now the preferred standard of care in the second-line (2L) setting. Optimal post-T-DXd treatment pathways are unclear due to the lack of data for regimens in the 3L or 4L setting for patients with HER2+ mBC. This study aimed to better characterize real-world (rw) treatment patterns and clinical outcomes in patients with HER2+ mBC who were previously treated with T-DXd in the rw practice setting. Methods: This was a noninterventional, retrospective, observational cohort study of patients diagnosed with HER2+ mBC who initiated a subsequent treatment after receiving T-DXd in the metastatic setting, using the US-based Flatiron Health Enhanced Datamart. The study observation period was from January 1, 2011, to March 31, 2024. Patients with HER2+ mBC treated with ≥1 T-DXd-containing line of therapy (LOT) and a subsequent LOT, with an index date ≥90 days prior to the end of the observation period, were included. Index date was defined as the start of the LOT that followed the first T‑DXd-containing LOT. Patient demographics, clinical characteristics, and treatment utilization were assessed descriptively. 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. Results: The study included 228 patients (mean age, 60.3 years; 99.6% were female and 66.7% were White). Most patients initiated their index treatment in the 4L setting (median of 3 LOTs prior to index date; line of index treatment: 2L [4.4%], 3L [22.4%], 4L [31.1%], 5L [22.8%], 6L [11.0%], 7L+ [8.3%]). 64.5% of patients had ≥3 metastatic sites at the index date; 31.6% had CNS metastases. With a median follow-up of 10.4 months (range, 0.5-42.4 months), the median rwPFS was 4.3 months (95% CI, 3.5-5.0 months) and the median rwOS was 15.7 months (95% CI, 12.1-20.0 months). The median rwTTD/D and rwTTNT/D were 6.2 and 6.9 months, respectively. The most common regimens received as the index LOT were trastuzumab + tucatinib + chemotherapy (TTC; 34.2%), other anti-HER2 (18.9%), trastuzumab + chemotherapy (12.7%), other (12.7%), margetuximab + chemotherapy (8.8%), trastuzumab emtansine (T-DM1; 6.6%), and anti-HER2 TKI + chemotherapy (6.1%). Treatment sequencing varied by index line number. T-DM1-based regimens were more common when the index LOT was 3L (15.7%) vs 4L (5.6%) or 5L (3.8%), and 21% to 22% of patients with index 3L or 4L treatment received other anti-HER2 therapies immediately post T-DXd vs 13.5% with index 5L treatment. Conclusions: Treatment sequencing data suggest that TTC and other anti-HER2 therapies are being used in the post-T-DXd rw setting, though potential differences by line exist. This study demonstrates an ongoing and substantial unmet need for more effective treatments among patients with HER2+ mBC previously treated with T-DXd, as shown by the short rwPFS/OS and short times on subsequent treatments following T-DXd.
Presentation numberPS5-05-22
A Multicenter Retrospective Cohrt Study on Long-Term Outcomes after Partial Mastectomy in Japan.
Goro Kutomi, Juntendo University School of Medicine, Tokyo, Japan
G. Kutomi1, K. Kida2, Y. Kajiura2, K. Narui3, K. Kataoka4, M. Taguri5, K. Harada5, H. Shima6, R. Nakamura7, T. Ohnishi8, T. Nakayama9, M. Nagahashi10, T. Sankai11, K. Orimoto12, Y. Horimoto12, T. Ishikawa12; 1Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, JAPAN, 2Department of Breast Surgical Oncology, St. Luke’s International Hospital, Tokyo, JAPAN, 3Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Kanagawa, JAPAN, 4Advanced Medical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, JAPAN, 5Department of Medical Data Science, Tokyo Medical Univesity, Tokyo, JAPAN, 6Department of Surgery, Division of Breast Surgery, Sapporo Medical University School of Medicine, Hokkaido, JAPAN, 7Department of Breast Surgery, Chiba Cancer Center Hospital, Chiba, JAPAN, 8Department of Breast Surgery, National Cancer Center Hospital East, Chiba, JAPAN, 9Department of Breast and Endocrine Surgery, Osaka International Cancer Institute, Osaka, JAPAN, 10Department of Surgery, Division of Breast and Endocrine Surgery, Hyogo Medical University School of Medicine, Hyogo, JAPAN, 11Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, Kanagawa, JAPAN, 12Department of Breast Oncology and Surgery, Tokyo Medical Univesity, Tokyo, JAPAN.
Background: With recent advances in breast cancer treatment, changes in local recurrence and survival following breast-conserving surgery (BCS) are also anticipated over time. Techniques for BCS vary across countries, and anatomical differences among ethnic groups may influence patterns of local recurrence risk. Objective of this study: To investigate the recent rates of local recurrence and prognosis following BCS in Japan using multi-institutional real-world data, with a particular focus on identifying trends and risk factors for local recurrence through comparison with historical data. Methods: This multicenter retrospective cohort study included 8,897 breast cancer patients who underwent BCS between January 2014 and December 2018 at 25 participating institutions across Japan. Data on patient demographics, surgical details, pathological factors including margin status, adjuvant therapy, and outcomes were collected. The primary endpoint was local recurrence rate. Secondary endpoints included distant recurrence rate, overall mortality, and breast cancer-specific mortality. Local recurrence rate (LRR), disease-free survival (DFS) and overall survival (OS) were estimated by Kaplan-Meier analysis; Results: Median follow-up was 6.6 years. In the overall population (N=8,897), Clinical stages at diagnosis were Stage 0 (n=918:10.3%), Stage I (n=4,539:51.0%), Stage II (n=2,566: 28.8%), and Stage III (n=360:4.0%). 5 year LRR, DFS and OS were 2.0%(1.7-2.3: 95%Cl), 94.1%(93.6-94.6: 95%Cl) and 97.8%(97.4-98.1: 95%Cl), respectively. 10 year LRR, DFS and OS were 4.6%(3.6-5.5: 95%Cl), 86.2%(84.1-88.3: 95%Cl) and 94.1%(93.1-95.1: 95%Cl), respectively. Potential risk factors for local recurrence are younger age of onset, tumor size, high Ki-67 , LV invasion, lack of adjuvant therapy (Table 1). Adjuvant systemic treatment was suggested to contribute to a reduced risk of local recurrence in all subtypes. Conclusions: Our recent data suggest that outcomes of BCS in Japan have improved compared to the past. As a growing number of new systemic therapies continue to be introduced each year, it is crucial to incorporate contemporary data—rather than relying solely on historical studies—when considering optimal surgical strategies.
Presentation numberPS5-05-23
Real-world Treatment Patterns of Capivasertib in Metastatic Breast Cancer in the US
Kelsey H Natsuhara, University of California, San Francisco/Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
K. H. Natsuhara1, L. Park2, S. Udayachalerm3, R. Mireia4, B. Murphy5, B. Nordstrom3, L. A. Huppert1; 1Oncology, University of California, San Francisco/Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 2Oncology outcomes research, Astrazeneca, Gaithersburg, MD, 3RWE, PPD Evidera Real World Data & Scientific Solutions, Thermo Fisher Scientific, Wilmington, NC, 4RWE, PPD Evidera Real World Data & Scientific Solutions, Thermo Fisher Scientific, Sundbyberg, SWEDEN, 5RWE, PPD Evidera Real World Data & Scientific Solutions, Thermo Fisher Scientific, Wilmington, NC, USA, Wilmington, NC.
BACKGROUND: In patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative metastatic breast cancer (MBC) with ≥ 1 PIK3CA, AKT1, or PTEN tumor alterations, capivasertib plus fulvestrant is approved in the US for use after progression on at least one endocrine-based treatment. This study assessed real-world treatment patterns of capivasertib in the US. METHODS: This retrospective cohort study used Flatiron Health electronic health record-derived database. Eligible patients were ≥18 years, diagnosed with MBC, initiated capivasertib between November 16, 2023 and July 31, 2024, and had at least 6 months (mos) of potential follow-up time from capivasertib initiation (index date). Real-world time to treatment discontinuation (rwTTD) and time to next line of treatment (rwTTNT) were assessed from the index date. Summary statistics were used to describe patient characteristics at baseline and treatment patterns. rwTTD and rwTTNT were estimated using Kaplan-Meier methods. The line of therapy (LOT) was defined using an algorithm that incorporated prior treatment use. RESULTS: This study identified 412 patients with MBC who received capivasertib. The median age was 67 years (IQR 60, 75). The majority were female (98.8%), white (71.6%), and from community practice settings (83.0%). Prior to capivasertib initiation, sites of disease included: bone (91.0%), liver (44.2%), lung (28.2%), and brain (7.0%). PIK3CA, AKT1, or PTEN alterations were documented in 90.3% of patients: PIK3CA (75.0%), PTEN (14.3%), and AKT1 (10.0%) alterations. Co-mutations with ESR1 mutations were observed in 25.5% of patients. Prior therapies for MBC included: CDK4/6 inhibitors (87.1%), fulvestrant (66.0%), chemotherapy (31.8%), alpelisib (20.6%), and antibody-drug conjugates (14.1%) in any prior LOT. Most patients received capivasertib with fulvestrant (n=387, 93.9%); in 25 patients (6.1%), use with another endocrine therapy/combination or alone was recorded in the chart, but could not be verified due to data limitations. Among patients recorded as having received capivasertib with fulvestrant (n=387/412, 93.9%), more than half of patients received capivasertib in second-line (2L) (n=115, 29.7%) or 3L (n=102, 26.4%), though many were treated in 4L+ (n=146, 37.7%). 2L and 3L median rwTTNT was 7.1 mos (IQR 5.8, NE) and 6.9 mos (IQR 6.1, 7.8), respectively. 2L and 3L median rwTTD was 6.9 mos (IQR 5.3, NE) and 6.6 mos (IQR 5.2, 7.1), respectively (Table 1). CONCLUSIONS: Findings from this large database analysis of US patients demonstrate the effectiveness of capivasertib + fulvestrant in real-world practice. Clinical outcomes in 2L and 3L closely match those observed in the CAPItello-291 Phase 3 randomized controlled trial, which supported FDA approval of the capivasertib + fulvestrant regimen. Numerically improved outcomes were observed in patients who used capivasertib in earlier vs later line settings.
| Capivasertib+fulvestrant (N=387) |
rwTTNT
median (95% CI), months
|
rwTTD
median (95% CI), months
|
| 1L (n=24) | 10.0 (6.5 – NE) | 9.5 (5.6 – NE) |
| 2L (n=115) | 7.1 (5.8 – NE) | 6.9 (5.3 – NE) |
| 3L (n=102) | 6.9 (6.1 – 7.8) | 6.6 (5.2 – 7.1) |
| 4L (n=45) | 6.2 (4.8 – 9.9) | 5.5 (4.3 – 7.4) |
| 5L+ (n=101) | 5.0 (3.9 – 6.7) | 4.6 (3.5 – 5.9) |
|
median follow-up of 7.8 months (IQR 6.2, 10.1)
|
Presentation numberPS5-05-24
Real-world validation of the 21-gene Recurrence Score in Node Negative Breast Cancer: A Comparative Analysis with TAILORx.
Ryan Rodgerson, University of East Anglia, Norwich, United Kingdom
R. Rodgerson1, M. Youssef2; 1Norwich Medical School, University of East Anglia, Norwich, UNITED KINGDOM, 2Breast Surgery/General Surgery, Norfolk and Norwich University Hospital, Norwich, UNITED KINGDOM.
¹Sparano JA et al. TAILORx trial: endocrine vs chemoendocrine therapy in HR+/HER2− breast cancer Background: The TAILORx¹ trial established the 21-gene Recurrence Score as a key tool for guiding chemotherapy decisions in node negative, hormone receptor-positive, HER2-negative breast cancer. While its prognostic utility is well supported in clinical trials, its real-world validation remains limited. This study evaluates the 21-gene Recurrence Score’s performance in a real-world cohort and compares outcomes with TAILORx¹-defined risk groups. Methods: We conducted a retrospective cohort study using prospectively collected data of 388 node-negative patients who underwent 21-gene Recurrence Score testing between 2015 and 2024 in a tertiary referral cancer centre. Patients were stratified into five groups based on TAILORx¹ criteria: Group A (Age > 50, RS 11-25), Group B (Age ≤ 50, RS 11-15), Group C (Age ≤ 50, RS 16-25), Group D (RS 0-10 all ages), Group E (≥ 26 all ages). Treatment data (Endocrine Therapy [ET] vs. Chemotherapy + ET [CET]) and survival outcomes were analysed using Kaplan-Meier, log-rank tests and cox regression. Results: Group A (n=151) showed slightly more events than expected (10 observed vs. 8.2 expected), but outcomes remained consistent with TAILORx¹, which found no chemotherapy benefit in this group. Group B (n=22) had significantly worse outcomes than expected (HR 6.51, p=0.021), contrasting with TAILORx¹, where this group had excellent survival with ET alone. Group C (n=42) had no events, suggesting better-than-expected outcomes. Group D (RS 0-10) also showed fewer events than predicted. Group E (RS ≥26) outcomes aligned with TAILORx¹, supporting chemotherapy use. Multivariable analysis identified CET (HR 4.25, p=0.034) and intermediate risk (HR 9.31, p=0.036) as significant predictors of poorer outcomes, likely reflecting treatment selection bias. Conclusions: Our real-world data largely align with TAILORx¹, confirming the prognostic value of the 21-gene Recurrence Score in node-negative patients. However, worse-than-expected outcomes in group B suggest biological heterogeneity or treatment adherence issues. These findings support continued use of the 21-gene Recurrence Score for risk stratification and highlight the need for careful clinical judgement in younger patients. Larger studies are warranted to validate these trends.
| Group | RS Range | Observed Events | Expected Events | Interpretation |
| Group A | 11-15 | 10 | 8.2 | Slightly more events – Consistent |
| Group B | 16-20 | 5 | 2 | Worse outcomes (p = 0.021) – Divergent |
| Group C | 21-25 | 0 | 3 | No events – better than expected |
| Group D | 0-10 | 1 | 4 | Fewer events – Consistent |
| Group E | ≥26 | 8 | 7.5 | Matches TAILORx – Consistent |
Presentation numberPS5-05-25
Is the 21-Gene Recurrence Score Validated for Real-World use in Node Positive Breast Cancer? A Retrospective Cohort Study
Ryan Rodgerson, University of East Anglia, Norwich, United Kingdom
R. Rodgerson1, M. Youssef2; 1Norwich Medical School, University of East Anglia, Norwich, UNITED KINGDOM, 2Norfolk and Norwich University Hospital, University of East Anglia, Norwich, UNITED KINGDOM.
¹Kalinsky K et al. RxPONDER (SWOG S1007), RS ≤25, HR+/HER2−, node+ BC. Cancer Res. 2023;83(5 Suppl):GS1-01 Background: The 21-gene Recurrence Score is a validated prognostic and predictive tool for breast cancer, with evidence supporting its utility over a 9-year follow-up. The RxPONDER¹ trial further confirmed its clinical relevance in a controlled setting. However, its performance in real-world clinical practice remains underexplored. This study evaluates the application of the 21-gene Recurrence Score in routine care using RxPONDER¹ criteria and determines whether its prognostic value remains in real-world data. Methods: We conducted a retrospective cohort study using prospectively collected data from Node-positive patients at the Norfolk and Norwich University, a tertiary referral cancer centre treating 650 breast cancer patients per year (2015-2024). Eligible patients were hormone receptor-positive, HER2-negative breast cancer patients with 1-3 positive lymph nodes and available 21-gene Recurrence Scores. Treatment and survival data were extracted from hospital records. Patients were stratified by menopausal status and recurrence score. Kaplan-Meier curves and Cox regression analyses assessed outcomes and compared with RxPONDER¹ findings. Results: 282 patients were included. Chemotherapy was administered to all high-risk premenopausal patients (24.1%) and selectively to intermediate-risk patients based on additional clinical factors. In post-menopausal patients (75.9%), chemotherapy was primarily reserved for high-risk cases. High 21-gene Recurrence Scores (>25) were significantly associated with poorer outcomes (HR 6.59, 95% CI 1.39-31.15, p = 0.017), intermediate scores (11-25) showed a non-significant trend (HR 2.38, p = 0.265). Endocrine therapy adherence was the strongest independent predictor of survival (HR 0.012, p = 0.004). Chemotherapy plus endocrine therapy (CET) showed no statistically significant benefit after adjustment (HR 1.82, p = 0.5) These findings align with RxPONDER¹: chemotherapy showed no significant benefit in postmenopausal patients, but some benefit in premenopausal patients. Conclusions: The 21-gene Recurrence Score remains prognostic in node-positive patients and real-world outcomes mirror RxPONDER¹ findings. Our study highlights the importance of endocrine therapy adherence and supports continued use of the 21-gene Recurrence Score for guiding chemotherapy decisions in clinical practice. Larger cohort real-world studies are needed to further validate these findings.
| Risk Group | Treatment Type | Number of Events |
| Low | Endocrine Therapy only (ET) | 1 |
| Intermediate | Endocrine Therapy only (ET) | 7 |
| Intermeidate | Chemotherapy + ET | 3 |
| High | Endocrine Therapy only (ET) | 1 |
| High | Chemotherapy + ET | 6 |
| High | Chemotherapy only | 1 |
Presentation numberPS5-05-26
Analyzing Age-Related Patterns of Second Primary Cancer Sites Following a Primary Breast Cancer Diagnosis
Vontyna Smith, Q-Centrix, Chicago, IL
B. Zigler, V. Wang, V. Smith; Real World Data, Q-Centrix, Chicago, IL.
Objective: This study aims to evaluate age-related patterns pertaining to the anatomical sites of second primary cancers occurring within five years of an initial breast cancer diagnosis. Methods: Our analysis focuses on patients (n=68,109) whose first recorded cancer diagnosis was early-stage breast cancer, diagnosed between 2008 and 2019. Age, diagnosis dates, and all anatomical cancer sites of these 68,109 patients were extracted from the Q-Centrix Clinical Data Warehouse. The Q-Centrix Clinical Data Warehouse is a proprietary database of de-identified clinical data produced through expert-driven human abstraction. The subset sourced from the Clinical Data Warehouse includes information from 78 hospitals, health systems, and cancer centers across the country. Patients were split into 6 age groups: 40 years of age compared to those < 40 years of age. A p-value cutoff of <0.05 was considered significant. A second analysis was conducted on the subset of patients who developed a second non-simultaneous cancer within five years of the initial breast cancer diagnosis. Of the 68,109 patients who had breast cancer logged as their first cancer in our data warehouse, 4680 (~7%) fit these criteria. The site of the second cancer was categorized as breast or non-breast. 29 patients in this group did not have a second cancer site stored in the data warehouse. 2 additional patients did not have their age stored in the data warehouse. These patients were dropped from the final set, bringing the total to 4,649. A logistic regression model was then run to determine the odds of the second primary site being located outside the breast versus inside the breast in patient age-groups >40 years of age compared to those <40 years of age. A p-value cutoff of <0.05 was considered significant. The age distribution of this group included 119 patients <40 years of age, 40-49 (n=561), 50-59 (n=1070), 60-69 (n=1426), 70-79 (n=1038), and 80+ (n=435). Results: The odds of developing a second non-simultaneous cancer within 5 years of an initial early-stage breast cancer diagnosis relative to the <40 age group was .68 in the 50-59 age group, .60 in the 60-69 age group, .55 in the 70-79 age group and .59 in the 80+ age group (p-value <0.05). The odds of the 40-49-year age group relative to the <40-year age group was not statistically significant. In the group that did develop a second cancer diagnosis within 5 years, the odds of the tumor location being outside the breast versus inside the breast were 2.05 times greater for patients aged 50-59 years. A 60-69-year-old patient was 3.49 times likelier, a 70-79-year-old patient was 4.76 times likelier, and a patient 80 years or older was 5.05 times likelier to have a second cancer site outside of the breast relative to a patient who was <40 years of age (p-value<.05). The odds of the 40-49-year age group developing a cancer located outside the breast versus inside the breast relative to the <40-year age group was not statistically significant. Conclusions: The odds ratios in the second analysis suggest that the odds of developing a second cancer outside the breast, relative to within the breast, within five years of an initial early-stage breast cancer diagnosis, increase progressively with patient age. Further analyses are required to determine how this relationship differs with respect to the original breast cancer diagnosis receptor type, stage, and anti-neoplastic treatment modality.
Presentation numberPS5-05-27
Real-world Clinical Outcomes in Patients with HER2+ Metastatic Breast Cancer Treated with Trastuzumab Deruxtecan After One or More Prior Lines of Therapy: Data from U.S Community Oncology Practices
David Peevyhouse, ONCare Alliance, Tacoma, WA
S. Mehta1, H. Neuhalfen2, A. Peevyhouse2, D. Adhikari2, C. Williams2, M. Garretson3, S. Blau2, M. Walker2, D. Peevyhouse4; 1HEOR, Daiichi-Sankyo, Basking Ridge, NJ, 2RWD, ONCare Alliance, Tacoma, WA, 3HEOR, Aaron Peevyhouse, Basking Ridge, TN, 4RWE, ONCare Alliance, Tacoma, WA.
Background: Trastuzumab deruxtecan (T-DXd) was initially (12/2019) approved by the FDA for treatment of patients (pts) with HER2+ unresectable or metastatic breast cancer (mBC) with two or more prior anti-HER2 regimens. T-DXd was later (5/2022) approved for use by pts with one prior anti-HER2 regimen. This study examined clinical outcomes in pts with HER2+ mBC treated with T-DXd 2L and 3L+ settings. Methods: This retrospective cohort study used deeply-curated electronic medical record data from practices affiliated with ONCare Alliance. Adult pts with mBC diagnosis, confirmed HER2+ status, who received T-DXd in the 2L or later setting 1/2020 to 8/2024 were included. Pt characteristics, treatment utilization and medical events of interests were assessed descriptively by 2L and 3L+ cohorts. Clinical outcomes (duration of therapy [DoT], real-world time to treatment failure [rwTTF], overall survival [OS] and real-world progression-free survival [rwPFS]) were assessed by 2L and 3L+ cohorts using Kaplan-Meier methods. Clinical outcomes of 1L and 2L rwTTF were examined in an exploratory subgroup of pts with 1L taxane+trastuzumab+pertuzumab based regimen (THP) followed by 2L T-DXd vs. 2L trastuzumab emtansine (T-DM1). Results: A total of 300 pts (70 2L and 230 3L+ pts [61.3% in 3L/4L]) treated with T-DXd were included (overall mean age = 59.3 yrs; 82.3% White). At mBC diagnosis, a greater proportion of pts in the 2L T-DXd cohort vs. 3L+ cohort had distant bone / visceral metastasis (45.7% vs. 38.3%) and visceral only metastasis (47.1% vs. 42.2%), where a significantly lower proportion had bone only metastasis (7.1% vs. 19.6%; p = 0.0493). Fewer 2L pts had brain metastasis at initiation of T-DXd (14.3% vs. 37.0% in 3L+, p = 0.0031), but a higher rate of baseline comorbidities was seen in 2L pts (50.0% vs. 30.4%, p = 0.0027). Prior to the initiation of 2L T-DXd, 64.3% (n=45) pts received a trastuzumab+pertuzumab based regimen and 22.8% (n=16) received T-DM1 in 1L; prior to 3L+ T-DXd use, 38.7% (n=89) received T-DM1 in 2L. Within a median follow-up of 17.0 months (mo), pts receiving T-DXd in 2L had longer median DoT than 3L+ pts (20.5 mo vs. 12.5 mo, p = 0.0390), longer rwPFS (15.3 mo in 2L vs. 12.4 mo in 3L+, p = 0.0179), longer OS (median NR in 2L vs. 29.0 mo in 3L+, p = 0.0050), and numerically longer rwTTF (13.6 mo in 2L vs. 10.4 mo in 3L+, p = 0.0565). Of pts with a response assessment (66 2L, 205 3L+ pts), an overall objective response was observed in 80.3% 2L vs. 71.7% 3L+ pts (p = 0.1672). The overall rates of any grade medical event of interest were consistent across 2L and 3L+ cohorts (fatigue [72.9% vs. 73.0%], nausea/vomiting [78.6% vs. 72.2%], diarrhea [55.7% vs. 43.9%], and interstitial lung disease (ILD)/pneumonitis [8.6% vs. 10.4%], except respiratory infection, which was higher in 2L T-DXd [38.6% vs. 18.7%, p = 0.0006]). Of the pts who had discontinued T-DXd at any point (45 2L and 180 3L+), the most common reason for discontinuation was disease progression (51.1% vs. 55.0%), followed by toxicity other than nausea/vomiting/ILD (17.8% vs. 11.7%), pt choice (8.9% vs. 6.1%) and toxicity related to ILD (6.7% vs. 6.7%). Among exploratory subgroups, median rwTTF was 1L THP 11.6 mo à 2L T-DXd 14.2 mo (n=38) vs. 1L THP 11.6 mo à 2L T-DM1 8.4 mo (n=60) (2L p = 0.0367). Conclusion: The study findings affirm the real-world clinical effectiveness and safety of T-DXd in pts with HER2+ mBC as observed in the DESTINY clinical trials. Despite higher disease and comorbidity burden in this small sample of pts with 2L T-DXd use, longer delays in time to progression and discontinuation were observed compared to later line use of T-DXd; and longer rwTTF compared to 2L T-DM1 use, reinforcing the importance of utilizing T-DXd as early as possible in the treatment pathway to improve long-term pt outcomes.
Presentation numberPS5-05-28
Efficacy of systemic therapy in HER2-low breast cancer with CNS metastases: a “real-world” 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.
Background: The incidence of central nervous system (CNS) metastases in breast cancer is increasing. While local treatments such as surgery and radiation remain standard, systemic therapies have shown promise in HER2-positive disease. However, data on the efficacy of systemic therapy in HER2-low breast cancer with CNS involvement remain limited. This study describes outcomes of upfront systemic therapy in HER2-low breast cancer patients with CNS metastases treated at a single institution.Methods: We retrospectively reviewed breast cancer patients with CNS metastases treated at Henry Ford Health between Jan 2014 and July 2024. HER2-low status was defined as: HER2 immunohistochemistry 1+, or 2+ with negative fluorescent in situ hybridization (FISH). Eligible patients received no concurrent local CNS therapy; prior local treatment was permitted if unrelated to the studied lesions. CNS responses were to be assessed using the Response Assessment in Neuro-Oncology for brain metastases (RANO-BM) or the modified RANO-LM (leptomeningeal metastases) criteria. Results: Sixteen HER2-low patients were included. The median age was 56 years (range, 38-93); 63% (n = 10) were African American. Most (75%) had hormone receptor-positive disease, and 53% presented with de novo metastatic disease. Thirteen patients (81%) had HER2 IHC 1+, and only three had HER2 IHC 2+/FISH-negative disease. The majority (88%) had parenchymal CNS metastases; one patient had leptomeningeal disease, and one had both. Eight patients (50%) had prior CNS metastases, and 38% received local therapy before study inclusion. Notably, a significant proportion of patients (88%) had non-measurable disease by RANO-BM criteria (lesions <10 mm), with 69% having lesions smaller than 5 mm. Systemic therapies for the CNS disease included cytotoxic chemotherapy in seven patients (44%), trastuzumab deruxtecan (T-DXd) in four (25%), abemaciclib in two (13%), other CDK4/6 inhibitors in two (13%), and sacituzumab govitecan in one (6%). Commonly utilized chemotherapy included taxanes (paclitaxel or nab-paclitaxel) in 4 of 7 patients. In the 16 patients, the overall response rate (ORR) was 31.3%, the disease control rate (DCR) was 56.3%, the median duration of response (DOR) was 7 months, and the median progression-free survival (PFS) was 2 months. In a comparison cohort of HER2-positive patients (n=17), ORR was 53%, DCR was significantly higher at 94% (p=0.017), with similar DOR (5 months), and numerically longer PFS (3 months, p=0.351). Outcomes among the African American patients (n=10) mirrored the overall HER2-low group, with ORR 30%, DCR 50%, DOR 5 months, and PFS 1 month. Notably, the patients with HER2 IHC 2+ tumors (n=3; all HR+) experienced higher benefit (ORR 67%, DCR 100%) with no disease progression at last follow-up. HER2-low patients treated with T-DXd (n=4) demonstrated a DCR of 75%, with only one progression. Chemotherapy yielded a poor ORR of 29%, though two patients achieved complete CNS responses with taxane-based regimens. Conclusion: This “real-world” experience highlights modest intracranial activity of systemic therapy in HER2-low breast cancer with CNS metastases. Although response rates were lower than in HER2-positive counterparts, HER2 2+ tumors and patients treated with T-DXd showed encouraging outcomes. These findings support the need for prospective studies focusing on HER2-low CNS disease and suggest T-DXd as a promising therapeutic option, even in the absence of concurrent local treatment. Additionally, our study underscores a unique challenge in the management of breast cancer and brain metastases: CNS lesions are often smaller than the >10 mm threshold typically required for clinical trial eligibility, necessitating special attention and adapted approaches for response assessment.
Presentation numberPS5-05-29
Long-term impact of early-onset breast cancer in brazilian women: a real-world study of years of life lost and clinical outcomes
Diego Lopes Paim Miranda, Einstein Hospital Israelita, São Paulo, Brazil
D. L. Miranda, D. Dias e Silva, B. Viesser Miyamura, U. Donizeti Rocioli Junior, P. Serrano Uson Junior, J. Rodrigues Beal, P. Taranto, J. Pontes Batista Cassoli, F. Moura; Department of Oncology, Einstein Hospital Israelita, São Paulo, BRAZIL.
Introduction: Breast cancer is the leading cause of cancer-related death among women in Brazil and worldwide. However, its impact is not evenly distributed, with significant variations in incidence, mortality, and survival across different countries and regions. The incidence of breast cancer among young adults under the age of 50 has been increasing globally. This rise carries important implications both at the individual and societal levels, including a greater disease burden, reduced economic productivity, and an increased risk of long-term adverse clinical outcomes. There is a scarcity of data in the literature regarding this trend in Brazilian and Latin American populations. Objectives: We aimed to investigate the distribution profile of breast cancer diagnoses in adults under 50 years of age at two tertiary cancer centers in Brazil, as well as to compare it with the profile of patients aged 50 years and older, with a focus on clinical presentation and outcomes. Methods: We used real-world data from Einstein Hospital Israelita and Hospital Municipal Vila Santa Catarina between 2018 and 2023, including patients diagnosed with malignant breast neoplasms, according to the International Classification of Diseases – 10th Revision (ICD-10). Early-onset breast cancer was defined as a diagnosis in adults aged 18 to 49 years. Data extraction was automated using electronic medical record (EMR) systems and big data sources to obtain information related to ICD codes, demographic data, and medical history. For the calculation of years of life lost (YLLs), projected life expectancy was estimated for each individual based on their adjusted year of birth, sex, and place of residence, allowing for more precise and personalized estimates. Results: A total of 3,102 patients were analyzed, of whom 1,752 (56%) were aged 50 years or older and 1,350 (44%) were between 18 and 49 years old. Among the latter group, 28% reported their ethnicity, with the majority identifying as white (20%). Women accounted for 99.7% of the cases. Male sex was more frequent in the group aged 50 and older (1% vs. 0.3%; p=0.023). In the 18 to 49-year-old group, 61% of patients presented with localized or locally advanced disease (clinical stages I to III), while 39% had metastatic disease (clinical stage IV). Similarly, among patients aged 50 years or older, 59% were diagnosed at stages I to III, and 41% at stage IV. The median body mass index was lower in the younger group (24.6 kg/m² vs. 26.1 kg/m²; p<0.001). Over the study period, there was an observed increase in breast cancer diagnoses among individuals aged 18 to 49, with a 10% rise in rates, from 4.47/10,000 in 2018 to 4.92/10,000 in 2023. In contrast, a 7.1% decrease was observed in the group aged 50 and older, from 11.97/10,000 in 2018 to 11.11/10,000 in 2023. Additionally, there was a higher proportion of germline testing among patients aged 18 to 49 (21% vs. 11%; p<0.001). When assessing years of life lost (YLLs), patients under 50 years of age exhibited a significantly higher burden, with a median of 42 years lost (IQR 37.5-46.0), in contrast to a median of 21 years (IQR 13.0-26.0) among those aged 50 and older. Conclusions: In line with global trends, the apparent increase in breast cancer cases among individuals under 50 years of age in Brazil raises hypotheses regarding both the rise in incidence and the possibility of distinct characteristics compared to those aged 50 and older. This pattern highlights the need for further investigations to better understand risk factors and to improve screening and management strategies for this age group.
Presentation numberPS5-05-30
Cycline Dipendent Kinase inhibitors 4/6 and endocrine therapy in the first line of treatment for HR+Her2- advanced breast cancer: a meta-analysis of the published real-world studies
Angela Toss, Azienda Ospedaliero-Universitaria di Modena Policlinico di Modena, Modena, Italy
L. Moscetti1, I. Sperduti2, S. Zaniboni1, L. Belluzzi1, E. Zattarin1, L. Cortesi3, P. Vici4, F. Piacentini1, C. Omarini1, E. Barbieri1, F. Canino1, M. Barbolini1, F. Caggia1, M. Dominici1, A. Toss1; 1Oncology, Azienda Ospedaliero-Universitaria di Modena Policlinico di Modena, Modena, ITALY, 2Biostatistical Unit—Clinical Trials Center,, IRCCS Regina Elena National Cancer Institute, Roma, ITALY, 3Oncology, Azienda USL-IRCCS Reggio Emilia, Reggio Emilia, ITALY, 4Oncology, IRCCS Regina Elena National Cancer Institute, Roma, ITALY.
Background: First-line treatment for HR+/HER2- advanced breast cancer (ABC) is based on CDK4/6 inhibitors (CDK4/6i) and endocrine therapy (ET). The three available CDK4/6i – palbociclib (P), abemaciclib (A), and ribociclib (R) – demonstrated comparable efficacy in pivotal Phase III trials in terms of progression-free survival (PFS). However, only R achieved a significant overall survival (OS) benefit. Most previous analyses comparing these CDK4/6i focused on efficacy, safety, and tolerability, generally showing no significant differences. Limited real-world (RW) data exist regarding inter-drug comparisons in the RW setting.Methods: We conducted a meta-analysis of the RW studies in the first-line HR+/HER2- ABC setting (with P, A, and R) to assess potential outcome differences between the three drugs. A systematic literature search was performed using PubMed with the terms: “breast cancer,” “real world,” “palbociclib,” “abemaciclib,” “ribociclib” “dalpiciclib”. Treatment comparisons utilized as effect size the percentage (%) of 2-years real-world PFS (rwPFS) and 95% confidence intervals (CI). The % of 2-year rwPFS (%2yPFS) was estimated from the median rwPFS for this analysis Heterogeneity was assessed using the χ2 Q test (p50% considered significant). The pooled effect size estimate was calculated with a random-effect model using the 2-year PFS as endpoint.Results: 67 studies were identified 56 reported the median rwPFS as the primary outcome, one reported time to next treatment (TTNT), 8 reported time to chemotherapy or time to treatment discontinuation, 2 did not reported any outcome. Considering 56 studies involving four CDK4/6 inhibitors reporting rwPFS, no significant differences were observed when comparing all three drugs (%2yPFS 49, 95% IC 46-51, p=0.39). High heterogeneity was observed across the included studies (I2=81%, p<0.00001). Four out of 56 trials were excluded from the final analysis. The sensitivity analysis conducted in the 52 studies involving four trials with A (%2yPFS 49, CI 95% 40-59), thirty-nine trials with P (%2yPFS 48, CI 95% 45-50) and nine with R (%2yPFS 54, CI 95% 48-60) confirmed the effect size observed. In the analysis comparing the three CDK 4/6 inhibitors, no differences were observed when comparing A and P (p=0.751) or A and R (p=0.376), but a possible difference was observed when comparing R and P (p=0.045). Conclusion: This analysis did not reveal significant differences in 2-year rwPFS, the primary outcome in most real-world studies, among the CDK4/6 inhibitors. Any observed differences between R and P should be interpreted with consideration for the patient characteristics within the studies analysed. Further adjustment of the analysis using multiple variables is planned to assess inter-trial differences and mitigate bias inherent in real-world settings.
Presentation numberPS5-04-01
Survival Outcomes in Metastatic Breast Cancer: An analysis of the US population-based SEER database
Alexander Niyazov, Pfizer, New York, NY
A. Niyazov1, R. Sharma2, S. Bilthare2, S. Iyer2; 1Value&Evidence, Pfizer, New York, NY, 2Evidence, HEOR&RWE, Putnam, Inizio Advisory, New York, NY.
Background Over the last 15 years, several therapies have been approved for the treatment of metastatic breast cancer (mBC) in the United States (U.S.). Given the evolving treatment landscape, it is important to assess and understand overall survival (OS) and related predictors in the real-world setting. Methods Data were obtained from the Surveillance, Epidemiology, and End Results (SEER), which covers approximately 48% of the U.S. population. Patients diagnosed with mBC (ICD-10: C50.0 to C50.9) between 2010 and 2021 were included. Follow-up continued until death or December 2021. Overall survival (OS) and cancer-specific survival (CSS) were estimated using Kaplan-Meier methods. Median survival and 5-year survival rates were reported. Predictors of survival were assessed using multivariate Cox proportional hazards modeling. Results The cohort included 37,017 patients with mBC. The majority (58%) were aged <65 years, with a mean age of 61 years. 54% of the patients had liver or lung metastases. Prior surgery was reported for 25% of the patients. Among patients with subtypes reported (n=31,656), the proportion of patients with HR+/HER2-, HER2+ (HR+/HER2+, HR-/HER2+), and triple negative breast cancer (TNBC) subtypes were 60%, 26%, and 14% respectively. In the overall mBC cohort, median OS was 28 months, with a 5-year OS rate of 28% and median CSS was 32 months, with a 5-year CSS rate of 32% (Table 1). Among patients aged ≥65 years in the overall mBC cohort, median OS and CSS were 18 and 23 months, respectively, with 5-year survival rates of 20% and 26%. In the HR+/HER2- mBC subgroup, median OS and CSS were 35 and 39 months, with a 5-year OS rate of 30% and 34% respectively. In the HER2+ mBC subgroup, median OS and CSS were 44 and 49 months, with a 5-year OS rate of 41% and 44% respectively. In the triple negative mBC subgroup, median OS and CSS was 12 and 13 months, with a 5-year OS rate of 12% and 14% respectively. Significant predictors of OS and CSS included age, race, histology, breast cancer subtype and prior surgery status (p < 0.05). Overall survival prognosis was significantly (p<0.001) worse for TNBC compared to HR+/HER2- mBC patients (HR;95% CI:2.38;2.29,2.48). Conclusion Overall survival in patients diagnosed with mBC between 2010 to 2021 varied by breast cancer subtype with prognoses relatively worse in TNBC patients compared to HR+/HER2- patients. Continued development of novel therapies for TNBC patients will be key to achieving further improvements in survival outcomes.
Presentation numberPS5-04-02
Interim results: Real world study of treatment discontinuations and modifications for patients with HER2-positive 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-positive (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+ mBC who enrolled in AstraZeneca Canada’s patient support program (PSP) for treatment with T-DXd, following regulatory approval based on the DESTINY-Breast03 (DB-03) trial. Methods: This interim analysis included all patients enrolled in the PSP from June 2022 to August 2023 and initiated T-DXd therapy. The 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 347 patients were included with a mean age of 60 years (SD, 13.2); 6% (n=19) had no prior lines of therapy in the metastatic setting, 64% (n=222) had 1, 9% (n=30) had 2, and 4% (n=14) had 3+, with 18% (n=62) missing data; 30% (n=103) had stable brain metastases at enrollment. Median follow-up was 5.6 months (range 0.2-29.7). Among patients that started at a dose of 5.4 mg/kg (84.4%, n=293) or a lower dose of 4.4 or 3.2 mg/kg (n=20, 5.8%), 32% (n=101) had a dose reduction, 28% (n=87) had a discontinuation, 51% (n=161) had either a dose reduction or discontinuation, and 51% (n=161) had a dose interruption. Some patients (n=34, 9.8%) received a dose not specified in the label (neither 5.4, 4.4, or 3.2 mg/kg). Cumulative probabilities of treatment discontinuation at 3, 6 and 9 months were 10.2% (95% CI, 6.8-13.4), 18.2% (95% CI, 13.6-22.6), and 29.7% (95% CI, 23.3-35.6), respectively. Overall median TTD was 14.3 months (95% CI, 12.4-18.4); median TTD among patients with stable brain metastases was 15.2 months (95% CI, 15.2-not reached). Median duration of treatment (excluding dose interruptions) was 12.7 months (95% CI, 10.3-18.4). Of the total observed discontinuations (28%, n=98) progression was the most common reason (44%, n=43) followed by prescriber decision (23%, n=22), death (16%, n=15), patient decision (12%, n=12), or other/unknown/missing data (6%, n=6). Conclusion: RW use of T-DXd in the Canadian PSP, designed for patients eligible under the regulatory authorization based on DB-03, reflected a largely second-line setting, with a median TTD consistent across the overall population and patients with stable brain metastases. 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 numberPS5-04-03
Real world outcomes of use of Capivasertib in patients with HR positive HER2 negative metastatic breast cancer: A single center study
Vinit Singh, Roswell Park Comprehensive Cancer Center, Buffalo, NY
V. Singh1, A. Al-Alwan1, S. Kabraji2, E. Levine2, V. Gupta2; 1Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Breast Oncology, Deparment of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY.
Introduction: Capivasertib in combination with Fulvestrant is approved for a second or later line treatment option for metastatic ER/PR positive HER2 negative breast cancer, for patients harboring one or more PIK3CA, AKT1, and PTEN alterations, especially after use of CDK4/6 inhibitors. These mutations are seen in ~55% patients with HR+ mBC. There is limited real-world data on the use of the combination. The purpose of this analysis is to assess the real-world efficacy and safety of this regimen in patients with HR positive metastatic breast cancer. Methods: We conducted a single-center, IRB-approved retrospective chart review of patients diagnosed with metastatic breast cancer (mBC) who received capivasertib in combination with Fulvestrant, with the last follow-up date as of June 30, 2025. Patient-specific demographic, clinical, pathological, and treatment data were collected. Data was summarized using descriptive statistics. Time to treatment failure (discontinuation either because of progression or adverse effects) and PFS were calculated by the Kaplan-Meier method. Results: We identified 28 patients who received Capivasertib; median age – 57 years (IQR 49-66.5). 21 (75%) had PICK3CA, 5 (17.9%) PTEN, and 4 (14.3%) AKT1 mutation with 2 patients had both PICK3CA and PTEN mutation. All the patients had received prior CDK4/6 inhibitors and 11 had exposure to prior chemotherapy. The median follow-up was 7.6 months (IQR, 4.1-12.3) compared to 13 months (IQR 9·1-16·7) in CAPItello-291, with a median time to treatment failure of 5 +/- 0.5 months (5.8 months in CAPItello-291). Median PFS was 6 +/- 0.5 months (7.2 months in CAPItello-291). Treatment was discontinued in 19 (67.9%) patients due to disease progression and in 3 (10.7%) patients because of adverse effects. The reason for discontinuation was hyperglycemia, liver injury, and skin rashes. Medications were either discontinued or reduced for 10/28 (34%). Skin rash was the most common reason for dose reduction. 7/28 (25%) patients died by the last follow-up date, with 4/7 (57.1%) having prior exposure to chemotherapy. 4/7 (57.1%) Patients had either visceral or treated brain metastases before starting capivasertib. 1/4 (25%) of patients with treated brain metastasis and 3/15 (20%) with one or more visceral metastases died during the study period. Conclusion: In this single-center experience, the findings of CAPItello-291 was validated, that a combination of capivasertib and fulvestrant is an effective treatment in second or later-line therapy for metastatic HR+ HER2- breast cancer patients harboring AKT pathway alterations, with a similar discontinuation rate.
Presentation numberPS5-04-04
Design of a Prospective Registry Study to Evaluate the Decision Impact and Clinical Utility of the Lipidomic-based Blood Test BREASTEST Plus in Breast Cancer Screening
Simon Preston, BCAL Diagnostics, Sydney, Australia
S. Preston1, S. Ryan2, D. Speakman3; 1Clinical Research, BCAL Diagnostics, Sydney, AUSTRALIA, 2Management, BCAL Diagnostics, Sydney, AUSTRALIA, 3Surgery, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA.
Background: BREASTEST Plus is a novel blood-based diagnostic assay used alongside imaging to support breast cancer screening decision-making. While prior validation studies have demonstrated the analytical and clinical performance of the assay, real-world evidence (RWE) on its clinical utility and long-term impact on patient outcomes remains essential. To address this, BCAL Diagnostics has initiated a prospective, observational registry study aimed at evaluating how BREASTEST Plus influences physician decision-making and patient outcomes in routine clinical practice. Methods: This multi-centre, prospective registry will enroll a minimum of 2,000 women across Australia who undergo BREASTEST Plus testing as part of their clinical assessment. Inclusion criteria are broad, reflecting routine clinical use, and will include both symptomatic and asymptomatic women aged ≥30 years. Treating clinicians will complete a structured decision-impact survey following receipt of results to assess how the assay influenced diagnostic or management decisions. Patients will be followed for up to five years, with key endpoints including one and two year sensitivity and specificity. Results: Primary endpoints are: (1) proportion of cases in which BREASTEST Plus results changed clinical management, and (2) concordance of assay results with cancer diagnoses over time. Secondary endpoints include reduction in unnecessary procedures, and patient-reported metrics. Interim analyses will be performed at 12-month intervals to inform clinical and regulatory stakeholders. Conclusion: This RWE registry is the first of its kind for a lipid-based breast cancer diagnostic and will provide critical insights into the utility, impact, and long-term value of BREASTEST Plus in everyday practice. Results will inform guidelines, reimbursement decisions, and broader adoption strategies across healthcare systems.
Presentation numberPS5-04-05
Long term Breast Cancer real-world overall survival in France
Olivier Tredan, CENTRE LEON BERARD, LYON, France
O. Tredan1, Y. Delpech2, S. Bara3, M. Lotz4, M. Moreau5, D. Stamenic5, M. Le Foll-Elfounini5, L. Mansi6; 1Gynecology, Breast Oncology, Medical Oncology, CENTRE LEON BERARD, LYON, FRANCE, 2Breast, Oncogynecologic and Reconstructive Surgery, Centre Antoine LACASSAGNE, NICE, FRANCE, 3Registre des Cancers de la Manche, Centre Hospitalier du Cotentin, Cherbourg-en-Cotentin, FRANCE, 4Medical Affairs, ROCHE, Boulogne-Billancourt, FRANCE, 5Medical Evidence & Data Science, ROCHE, Boulogne-Billancourt, FRANCE, 6Medical Oncology, CHRU Jean Minjoz, Besançon, FRANCE.
Objectives Assess the Overall survival (OS) rates at 5,10 and 12 years in French women with breast cancer (BC) by BC subtype Material and methods Data were extracted from a 2% representative sample of the French National Claims Database which covers around 98% of the total French population. All French women aged ≥18 years diagnosed with BC between 2010 and 2023 were identified using markers of BC management. HER2+ and HR+ status were determined based on the reimbursement of HER2-targeted therapy and endocrine therapy, respectively. The BC subtype of patients without HER2-targeted nor endocrine therapy could not be determined because of the lack of tracking drugs. These patients constituted the “undefined” group with regard to their BC subtype and were composed of 2 main subgroups: ÏPatients treated with systemic treatment(s) (potentially composed among others of TNBC or patients with visceral crisis)ÏPatient with BC in situ diagnosis without systemic treatment A one year historical period has been used. OS was defined as the time from the BC diagnosis date, or otherwise from the date of the first evidence of BC specific treatment until death from any cause. Results A total of 12 219 incident BC cases were identified between 2010 and 2023, of which 7 878 (64,5%) were HR+/HER2- and, 1 414 (11.6%) HER2+ including 895 (7,3%) HER2+/HR+ and 519 (4,3%) HER2+/HR-. The “undefined” group (N=2 927, 24.0% of total incident BC cases) was composed of 1 188 (9.7%) patients treated with systemic treatment(s), 768 (6.3%) patients with BC in situ diagnosis without systemic treatment and 971 patients with invasive cancer diagnosis without systemic treatment (8.0 %) In the HER2+ BC subtype, the 5-years, 10-years and 12-years OS rates were 87.7% (95% CI: 85.6%, 89.4%), 80.4% (95% CI: 77.6%, 82.8%) and 74.4% (95% CI: 70.5%, 77.8%). Among HER2+, the corresponding OS rates were 90.4% (95% CI: 88.1%, 92.4%), 81.0% (95% CI: 77.3%, 84.2%) and 73.7% (95% CI: 68.3%, 78.3%) in HER2+/HR+ and 82.7% (95% CI: 78.8%, 86.0%), 79.2% (95% CI: 74.7%, 82.9%) and 75,4 (95% CI: 69.6%, 80.3%) in HER2+/HR-, showing a better survival in HER2+/HR+ at 5 years but quite similar OS rates at 12 years. In the HR+/HER2- BC subtype, the 5-years, 10-years and 12-years OS rates were 88.8% (95% CI: 88.0%, 89.5%), 77.6% (95% CI: 76.3%, 78.8%) and 73.7% (95% CI: 72.1%, 75.1%). Among the undefined group, the 5-years, 10-years and 12-years OS rates were 73.0% (95% CI: 70.1%, 75.6%), 66.7% (95% CI: 63,4.%, 69.8%) and 65.1% (95% CI: 61,5.%, 68.5%) in patients treated with systemic treatment(s), in patients treated with systemic treatment(s), which fits with the bad prognosis of TNBC and patients with visceral crisis. OS rates were 98.0% (95% CI: 96.6%, 98.8%), 93.1% (95% CI: 90.2%, 95.1%) and 91.4% (95% CI: 87.9%, 94.0%) in patients with an in situ BC diagnosis without systemic treatment, demonstrating as expected higher survival rates than other groups. ConclusionAs of today, 12-year OS rates tend to be similar in HER2+ (regardless of the HR status) and HR+/HER2- groups, demonstrating an efficient BC management evolution, especially in the HER2+ group. These results also highlight a significantly higher 5-year survival for HR+ compared to HER2+. However, this difference tends to equalize at 12 years, in line with the long-term recurrence risk of HR+.
Presentation numberPS5-04-06
Current and Emerging Treatment Patterns for Patients with Residual Disease Post-Neoadjuvant Therapy in Early-Stage Triple Receptor-Negative Breast Cancer
Amin Haiderali, Merck, North Wales, PA
A. Haiderali1, J. R. Earla2, P. Kondaparthi3, J. Nathani4, S. Sharma5; 1Value & Implementation, Merck, North Wales, PA, 2Value & Implementation, Merck, Rahway, NJ, 3HEOR, Parexel International, Hyderabad, INDIA, 4HEOR, Parexel International, Bengaluru, INDIA, 5HEOR, Parexel International, Chandigarh, INDIA.
Background: Triple-negative breast cancer (TNBC) is associated with a poorer prognosis compared to other subtypes. Neoadjuvant chemotherapy with anthracyclines and taxanes aims to improve pathological complete response (pCR) rates. Patients without pCR face higher relapse risks and worse outcomes, necessitating escalation strategies for residual disease. Objective: This review aimed to summarize real-world treatment patterns for patients with early-stage TNBC focusing on adjuvant therapy for those with residual disease after neoadjuvant chemotherapy. Methods: A systematic literature search was conducted using Embase and MEDLINE databases (2014-2024) and relevant conference proceedings (2021-2024). Real-world studies on treatment utilization rates, discontinuation rates, efficacy, and safety were included. Results: Six studies from the USA, Brazil, Italy, Turkey, and Middle East Africa region were identified. Capecitabine alone or in combination with radiotherapy emerged as the primary adjuvant pharmacological therapy for residual TNBC after neoadjuvant chemotherapy. This was reported in three studies, with utilization rates ranging from 31.7% to 42.1%. Additionally, adjuvant radiotherapy was reported in two studies, with higher usage rates ranging from 71.1% to 94.7%. Most patients received sequential rather than concurrent radiotherapy and capecitabine treatment. The rates of discontinuation varied by cause, with toxicity accounting for 6.0% to 17.0% of discontinuations and disease progression leading to 12.6% to 17.0% of discontinuations. Two-year disease-free survival (DFS) rates after adjuvant capecitabine ranged from 62.0% to 88.9%. The three-year overall survival (OS) rate for patients with residual disease treated with adjuvant capecitabine, either alone or in combination with radiotherapy, was 76.2%. The proportion of patients experiencing disease progression (PD) after adjuvant therapy ranged from 28.9% to 64.2%. Conclusion: This review highlights significant unmet needs in early TNBC patients with residual disease after neoadjuvant therapy. While guidelines recommend capecitabine for this patient population, they also recommend exploring newer therapies like pembrolizumab and olaparib for specific patient sub-populations. There are treatment strategies available for the peri-operative setting, but the associated evidence was not captured in this review. The high rates of residual disease, treatment discontinuation, and disease progression underscore the need for more effective and tolerable adjuvant therapies. Currently, multiple novel therapies are being evaluated for this patient population including antibody drug conjugates (ADCs) and other targeted immunotherapies to improve outcomes and mitigate the unmet need.
Presentation numberPS5-04-07
Real world experience with trastuzumab deruxtecan for metastatic breast cancer at a major Australian cancer centre
Michelle Li, Peter MacCallum Cancer Centre, Melbourne, Australia
M. Li1, C. T. Van Geelen2, L. E. Lara Gonzalez2, K. Clarke2, S. Lingaratnam3, P. A. Francis1, S. Loi1; 1Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 2Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, AUSTRALIA, 3Department of Pharmacy, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA.
BACKGROUND: Trastuzumab deruxtecan (T-DXd) is approved for HER2-low and HER2+ metastatic breast cancer (MBC), offering significant clinical benefit. However, treatment-related toxicities—particularly nausea, vomiting and weight loss—can substantially impair patient quality of life and lead to early discontinuation. We evaluated real world efficacy and tolerability of T-DXd in an Australian context. METHODS: We conducted a retrospective cohort study of patients aged ≥18 years with MBC treated with T-DXd between May 2019 and December 2024 at a single tertiary Australian cancer centre. Demographic, clinicopathologic, treatment and toxicity data were extracted from medical records. Kaplan-Meier analysis was utilized for progression-free survival (PFS), with censoring of patients still on treatment. RESULTS: Data from 75 patients was analyzed with a median follow-up of 15.5 months (95% CI 16.3-23, range: 0.8-68.7). Median age was 46 years (IQR 39-56). 64% had recurrent metastatic disease; 36% were de novo metastatic. 36 patients (48%) were HER2-low and 39 patients (52%) were HER2+. Hormone receptor (HR) positivity was observed in 60% (36% HER2-low and 24% HER2+). 31% had liver metastases. Median PFS1 was 6.5 months (95% CI 5.8-10.3, range: 0.2-31) in the HER2-low cohort and 19.3 months (95% CI 15.6-23.6, range: 1.1-52.4) in the HER2+ cohort. Median PFS1 was higher in HR+ HER2-low (7.23 months, 95% CI 6-11.7, range: 1.3-31) compared to HR- HER2-low (5.5 months, 95% CI 2.5-8.6, range: 0.2-12.9). Number of prior lines of therapy was higher in HER2-low (median 2, range: 0-14) than HER2+ (median 1, range: 0-6). Common toxicities were nausea, vomiting and weight loss, at rates higher than reported in trial data (Table 1). Dose reductions (DRs) occurred in 33 patients (44%), with 6 (8%) receiving upfront DR for performance status and 27 (36%) requiring at least one subsequent DR, most often due to nausea/vomiting (21.3%). DR typically occurred after a median of 6 cycles (range: 1-40). Treatment discontinuation due to toxicity occurred in 7 patients (9.3%), most often due to interstitial lung disease (ILD; 5 patients, 6.7%). 1 patient (1.3%) discontinued due to hepatotoxicity resulting in thrombocytopenia and 1 patient (1.3%) due to fatigue. 48 patients (64%) had progressed on T-DXd. 32 patients (42.7%) received next line therapy, most commonly capecitabine in HER2-low (7 patients, 9.3%) and trastuzumab emtansine in HER2+ (4 patients, 5.3%). Median PFS2 was 3.4 months (95% CI 2.8-7, range: 0.7-20.5). CONCLUSION In this real world cohort, T-DXd demonstrated substantial activity in HER2-low and HER2+ MBC, consistent with trial data. However, treatment-related toxicities—especially nausea, vomiting, weight loss and ILD—often led to dose modifications and drove discontinuation in ~10% of patients. Interventions targeting emesis and cachexia may help improve tolerability and duration of treatment.
| All patients (n=75) | HER2-low (n=36) | HER2+ (n=39) | |||||||
| Toxicity | Any grade | G2 | ≥G3 | Any grade | G2 | ≥G3 | Any grade | G2 | ≥G3 |
| Nausea | 71 (94.7%) | 25 (33.3%) | 7 (9.3%) | 36 (100%) | 14 (38.9%) | 4 (11.1%) | 35 (89.7%) | 11 (28.2%) | 3 (7.7%) |
| Vomiting | 45 (65%) | 3 (4%) | 5 (6.7%) | 19 (52.8%) | 3 (8.3%) | 2 (5.6%) | 26 (66.7%) | 0 (0%) | 3 (7.7%) |
| Weight loss | 29 (38.7%) | 13 (17.3%) | 3 (4%) | 14 (38.9%) | 7 (19.4%) | 1 (2.8%) | 15 (38.5%) | 6 (15.4%) | 2 (5.1%) |
Presentation numberPS5-04-10
Comparative analysis of second primary malignancies in invasive lobular versus ductal breast carcinoma: A SEER-based study
Sneha Singh, Sunrise Hospital and Medical Center, Las Vegas, NV
S. Singh1, S. Sangam2, B. Zengin3, S. Modi4, H. Jain5, A. MariamRoy6; 1Internal Medicine, Sunrise Hospital and Medical Center, Las Vegas, NV, 2Internal Medicine, SBH Health System, Bronx, NY, 3Internal Medicine, Hamilton Medical Center, Dalton, GA, 4Internal Medicine, Jefferson Einstein Healthcare Network, Philadelphia, PA, 5Internal Medicine, Allegheny Health Network, Pittsburg, PA, 6Department of Medicine, The Ohio State University, Columbus, OH.
Background: Invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC) are the two most common histologic subtypes of breast cancer (BC), with IDC accounting for approximately 70-80% of cases and ILC comprising 10-15%. Improved screening and advances in breast cancer treatment have increased survivorship but also heightened the risk of second primary malignancies (SPMs) among long-term survivors. Our study presents a comparative analysis of the types of SPMs among ILC and IDC survivors. Methods: Using the National Cancer Institute’s Surveillance, Epidemiology, and End Results(SEER) 8 database registry, we identified a cohort of female patients diagnosed with ILC and IDC between 1975 and 2022. We analyzed the incidence of SPM using the SEER*Stat MultiplePrimary-Standardized Incidence Ratio (MP-SIR) session for the overall cohort of females with ILC and IDC. The cohort was further stratified by age (/= 40 years). Standardized incidence ratios (SIRs) were calculated to compare the observed rates with the expected rates between ILC/IDC and the general population. Results: Both ILC and IDC had an increased risk of all types of SPMs, with SIRs of 1.09 (95%CI: 1.06-1.11) for ILC and 1.14 (95% CI: 1.14-1.15) for IDC. Some sites emerged as commonsites of SPMs for both ILC and IDC, and those with highest SIRs include soft tissue (ILC: SIR =1.72; IDC: SIR = 1.55, p < 0.05), thyroid (ILC: SIR = 1.42; IDC: SIR = 1.22, p < 0.05), skinexcluding basal and squamous (ILC: SIR = 1.19; IDC: SIR = 1.06, p < 0.05), and acute myeloidleukemia (ILC: SIR = 1.90; IDC: SIR = 1.78, p < 0.05). Sites for SPM unique to ILC include mainly the stomach (SIR = 1.58, CI 1.33 – 1.86) and melanoma of the skin (SIR = 1.18, CI 1.05 -1.32). Sites for SPM unique to IDC with p < 0.05 include salivary gland, esophagus, pleura, lung, bones and joints, corpus uteri, and acute lymphocytic leukemia (Table). Greater incidence of SPMs was observed for patients age< 40 for both ILC and IDC (ILC: age /= 40 SIR = 1.08; IDC: age /= 40 SIR = 1.11). For ILC, SPM with involvement of the stomach is seen in the cohort of both age categories, but did not reach statistical significance for age < 40 (age /= 40: SIR = 1.56, CI 1.31-1.84). Conclusion: Our study provides a detailed understanding of the variation in the pattern of SPMs for both ILC and IDC. Patients with ILC exhibited a higher propensity to develop SPM at specific sites, such as the stomach and skin melanoma, highlighting the need for further investigation into the underlying biological mechanisms. Conversely, IDC has a broader distribution of unique SPMs involving the lung, pleura, salivary gland, esophagus, uterus, and bones. Younger patients exhibited a higher risk of SPMs in both ILC and IDC cohorts. These findings underscore the importance of a tailored surveillance strategy based on histological subtype.
| Site of SPM | ILC SIR (95% CI) | IDC SIR (95% SIR) |
| Common sites | ||
| Soft tissue | 1.72 (1.31-2.22) | 1.55 (1.41-1.69) |
| Thyroid | 1.42 (1.20-1.66) | 1.22 (1.16-1.29) |
| Skin | 1.19 (1.07-1.33) | 1.06 (1.02-1.10) |
| Acute myeloid leukemia | 1.90 (1.55-2.30) | 1.78 (1.67-1.90) |
| Unique to ILC | ||
| Stomach | 1.58 (1.33-1.86) | |
| Melanoma of skin | 1.18 (1.05-1.32) | |
| Unique to IDC | ||
| Salivary gland | 1.65 (1.44-1.87) | |
| Esophagus | 1.19 (1.08-1.30) | |
| Pleura | 2.86 (1.43-5.11) | |
| Lung | 1.02 (1.00-1.14) |
Presentation numberPS5-04-11
Real-world incidence and management of trastuzumab deruxtecan-associated interstitial lung disease
Jasmin Hundal, Cleveland Clinic, Cleveland, OH
J. Hundal1, N. Rehman2, A. Abushamma3, S. Haddad2, M. Velimirovic1, W. Wei1, B. Honnekeri2, X. Chen1, M. Smith1, M. Kruse1, A. Ali1; 1Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 2Internal Medicine, Cleveland Clinic, Cleveland, OH, 3Internal Medicine, Cleveland Clinic, Akron, OH.
Background:Trastuzumab deruxtecan (T-DXd) is an anti-human epidermal growth factor receptor 2 (HER2) antibody-drug conjugate that has demonstrated significant clinical benefit in patients with HER2-positive breast cancer and, more recently, in HER2-low and hormone receptor-positive (HR+), HER2 ultra-low metastatic breast cancer (MBC), thereby expanding the spectrum of targeted therapies. However, treatment-related interstitial lung disease (ILD) remains a clinically significant and potentially fatal adverse event. Real-world data on the incidence, clinical course, and risk factors associated with pneumonitis in patients receiving T-DXd remain limited.Methods:We conducted a real world (RW) single-institution, multi-site at Cleveland Clinic review of patients with MBC treated with T-DXd from December 2019 to December 2023. Demographic, clinical, and treatment-related variables were collected, including tumor characteristics, prior therapies, and outcomes. The primary endpoint was the incidence of pneumonitis, identified through clinical and radiographic assessments. Descriptive statistics were used to summarize patient characteristics and outcomes.Results:Among 76 patients, most were female (98.7%) and White (86.8%). Over half of patients had de novo metastatic disease (51.3%) and positive HR receptor status (52.6%). Prior treatments included chemotherapy, novel targeted therapies, and immunotherapy. At the time of data cutoff, 36.8% remained on T-DXd, while 42.1% had progressed. The majority (61.8%) were alive at last follow-up, with minimal loss to follow-up (2.6%).Treatment-related all-grade ILD occurred in 6 patients (7.9%). Management and outcomes varied by grade (Table 1). Additionally, 2 patients developed suspected pneumonitis, characterized by inflammatory changes on imaging after initiating T-DXd. Both cases were resolved without treatment with steroids and T-DXd was resumed. Conclusion: In this RW cohort of MBC treated with T-DXd, all-grade ILD occurred in 7.9% of patients, which is lower than the rates noted in all DESTINY trials (10-17.4%). Rate of Grade 3 or higher ILD in our study was 5.2% which is higher than reported in DESTINY trials (2.4-2.7%). 1 ILD-related death was observed in our study. Of all the 6 patients with confirmed ILD, none of the patients obtained a bronchoscopy before start of steroids. These findings underscore the importance of screening, early detection, and the need for individualized management strategies. The low rate of bronchoscopy diagnosis also highlights opportunities to engage pulmonary medicine teams in a timely manner. Further research is needed to identify predictive factors and optimize risk mitigation in patients receiving T-DXd.
| Toxicity Grade | Management | Steroid Duration (days) | Time to Resolution | T-Dxd Management | Prior Treatment with CDK4/6 inhibitor |
HER 2 Status (IHC)
|
T-Dxd Line of Therapy
|
Prior T-DxD Cycles |
| 2 | Prednisone | 60 | N/A | Discontinued | Palbociclib | 2+ | 3rd | 29 |
| 2 | Prednisone | 68 | 42 | Discontinued | None | 3+ | 3rd | 16 |
| 2 | Prednisone | 21 | N/A | Discontinued due to second episode of pneumonitis | None | 1+ | 3rd | 1 |
| 3 | Prednisone | 106 | <3 month | Discontinued | None | 3+ | 5th | 2 |
| 3 | Prednisone | 33 | N/A | Discontinued | Ribociclib | 2+ | 4th | 6 |
| 5 | Methylprednisolone | 23 | N/A | Discontinued | None | 3+ | 1st | 18 |
Presentation numberPS5-04-12
Inflammatory breast cancer : Monocentric epidemiologic, anatomiclinical features and therapeutic results about 68 patients treated from 2004 to 2024
Hamouda Boussen, Hospital of Ariana, Tunis, Tunisia
M. Boujnah1, S. Myriam1, A. Haouari1, B. Saif1, N. Mejri2, M. Manai3, H. Boussen2; 1Medical oncology, Hospital of Ariana, Ariana, TUNISIA, 2Medical oncology, Hospital of Ariana, Tunis, TUNISIA, 3Immunology, Pasteur Institute of Tunis, Tunis, TUNISIA.
Objective : To report the results of a monocentric retrospective serie of inflammatory breast cancers cases observed and treated during a period of 20 years Patients and methods : We collected in a cohort of breast cancers, the cases of inflammatory breast cancer(IBC) subtype diagnosed as T4d AJCC with an evolution of local symptoms in less than 6 months. We analyzed the epidemiologic, anatomic-clinical features, treatments and results. Results : We collected inside a cohort of 2400 breast cancer cases, 68 patients(2.8%), treated for an IBC, having a median age of 42.2 years(27 to 67) with a mean delay of evolution of 5.8 (3 to 6) months. Molecular subtypes are as followed : Luminal= 39/68(58.5%), Her 2 positive 18/69 (26%) and triple negative 11/68(15.5 %). Mean radiological tumor size was 32.5mm(25 to 70mm). Within the 55 patients operated, we observed 10 pathologic complete responses(19%) in 6 cases of luminal B subtype(3 of them her 2+), 3 her 2 positive subtype and 1 triple negative case. We observed 22 relapses with a mean delay of 24 months, msotly metastatic. Nombre de pCr chez les malades opérées =10 (6 luminal , 3 HER2 surexprimé , 1 TN). Overall survival was at 87,14 months, 108 for luminal B subtype, 96,4 for her 2 positive cases and 15.1 months for triple negative cases. Conclusion : Our monocentric serie from Tunisia, reflet’s the epidemiologic and anatomo-clinical features of IBC in our coutry. Patients remain younger than those reported in western countries. We observed a positive trend in terms of survival between patients treated before and after 2010, probably due to the multidisciplinary management and the impact of targeted therapies in her 2 positive cases.
Presentation numberPS5-04-13
Intraoperative Radiotherapy in Early-Stage Breast Cancer:Long-Term Survival and Real-World Outcomes from Community Hospitals in Baltimore.
Angel “Luis” OROSCO TTAMINA, MedStar Health Georgetown University (Baltimore) Internal Medicine Program., Baltimore, MD
A. OROSCO TTAMINA1, P. Fowler2, M. Farha3; 1Internal Medicine Resident, MedStar Health Georgetown University (Baltimore) Internal Medicine Program., Baltimore, MD, 2Chief of Radiation Oncology, MedStar Good Samaritan Hospital, Baltimore, MD, 3Medical director of the Breast Centre, MedStar Good Samaritan Hospital., Baltimore, MD.
Background: Breast-conserving surgery followed by whole-breast radiotherapy (WBRT) is the standard of care for early-stage breast cancer, significantly reducing the risk of local recurrence. However, WBRT requires multiple treatment sessions over several weeks, imposing logistical, financial, and compliance burdens that can particularly impact socioeconomically disadvantaged populations. Intraoperative radiotherapy (IORT), which delivers a single fraction of radiation during surgery, has been proposed as an alternative that may improve treatment adherence and reduce toxicity. While randomized trials have demonstrated non-inferiority in selected patients, real-world data on long-term oncologic outcomes in community hospital settings remain limited. This study evaluates overall survival (OS) and progression-free survival (PFS) in patients treated with IORT at MedStar community hospitals in Baltimore. Methods: In this prospective cohort study, patients with early-stage breast cancer who underwent breast-conserving surgery with IORT at four MedStar community hospitals in Baltimore between 2013 and 2019 were included. Clinical outcomes were assessed through medical records, imaging, and follow-up visits. OS and PFS at 5 years were estimated using Kaplan-Meier survival analysis, and recurrence rates were determined through clinical and radiologic evaluations. Results: A total of 115 patients were initially identified; 10 were excluded due to loss to follow-up (n=2), undocumented date of death (n=5), or receipt of adjuvant WBRT (n=3), leaving 105 patients for the final analysis. The median age was 65 years (range, 45-82), with most patients having hormone receptor-positive, HER2-negative tumors, and surgical tumor sizes ranging from 0.4 to 3.7 cm. The 5-year OS was 88%, and the 5-year PFS was 87%, demonstrating durable local disease control. Most recurrences occurred within the first five years, consistent with prior IORT studies. No severe late toxicities or unexpected adverse events were reported. Conclusions: IORT demonstrated comparable long-term OS and PFS to historical WBRT data while reducing treatment duration and eliminating the need for prolonged hospital visits. These results align with previous IORT trials, including TARGIT-A and ELIOT, which reported similar overall survival outcomes, supporting the efficacy of IORT in early-stage breast cancer. This study supports the feasibility of implementing IORT in community hospital settings, where access to prolonged radiation therapy can be challenging, and highlights its potential to improve accessibility and mitigate disparities in radiotherapy completion for socioeconomically disadvantaged populations. Further studies with larger cohorts and longer follow-up are warranted to refine patient selection and optimize treatment protocols for broader clinical application.
Presentation numberPS5-04-14
Real-world cardiotoxicity in HER2-positive breast cancer patients in Peru
Jorge Sanchez A, Instituto Nacional de Enfermedades Neoplasicas, Lima, Peru
J. Sanchez A1, W. Cruz-Diaz1, J. Luque1, V. Tuesta1, E. Ruiz-Mori2, Z. Morante1, G. Valencia1, P. Rioja1, S. Neciosup1, T. Vidaurre1, K. Roque1, C. Castañeda1; 1Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplasicas, Lima, PERU, 2Cardio-oncology Unit, Instituto Nacional de Enfermedades Neoplasicas, Lima, PERU.
Background: Anti-HER2 therapies have significantly improved survival in HER2-positive breast cancer and are the standard of care. Early identification of Trastuzumab-related cardiotoxicity is essential to guide timely interventions and preserve cardiac function. While left ventricular ejection fraction (LVEF) remains the standard monitoring tool, it often misses subclinical dysfunction. Global longitudinal strain (GLS), derived from speckle-tracking echocardiography, offers greater sensitivity for early detection. Although Trastuzumab is used across Latin America, published data on cardiotoxicity remain limited. This study analyzes cardiotoxicity prevalence and evaluates the role of LVEF and GLS in cardiac surveillance in a real-world Peruvian cohort.Methods: We performed a retrospective observational study among 144 women diagnosed with HER2-positive breast cancer who were treated with Trastuzumab ± Pertuzumab in adjuvant, neoadjuvant, or metastatic settings. Clinical-pathological variables and patterns of treatment were collected. Cardiac function was assessed using electrocardiogram (EKG), serum troponins, echocardiographic measurements of LVEF, and GLS at baseline, during and at treatment completion. Cardiotoxicity was defined as either a ≥10% absolute reduction in LVEF or a ≥15% absolute reduction in GLS.Results: The median age was 49 years (range 24-73). Of the total cohort, 112 patients received neoadjuvant treatment, 93 continued with adjuvant therapy, and 37 were treated for metastatic disease. Left-sided radiotherapy was administered in 28% of cases. Pertuzumab was co-administered in 39% of patients. The median number of Trastuzumab cycles was 15, over a median treatment duration of 45 weeks. Median dose density was 0.33 doses/week for Trastuzumab and 0.23 doses/week for Pertuzumab. All patients underwent baseline electrocardiogram and troponin assessment. Except for one case of chemotherapy-related tachyarrhythmia, all patients were asymptomatic throughout treatment. LVEF measurements were available in 136 patients (94.4%) and GLS in 118 patients (81.9%). Cardiotoxicity was observed in 12 patients (8.3%): 9 patients experienced a ≥10% reduction in LVEF and 3 experienced a ≥15% reduction in GLS, with no overlap between the two groups. Subgroup analysis showed that all GLS alterations occurred in neoadjuvant patients, while LVEF reductions were observed in 4 neoadjuvant, 1 adjuvant, and 4 metastatic cases. Among those with LVEF decline, 2 were obese and 2 had received left-sided radiotherapy. Among GLS-declining patients, 1 had also received left-sided radiotherapy. One patient with GLS decline also presented with elevated troponin, suggesting subclinical myocardial injury.Conclusions: Cardiotoxicity affected 8.3% of patients in this Peruvian real-world cohort, with no overlap between LVEF and GLS-defined events. GLS enabled detection of subclinical myocardial dysfunction not identified by LVEF. The presence of known risk factors such as obesity and left-sided radiotherapy in several cases underscores the need for comprehensive cardiac monitoring. These findings support the integration of GLS into routine surveillance, particularly for high-risk patients, to enable early cardioprotective interventions and ensure continuity of oncologic treatment.
Presentation numberPS5-04-15
Improving Adrenal Insufficiency Screening in Patient Receiving Keynote-522</b>
Mara Hofherr, Washington University, St. Louis, MO
M. Hofherr, A. Golden, K. Clifton, H. Al-Bahadili; Medical Oncology, Washington University, St. Louis, MO.
Abstract Title: Improving Adrenal Insufficiency Screening in Patient Receiving Keynote-522 Investigators: Amanda Golden, MD, Katherine Clifton, MD, Mara Hoefherr, PharmD, Huda Al-Bahadili, MD Background: Endocrine toxicities of immune checkpoint inhibitors, such as the pembrolizumab, are well known and documented. Immune-related adverse events from checkpoint inhibitors can present as endocrinopathies involving any organ system. In KEYNOTE-522, 18 patients (2.3%) of any grade and 10 patients (1.3%) with grade 3 adrenal insufficiency (AI) were reported in the pembrolizumab arm. Testing for adrenal insufficiency in this population is imperative as the symptoms of this toxicity can be very vague and nonspecific. Current cortisol testing is recommended by the pembrolizumab package insert, however is not well established in clinical practice. This study investigates improved screening for adrenal insufficiency in KEYNOTE-522 patients. Methods: In this retrospective, single center quality improvement project at baseline, at 8 and at 16 weeks of therapy with the Keynote-522 regimen. Any value greater than 12 is considered normal, 12 or below is considered abnormal and had further testing done. System plans were updated. Patients were included if they received treatment from 3/1/2024 – 9/1/2025. The authors will collect baseline endocrine data, pertinent variables (i.e, symptoms of endocrine disorders, recent steroid use, amount of pembrolizumab received, i.e.), and outcomes. Results: A total of 70 patients were given KEYNOTE-522 in the specified timeframe and had baseline cortisol drawn. We plan to collect baseline endocrine laboratory parameters. We will Strengths include the resources and protocols already in place for patients undergoing the Keynote-522 regimen. We would be able to incorporate the testing for adrenal insufficiency into already planned labs draws. Potential limitations or confounding factors could be if a patient has recently gotten any treatment with dexamethasone or another steroid treatment. This could affect the values listed above. If the data would prove to be inconclusive for a patient, then next steps would be a chart review to ensure they did not have glucocorticoid therapy prior to lab testing. Results will be collected after 9/1/2025 and incorporated into the poster presentation and subsequent manuscript.
Presentation numberPS5-04-16
Analysis of Efficacy, Safety and Cost of Datopotamab Deruxtecan and Sacituzumab Govitecan for Advanced Hormone Positive Breast Cancer Patients
Manaswini Krishnakumar, Saint Vincent hospital, Worcester, MA
A. Reddy1, M. Krishnakumar2, M. Zafar3; 1Hematology and Oncology, Mercy Hospital, Fort Smith, AR, 2Internal Medicine, Saint Vincent hospital, Worcester, MA, 3Internal Medicine, Mercy Hospital, Fort Smith, AR.
Analysis of Efficacy, Safety and Cost of Datopotamab Deruxtecan and Sacituzumab Govitecan for Advanced Hormone Positive Breast Cancer Patients BackgroundTROP2 antibody drug conjugate (ADC) broadens therapeutic option for metastatic breast cancer patients particularly in patients with no HER2 expression. Sacituzumab govitecan (SG) received FDA approval in 2023 based on TROPiCS02 trial, while datopotamab deruxtecan (DD) secured approval in January 2025 following TROPIONBreast01. Given the absence of head-to-head trials, we aim to analyze the safety and cost effectiveness of these drugs which could guide in better treatment selection.Methods A systematic search was conducted across PubMed, Scopus, and the Cochrane to identify RCTs evaluating SG or DD in hormone receptor-positive, HER2-negative (HR+/HER2-) metastatic breast cancer. Trials were eligible if they included post-endocrine therapy or chemotherapy-pretreated patients with HR+/HER2- disease and reported survival or safety outcomes. .Results TROPIONBreast01 randomized 365 patients to DD and 367 to chemotherapy. Median PFS improved from 4.9 to 6.9 months (HR 0.63) without statistically significant OS (HR 1.01). TROPiCS02 randomized 272 patients to SG and 271 to chemotherapy and showed improved median OS from 11.2 to 14.4 months (HR 0.79). Neutropenia and diarrhea were the most common grade 3 side effects with SG. Stomatitis (6.4%) and fatigue (1.7%) were the most common grade 3 side effects with DD (Table 1). Exclusive side effects with DD included ophthalmological adverse effects including dry eye 0.6% and keratitis 0.6%. No cases of interstitial lung disease were reported with SG in TROPiCS 02 study. From available data on drug cost and median treatment duration from these studies we estimated the total cost of treatment as $165000 and $142000 for DD and SG respectively. The total cost will be higher for SG if adverse event management costs are included. DiscussionTROP2-directed ADCs represent a promising treatment for heavily pretreated HR+/HER2- metastatic breast cancer patients. In the TROPiCS-02 trial, SG significantly improved both PFS and OS while DD in the TROPION-Breast01 trial improved PFS but showed no OS benefit. SG had significantly higher adverse events and hematologic toxicities compared to DD. Though cost of these treatments is similar SG’s higher toxicity may lead to greater health care utilization and poor quality of life. DD should be used with caution in patients with preexisting eye disorders. Real-world comparative analysis and biomarker-driven studies are needed to guide optimal sequencing. Abbreviations: PFS, progression free survival; OS, overall survival; DCR, disease control rate; ORR, overall response rate; G3AER, grade 3 adverse events rate; DDR, drug discontinuation rate; HR, hazard ratio.
| Endpoints and safety data | Datopotamab Deruxtecan TROPION-Breast01 | Sacituzumab Govitecan TROPiCS-02 |
| Median PFS (months) | 6.9 vs 4.9 HR 0.63 (0.52–0.76) | 5.5 vs 4.0 HR 0.66 (0.53–0.83) |
| Median OS (months) | 18.6 vs 18.3 HR 1.01 (0.83–1.22) | 14.4 vs 11.2 HR 0.79 (0.65–0.96) |
| ORR | 36 % | 21 % |
| DCR | 75 % | 73 % |
| G3AER | 20.8 % | 74 % |
| DDR | 3.1 % | 6 % |
Presentation numberPS5-04-17
Male Breast Cancer in Mexico: Experience of a reference breast cancer center in Northeastern Mexico.
Valentina Leitzelar-Bueso, Tecnológico de Monterrey, Monterrey, Mexico
V. Leitzelar-Bueso, L. F. Martinez-Caudillo, M. S. Guzman-Garcia, J. E. Guzman-Garcia, D. Aguilar-Y-Mendez; Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey, MEXICO.
Male breast cancer (MBC) is rare and often diagnosed late due to limited awareness. This retrospective study describes the clinical presentation, histopathological profile, risk factors, and treatment in a cohort of male patients from a reference breast cancer center in Northeastern Mexico. Methods: In the period from 2015-2025, a total of 11 male patients were diagnosed with MBC. Data collected from clinical records included demographic characteristics such as age of diagnosis, as well as clinical presentation (including TNM classification), associated symptoms, histopathological findings, genetic tests, initial therapeutic approach, and relevant risk factors such as obesity, alcohol use, smoking, and family history among others using a checklist designed after an extense review of literature. (Table 1). Results: The median age of diagnosis was 56 years old. Over 80% presented with a palpable mass; 20% were incidental findings. Some patients had inflammatory signs (erythema, peau d’orange), pain (5), ulceration (1), lymphadenopathy (3), or skin retraction (1). In terms of risk factors, obesity was documented in two patients, and alcohol consumption was positive in 75% of the cohort. Active smoking and family history of prostate and breast cancer was noted in four individuals. Histologically, the predominant subtype was invasive ductal carcinoma of no special type (NST). Hormone receptor status was positive in all, with high estrogen receptor expression (ER >90%) and variable PR/HER2. Ki-67 ranged from 0-40%. TNM classification at diagnosis varied, with presentations spanning from stage IA (10%) to stage IV (10%) , although locally advanced disease (stage II and III, 20%) was common. Two patients had metastases at diagnosis (lung, bone, liver). Multigene panel testing (performed in 6 patients) revealed BRCA1 (1), BRCA2 (2), and PALB2 (1) mutations. All underwent mastectomy. Chemotherapy (7), radiotherapy (8), and hormonal therapy with tamoxifen (9) were common therapeutic approaches. One patient received paclitaxel, while another with a BRCA2 variant received treatment with olaparib. Conclusion: MBC typically presents as a palpable mass, often with inflammatory skin changes. Hormone receptor positivity and BRCA2 mutations were frequent. Modifiable risk factors like alcohol use and smoking were prevalent. Inconsistent recording of risk factors limits data comparability, underscoring the need for standardized collection tools. Furthermore, the presence of familial cancer syndrome reinforces the need for genetic testing and counseling in high risk populations. Additionally, public health efforts should promote MBC awareness, early diagnosis, and timely treatment to reduce mortality and improve outcomes.
| Risk Factor | Total | Percentage |
| Obesity | 2 | 25% |
| Alcohol | 6 | 75% |
| Smoking | 5 | 63% |
| Advanced age | 3 | 27% |
| Family history | 3 | 38% |
| Pathogenic variants | 4 | 36% |
| Exogenous hormone use | 0 | 0% |
| Sedentarism | 0 | 0% |
| Exposure to exogenous radiation | 0 | 0% |
| Gynecomastia | 0 | 0% |
| Fractures >45 yo | 0 | 0% |
| Cryptorchidie | 0 | 0% |
| Klinefelter syndrome | 0 | 0% |
Presentation numberPS5-07-02
NRG-BR008: A phase III randomized trial of radiotherapy optimization for low-risk HER2-positive breast cancer (HERO)
Melissa P Mitchell, The University of Texas MD Anderson Cancer Center, Houston, TX
M. P. Mitchell1, L. Z. Braunstein2, H. Bandos3, W. M. Sikov4, A. J. Khan5, P. Y. Chen6, P. A. Ganz7, R. Jagsi8, J. R. White9, R. S. Cecchini3, H. Kang10, S. L. Puhalla11, K. L. Bolton12, E. P. Connolly13, E. Stringer-Reasor14, K. R. Gergelis15, T. B. Julian16, E. P. Mamounas17, N. Wolmark18; 1Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 3Department of Biostatistics and Health Data Science, University of Pittsburgh; NRG Oncology Statistical and Data Management Center, Pittsburgh, PA, 4Department of Hematology/Oncology, Women and Infants Hospital of Rhode Island Breast Health Center; Warren Alpert Medical School of Brown University, Providence, RI, 5Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center Monmouth, Middletown, NJ, 6Department of Radiation Oncology, Corewell Health System – Beaumont University Hospital, Royal Oak, MI, 7Medicine/Health and Policy Management, David Geffen School of Medicine at UCLA; UCLA Fielding School of Public Health; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 8Department of Radiation Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, 9Department of Radiation Oncology, University of Kansas Medical Center Comprehensive Cancer Center, Kansas City, KS, 10Department of Radiation Oncology, Stritch School of Medicine at Loyola University, Maywood, IL, 11Department of Medical Oncology/Hematology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, 12Division of Oncology, Washington University School of Medicine, St. Louis, MO, 13Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY, 14Department of Hematology/Oncology, O’Neal Comprehensive Cancer Center at University of Alabama, Birmingham, AL, 15Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 16Department of Surgical Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, 17Surgical Oncology, AdventHealth Cancer Institute, Orlando, FL, 18Department of Surgical Oncology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine; NSABP Foundation, Inc, Pittsburgh, PA.
Background Breast radiotherapy (RT) is the standard of care for patients with early-stage breast cancer (BC) who undergo breast-conserving surgery (BCS). However, the magnitude of benefit of RT is less clear in BCS patients with low-risk disease who receive effective systemic therapy. Among patients with early-stage HER2-positive (HER2+) BC, 10-year locoregional recurrence has been reported as low as 1.5% following BCS, adjuvant chemotherapy and HER2-targeted therapy, and RT. Given these exceedingly favorable outcomes, with the addition of HER2-directed therapy, we seek to evaluate the feasibility of omitting RT among patients with early-stage HER2+ BC following BCS and appropriate systemic therapy. Methods This is a phase III randomized trial for patients ≥18 years with early-stage, node-negative, HER2+ (IHC/FISH) BC treated with BCS with negative margins and sentinel lymph node biopsy or axillary dissection. Patients undergoing primary surgery must have pathologic T1 (≤3 cm) N0 disease, whereas patients receiving neoadjuvant therapy must have clinical T1-2 (≤5 cm) N0 disease and exhibit a pathologic complete response (ypT0N0) at surgery. Patients with residual non-invasive disease (DCIS) in the surgical specimen (ypTis) are eligible if residual DCIS spans ≤1 cm and surgical margins are negative for DCIS. All patients must receive systemic therapy and HER2-targeted therapy, either in the adjuvant or neoadjuvant setting. Stratification is by age (1 cm), estrogen-receptor status (positive; negative), and systemic therapy sequencing (adjuvant v neoadjuvant). Patients will be randomized to standard breast RT in addition to continuation of HER2-targeted therapy to complete one year of treatment (Arm 1), or HER2-targeted therapy alone (Arm 2). Endocrine therapy will be recommended for patients with hormone-receptor positive tumors. The primary endpoint is the recurrence-free interval (RFI). Secondary endpoints include time to ipsilateral breast recurrence, locoregional recurrence, disease-free survival, and overall survival, in addition to the 7-year ipsilateral breast recurrence rate among those not receiving RT. A health-related quality of life sub-study will assess differences in patient-reported breast pain and worry. We estimate a 7-year RFI of 97.5% with RT and allow for a clinically acceptable decrement of 3.63% without RT (7-year RFI of 93.87%; HR 2.5) to establish omission of RT as non-inferior. NRG-BR008 aims to enroll 1,300 patients over 7.25 years, yielding 80% power to detect the non-inferiority of RT omission with a one-sided α=0.05. We expect to observe the required 38 RFI events within 4.5 years of additional follow-up. The NRG-BR008/HERO trial opened to accrual in March 2023. Accrual is 103/1,300 as of July 9, 2025.
Presentation numberPS5-07-03
Capitrue, capicorn, and capitana: three phase iiib studies to evaluate the use of capivasertib in combination with fulvestrant in patients with advanced breast cancer who have relapsed/progressed on endocrine therapy and cdk4/6 inhibitors reflecting real-world clinical practice in china, germany, belgium, portugal and spain.
Rodrigo Sánchez Bayona*, Hospital Universitario 12 de Octubre, Madrid, Spain
R. Sánchez Bayona*1, S. Kuemmel*2, Z. Jiang*3, M. Oliveira4, O. Gluz5, S. Zhang6, T. Pascual7, R. Wuerstlein8, P. Yuan9, M. Vidal Losada7, M. Graeser10, B. Zhao11, F. Henao Carrasco12, R. Kates13, F. Ju14, G. Viñas15, C. zu Eulenburg13, Z. Chen16, A. Perelló Martorell17, E. de Azambuja18, F. Fu19, A. Antón20, H. Gouveia21, C. Hao22, C. San Millán Martín23, T. Schinkoethe24, Y. He25, Z. Jian26, C. Saura*4, N. Harbeck*27; 1Oncology Department, Hospital Universitario 12 de Octubre, Madrid, SPAIN, 2Oncology Department, Interdisciplinary Breast Center, KEM Evangelical Hospital Essen-Mitte, Essen, GERMANY, 3Breast Cancer, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA, 4Medical Oncology Dept, Vall d’Hebron University Hospital, Barcelona, SPAIN, 5Oncology Department, Breast Center Niederrhein, Evangelical Bethesda Hospital, Moenchengladbach, GERMANY, 6Oncology Department, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA, 7Oncology Department, Hospital Clinic de Barcelona, Barcelona, SPAIN, 8Breast Center, Dept. of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, SPAIN, 9Oncology Department, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, CHINA, 10Dept. of Obstetrics and Gynecology, University Witten-Herdecke, Witten, GERMANY, 11Oncology Department, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, CHINA, 12Oncology Department, Hospital Universitario Virgen Macarena, Seville, SPAIN, 13Oncology Department, Westdeutsche Studiengruppe GmbH, Moenchengladbach, GERMANY, 14Oncology Department, Qingdao Central Hospital, Qingdao, CHINA, 15Oncology Department, ICO Girona, Girona, SPAIN, 16Oncology Department, Zhejiang Cancer Hospital, Hangzhou, CHINA, 17Oncology Department, Hospital Universitari Son Espases, Palma de Mallorca, SPAIN, 18Dept. Medical Oncology, Institut Jules Bordet,, Anderlecht, BELGIUM, 19Oncology Department, Fujian Medical University Union Hospital, Fuzhou, CHINA, 20Oncology Department, Hospital Universitario Miguel Servet, Zaragoza, SPAIN, 21Breast Unit, Breast Unit of the Champalimaud Foundation, Lisbon, PORTUGAL, 22Oncology Department, Tianjin Cancer Hospital Airport Hospital, Tianjin, CHINA, 23OBU Medical Department,, AstraZeneca Spain, Madrid, SPAIN, 24Oncology Department, CANKADO GmbH, Munich, GERMANY, 25Oncology Department, Huizhou Central People’s Hospital, Huizhou, CHINA, 26OBU Medical Department, AstraZeneca China, Beijing, CHINA, 27Breast Center, Dept. of Obstetrics and Gynecology and CCC Munich, LMU University Hospital, Munich, GERMANY.
* These authors contributed equally to this abstract. Background HR+/HER2− advanced breast cancer (ABC) remains a therapeutic challenge due to resistance to endocrine therapy (ET) and CDK4/6 inhibitors. Alterations in the PI3K/AKT pathway, including PIK3CA, AKT1, and PTEN alterations, are associated with poor prognosis and with the activation of PI3K-AKT signaling. Capivasertib, a selective AKT inhibitor, improved progression free survival (PFS) statistically significant and clinically meaningful in the CAPItello291 trial (NCT04305496), when combined with fulvestrant, both in the overall population (PFS in Capivasertib+fulvestrant group 7.2 vs 3.6 in placebo+fulvestrant group, HR=0.60) and in patients with PIK3CA/AKT1/PTEN altered tumors (PFS in Capivasertib+fulvestrant group 7.3 vs 3.1 in placebo+fulvestrant group, HR=0.50). Building on these findings, the CAPItrue (China-NCT06635447), CAPIcorn (Germany, Belgium, and Portugal), and CAPItana (Spain-NCT06764186) trials aim to evaluate the real-world (RW) effectiveness and safety in five countries, addressing the need for region specific data to inform clinical practice and treatment strategies in diverse healthcare settings. Trial Design Based on CAPItello291 trial protocol, the CAPItrue (China), CAPIcorn (Germany, Belgium, and Portugal), and CAPItana (Spain) studies are Phase IIIb, single arm, multicenter trials designed to evaluate in a RW setting the effectiveness and safety of the combination. All studies administer capivasertib 400 mg BID (4 days on, 3 days off) together with fulvestrant (FUL) 500 mg IM every 28 days, with a loading dose given on Day 15 of Cycle 1, in patients with HR+/HER2- advanced breast cancer and alterations in PIK3CA/AKT1/PTEN who have progressed on ET in combination with CDK4/6 inhibitors. CAPItrue uniquely allows inclusion of patients without PI3K/AKT pathway alterations. Inclusion criteria allow for up to two previous lines of ET (and one line of chemotherapy for advanced disease), include diabetic or prediabetic patients (HbA1c <8%). Eligibility criteria have been expanded to include with prior SERDs treatment in the three studies, and specifically for CAPItana up to 20% of patients with an ECOG PS2. In the three studies, the primary endpoint is time to next/first subsequent treatment (TTNT/TFST) as the main measure of effectiveness. CAPItrue includes two cohorts: patients without prior FUL (cohort 1) and with 1L FUL (cohort2, not included in CAPItello-291). Regarding health-related quality of life, different questionnaires are employed according to the region: the EORTC QLQ BR42 in Spain, FACT G in Germany, and an adaptation of the EORTC QLQ BR23 and EORTC QLQ-C30 in China, reflecting local practices. To date, in the CAPItrue study, 81 sites have been activated, and 203 out of 560 patients have been enrolled (First Subject In (FSI): 26 Sep 2024 Huizhou Central People’s Hospital). In Spain, 17 sites are active with 53 out of 100 patients enrolled ((FSI: 7Jan2025 in Hospital Universitario Miguel Servet). Meanwhile, CAPIcorn plans to open 23 sites (15 in Germany, 5 in Belgium, and 3 in Portugal) with an expected recruitment of 250 patients. The parallel execution of these studies across five countries (China, Germany, Belgium, Portugal, and Spain) will provide robust, region-specific RW data from approximately 910 patients. This will enable the adaptation of CAPItello‑291 findings to diverse clinical practices and contribute to the development of globally and locally relevant oncology strategies. A joint analysis of the data gathered from the three studies is planned.
Presentation numberPS5-07-04
NRG-BR009: A phase III trial evaluating addition of adjuvant chemotherapy to Ovarian Function Suppression + Endocrine Therapy in premenopausal women with pN0-1, HR+/HER2- breast cancer and recurrence score ≤25 (OFSET)
Sandra Swain, Georgetown Lombardi Comprehensive Cancer Center; Georgetown University Medical Center; and MedStar Health, Washington, DC
S. Swain1, G. Tang2, S. L. Puhalla3, P. A. Ganz4, N. L. Henry5, R. S. Cecchini2, S. A. Reid6, P. Rastogi3, C. E. Geyer, Jr.3, J. R. White7, A. S. Clark8, T. C. Haddad9, G. A. Vidal10, N. Wolmark11, E. P. Mamounas12; 1Department of Medicine/Research Development, Georgetown Lombardi Comprehensive Cancer Center; Georgetown University Medical Center; and MedStar Health, Washington, DC, 2Department of Biostatistics and Health Data Science, University of Pittsburgh; NRG Oncology Statistical and Data Management Center, Pittsburgh, PA, 3Department of Hematology/Medical Oncology, UPMC Hillman Cancer Center; University of Pittsburgh School of Medicine, Pittsburgh, PA, 4Medicine/Health and Policy Management, David Geffen School of Medicine at UCLA; UCLA Fielding School of Public Health; UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 5Department of Internal Medicine, University of Michigan Medical School; SWOG, Ann Arbor, MI, 6Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, 7Department of Radiation Oncology, University of Kansas Medical Center Comprehensive Cancer Center, Kansas City, KS, 8Department of Medical Oncology/Hematology, Perelman School of Medicine at the University of Pennsylvania; Abramson Cancer Center, Philadephia, PA, 9Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 10Department of Medical Oncology/Hematology, West Cancer Center and Research Institute, Germantown, TN, 11Department of Surgical Oncology, UPMC Hillman Cancer Center; University of Pittsburgh School of Medicine; NSABP Foundation, Inc., Pittsburgh, PA, 12Department of Surgical Oncology, AdventHealth Cancer Institute, Orlando, FL.
Background As shown by the TAILORx and RxPONDER trials, recurrence score (RS) identifies many postmenopausal pts who do not benefit from addition of ACT to endocrine therapy (ET); however, it also identifies certain subsets of premenopausal pts who do benefit (node-neg/high clinical risk/RS 16-20, node-neg/RS 21-25, and node-pos/RS ≤25). Most premenopausal pts in these trials did not receive ovarian function suppression (OFS) as part of their ET regimen. Given the observed benefit from OFS in high-risk premenopausal pts with HR+/HER2- BC in the SOFT/TEXT trials, we question whether the noted ACT benefit in TAILORx/RxPONDER may have been the result of chemotherapy-induced OFS. To address this, we developed OFSET, a phase III clinical trial comparing OFS+ET v ACT+OFS+ET. Methods We hypothesize that addition of ACT to OFS+ET is superior to OFS+ET in improving invasive breast cancer-free survival (IBCFS) among premenopausal, early-stage BC pts with HR+/HER2- tumors and a 21-gene RS between 16-25 (for pN0 pts) and 0-25 (for pN1 pts). Secondary objectives include invasive disease-free survival, overall survival, distant recurrence-free interval, breast cancer-free interval, and health-related quality of life (HRQOL). Pts must be node-neg with RS 16-20 (plus high clinical risk), or RS 21-25, or have 1-3 positive nodes with RS ≤25. Stratification is by nodal status/RS status (pN0 RS 16-25 v pN1 RS 0-15 and pN1 RS 16-25), intent to receive CDK4/6 inhibitor (yes; no), and age (18-39 v ≥40). Pts are randomized after surgery to either OFS+ET or ACT+OFS+ET. ET is an aromatase inhibitor (AI) per physician discretion; or tamoxifen if AI is not tolerated or if OFS is incomplete. Radiotherapy will be administered per protocol guidelines. The HRQOL substudy will assess differences between arms in severe menopausal symptoms (measured by FACT ESS-19) and pain severity (PROMIS). Blood and tumor specimens will be collected for future research. We anticipate accrual of 3,960 pts in 7 yrs, 7 mos. Per NSABP B-28/RxPONDER data, 5-yr IBCFS of pN1 pts on the ACT+OFS+ET arm is estimated at 92.3%. Based on TAILORx data, 5-yr IBCFS of pN0 pts on the ACT arm is ~95%. Assuming 56% of pts to be pN0 and 44% pN1, and a 0.5% annual loss-to-follow-up rate, the definitive analyses to detect a hazard ratio: 0.75 with ACT+OFS+ET v OFS+ET, with 1-sided α of 0.025 and 80% power, will require 380 IBCFS events, expected to occur ~11 yrs after study initiation. Accrual is 292/3,960 (from 8/2023 to 5/2025). Among 148 pts with OFS data available, 79 (53.4%) were prescribed goserelin and 69 (46.6%) leuprolide. A monthly dosing schedule was prescribed for 86.1% of pts receiving goserelin and 73.5% of those receiving leuprolide; the remaining received a 3-monthly schedule. NCT05879926 Support: U10 CA180868, -80822, UG1 CA189867, U24 CA196067
| Age | |||
| Median (Min-Max) | 44 (22-54) | ||
| <40 years old | 42 (14.4%) | ||
| ≥40 years old | 250 (85.6%) | ||
| Nodal status and RS | CDK inhibitor planned | No plan of CDK inhibitor | Total |
| pN0, RS 16-25 | 11 (19.3%) | 46 (80.7%) | 57 |
| pN1, RS 0-15 | 80 (54.1%) | 68 (45.9%) | 148 |
| pN1, RS 16-25 | 48 (55.2%) | 39 (44.8%) | 87 |
Presentation numberPS5-07-05
A prospective multicenter study on the clinical utility of virtual sonography for metastatic axillary lymph nodes during neoadjuvant chemotherapy in breast cancer patients (FUSION-03)
Manabu Futamura, Gifu University Hospital, Gifu, Japan
M. Futamura1, S. Junta2, I. Kazuhiro3, S. Takehiko4, K. Keitaro5, N. Shogo6, Y. Miwa7, N. Akira1, O. Mai1, M. Nobuhisa8, U. Takayoshi9, A. Sadako10; 1Breast Surgery, Gifu University Hospital, Gifu, JAPAN, 2Breast Surgery, Chiba University Hospital, Chiba, JAPAN, 3Surgery, Gihoku Kosei Hospital, Yamagata, JAPAN, 4Breast Surgical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 5Breast Surgery, Ogaki Municipal Hospital, Ogaki, JAPAN, 6Breast and Endocrine Surgery, Aichi Medical University Hospital, Nagakute, JAPAN, 7Breast Surgical Oncology, Showa Medical University Koto Toyosu Hospital, Tokyo, JAPAN, 8Gastroenterological Surgery, Gifu University Hospital, Gifu, JAPAN, 9Breast Imaging and Breast Interventional Radiology, Shizuoka Cancer Center Hospital, Shizuoka, JAPAN, 10Breast Surgery, Tokyo Women’s Medical University Hospital, Tokyo, JAPAN.
Background: Neoadjuvant chemotherapy (NAC) for breast cancer (BC) facilitates minimally invasive surgical approaches for both the primary tumor and axillary lymph nodes (LNs). When NAC is effective, breast-conserving surgery may be feasible, and axillary lymph node dissection (ALND) may be avoided. However, ALND is still commonly performed in node-positive patients, even when post-NAC imaging shows no evidence of residual nodal metastases. Recently, tailored axillary surgery/dissection (TAS/TAD) has gained attention. TAS involves pre-NAC placement of a marker in biopsy-proven metastatic LNs, followed by targeted excision of the marked LN, sentinel LNs, and any palpable nodes after NAC. This less invasive strategy may provide sufficient local control if the false-negative rate (FNR) of axillary evaluation is kept below 10%. Virtual sonography (VS)—a fusion technique combining real-time ultrasound (US) with pre-acquired imaging such as CT or MRI—enables accurate localization of target lesions. Commercial US systems already integrate this technology, offering benefits including minimal invasiveness, repeatability, and cost-effectiveness. In this ongoing study, we apply VS to enhance TAS precision during surgery following NAC. Objective: To evaluate the clinical utility of VS in TAS for patients with axillary LN metastases undergoing NAC in a prospective, multicenter observational setting. Study Design: In TAS, a marker is inserted into the metastatic LN prior to NAC as a surgical guide. While US is standard for identifying the marker, detection rates vary. TAS is considered feasible when the marker identification/excision rate reaches ≥ 95%. To enhance detection accuracy, this study utilizes VS for marker localization. Approved by the Central Ethics Committee of Gifu University (Approval No. 2024-005), this study follows these procedures: 1) Histopathological confirmation of axillary LN metastasis prior to NAC. 2) US-guided placement of an UltraCor™ Twirl™ marker in the metastatic LN. 3) Acquisition of axillary US volume data and longitudinal marker tracking using VS at three key time points: before NAC, after the second cycle, and preoperatively. 4) Measurement of the time required for marker identification using fusion imaging at the final preoperative session. 5) US-guided injection of blue dye (0.1-0.2 mL) into/around the marker-identified LN based on fusion images. 6) TAS during BC surgery, followed by radiographic confirmation of marker retrieval in the excised specimen. Marker presence confirms TAS success. Backup ALND will be performed thereafter. Eligibility Criteria:-Inclusion: 1) Primary BC with histologically or cytologically confirmed axillary LN metastasis. 2) Clinical stage cT0-T4, cN1-N3, M0 (Stage IIA-IIIC). 3) Planned NAC ( ≥ 3 cycles). 4) Age ≥ 20 years. 5) ECOG performance status 0-2. 6) Written informed consent. -Exclusion: 1) Inability to insert a marker into/around the metastatic LN. 2) Contraindications to NAC for systemic or medical reasons. Primary Endpoint: Marker identification rate of metastatic axillary LNs using US-US fusion (VS), confirmed by radiography of the excised specimen post-TAS. Secondary Endpoints: 1) Marker identification rate at NAC completion via US-US fusion. 2) Time required for marker identification via fusion imaging. 3) Number of metastatic LNs retrieved via TAS and ALND. 4) FNR of TAS (i.e., cases with metastases found only in ALND among those with any positive nodes). Target Sample Size: Assuming a 95% expected rate and an 83% threshold, a one-sided test with 2.5% significance and 80% power requires 58 patients. Allowing for a 5% dropout rate, the target enrollment is 62 patients.
Presentation numberPS5-07-06
BREAKER-101: a phase 1a/1b open-label study evaluating the safety, tolerability, pharmacokinetics, and efficacy of BBO-10203 in patients with advanced solid tumors
Andreas Varkaris, Massachusetts General Hospital, Boston, MA
A. Varkaris1, M. Lipsyc-Sharf2, M. Barve3, M. Oprychal4, A. Giordano5, E. Hamilton6, J. Rodon7, K. Jhaveri8, V. Kaklamani9, C. Lemech10, L. Wu11, R. M. Shah12, J. Rhee12, Y. Ben12, S. J. Luen13; 1Medical Oncology, Massachusetts General Hospital, Boston, MA, 2Medical Oncology, UCLA Health, Burbank, CA, 3Medical Oncology, Sarah Cannon Research Institute at Mary Crowley, Dallas, TX, 4Medical Oncology, Indiana University, Bloomington, IN, 5Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 6Medical Oncology, Sarah Cannon Research Institute, Nashville, TN, 7Medical Oncology, MD Anderson Cancer Center, Houston, TX, 8Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 9Medical Oncology, University of Texas Health Science Center, Houston, TX, 10Medical Oncology, Scientia Clinical Research, New South Wales, AUSTRALIA, 11Medical Oncology, Columbia University Irving Medical Center, New York, NY, 12Medical Oncology, BBOT, San Francisco, CA, 13Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA.
Background: Activation of phosphatidylinositol 3-kinase alpha-protein kinase B (PI3Kα/AKT) signaling induced by canonical-upstream RTK or RAS interation has been implicated in resistance to hormonal and HER2-directed targeted therapies in advanced breast cancer (aBC). Although commercially available orthosteric PI3Kα inhibitors can suppress pathway activation, their clinical utility as monotherapy and in combination regimens has been hindered by a narrow therapeutic window due to on-target PI3Kα toxicities such as hyperglycemia. Previous studies have established that rat sarcoma (RAS)-mediated activation of PI3Kα plays a critical role in promoting oncogenic signaling while preserving normal cellular functions such as glucose metabolism. BBO-10203 is a novel, first-in-class, covalent small molecule with blood-brain barrier penetration, designed to break the PI3Kα:RAS interaction and inhibit RAS-mediated activation of the PI3K-AKT pathway while preserving the normal catalytic function of PI3Ka involved in cell growth, metabolism, and glucose homeostasis. In preclinical models of HER2-positive (HER2+) aBC, BBO-10203 demonstrated potent antitumor activity as monotherapy and exhibited strong synergy when combined with HER2-directed therapies. Similarly, BBO-10203 showed synergy with hormonal and CDK4/6 directed therapies in hormone receptor-positive, HER2-negative (HR+/HER2-) aBC models. Notably, treatment with BBO-10203 did not result in hyperglycemia or other toxicities typically associated with PI3Kα kinase inhibition. These findings support the further development and clinical evaluation of BBO-10203 as a therapeutic strategy for HR+/HER2- and HER2+ aBC. Methods: BREAKER-101 (NCT06625775) is a first-in-human, multicenter, open label Phase 1a/1b study. During Phase 1a dose escalation, this study will primarily evaluate the safety and tolerability of BBO-10203 as monotherapy in patients with HER2+ aBC, HR+/HER2- aBC, KRAS-mutant advanced non-small cell lung cancer (aNSCLC) and KRAS-mutant advanced colorectal cancer (aCRC). Secondary objectives are to characterize the pharmacokinetics (PK) of BBO-10203 and to evaluate preliminary antitumor activity by RECIST v1.1 including objective response, clinical benefit rate (CBR), progression-free survival (PFS), and overall survival (OS). At recommended dose(s) for expansion, additional safety, PK, and antitumor activity will be evaluated for BBO-10203 monotherapy and in combination with trastuzumab in HER2+ aBC, as well as in combination with fulvestrant ± ribociclib in HR+/HER2- aBC, and with FOLFOX + bevacizumab in aCRC. As an exploratory endpoint, intracranial activity of BBO-10203 will be evaluated using Response Assessment in Neuro-Oncology Brain Metastases (RANO-BM) criteria. Patients must have previously received standard of care therapy to be eligible for the study and prior treatment with a PI3Kα or AKT inhibitor is permitted during dose escalation. BBO-10203 will be taken orally, once daily, in a 21-day or 28-day treatment cycle depending on the cohort. This study is currently open for enrollment.
Presentation numberPS5-07-07
Toward optimization of treatment strategies for early breast cancer: a multicenter observational study integrating whole genome and transcriptome sequencing within the National Whole Genome Project in Japan (WJOG16822B)
Jun Masuda, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
J. Masuda1, Y. Ozaki1, M. Kubo2, Y. Takano3, N. Tomioka4, K. Matsumoto5, H. Bando6, N. Hayashi7, T. Iwasa8, M. Takahashi9, N. Kawaguchi-Sakita10, A. Fushimi11, T. Mukohara12, M. Futamura13, M. Doi14, M. Miyashita15, M. Baba16, M. Tsukabe17, N. Yamashita1, K. Nonogaki1, S. Amino18, M. Yamashita19, E. Habano20, M. Akiya21, M. Takamatsu21, T. Osako21, A. Ueki20, K. Takeuchi21, N. Tanaka18, S. Mori18, I. Fukada22, S. Takahashi22, S. Morita23, S. Imoto24, T. Ueno1, T. Takano1, S. Kitano19; 1Breast Oncology Center, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 2Department of Breast Surgical Oncology, Kyushu University Hospitall, Kyushu University, Fukuoka, JAPAN, 3Department of Breast and Endocrine Surgery, Nagoya University Hospital, Nagoya, JAPAN, 4Department of Breast Surgery, National Hospital Organization Hokkaido Cancer Center, Sapporo, JAPAN, 5Department of Medical Oncology, Hyogo Cancer Center, Akashi, JAPAN, 6Department of Breast and Endocrine Surgery, Institute of Medicine, University of Tsukuba, Tsukuba, JAPAN, 7Division of Breast Surgical Oncology, Department of Surgery, Showa Medical University, Tokyo, JAPAN, 8Department of Medical Oncology, Kindai University Faculty of Medicine, Osakasayama, JAPAN, 9Department of Breast Surgery, Hokkaido University Hospital, Sapporo, JAPAN, 10Department of Clinical Oncology, Kyoto University Hospital, Kyoto, JAPAN, 11Department of Surgery, The Jikei University School of Medicine, Tokyo, JAPAN, 12Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, JAPAN, 13Department of Breast Surgery, Gifu University Hospital, Gifu, JAPAN, 14Department of Genomic Medicine, Hiroshima Prefectural Hospital, Hiroshima, JAPAN, 15Department of Breast and Endocrine Surgical Oncology, Tohoku University School of Medicine, Sendai, JAPAN, 16Department of Breast Surgery, Sunagawa City Medical Center, Sunagawa, JAPAN, 17Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Osaka, JAPAN, 18Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, JAPAN, 19Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 20Clinical Genetic Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, JAPAN, 21Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, JAPAN, 22Department of Genomic Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 23Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, JAPAN, 24Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, JAPAN.
Background: Despite curative-intent treatment, approximately 30% of patients with operable breast cancer experience recurrence. In patients with early-stage triple-negative breast cancer (TNBC), the phase 3 KEYNOTE-522 trial showed significant improvement in overall survival (OS) with the addition of pembrolizumab to neoadjuvant chemotherapy. However, the prognosis of patients who do not achieve a pathologic complete response (pCR) remains poor, highlighting the need for novel therapeutic strategies. While most patients with hormone receptor-positive (HR+), HER2-negative (HER2-) early breast cancer do not experience recurrence under current standard therapies, a substantial proportion−particularly those with high-risk clinical or pathological features−are at risk of developing distant metastases, often within the first few years after treatment. Optimized treatment strategies are needed to reduce early relapse and prevent metastatic progression in this high-risk population. Study Design: This is a multicenter, prospective observational cohort study embedded within the National Whole Genome Project in Japan, conducted at 17 sites of the West Japan Oncology Group (WJOG). Tumor samples are collected prior to neoadjuvant chemotherapy and at the time of surgery. Blood samples are collected at baseline and prior to surgery. Tumor specimens and matched blood samples undergo whole genome sequencing (WGS) and RNA sequencing. Multiplex immunohistochemistry is performed to evaluate immune cell infiltration such as CD4+/CD8+ T cells, regulatory T cells, B cells and M2 macrophages, as well as the expression of immune markers including PD-L1, PD-L2, granzyme B. Peripheral blood mononuclear cells are analyzed for immune cell subset profiling and functional status. Circulating tumor DNA is analyzed for genomic profiling and detection of minimal residual disease. The results of WGS are reviewed by an institutional Molecular Tumor Board and returned to participants in a clinical report format. Eligibility Criteria: Key eligibility criteria includes age ≥18, ECOG PS 0-2, stage II-III TNBC or HR+, HER2- breast cancer, and a planned treatment with anthracycline- and taxane-based neoadjuvant chemotherapy, with or without pembrolizumab. Objectives: The primary objective is to identify genomic and immunologic biomarkers associated with pCR following neoadjuvant chemotherapy. Secondary objectives include identification of biomarkers associated with survival outcomes including invasive disease-free survival, breast cancer-specific survival, OS and treatment-related adverse events. Additionally, the study aims to identify novel treatment targets for patients who do not achieve pCR. Present and Target Accrual: Target accrual is 240 patients (TNBC cohort: 100; HR+/HER2- cohort: 140) over 2 years. Enrollment began in December 2023, and 150 patients have been enrolled at the time of submission. Clinical trial information: jRCT1030250152
Presentation numberPS5-07-08
CDK4/6 inhibitor (CDK4/6i) dosing knowledge (CDK) study: A pragmatic randomized trial of indicated vs. titrated CDK4/6i dosing in older adults with HR+/HER2- metastatic breast cancer (MBC)
Erica L Mayer, Dana-Farber Cancer Institute, Boston, MA
A. Gregory1, E. L. Mayer2, D. Hershman3, J. Cowden4, R. D. Harvey5, K. Kalinsky6, A. Magnuson7, P. Manohar8, T. Pollastro9, M. Sedrak10, P. A. Spears11, R. Thota12, D. A. Walker13, A. Boose1, E. Garrett-Mayer1, G. Grantham1, D. Hinshaw1, C. MacInnis1, P. Mangat14, J. Perez1, J. Gralow1; 1CENTRA, American Society of Clinical Oncology, ALEXANDRIA, VA, 2Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 3Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, 4Patient Advocacy, Patient-Centered Dosing Initiative, Brandenton, FL, 5Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 6Winship Cancer Institute, Emory University, School of Medicine, Atlanta, GA, 7Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 8Breast Oncology, Rush Medicine, Chicago, IL, 9Patient Advocacy, UW Medicine, Mercer Island, WA, 10Department of Medicine, University of California Los Angeles, Los Angeles, CA, 11UNC Patient Advocates for Research Council, University of North Carolina, Lineberger Comprehensive Cancer Center, Raleigh, NC, 12Hem/Onc, Intermountain Health, Salt Lake City, UT, 13Board Leadership, Young Survival Coalition, New York, NY, 14CENTRA, American Society of Clinical Oncology, Alexandria, VA.
Background Many cancer drugs have been FDA approved based on a drug development paradigm using a maximum tolerated dose (MTD) approach. For targeted anticancer agents, including CDK4/6is, the MTD approach is less relevant, as the optimal biological dose may be lower than the MTD. CDK4/6is are commonly combined with endocrine therapy (ET) for treatment (tx) of HR+/HER2- MBC. Clinical trials and real-world data show that CDK4/6is are commonly dose-reduced to manage side effects and improve tolerability, without loss of efficacy. Benefits of CDK4/6i therapy are similar in younger and older patients (pts); however, toxicity can be worse in older pts leading to more dose reductions, tx interruptions, and discontinuations. We hypothesize that titrated dosing (i.e., initiating tx at lower doses and escalating as tolerated) may enable older pts to remain on tx longer with fewer adverse events (AEs). The CDK Study (NCT06377852), a multicenter, pragmatic, randomized trial, was developed to determine if titrated dosing of palbociclib (P) or ribociclib (R) in pts aged ≥65 years with HR+/HER2- MBC would allow pts to stay on tx longer than the standard FDA-approved indicated dosing approach. This study is funded through a Patient-Centered Outcomes Research Institute (PCORI) Award (BPS-2022C3-26451). Trial Design For CDK4/6i dosing, pts are randomized 1:1 to: Arm 1 (Indicated): FDA-approved dose (P 125 mg or R 600 mg), or Arm 2 (Titrated): provider-choice lower starting dose (P 75 or 100 mg, or R 200 or 400 mg) with escalation to full dose as tolerated. All pts receive CDK4/6i (P or R) for 21 days in a 28-day cycle plus ET (aromatase inhibitor (AI) or fulvestrant (F)), agent selection is by physician choice. Subsequent dose reduction and discontinuation in both arms is per standard-of-care management. Main Eligibility Criteria Eligible pts are ≥65 years old with HR+/HER2- MBC, adequate organ function, and planned ET initiation in combination with first use of P or R in the metastatic setting. Specific Aims The primary aim is to compare time to discontinuation (TTD), on the indicated dosing arm vs. the titrated dosing arm. Secondary aims include evaluation of progression-free survival, overall survival, toxicity, dose intensity, quality of life (PROMIS-29 and FACT-G, AEs, health care utilization), and associations of baseline factors with outcomes. Statistical Methods Randomization is stratified by CDK4/6i (P vs. R), age (65-74 vs. >75 yrs), and ET (AI vs. F). The primary analysis will estimate the hazard ratio (HR) comparing TTD in the two arms using a stratified Cox proportional hazards model. The null hypothesis will be rejected if the 2-sided p-value for the HR is <0.05. A sample size of 500 was based on 80% power to detect a HR of 0.75. Present Accrual and Target Accrual As of July 7, 2025, 30 pts of 500 total pts have enrolled at 13 US clinical centers (80+ locations). Final analysis will occur either after 379 discontinuation events or 24 months after last enrollment. Conclusion The CDK Study will generate evidence to help oncologists and pts make decisions about the optimal dose and dosing strategy for CDK4/6is in older pts. Trial progress demonstrates the feasibility of trials evaluating alternative dosing strategies for FDA-approved anticancer regimens in older adults. Acknowledgements The authors thank Anne Loeser and Bridgette Hempstead, dedicated pt advocates, for their leadership in developing the CDK Study. The statements in this publication are solely the responsibility of the authors and do not necessarily represent the views of PCORI, its Board of Governors or Methodology Committee. Contact Information For more information about the CDK Study (NCT06377852), please contact CDKstudy@asco.org or visit www.cdkstudy.org.
Presentation numberPS5-07-09
A randomized clinical trial comparing ctDNA-directed therapy change with standard of care in patients with metastatic triple negative breast cancer (mTNBC)
Jessica Mezzanotte Sharpe, Vanderbilt University Medical Center, Nashville, TN
J. M. Sharpe, C. Brothers, B. H. Park, V. G. Abramson; Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN.
Background: Despite treatment advances in mTNBC, most patients presenting for first-line therapy will require cytotoxic chemotherapy (CT). Sacituzumab govitecan (SG) is an antibody-drug conjugate to the Trop-2 antigen that is expressed in the majority of TNBCs. SG is approved as second- or third-line therapy for patients with mTNBC per the phase III ASCENT trial, but its utility as a first-line treatment in PD-L1 negative patients is unknown. ctDNA correlates with cancer burden and response to local and systemic therapies, but there is not an approved use for quantitatively monitoring patients’ ctDNA levels during treatment. Previous studies showed a direct correlation between a patient’s ctDNA mean variant allele frequency (VAF) and tumor burden as well as response or resistance to treatment. We hypothesize that a lack of a significant decrease in ctDNA VAFs shortly after initiating a first-line CT for mTNBC could serve as a biomarker of de novo resistance and could warrant a change of therapy earlier than conventional imaging studies would normally dictate. This study evaluates switching to SG in PD-L1 negative mTNBC patients without a 50% decrease in ctDNA VAFs less than one month after starting a first-line CT with the goal of improving progression-free survival (PFS) and overall survival (OS) and of decreasing toxicity by stopping an ineffective treatment earlier. Methods: In this two-arm randomized clinical trial, 120 patients will receive standard of care (SOC) physician’s choice CT. Patients will be randomized 1:1. One group will undergo SOC treatment with conventional staging imaging to guide treatment decisions, while the other group will undergo ctDNA evaluation on cycle (C) 1 days (D) 1 and 15 of treatment. If the mean ctDNA VAF on-treatment does not decrease by 50% on C1D15, patients will change SOC CT to SG. Both groups will undergo radiological assessment every 9 weeks. ctDNA will be banked on C4D1, C7D1, and at the end of treatment. Patients with a biopsy-confirmed new diagnosis of mTNBC that is PD-L1 negative (CPS <10) or who are otherwise not eligible for immune checkpoint inhibitor therapy are eligible for this study. Patients will not have received prior treatment for mTNBC and must have an ECOG PS of 0-2 as well as measurable disease by RECIST. The primary endpoint is PFS. This study has 80% power to detect a 2-month difference in PFS between patients treated with a ctDNA-guided therapeutic approach compared to patients assessed by conventional imaging alone (one-sided type 1 error of 10%). Key secondary endpoints include evaluating the overall response rate (RECIST v1.1), PFS2, and OS. Correlative studies include assessing ctDNA-defined clonal mutation profiles and their predictive value and correlating changes in ctDNA with standard imaging. This study (NCT05770531) is open at Vanderbilt University Medical Center and will open at 7 other sites through the ACCRU network. Ten patients have been enrolled as of 6/2025.
Presentation numberPS5-07-10
Young HOPE/JCOG2402: A Randomized Phase III Study of Response-Guided Therapy Following Neoadjuvant Endocrine Therapy to Optimize Adjuvant Treatment in Premenopausal HR+/HER2- Breast Cancer
Makiko Ono, The Cancer Institue Hospital of JFCR, Tokyo, Japan
M. Ono1, Y. Sagara2, Y. Kikawa3, M. Kawamura4, I. Nishibuchi5, M. Yoshida6, Y. Koi7, N. Uehiro8, T. Tsukioki9, Y. Tanabe10, Y. Naito11, H. Tsuda12, N. Mitome13, K. Sasaki13, T. Shibata13, F. Hara14, T. Fujisawa15, H. Iwata16, T. Shien9; 1Department of Medical Oncology, The Cancer Institue Hospital of JFCR, Tokyo, JAPAN, 2Department of Breast Surgery, Sagara Hospital, Kagoshima, JAPAN, 3Department of Breast Surgery, Kansai Medical University Hospital, Osaka, JAPAN, 4Department of Radiology, Nagoya University Hospital, Nagoya, JAPAN, 5Department of Radiology, Hiroshima University Hospital, Hiroshima, JAPAN, 6Department of Pathology, National Cancer Center Hospital, Tokyo, JAPAN, 7Department of Breast Oncology, Kyushu Cancer Center, Fukuoka, JAPAN, 8Department of Breast Surgery, The Cancer Institue Hospital of JFCR, Tokyo, JAPAN, 9Department of Breast and Endocrine Surgery, Okayama University Hospital, Okayama, JAPAN, 10Department of Medical Oncology, Toranomon Hospital, Tokyo, JAPAN, 11Department of Medical Oncology, National Cancer Center Hospital East, Chiba, JAPAN, 12Department of Pathology, Chiba Medical Center, Chiba, JAPAN, 13JCOG Data Center/Operations Office, National Cancer Center Hospital, Tokyo, JAPAN, 14Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, JAPAN, 15Department of Breast Oncology, Gunma Prefectural Cancer Center, Gunma, JAPAN, 16Department of Advanced Clinical Research and Development, Nagoya City University, Graduate School of Medical Sciences, Nagoya, JAPAN.
Background:In Asia, the incidence of breast cancer (BC) is increasing, with premenopausal patients accounting for 30-40% of all cases. The TAILORx and RxPONDER trials demonstrated that premenopausal women with hormone receptor (HR)-positive/HER2-negative BC and intermediate risk (defined as node-negative with high clinical risk and Oncotype DX Recurrence Score [RS] of 16-20, node-negative with RS 21-25, or node-positive with RS ≤25) derive benefit from adding chemotherapy (CT) to endocrine therapy (ET). The ADAPT trial suggested that patients achieving endocrine response (Ki-67≤10%) after neoadjuvant endocrine therapy (NET) had excellent outcomes without CT, regardless of menopausal status. Thus, ET response may serve as a biomarker to identify premenopausal patients who could safely omit CT. We hypothesize that premenopausal HR+/HER2- BC patients with intermediate risk (pN0 with RS16-25 or pN1 with RS ≤25) who achieve an ET response after NET can safely omit CT. The Young HOPE/JCOG2402 study is a randomized, phase III, non-inferiority trial.Methods:Patients are randomized (1:1) to either an upfront surgery arm or a NET arm, stratified by clinical nodal status (cN0/cN1), histologic grade (HG 1-2 vs. 3), and institution. Key eligibility criteria include: 1) ECOG PS 0-1; 2) premenopausal status with spontaneous menstruation within 12 months; 3) ER and/or PgR-positive and HER2-negative BC with ≥10% ER expression; 4) cN0 with clinical high-risk tumor (HG 1 with 3 cm <T≤5 cm, HG 2 with 2 cm <T≤5 cm, or HG 1 with 1 cm <T≤5 cm) or cN1 with cT <5 cm and HG 1-2; and 5) adequate organ function. In the NET arm, patients receive NET for 3 months followed by surgery. Those with pN0 with RS 16-25 or pN1 with RS ≤25 achieving an ET response (Ki-67 ≤10%) will omit CT. In the upfront surgery arm, CT is administered for pN0 with RS ≥16 or pN+ disease. ET consists of an aromatase inhibitor (AI) plus ovarian function suppression (OFS), with tamoxifen permitted per investigator discretion. Radiotherapy will be administered per investigator discretion per protocol guidelines. The primary endpoint is event-free survival (EFS). Secondary endpoints include overall survival, relapse-free survival, distant relapse-free survival, patient-reported outcomes (EORTC QLQ-C30, QLQ-BR42), ET-alone proportion, and ET response rate. Based on prior data (TAILORx, RxPONDER, ADAPT), 5-year EFS in the upfront surgery arm is estimated at 92%. The non-inferiority margin is 3.4% (HR 1.45) with one-sided α=0.05 and 70% power, requiring enrollment of 928 patients. Target accrual is 950 patients over 5 years and 7 years follow-up. Translational research will be conducted alongside this trial. Patient accrual is scheduled to begin in 2025.
Presentation numberPS5-07-11
A phase 1 study of LY4257496, a novel GRPR-targeted radioligand therapy, in patients with GRPR-positive metastatic ER+ breast cancer and other advanced solid tumors – OMNIRAY (Trial in Progress)
Komal Jhaveri, Memorial Sloan Kettering Cancer Center, New York, NY
K. Jhaveri1, L. Bodei2, P. Bedard3, K. Jerzak4, D. Juric5, J. O’Shaughnessy6, E. Mayer7, N. Harbeck8, H. Rugo9, A. Bardia10, P. Veit-Haibach11, P. Heidari12, V. Prasad13, K. Herrmann14, C. Trieu15, F. Almaguel16; 1Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 3Division of Medical Oncology, Princess Margaret Hospital, University of Toronto, Toronto, ON, CANADA, 4Medical Oncology, Sunnybrook Health Sciences Centre, North York, ON, 5Department of Medicine, Termeer Center for Targeted Therapies, Boston, MA, 6Medical Oncology, Texas Oncology, Dallas, TX, 7Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 8Breast Center, Dept. OB&GYN and CCC, Munich, LMU University Hospital, Munich, GERMANY, 9Medical Oncology, City of Hope, Duarte, CA, 10Medical Oncology, University of California Los Angeles, Santa Monica, CA, 11Joint Department of Medical Imaging, University Health Network, Toronto, ON, CANADA, 12Department of Radiology, Massachusetts General Hospital, Boston, MA, US, Boston, MA, 13Division of Nuclear Medicine, Washington University in St. Louis, St. Louis, MO, 14Department of Nuclear Medicine, University Hospital Essen, Essen, GERMANY, 15Clinical Development, Eli Lilly & Company, Indianapolis, IN, 16Medical Oncology, Biogenix Molecular, Miami, FL.
Background: Gastrin-releasing peptide receptor (GRPR) is a G protein-coupled receptor overexpressed in estrogen receptor-positive (ER+) breast cancers (BC) and other solid tumors.1 LY4257496 is a novel GRPR antagonist with improved in vivo stability, radiolabeled with lutetium-177, enabling targeted delivery of beta-radiation to GRPR-overexpressing cancer cells leading to DNA damage and cell death. The OMNIRAY phase 1 trial is evaluating LY4257496 as monotherapy in patients (pts) with GRPR-positive advanced solid tumors and in combination with endocrine therapy (ET), CDK4/6 inhibitor (CDK4/6i) therapy, or cytotoxic chemotherapy in pts with GRPR positive, ER+, HER2- metastatic breast cancer (MBC). Trial Design: This is a global, first-in-human phase 1a/b trial of LY4257496 in pts with selected GRPR-positive advanced solid tumors, including dose escalation of LY4257496 monotherapy followed by dose expansion of LY4257496 alone and in combination with other anti-cancer therapies. Dose escalation will enroll pts with ER+ MBC (HER2- or HER2+), colorectal cancer (CRC), metastatic castration-resistant prostate cancer (mCRPC), and endometrial cancer in cohort A1 and pts with ER+, HER2- MBC in the randomized dose optimization cohort A2. Dose expansion will enroll pts with ER+, HER2- MBC treated with LY4257496 monotherapy (cohort B1) and in combination with ET (cohort B2), capecitabine (cohort B3), or ET + abemaciclib (cohort B4). Additional monotherapy cohorts will include CRC (cohort C) and other GRPR-positive solid tumors (cohort D). Monotherapy dose escalation will be evaluated using a modified toxicity probability interval-2 (mTPI-2) design. In dose expansion, each combination cohort will include a safety lead-in of 3-6 pts. LY4257496 will be administered intravenously on an every 4 to 6 weeks treatment cycle for up to 6 cycles. Split dosing (treating on D1 and D8 of each cycle) will also be explored. Eligibility criteria: Eligible pts must have a locally advanced, unresectable, or metastatic solid tumor that is GRPR-positive on screening nuclear medicine GRPR imaging (SPECT/CT or PET/CT) as assessed by the treating investigator based on imaging interpretation manual. In dose escalation, pts with ER+ MBC may have received up to 5 prior systemic treatment regimens. In dose expansion, prior therapy requirements are outlined in the Table below. Key exclusions include prior radiopharmaceutical (except 177Lu-PSMA-617 for mCRPC), recent pancreatitis, and untreated central nervous system metastases. Key study objectives: Evaluate safety, antitumor activity, optimal dose, PK, and biodistribution and dosimetry of LY4257496. Table
| Patient Subgroup | Key Eligibility | Study Drugs (Cohort) | |||
| MBC: ER+, HER2- |
|
|
|||
|
|
||||
| CRC |
|
|
|||
| Other GRPR+ solid tumors |
|
|
ETa, fulvestrant or imlunestrant; ETb, investigators’ choice of aromatase inhibitor, fulvestrant, or imlunestrant 1Stoykow C, et al. Theranostics 2016, 6:1641-50
Presentation numberPS5-07-12
Opt-pembro: a phase III randomized trial of adjuvant pembrolizumab omission in patients with early-stage triple-negative breast cancer achieving pathologic complete response after neoadjuvant chemo-immunotherapy
Joana Mourato Ribeiro, Institute Gustave Roussy, Paris, France
J. M. Ribeiro1, F. Duhoux2, E. Romano3, B. Verret1, F. Dalenc4, E. Azambuja5, S. Ladoire6, A. Rorive7, A. Mailliez8, P. Bénédicte9, C. Lefeuvre10, D. Taylor11, A. Gonçalves12, K. Punie13, M. Mouret-Reynier14, C. Fontaine15, C. Bailleux16, E. Naert17, P. Corbaux18, F. Derouane19, M. Leheurteur20, E. Jacquet21, M. Robert22, A. Lemoine23, T. Robinson24, R. Salgado25, M. Van Bockstaele26, M. Lambertini27, S. Tillaux28, T. Sara29, A. Partridge29, L. Carrey30, K. Pilger31, J. Lemonier32, S. Mardinian33, S. Mijonnet33, F. Clement-Bidard34, T. Bachelot35, V. Ines36, S. Michiels28, T. Olivier35; 1Breast Unit, Medical Oncology Department; IHU-National PRecISion Medicine Center in Oncology, Institute Gustave Roussy, Paris, FRANCE, 2Breast Unit, Medical Oncology Department; IHU-National PRecISion Medicine Center in Oncology, Institute Gustave Roussy, Brussels, BELGIUM, 3Department of Medical Oncology, Center for Cancer Immunotherapy, Institut Curie, Paris, France, Institut Curie, Paris, FRANCE, 4Medical Oncology, Institut universitaire du cancer Toulouse-Oncopole, Toulouse, FRANCE, 5Medical Oncology Department, Institut Jules Bordet and L’Université Libre de Bruxelles (U.L.B.), Brussels, BELGIUM, 6Medical Oncology Department, Centre Georges-François Leclerc, Dijon, FRANCE, 7Medical Oncology Department, CHU Sart Tilman Liege,, Liege, BELGIUM, 8Medical Oncology Department, Center Oscar Lambret, Lille, FRANCE, 9Medical Oncology Department, Chu Helora Hospital La Louvière Site Jolimont, Haine Saint Paul, BELGIUM, 10Medical Oncology Department, Centre Eugène Marquis, Rennes, FRANCE, 11Medical Oncology Department, CHU Namur Ste Elisabeth, Namur, BELGIUM, 12Medical Oncology Department, Institut Paoli-Calmette, Marseille, FRANCE, 13Department of Medical Oncology, ZAS Hospitals, Antwerp, FRANCE, 14Medical Oncology Department, Centre Jean Perrin, Clermont-Ferrand, FRANCE, 15Department of Medical Oncology, Academisch Ziekenhuis, Vrije Universiteit Brussel, Brussels, BELGIUM, 16Medical Oncology Department, Centre Antoine Lacassagne, Nice, FRANCE, 17Department of Medical Oncology, Ghent University Hospital, Ghent, BELGIUM, 18Medical Oncology Department, Institut de Cancérologie et d’Hématologie Universitaire de Saint-Étienne (ICHUSE), Centre Hospitalier Universitaire de Saint-Etienne, Saint-Priest-en-Jarez, FRANCE, 19Medical Oncology Department, UZ Leuven, Leuven, BELGIUM, 20Medical Oncology Department, Centre Henri Becquerel, Rouen, FRANCE, 21Medical Oncology Department, University of Franche Comte, FEMTO-ST Institute, CNRS, Besançon, FRANCE, 22Medical Oncology Department, Institut de Cancérologie de l’Ouest (ICO), Saint-Herblain, FRANCE, 23Medical Oncology Department, Institut Godinot, Paris, FRANCE, 24Medical Oncology Department, Bristol Haematology and Oncology Centre, Bristol, UNITED KINGDOM, 25Department of Pathology, GZA-ZNA Hospitals; Division of Research, Peter Mac Callum Cancer Centre, Melbourne, Australia, Antwerp, BELGIUM, 26Pathology Department, University Clinics St-Luc, Brussels, BELGIUM, 27Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genoa, ITALY, 28Service de Biostatistique et d’Epidémiologie, Oncostat Inserm U1018, Université Paris-Saclay, Equipe, Institute Gustave Roussy, Paris, FRANCE, 29Division of Breast Oncology, Dana-Farber Cancer Institute, Boston, MA, 30Medical Oncology, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 31Clinical Trial Center, Cliniques universitaires Saint-Luc, Brussels, BELGIUM, 32Unicancer Research Governance, UNICANCER, Paris, FRANCE, 33UCBG – French Breast Cancer Intergroup, UNICANCER, Paris, FRANCE, 34Department of Medical Oncology, Institut Curie, Paris, FRANCE, 35Medical Oncology Department, Centre Léon Bérard, Lyon, FRANCE, 36Cancer Survivorship Group, INSERM Unit 981, Molecular Predictors and New Targets in Oncology, Institute Gustave Roussy, Paris, FRANCE.
Background: In early-stage triple-negative breast cancer (eTNBC), the addition of pembrolizumab to neoadjuvant chemotherapy followed by continued adjuvant immunotherapy significantly improved pathological complete response (pCR), event-free and overall survival (OS) in the KEYNOTE-522 trial. However, the continuation of pembrolizumab post-surgery in all patients, irrespective of response, raises concerns of overtreatment, toxicity, and cost. This is particularly evident in patients achieving pCR, who exhibit excellent long-term outcomes. The OPT-PEMBRO trial addresses whether adjuvant pembrolizumab can be safely omitted in this favourable-risk subgroup. Trial Design: OPT-PEMBRO is an international, multicenter, randomized, open-label, phase III non-inferiority trial sponsored by UNICANCER. It will enroll 2454 patients with stage T1cN1-2 or T2-4N0-2 TNBC who achieve pCR (ypT0 ypN0) following standard neoadjuvant chemotherapy plus pembrolizumab. Patients are randomized 1:1 within 12 weeks post-surgery to either continue standard adjuvant pembrolizumab (6 months) or observation alone. The primary endpoint is recurrence-free survival (RFS). Secondary endpoints include invasive breast cancer-free survival (iBCFS), distant relapse-free survival (DRFS), OS, safety, quality of life (EORTC QLQ-C30, BR42, EQ-5D-5L), immune-related adverse events (irAEs), patient-reported outcomes (PRO-CTCAE), and fertility impact. Translational objectives include assessment of baseline stromal TILs and their interaction with outcomes. Patient Population: Eligible patients are adults (≥18 years) with histologically confirmed stage T1cN1-2 or T2-4N0-2 TNBC (ER/PR ≤10%, HER2-negative), treated with ≥6 cycles of neoadjuvant chemotherapy plus pembrolizumab and achieving pCR. Additional criteria include ECOG 0-2, adequate organ function, no prior invasive breast cancer, and no evidence of residual disease or distant metastasis. Statistical Considerations: OPT-PEMBRO is powered to demonstrate non-inferiority of observation versus standard adjuvant pembrolizumab in terms of RFS, using a non-inferiority margin of 2.5% (HR 1.44). Interim analyses for futility are planned. A prospective meta-analysis with the U.S.-based OptimICE-pCR trial is also anticipated. Current Status: Trial enrolment is ongoing across centres in France and Belgium. A total of 2454 patients will be enrolled over 4 years, with an additional 4 years of follow-up. The final analysis is event-driven and expected after 285 RFS events. Long-term follow-up will extend to 14.5 years to assess OS, late toxicity, and survivorship. Conclusion: OPT-PEMBRO is an international randomized trial to test the non-inferiority of omitting adjuvant immunotherapy in patients with eTNBC presenting pCR following standard neoadjuvant chemotherapy plus pembrolizumab. By targeting a good-prognosis population, it aims to personalize immunotherapy duration, reduce toxicity, preserve quality of life, and lower healthcare costs without compromising outcomes.
Presentation numberPS5-07-13
Impact of pegulicianine on radiotherapy decisions and long-term outcomes in patients undergoing breast conserving surgery
Jorge Ferrer, Lumicell, Inc., Newton, MA
K. Smith1, J. Ferrer2, B. Schlossberg3, S. F. Shaitelman4, G. Doros5, B. L. Smith6; 1Clinical Affairs, Lumicell, Inc., Newton, MA, 2Research and Development, Lumicell, Inc., Newton, MA, 3Product Development, Lumicell, Inc., Newton, MA, 4Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 5Biostatistics, Boston University School of Public Health, Boston, MA, 6Medicine, Massachusetts General Hospital, Newton, MA.
Introduction: Studies have shown that local recurrences typically occur near the site of the primary cancer. Additionally, interest in personalized radiotherapy after breast conserving surgery (BCS) continues to grow, with de-escalation or omission pursued when oncologically appropriate. Based on current guidelines, wider margins, among other factors, can influence recommendations for de-escalation or omission. Completed trials of pegulicianine fluorescence guided surgery (pFGS) for real time residual cancer assessment within the lumpectomy during BCS showed removal of additional tumor and reduction of second surgeries compared with standard lumpectomy surgery. Specifically, in one study, pFGS guided the removal of residual cancer in 27 of 357 (7.6%) patients undergoing BCS. These previous trials enrolled a diverse cohort, including patients with invasive ductal carcinoma, invasive lobular carcinoma, and/or ductal carcinoma in situ (DCIS), with receptor profiles (HER2, ER, PR) reflective of the broader population receiving lumpectomies for breast cancer. We hypothesize that removal of residual cancer not identified during the initial BCS will reduce 5-year local recurrence rates and that pFGS-guided excisions will provide appropriate margin gains or remove residual disease, altering radiotherapy planning. 5-year follow-up data on patient outcomes and information about the adjuvant radiotherapy patients received following pFGS-guided excisions is needed to evaluate the impact of pFGS on local recurrence rates and on radiotherapy treatment decisions. Study Design and Methods: Follow-up data will be collected from patients in prior prospective feasibility and pivotal studies to assess whether removal of residual cancer impacts local recurrence rates and radiotherapy treatments. Retrospective data collection from up to 640 breast cancer patients previously enrolled in parent trials NCT03686215 and NCT03321929 will be collected and analyzed, with a goal of demonstrating the long-term benefits of pFGS in improving surgical outcomes and informing radiotherapy decisions. Data collection will be conducted through systematic retrospective chart reviews, with rigorous quality control measures to ensure accuracy and reliability. A breast cancer radiation oncologist will determine the recommended radiotherapy treatment prior to pFGS use. The actual radiotherapy received following pFGS guided excisions will then be compared with these recommendations. Finally, radiotherapy treatments in the pFGS population will be compared with data drawn from contemporaneous publications. Data on cancer recurrence and survival will be collected. For each study participant that has developed a recurrent cancer, an evaluation of the location of the recurrent cancer compared to the index cancer will be performed. The 5-year median local recurrence rates and the 95% confidence interval (CI) will be calculated using Kaplan-Meier methods for time to event endpoints. Each patient’s follow-up period will be the time from index lumpectomy to last healthcare provider encounter. Local recurrence rates from these patients will be compared with rates reported in recent peer-reviewed literature.
Presentation numberPS5-07-14
The CROWN Study (CaRdiac Outcomes With Near-complete estrogen deprivation)
Sarah Hatcher, Duke University, Durham, NC
S. Hatcher1, R. B. D’Agostino, Jr.2, N. O’Connell2, R. Bansal1, C. Anders1, S. Telloni1, K. Westbrook1, A. Natarajan1, N. J. Pagidipati3, D. Wendell4, E. Douglas5, K. Ansley5, C. J. Park6, K. M. Richardson6, S. R. Sirkisoon7, M. Hackney8, H. Vachhani8, M. Ross8, L. N. Vélez-Torres9, W. Bottinor10, W. Hundley11, A. Thomas1, J. H. Jordan12; 1Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, 2Department of Biostatistics and Data Science, Wake Forest University Health Sciences, Winston-Salem, NC, 3Division of Cardiology, Department of Medicine, Duke University, Durham, NC, 4Division of Cardiology, Cardiovascular Magnetic Resonance Center, Duke University, Durham, NC, 5Department of Clinical Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 6Department of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 7Data Coordinating Center, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 8Division of Hematology, Oncology and Palliative Care, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, 9Division of Hematology, Oncology and Palliative Care, Massey Comprehensive Cancer Center, University of Puerto Rico Comprehensive Cancer Center and Virginia Commonwealth University, Richmond, VA, 10Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, 11Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University Health Sciences, Richmond, VA, 12Pauley Heart Center, Department of Internal Medicine and Department of Biomedical Engineering, Virginia Commonwealth University Health Sciences and Virginia Commonwealth University, Richmond, VA.
Background: Treatment for premenopausal women with high- or intermediate-risk hormone receptor (HR)-positive breast cancer (BC) often includes concurrent ovarian function suppression (OFS) and aromatase inhibitors (AIs) to induce near-complete estrogen deprivation (NCED). The long-term cardiovascular (CV) consequences of NCED are unknown. In non-cancer populations, premature menopause is associated with increased CV morbidity. Given the cardiotoxicity of standard BC therapies and the extended life expectancy of these women, the CV impact of NCED warrants focused investigation. The CROWN study integrates advanced imaging to assess cardiac dysfunction alongside biomarker and demographic analyses, aiming to better understand the progression of CV injury and develop tools to assess and mitigate CV risk. Methods: CROWN is an NIH-funded, prospective cohort study conducted across three NCI-designated cancer centers: Duke University, Virginia Commonwealth University, and Wake Forest University. Eligible participants include premenopausal women aged ≤55 years with Stage I-III BC, ECOG performance status 0-2, who have completed chemotherapy, surgery, and radiation. Women with HR-positive BC receive OFS plus an AI; those with HR-negative BC are included as comparators. CV assessments occur at baseline, 1 year, and 2 years, and include cardiac magnetic resonance imaging (CMR), coronary computed tomography angiography (CCTA), and laboratory biomarkers, including exploratory biomarkers (Table). The primary objective is to assess the 24-month difference in myocardial blood flow, measured by adenosine stress CMR, between the NCED and HR-negative groups. Correlative analyses will evaluate the relationship between CMR results and CCTA-based coronary plaque characteristics. Additional analyses will explore the impact of pre-existing CV risk factors and dynamic treatment-related risk changes on CV outcomes. Primary statistical analyses will include both between-group (NCED vs. HR-negative) and within-group (longitudinal changes within the NCED group) comparisons. Secondary analyses will develop predictive models using clinical, demographic, and biomarker data to identify factors associated with CV changes. To date, we have enrolled 90 participants (73 NCED, 17 HR-negative), with a 13% attrition rate. Six patients have completed their Year 2 imaging and 23 have completed Year 1 imaging. Participant retention is essential for endpoint completion and statistical power. Retention strategies include IRB-approved newsletters and small study-branded items (magnets, sticky notes, pens) provided to participants, fostering a sense of community and engagement.
| Evaluation/Procedure | Baseline | Year 1 | Year 2 |
| Vitals | X | X | X |
| Body measurements: (Height, Weight, BMI, BSA) | X | X | X |
| Labs/Biomarkers | X | X | X |
| CMR | X | X | X |
| CCTA | X | X | |
| EKG | X | X | X |
| Patient questionnaires | X | X | X |
Presentation numberPS5-07-15
A randomized phase II study to evaluate the efficacy and safety of Trastuzumab deruxtecan (T-DXd) versus CDK4/6 inhibitor-based endocrine therapy as first-line therapy of hormone receptor-positive (HR+) and HER2-low/ultralow advanced breast cancer (ABC) patients classified as non-luminal subtype according to gene expression profiling: the PONTIAC study
Javier Cortes, IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, Spain
J. Cortes1, Á. Guerrero-Zotano2, M. Gion3, M. Campolier4, M. Verbeni5, S. Iacobucci6, A. del Pino7, S. Santasusagna8, K. Jhaveri9, A. Bardia10, C. Barrios11, S. Franco Millan12, G. Antonarelli13, S. Kümmel14, P. Cottu15, J. Pérez-García16, A. Llombart-Cussac17; 1Oncology, IOB Madrid, Institute of Oncology, Hospital Beata Maria Ana, Madrid, SPAIN, 2Oncology, Instituto Valenciano de Oncología, Valencia, SPAIN, 3Oncology, Hospital Universitario Ramón y Cajal, Madrid, SPAIN, 4Trial, Medica Scientia Innovation Research (MEDSIR) – Jersey City (New Jersey), Barcelona, SPAIN, 5Data Analytics, Medica Scientia Innovation Research (MEDSIR) – Jersey City (New Jersey), Barcelona, SPAIN, 6Oncology, Medica Scientia Innovation Research (MEDSIR) – Jersey City (New Jersey), Barcelona, SPAIN, 7Business, Medica Scientia Innovation Research (MEDSIR) – Jersey City (New Jersey), Barcelona, SPAIN, 8Scientific Impact, Medica Scientia Innovation Research (MEDSIR) – Jersey City (New Jersey), Barcelona, SPAIN, 9Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10Oncology, University of California Los Angeles, Health Jonsson Comprehensive Cancer Center, Los Angeles, CA, 11Oncology, Oncoclinicas – Latin American Cooperative Oncology Group, Porto Alegre, BRAZIL, 12Oncology, Cancer Treatment and Research Center (CTIC), Bogotá, COLOMBIA, 13Oncology and Hemato-Oncology, University of Milan, Milan, ITALY, 14Gynecology, Breast Center Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, GERMANY, 15Medical Oncology, Institut Curie, Paris, FRANCE, 16Oncology, International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, SPAIN, 17Medicine, Arnau de Vilanova, FISABIO; Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, Valencia, SPAIN.
BackgroundHR-positive and HER2-negative breast cancers exhibiting non-luminal subtypes by PAM50 gene expression profiling are associated with a more aggressive clinical behaviour and reduced sensitivity to endocrine-based therapies. Trastuzumab deruxtecan (T-DXd) is a HER2-directed antibody-drug conjugate that has demonstrated to improve clinical outcomes in endocrine-resistant patients (pts) with HR+ and HER2-low/ultralow ABC as shown in the DESTINY-Breast04 and DESTINY-Breast06 phase III trials. However, there are currently no available data supporting the use of this therapy in the frontline setting.The aim of the PONTIAC study is to evaluate the safety and efficacy profiles of T-DXd compared with endocrine therapy plus a CDK4/6 inhibitor as first-line therapy in pts with HR+ and HER2-low/ultralow ABC classified as non-luminal according to PAM50 gene expression profiling.Trial DesignPONTIAC (NCT06486883) is an international, multicenter, open label, randomized, phase II trial.Adult pts with HR+ and centrally confirmed HER2-low (IHC 1+ or 2+ with negative in situ hybridization test) or ultralow (IHC 0 with faint membrane staining in ≤ 10% of tumor cells) ABC, classified as non-luminal by central PAM50 analysis and previously untreated for advanced disease, are eligible. Moreover, pts treated with a CDK4/6 inhibitor in the adjuvant setting with a treatment-free interval ≥ 12 months following CDK4/6 inhibitor treatment completion are allowed.Pre-screening central PAM50 analysis will be conducted in endocrine-resistant pts and in endocrine-sensitive pts who meet at least one of the following criteria: estrogen receptor expression ≤ 50% or presence of liver metastases, or high histological grade or Ki67 > 50% in the primary tumor or known non-luminal subtype as per local PAM50 analysis.Pts will be randomized 1:1 to either T-DXd (5.4 mg/kg intravenously once every 3 weeks) or endocrine therapy (fulvestrant or an aromatase inhibitor ± GnRH analogs for men and pre-/perimenopausal women) plus investigator’s choice of CDK4/6 inhibitor (palbociclib, abemaciclib, or ribociclib). Stratification factors are visceral disease (yes vs. no), PAM50 intrinsic subtype (basal-like vs. others), and HER2 protein expression (HER2-low vs. HER2-ultralow). Pts will receive study treatment until disease progression, death, unacceptable toxicity, or consent withdrawal.Primary endpoints are to assess progression-free survival (PFS) in HER2-low pts and in both HER2-low and ultralow pts (all population). Key secondary endpoints include overall survival, objective response rate, clinical benefit rate, duration of response, time to response, health-related quality-of-life, and safety and toxicity.PFS will be estimated using the Kaplan-Meier method. Stratified log-rank tests at an overall two-side significance level of 0.05 will be used to assess treatment-group differences. An interim PFS analysis with the possibility of stopping the trial for futility will be carried out when 36% of the information is available. The primary PFS endpoints will be assessed by two-sided log-rank tests in HER2-low and all pts, with 80% power to detect hazard ratios (HR) of 0.526 and 0.608, respectively. Two hundred pts (100 per arm) are needed to test the hypotheses with 80% power. Secondary endpoints including ORR, CBR, DoR, and OS will be analyzed using stratified methods with appropriate confidence intervals. Pts-reported outcomes and safety will be analyzed descriptively. Target enrollment is 200 pts, and initial patient enrollment is anticipated to begin in Q3 2025.
Presentation numberPS5-07-16
Cappa, a phase 2 study to evaluate capecitabine plus pembrolizumab as post-operative adjuvant therapy for triple-negative breast cancer with residual disease after neoadjuvant chemo-immunotherapy
Delphine Loirat, Institut Curie – Institute of Women’s Cancers, Paris, France
D. Loirat1, F. C. Bidard1, J. Grenier2, T. L’Haridon3, A. Kieffer4, F. Dalenc5, C. Goislard De Monsabert6, F. Ricci7, M. By8, D. Bello Roufai1, Y. Kirova9, T. Roque10, A. Savignoni11, J. Y. Pierga1; 1Medical Oncology department, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 2Department of medical oncology, Institut du cancer Avignon Provence (ICAP), Avignon, FRANCE, 3Medical Oncology department, Clinique Mutualiste de l’Estuaire, France, Saint-Nazaire, FRANCE, 4Medical Oncology department, Institut de Cancérologie de Lorraine, Vandoeuvre-Les-Nancy, FRANCE, 5Medical Oncology department, Institut Claudius Regaud, IUCT Oncopole, Toulouse, FRANCE, 6Onco-Hematology Department, CHD Vendée, La Roche-sur-Yon, FRANCE, 7Medical Oncology department, Clinique de La Croix du Sud, Quint-Fonsegrives, FRANCE, 8Medical Oncology department, CHU Bretonneau, Tours, FRANCE, 9Department of Radiation Oncology, Institut Curie – Institute of Women’s Cancers, Paris, FRANCE, 10R&D, Unicancer, Paris, FRANCE, 11Biometrics Department, DREH, Institut Curie, Paris, FRANCE.
Background: The KEYNOTE-522 trial demonstrated the benefit of adding pembrolizumab to neoadjuvant chemotherapy in patients with stage II-III triple-negative breast cancer (TNBC), showing improvements in both pathological complete response (pCR) rates and overall survival. However, a key limitation of the KEYNOTE-522 trial is that it did not incorporate the use of post-neoadjuvant capecitabine for patients who did not achieve a pCR, despite evidence that capecitabine improves survival in this population. Although post-neoadjuvant capecitabine combined with pembrolizumab is commonly discussed, there is no prospective data confirming its efficacy and tolerance in clinical practice. Trial Design: CAPPA (NCT0597386) is a phase II, open-label, multicenter trial evaluating the addition of capecitabine to pembrolizumab as adjuvant therapy in patients with stage II-IIIb TNBC and residual disease after neoadjuvant chemo-immunotherapy. Main inclusion criteria are: (i) Histologically confirmed TNBC, defined as HER2-negative (according to ASCO/CAP criteria) and <10% of cells staining positive for ER and PR by IHC; (ii) Patients who received standard neoadjuvant chemo-immunotherapy (minimum of 6 cycles); (iii) Absence of pCR, defined as RCB class I-III. This trial includes two distinct cohorts: (i) A prospective experimental cohort (N=220), patients will receive capecitabine (1000 mg/m² BID, 14 days on and 7 days off) combined with pembrolizumab (200 mg every 3 weeks) for 6 months. Capecitabine is reduced at a dose of 825 mg/m2 BID during radiotherapy, performed as per standard practice, if indicated. (ii) An external cohort (N = 220), reflecting the treatment received in the KN-522 trial, will be enrolled in an ambispective manner and will include patients treated with pembrolizumab as part of standard adjuvant treatment, with similar eligibility criteria. The primary endpoint is the 2-year invasive disease-free survival (iDFS) rate. Secondary endpoints include distant disease-free survival (DDFS), overall survival (OS), and safety. Ancillary studies will be conducted on tumor biopsies, surgical specimens, and blood samples. The first patient was enrolled in March 2025. As of July 9, N=14 and 17 pts have been included in the experimental and external cohort, respectively. The inclusion period is expected to last 18 months. Funding: PHRC-K (grant PHRC-K22-084); Women’s Cancer Institute of Institut Curie (grant ANR-23-IAHU-0006)
Presentation numberPS5-07-17
A prospective, direct-to-patient study to evaluate clinical and molecular mechanisms of resistance to capivasertib in estrogen receptor-positive metastatic breast cancer
Jacqueline Jialin Tao, Columbia University Irving Medical Center, New York, NY
J. J. Tao1, E. Harden1, A. Johnston2, C. M. Sathe1, J. E. McGuinness1, M. S. Trivedi1, M. K. Accordino1, K. D. Crew1, D. L. Hershman1, N. Vasan3; 1Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, 2NA, Surviving Breast Cancer, Boston, MA, 3Department of Medicine, NYU Langone Health, New York, NY.
Background: The PI3K pathway is activated in approximately 40% of estrogen receptor-positive (ER+) breast cancers and is a key therapeutic target in ER+ metastatic breast cancer (MBC). PI3Kα inhibitors including alpelisib and inavolisib improve progression free survival (PFS) in PIK3CA-mutated ER+ MBC, but are often associated with high-grade toxicities such as hyperglycemia. AKT is a key downstream effector in the PI3K pathway, and the AKT inhibitor capivasertib was FDA approved in November 2023 for patients with ER+ MBC harboring mutations in PIK3CA, AKT1, and/or PTEN. In the CAPItello-291 trial, capivasertib combined with fulvestrant more than doubled PFS to 7.3 months versus 3.1 months with fulvestrant alone, and exhibited a favorable toxicity profile. However, despite initial disease responses, almost all patients eventually progress on therapy. Preclinical studies have identified several resistance mechanisms to AKT inhibitors including reactivated PI3K pathway signaling and increased compensatory signaling of parallel pathways. However, there are no validated clinical biomarkers of resistance to capivasertib. Understanding the clinical mechanisms of capivasertib resistance is critical to identifying patients who may not benefit from capivasertib and to informing subsequent therapeutic strategies. Methods: We are conducting a pragmatic, decentralized study enrolling patients treated at any medical institution who are starting capivasertib (n = 20). Eligibility criteria include 1) ER+ MBC with mutations in PIK3CA, AKT1, and/or PTEN, 2) no prior treatment with a PI3K pathway inhibitor, and 3) plans to initiate capivasertib as the next line of therapy. Participants are asked to collect circulating tumor DNA (ctDNA) at two time points: 1) before or within 2 weeks of starting capivasertib and 2) after progression on or discontinuation of capivasertib. The study uses a decentralized design in which participants are mailed liquid biopsy kits to their home and collect blood samples locally, with the goal of facilitating more rapid and inclusive enrollment. Recruitment is under way using social media channels, patient advocacy groups, and multi-institutional collaborative groups. Interim clinical data is collected from local medical records while participants remain on capivasertib. After sample collection is complete, pre- and post-treatment samples will be analyzed using whole exome and whole transcriptome sequencing via the Caris Assure platform. Paired ctDNA samples from each participant will be compared to identify acquired genomic and transcriptomic alterations on capivasertib. We hypothesize that alterations in negative regulators of mTORC1 and parallel pathways such as PIM signaling will drive clinical resistance to capivasertib. The study is approved by the Columbia University Irving Medical Center (CUIMC) Institutional Review Board and was developed in collaboration with the patient advocacy group PIK3CA Pathbreakers. Since January 2025, 10 patients have been enrolled and provided pre-treatment ctDNA samples. Further study details are available at https://contributeher.wixsite.com/capivaresistance.
Presentation numberPS5-07-18
Advancing personalized treatment of patients with metastatic breast cancer by functional drug screening and IMC profiling
Marcus Vetter, Cantonal Hospital Baselland, Liestal, Switzerland
M. Vetter1, C. Jehanno2, S. Foo2, A. Rouchon2, Y. Blum2, A. Mock2, M. Diepenbruck2, E. Bartoszek3, L. Toniato2, M. Kloc2, J. Gomez-Miragaya4, G. Boot5, S. Muenst-Soysal5, K. Mertz6, V. Kölzer6, R. Bill7, A. Ring8, B. Kasenda9, C. Zech10, A. Oseledchyk9, C. Kurzeder11, W. Weber11, B. Bodenmiller12, A. Wicki13, M. Bentires-Alj14, National Data Stream Consortium Switzerland; 1Cancer Center and Center for Oncology and Hematology, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 2University Basel, Department of Biomedicine, Basel, SWITZERLAND, 3University Hospital Basel, Department of Biomedicine, Basel, SWITZERLAND, 4Cancer Center and Center for Oncology and Hematology, Department of Biomedicine, Basel, SWITZERLAND, 5University Basel, Institute of Pathology and medical Genetics, Basel, SWITZERLAND, 6University Hospital Basel, Institute of Pathology and medical Genetics, Basel, SWITZERLAND, 7Medical Oncology, Inselspital Bern, Bern, SWITZERLAND, 8Medical Oncology, University Hospital Zürich, Zürich, SWITZERLAND, 9Medical Oncology, University Hospital Basel, Basel, SWITZERLAND, 10Radiology, University Hospital Basel, Basel, SWITZERLAND, 11Breast Center, University Hospital Basel, Basel, SWITZERLAND, 12a) Department of quantitative Biomedicine b)Institute of molecular health sciences, a) University of Zürich b)ETH Zürich, Zürich, SWITZERLAND, 13Comprehensive Cancer Center, University Hospital Zürich, Zürich, SWITZERLAND, 14Department of Biomedicine, Department of Surgery, University Hospital Basel, Basel, SWITZERLAND.
Background: Worldwide, nearly 2.6 million women are diagnosed annually with breast cancer and 685 000 lives are lost to the disease, the vast majority due to drug-resistant metastases. Although new therapies significantly increased cure rates for early-stage breast cancer, the situation for metastatic disease has hardly changed. Cutting-edge technologies may help characterize the cancer better and tailor therapy decision. Testing drugs on ex-vivo avatar models of patients’ tumors (i.e., personalized treatment) coupled with their in-depth characterization raises the hope of identifying more efficacious treatments. This should also spare the patients the side effects of ineffective treatments. Methodes: We have initiated the nation-wide Swiss Personalized Oncology National Data Stream (SPO-NDS) study that uses a variety of OMICs to inform clinical decision making for patients with advanced breast cancer. We have been using high-resolution image-based functional drug screening on patient-derived organoids (PDO). We pre-emptively tested drug’s efficacy, alone or in combination, on tissue biopsies taken upon disease progression on standard-of-care treatment (any line). Freshly isolated tumour cells were grown in 3D-culture, which retain the patient’s molecular and cellular characteristics. This has been combined with high-resolution imaging mass cytometry (IMC) which enables the analysis of dozens of markers at single-cell resolution while preserving spatial tumor architecture, and informing about tumor content, immune cell infiltration, presence of a specific target, or activation of specific pathways. The results of our studies were integrated into a molecular summary report, discussed at a Swisswide interdisciplinary molecular tumour board, yielding patient-specific cancer vulnerabilities and informing highly-personalized treatment recommendations. The future development of these technologies as a routine tool in the clinics could be of paramount importance to provide tailored therapies to the unique genomic, molecular, and clinical characteristics of each patient’s disease. Conclusion: This trial-in-progress supports the feasibility of using functional drug screening and multi-OMIC profiling to inform personalized treatment in advanced breast cancer.
Presentation numberPS5-07-19
ECOG-ACRIN tomosynthesis mammographic imaging screening trial (TMIST): Update for 2025
Etta Pisano, American College of Radiology, Philadelphia, PA
E. Pisano1, C. Gatsonis2, M. D. Schnall3, M. A. Troester4, E. Cole5, J. Cormack2, J. Steingrimsson2, I. F. Gareen6, M. Yaffe7, L. C. Collins8, A. Curtis9, R. Carlos10, K. D. Miller11, C. Comstock12; 1Center for Research and Innovation, American College of Radiology, Philadelphia, PA, 2Biostatistics, Brown University School of Public Health, Providence, RI, 3Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 4Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 5Center for Research and Innovation, Diagnostic Imaging, American College of Radiology, Philadelphia, PA, 6Center for Statistical Sciences, Brown University,, Providence, RI, 7Medical Biophysics, University of Toronto, Toronto, ON, CANADA, 8Pathology, Weill Cornell Medicine, New York, NY, 9Gibbs Cancer Center & Research Institute, Spartanburg Regional Healthcare System, Spartanburg, SC, 10Radiology, Columbia University Irving Medical Center, New York, NY, 11Medicine, Indiana University School of Medicine, Indiana, IN, 12Radiology, Weill Cornell Medicine, New York, NY.
Background: The ECOG-ACRIN Tomosynthesis Mammographic Imaging Screening Trial is a clinical trial being conducted to determine if tomosynthesis (TM) should replace digital mammography (DM) for breast cancer screening based on the impact of the use of each technology on the number of advanced breast cancers in the population of women being screened over several years. If TM is better at finding the types of cancers that are most likely to lead to mortality, the population screened with TM would be expected to have fewer of these types of cancers over time compared with the population screened with DM. TMIST also includes secondary aims in the areas of imaging assessment, medical physics, breast biology and pathology, long-term follow-up, and health care utilization. Besides collection of demographic and clinical information, the study also has a collection of TM and DM screening mammograms, blood and buccal samples, and pathology diagnostic slides and tissue blocks. Methods: Asymptomatic women ages 45 to 74 were enrolled into TMIST across 133 sites located in the United States, Canada, Argentina, Italy, Peru, Chile, South Korea, Spain, Taiwan, and Thailand. At the time of enrollment, women were randomized to undergo screening mammography with either TM or DM annually or biennially based on risk factors for their first 5 years on the study with up to 3 additional years of long-term follow-up under their routine breast screening protocols. Pathology materials for all enrolled women who undergo biopsy or surgery are being collected. Tissue blocks collected will undergo PAM-50 analysis, plus an immune signature. TMIST participants also voluntarily can contribute blood and/or buccal smears to the TMIST biorepository. Update: TMIST met the study accrual goal of 108,508 in December 2024. 21% of the United States participants are African American, with 49% Hispanic participation worldwide. 68.1% of TMIST participants have opted to provide buccal smears, and 66.5% have opted to provide blood to the biorepository. The study will complete follow-up in December 2027. Two ancillary studies not dependent on study endpoints with separate external funding are underway. The first active ancillary project is a study aimed at increasing participation of African American TMIST participants in the biospecimen sub-study of TMIST. New educational materials were developed to provide more information about the purpose of biospecimen collection within TMIST and the process of collecting blood and buccal samples. We are tracking the number of women approached with the new information and providing a patient incentive for those participants who re-consent and provide the samples in this sub-study. Three sites are currently participating that have large numbers of African American TMIST participants who initially declined to participate in the biospecimen collection sub-study. The first site began approaching eligible TMIST participants in April 2025. The sub-study continues through 2026. The second active ancillary project utilizes a case-control design to determine whether lower compression pressure applied during screening mammography correlates with interval cancers. The study is the first in TMIST to utilize selected imaging data and limited clinical information in a protected enclave. No images or data leave EA systems, with an external algorithm being applied to images by staff managing the TMIST imaging archive and statistical analysis being performed by the EA TMIST statistical team. There are multiple additional ancillary projects that are in development that are using TMIST data to 1) estimate the value of AI algorithms that predict short-term risk of breast cancer, 2) assess the rate of overdiagnosis, and 3) aid in the development of individualized screening recommendations (which also will use data from All of US), and others.
Presentation numberPS5-07-20
Trial in progress: Phase 1 Study Targeting DNA Methyltransferases in Metastatic Triple-Negative Breast Cancer (NCI Protocol #10546)
Roberto Leon-Ferre, Mayo Clinic, Rochester, MN
R. Leon-Ferre1, D. M. Zahrieh2, P. P. Advani3, D. Quiroga4, K. V. Giridhar1, P. J. Dizona1, D. Stover4, X. Wang5, A. J. Tevaarwerk1, G. M. Choong1, S. Yasir1, K. R. Kalari2, J. Foldi6, R. Parajuli7, M. H. Hackney8, H. K. Chew9, R. Wajeeha10, H. Bear11, E. Sharon12, B. Ko13, G. I. Shapiro12, S. Gore14, L. Pelosof14, L. Wang15, M. P. Goetz1; 1Medical Oncology, Mayo Clinic, Rochester, MN, 2Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 3Hematology/Oncology, Mayo Clinic, Jacksonville, FL, 4James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 5Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 6Malignant Hematology and Medical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 7Irvine Chao Family Comprehensive Cancer Center & Ambulatory Care, UCI Health, Irvine, CA, 8Internal Medicine, Virginia Commonwealth University, Richmond, VA, 9Internal Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA, 10Internal Medicine, Oklahoma University Health Stephenson Cancer Center, Oklahoma City, OK, 11Surgical Oncology, Virginia Commonwealth University, Richmond, VA, 12Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 13Oncology, National Cancer Institute, Bethesda, MD, 14Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, 15Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN.
Background: DNA methyltransferase (DNMT) isoforms 1, 3A and 3B are enzymes that regulate DNA methylation, controlling gene expression, chromatin stability and genetic imprinting. We have previously shown that DNMT3A protein expression in early triple negative breast cancer (TNBC) is associated with more aggressive clinicopathologic features (higher tumor grade, tumor proliferation and frequency of nodal metastases) and worse clinical outcomes in the absence of chemotherapy (Leon-Ferre et al, SABCS 2024). Pre-clinically, we have shown that DNMT3A protein expression predicts sensitivity to DNMT inhibitors (Yu J et al., J Clin Invest 2018), and that DNMT inhibitor treatment leads to downregulation of DNMT3A, enhanced expression of immune-related pathways, and halted tumor growth, particularly in combination with immunotherapy and paclitaxel. Methods: This is a phase 1, open-label, multicenter study evaluating the safety and recommended phase 2 dose (RP2D) of the oral DNMT inhibitor ASTX727 + paclitaxel + pembrolizumab in metastatic TNBC. ASTX727 is an oral fixed dose combination (FDC) tablet of 35 mg decitabine (active agent) and 100 mg cedazuridine (a cytidine deaminase inhibitor that prevents decitabine degradation in the bowel and liver, allowing oral delivery). The study consists of two parts: Part 1 is a dose-finding cohort, using a 3+3 design, with 5 dose levels (Table), and its primary objective is to determine the RP2D. Part 2 is an expansion cohort (n=6), with the primary objective to confirm tolerability of the regimen at the RP2D. Secondary objectives are to record the antitumor activity of the regimen, describe the AE profile, explore the association of baseline gene expression profiles with clinical benefit, and to evaluate impact of treatment on tumor and ctDNA methylation. Exploratory objectives will evaluate the association of baseline DNMT3A protein expression and antitumor activity, and the impact of treatment on the tumor and peripheral blood immune phenotype. Eligible patients are adults 18 years or older with metastatic TNBC (ER/PR ≤10%, and HER2-negative per ASCO/CAP guidelines), independent of PD-L1 expression. Patients are allowed to have received 0-3 prior lines of systemic therapy in the metastatic setting, including prior immune checkpoint inhibitors in the early and/or metastatic settings. Patients must be eligible for taxanes and have adequate organ and bone marrow function. Patients with treated brain metastases are eligible if previously treated without evidence of progression after 4 weeks. Leptomeningeal carcinomatosis is not allowed. Patients must have an ECOG performance status of ≤2 and have no gastrointestinal disorders impacting absorption of oral medications. As of June 20, 2025, the study is enrolling patients, with 10 patients dosed in the dose-finding phase.
| Dose Level | ASTX727 | Paclitaxel | Pembrolizumab | Cycle Duration |
| 1 | 1 FDC tablet D1-5 q 28 days |
90 mg/m2
IV D1, 8, 15
q 28 days
|
400 mg IV q6w | 28 days |
|
0
(Start)
|
1 FDC tablet D1-4 q 28 days |
90 mg/m2
IV D1, 8, 15
q 28 days
|
400 mg IV q6w | 28 days |
| -1 | 1 FDC tablet D1-3 q 28 days |
80 mg/m2
IV D1, 8, 15
q 28 days
|
400 mg IV q6w | 28 days |
| -2 | 1 FDC tablet D1, 3, 5 q 28 days |
60 mg/m2
IV D1, 8, 15
q 28 days
|
400 mg IV q6w | 28 days |
| -3 | 1 FDC tablet D1, 3, 5 q 28 days |
60 mg/m2
IV D1, 8
q 21 days
|
400 mg IV q6w | 21 days |
Presentation numberPS5-07-21
TROPION-Breast06: Multicenter, multinational, open-label, single-arm, phase 3b study of datopotamab deruxtecan (Dato-DXd) in patients (pts) with locally advanced inoperable or metastatic HR+/HER2 IHC 0 breast cancer (BC) refractory to endocrine therapy
Komal Jhaveri, Memorial Sloan Kettering Cancer Center, New York, NY
K. Jhaveri1, A. C. Garrido-Castro2, A. Decque3, F. Lujan3, M. Prahladan3, R. Taylor4, N. Toms3, F. Bidard5; 1Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 3Global Medical Affairs, Oncology & Hematology Business Unit, AstraZeneca, Cambridge, UNITED KINGDOM, 4Oncology Biometrics, Oncology R&D, AstraZeneca, Macclesfield, UNITED KINGDOM, 5Department of Medical Oncology, Institute Curie, Paris and Saint-Cloud, Paris, FRANCE.
Background: Chemotherapy (CT) is the main treatment in endocrine-refractory HR+/HER2− metastatic BC but has limited efficacy and substantial toxicities. Dato-DXd is a TROP2-directed antibody-drug conjugate (ADC) that consists of a humanized anti-TROP2 IgG1 monoclonal antibody conjugated to a potent topoisomerase I inhibitor via a plasma-stable cleavable linker. It is approved in multiple countries for adult pts with unresectable or metastatic HR+/HER2− (IHC 0, IHC 1+, or IHC 2+/ISH−) BC who have received prior endocrine therapy and CT for advanced disease, based on the findings of the TROPION-Breast01 study (NCT05104866). In the endocrine-refractory setting, the HER2-directed ADC trastuzumab deruxtecan significantly improved outcomes vs CT in pts with HR+/HER2-low (IHC 1+ or IHC 2+/ISH−) or HER2-ultralow (IHC 0 with membrane staining) unresectable or metastatic BC who had received ≥1 line of endocrine-based therapy and no prior CT in the advanced setting (DESTINY-Breast06; NCT04494425). TROPION-Breast06 aims to assess the efficacy and safety of Dato-DXd in the pre-CT endocrine-refractory setting for pts with HR+/HER2 IHC 0 (defined as no staining or incomplete and faint/barely perceptible membrane staining in ≤10% of tumor cells) inoperable or metastatic BC. Adverse events of special interest including Dato-DXd-related oral mucositis/stomatitis (OM/S) and ocular surface events (OSE) will be assessed. Trial Design: TROPION-Breast06 (EudraCT 2025-521904-23-00) is an open-label, single-arm, phase 3b study of Dato-DXd in adults (aged ≥18 years) with locally advanced inoperable or metastatic HR+, HER2 IHC 0 (per ASCO/CAP guidelines) BC who have not received any prior ADC or CT in the inoperable/metastatic setting, with progression on and not suitable for further endocrine therapy per investigator assessment. HER2 status will be determined on the most recent biopsy (collected within 6 weeks of treatment start) by local testing, and may include central confirmation. At time of enrolment, eligible pts must have an ECOG performance status of 0 or 1 and measurable disease per RECIST v1.1 or evaluable disease. Pts with clinically stable brain metastases are permitted. Sample size estimation is based on achieving a planned precision in the 95% confidence interval for median PFS, not hypothesis testing, with 100 participants chosen. Eligible pts from approximately 35 sites in North America, Europe and the Asia Pacific region will receive Dato-DXd (6 mg/kg IV) every 3 weeks until disease progression, unacceptable toxicity, withdrawal of consent or other discontinuation criteria are met. A prophylactic oral care plan will be started prior to study drug initiation and throughout treatment, including daily use of steroid-containing mouthwash and consideration of prophylactic cryotherapy; adherence to these measures and reports of mouth and throat symptoms will be captured daily in a digital tracker. Scanning/tumor assessments will be performed every 8 weeks from the first dose of study drug for 48 weeks and then every 12 weeks until RECIST v1.1-defined radiological progression by investigator assessment. Ophthalmologic assessments are mandated at regular intervals and daily use of artificial tears (4-8 times daily) and avoidance of contact lenses is recommended. The primary endpoint is investigator-assessed PFS per RECIST v1.1. Key secondary endpoints are the proportion of pts with OM/S, OSE, and treatment-related Grade ≥3 adverse events. Other secondary endpoints include clinical benefit rate, objective response rate, duration of response, overall survival, and safety/tolerability. Tumor tissue and blood samples will be collected for exploratory analyses.
Presentation numberPS5-07-22
Herthena-breast04: a phase 3, randomized, open-label study evaluating the efficacy and safety of patritumab deruxtecan (HER3-DXd) versus treatment of physician’s choice in hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2−) unresectable locally advanced or metastatic breast cancer
Barbara Pistilli, Gustave Roussy, Villejuif, France, and IHU PRISM National PRecISion Medecine Center in Oncology, Gustave Roussy, Villejuif Cedex, France
B. Pistilli1, S. Hou2, J. M. Collins3, P. K. Sudheendra2, V. Kaklamani4; 1Department of Medical Oncology, Gustave Roussy, Villejuif, France, and IHU PRISM National PRecISion Medecine Center in Oncology, Gustave Roussy, Villejuif Cedex, FRANCE, 2Oncology, Merck & Co., Inc., Rahway, NJ, 3Oncology, Daiichi Sankyo, Inc, Basking Ridge, NJ, 4Mays Cancer Center, University of Texas Health Science Center San Antonio, San Antonio, TX.
Background: There is an unmet therapeutic need for patients with HR+/HER2− metastatic breast cancer who experience disease progression and recurrence after first-line standard of care treatment with a CDK4/6 inhibitor and endocrine therapy (ET). The main therapy options, for those patients who are not considered suitable for additional ET-based therapy, are chemotherapy and antibody-drug conjugates (ADCs). Human epidermal growth factor receptor 3 (HER3) is overexpressed in 50-70% of breast cancers, with highest expression in HR+/HER2− breast cancer, and its overexpression is associated with poor prognosis and drug resistance. Patritumab deruxtecan (HER3-DXd), a novel ADC that selectively binds HER3, is composed of a fully human anti-HER3 IgG1 antibody linked to a cytotoxic topoisomerase I inhibitor via a stable tetrapeptide-based linker that is selectively cleaved within tumor cells. In the phase 2 ICARUS-Breast01 study, HER3-DXd showed clinically meaningful antitumor activity (ORR, 53.5% [95% CI, 43.2-63.6]; median PFS, 9.4 months [95% CI, 8.1-13.4]) and manageable safety in patients with HR+/HER2− advanced breast cancer who progressed on CDK4/6 inhibitor treatment, and 1 line of chemotherapy. The phase 3, multicenter, open-label, HERTHENA-Breast04 study (NCTXXXXXXXX) evaluates efficacy and safety of HER3-DXd monotherapy vs treatment of physician’s choice (TPC) in participants with HR+/HER2− unresectable locally advanced or metastatic breast cancer after progression on one line of CDK 4/6 inhibitor treatment. Methods: Participants must be aged ≥18 y and have centrally confirmed HR+/HER2− (per most recent ASCO/CAP guidelines: HER2 IHC 0 or 1+ or IHC 2+/in situ hybridization negative) unresectable locally advanced or metastatic breast cancer. Participants must have experienced disease progression or recurrence following prior treatment with a CDK4/6 inhibitor and ET and must be eligible for at least 1 TPC option, as determined by the investigator. Participants must have measurable disease per RECIST version 1.1 and ECOG PS of 0 or 1. Participants who have received prior chemotherapy or are candidates for an additional line of ET-based therapy in the advanced setting are ineligible for the study. Participants are randomized 1:1 to Arm 1 or 2, with approximately 500 participants assigned to each arm. Participants in Arm 1 will receive HER3-DXd 5.6 mg/kg IV on day 1 Q3W, and participants in Arm 2 will receive TPC consisting of 1 of the following options: 1) paclitaxel 80 mg/m2 IV on days 1, 8, 15, and 22 Q4W; 2) paclitaxel 90 mg/m2 IV on days 1, 8, and 15 Q4W; 3) nab-paclitaxel 100 mg/m2 IV on days 1, 8, and 15 Q4W; 4) capecitabine 1000 mg/m2 orally twice daily on days 1 to 14 Q3W; 5) liposomal doxorubicin 50 mg/m2 IV on day 1 Q4W; or 6) trastuzumab deruxtecan 5.4 mg/kg IV on day 1 Q3W. Treatment will continue until radiographic disease progression, unacceptable toxicity, or participant withdrawal. The dual primary endpoints are PFS per RECIST version 1.1 by BICR and OS. Secondary endpoints are ORR and DOR per RECIST version 1.1 by BICR, safety, and patient-reported outcomes. AEs are graded per National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Imaging assessments occur Q6W from randomization through week 60 and Q12W thereafter. Enrollment is planned to begin in Q3 2025 across North America, Latin America, Europe, and Asia Pacific.
Presentation numberPS5-07-23
Novel Targeting of the Microenvironment to Decrease Metastatic Recurrence of High-Risk TNBC: A Randomized Phase 2 Study of Tetrathiomolybdate (TM), an oral copper depletion agent plus capecitabine +/- pembrolizumab in patients with breast cancer at high risk of recurrence (NCT06134375).
Linda Vahdat, Dartmouth-Hitchcock, Lebanon, NH
N. Kornhauser1, N. Chan2, B. Park3, K. Miller4, K. Kalinsky5, N. Vidula6, M. Robson7, C. Seymour1, V. Mittal8, L. Vahdat1; 1Dartmouth Cancer Center, Dartmouth-Hitchcock, Lebanon, NH, 2Langone Perlmutter Cancer Center, New York University, New York, NY, 3Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 4Melvin and Bren Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, 5Emory Winship Cancer Institute, Emory University, Atlanta, GA, 6Hematology/Oncology, Massachusetts General Hospital, Boston, MA, 7Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, 8Neuberger Berman Foundation Lung Cancer Laboratory, Weill Cornell Medicine, New York, NY.
Triple-negative breast cancer (TNBC) accounts for only 17% of all breast cancer (BC) patients (pts), but 50% of metastasis-related deaths. Pts who complete neoadjuvant therapy and have significant residual disease at surgery are at highest risk with >60% relapse at 5 years (yrs) due to resistant disease. We and others have demonstrated that copper (Cu), an essential trace element, plays key roles in supporting TNBC resistance pathways [Ramchandani et al. Nat Comm 2021; Shanbhag et al PNAS 2019]. We completed a pilot phase 2 clinical trial of Cu-depletion with TM in 75 BC pts at a high-risk of relapse. We found that TM was safe, well tolerated and event-free survival (EFS) for TNBC pts is 88% for high-risk adjuvants (stage 3 BC) and 59.3% for stage 4 NED TNBC at a median follow-up of 10.4 yrs. We found that 3 components of the tumor microenvironment were significantly affected specifically VEFGR2+ endothelial progenitor cells (EPCs) and Cu-dependent LOXL-2 were significantly reduced, and the collagen microenvironment was normalized in Cu-depleted pts [Chan et al Clin Cancer Res 2017, Liu NPJ Breast 2021], recapitulating our observations in pre-clinical models. These findings have led to the hypothesis that Cu contributes to 3 key aspects of metastasis: (1) cancer cell intrinsic mitochondrial bioenergetics that mediates invasion/metastasis/chemoresistance; (2) the “pre-metastatic niche” that supports colonization, and outgrowth of disseminated metastatic tumor cells, and (3) stromal remodeling that promotes immune evasion and immunotherapy resistance. We expect that complementing standard chemo-immunotherapy with a Cu depletion strategy will overcome resistance and improve outcome. We will test this through a randomized phase 2 trial and seek mechanistic insights through comprehensive correlative studies. Our study design is a randomized phase 2 trial of TM with capecitabine vs capecitabine alone +/- pembrolizumab, in TNBC pts with significant residual invasive disease after completion of standard neoadjuvant therapy and surgery (RCB 2,3). The primary endpoint is distant relapse free survival and secondary endpoints are (i) safety, (ii) invasive disease-free survival (iDFS), and OS for the entire cohort and (iii) those who complete 6 months of TM therapy, (iv) Pt reported outcomes and (v) effect of therapy on biomarkers. Preceding this randomized phase 2 study will be a phase Ib clinical trial in 6 to 18 pts to establish the safety of the combination of adjuvant TM, capecitabine and pembrolizumab. There is a robust scientific correlative and exploratory component including evaluating the effect of Cu depletion on serial blood-based biomarkers including: (i) VEGFR2+ EPC (by flow cytometry), (ii) LOXL 2 (by ELISA), (iii) ctDNA (by PhasED-Seq, Foresight Diagnostics), (iv) immune monitoring. We plan to enroll 186 pts in this study across 8 sites. The study is open and actively accruing since November 2024.
Presentation numberPS5-07-24
A randomized phase III trial for evaluating the efficacy of adjuvant abemaciclib in patients with locoregional recurrence of hormone receptor-positive, HER2-negative breast cancer: JCOG2313 (AURA study)
Emi Tokuda, Fukushima Medical University, Fukushima, Japan
E. Tokuda1, Y. Ozaki2, F. Hara3, C. Funasaka4, Y. Naito5, A. Shimomura6, A. Yoshida7, N. Maeda8, T. Sangai9, T. Ishiba10, S. Minami11, Y. Sagara12, Y. Kajiwara13, N. Mitome14, K. Sasaki14, T. Shibata14, T. Fujisawa15, H. Iwata16, T. Shien17; 1Department of Medical Oncology, Fukushima Medical University, Fukushima, JAPAN, 2Department of Breast Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 3Department of Breast Oncology, Aichi Cancer Center Hospital, Aichi, JAPAN, 4Department of Experimental Therapeutics/ Medical Oncology, National Cancer Center Hospital East, Chiba, JAPAN, 5Department of Medical Oncology, National Cancer Center Hospital East, Chiba, JAPAN, 6Department of Breast and Medical Oncology, National Center for Global Health and Medicine, Tokyo, JAPAN, 7Department of Breast Surgical Oncology, St. Luke’s International Hospital, Tokyo, JAPAN, 8Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, JAPAN, 9Department of Breast and Thyroid Surgery, Kitasato University School of Medicine, Kanagawa, JAPAN, 10Department of Breast Surgery, Institute of Science Tokyo Hospital, Tokyo, JAPAN, 11Department of Surgery, NHO Nagasaki Medical Center, Nagasaki, JAPAN, 12Department of Breast and Thyroid Surgical Oncology, Hakuaikai Medical Corp. Sagara HP, Kagoshima, JAPAN, 13Department of Breast Surgery, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, JAPAN, 14Japan Clinical Oncology Group Data Center/Operations Office, National Cancer Center Hospital, Tokyo, JAPAN, 15Department of Breast Oncology, Gunma Prefectural Cancer Center, Gunma, JAPAN, 16Department of Advanced Clinical Research and Development, Nagoya City University, Graduate School of Medical Sciences, Aichi, JAPAN, 17Department of Breast and Endocrine Surgery, Okayama University Hospital, Okayama, JAPAN.
BackgroundHormone receptor (HR)-positive, HER2-negative breast cancer is the most common subtype, accounting for about 70% of all breast cancer cases. Although curative therapy with surgery, radiation, and systemic treatment achieves long-term remission, locoregional recurrence (LRR) occurs in 3-16% of patients, including ipsilateral breast tumor recurrence (IBTR), chest wall recurrence, or regional lymph node recurrence. Patients with LRR are at increased risk for subsequent distant metastasis and death. However, systemic treatment strategies in this setting remain poorly defined, and no standard adjuvant therapy has been established for patients who develop LRR after initial curative therapy. Abemaciclib, a selective CDK4/6 inhibitor, has improved invasive disease-free survival (IDFS) when added to endocrine therapy in high-risk early-stage HR+/HER2− breast cancer, as shown in the monarchE trial. However, its benefit in patients with isolated LRR remains unclear. Whether escalation with abemaciclib improves outcomes in this population requires investigation. Trial Design JCOG2313 (AURA; Abemaciclib Utility for locoregional Recurrence of breast cAncer) is a randomized, open-label, multicenter phase III trial conducted by the Japan Clinical Oncology Group (JCOG). The trial evaluates the efficacy of adding adjuvant abemaciclib to endocrine therapy compared with endocrine therapy alone in patients with first LRR of HR-positive, HER2-negative breast cancer after curative local therapy. Eligible patients are aged 18 years or older with isolated LRR occurring after initial surgery for primary breast cancer, who have undergone complete resection or definitive radiotherapy for LRR with curative intent.Chemotherapy administered before or after local therapy for LRR is permitted. Patients are randomized 1:1 to: Arm A: endocrine therapy alone (tamoxifen or aromatase inhibitor ± LHRH agonist) Arm B: endocrine therapy plus abemaciclib for 2 years The primary endpoint is IDFS. Secondary endpoints include overall survival, distant relapse-free survival, breast cancer-specific survival, and adverse events. The planned sample size is 290 patients to provide 70% power to detect a hazard ratio of 0.59 at a one-sided alpha level of 5%, assuming an accrual period of 4 years and a follow-up period of 5 years. Current Status The trial opened in February 2025 and is currently enrolling patients at participating 51 sites in Japan. A prospective circulating tumor DNA (ctDNA) ancillary study will assess minimal residual disease dynamics before, during, and after treatment using serial plasma samples. This ancillary study aims to evaluate the utility of ctDNA as a biomarker for recurrence risk stratification and treatment response monitoring. Significance This trial addresses a critical unmet need in the management of HR-positive, HER2-negative breast cancer patients with LRR. By evaluating the clinical benefit of abemaciclib in this setting, JCOG2313 may provide evidence to support treatment escalation and improve long-term outcomes. The findings may contribute to establishing future standards of care for patients with LRR. Trial registration:Japan Registry of Clinical Trials (jRCT): jRCTs031240666
Presentation numberPS5-07-25
Phase I Study of TAS0728 in Patients with Advanced Solid Tumors with HER2 Aberration (TAIBRAKHER Study)
Kazuki Nozawa, Nagoya City University, Nagoya, Japan
K. Nozawa1, C. K. Imamura2, S. Watanabe3, T. Kogawa4, M. Furuta5, T. Toyama6, H. Hayashi3, J. Tsurutani7; 1Advanced Clinical Research and Development, Nagoya City University, Nagoya, JAPAN, 2Advanced Cancer Translational Research Institute,, Showa University, Tokyo, JAPAN, 3Medical Oncology, Kindai University Faculty of Medicine, Osaka, JAPAN, 4Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 5Gastroenterology, Kanagawa Cancer Center, Yokohama, JAPAN, 6Breast Surgery, Nagoya City University, Nagoya, JAPAN, 7Advanced Cancer Translational Research Institute, Showa University, Tokyo, JAPAN.
Background: TAS0728 is a novel oral small-molecule inhibitor targeting HER2’s tyrosine kinase domain via covalent binding, showing significant antitumor activity in preclinical models. HER2 aberration including mutation and amplification drives progression of various solid tumors. Despite advances in HER2-targeted therapies, unmet needs remain, particularly for advanced-stage disease and post-standard therapy settings. In the first-in human phase I study of TAS0728 at escalating doses from 50 to 200 mg BID for 21-day cycles, dose-limiting toxicity of Grade 3 diarrhea was observed at the doses of 150 mg and 200 mg BID. And partial responses were observed in 2 of 14 evaluable patients. However, the maximum tolerated dose was not determined due to the occurrence of unacceptable toxicity at 150 mg BID. This study aims to assess the safety, tolerability, and pharmacokinetics, and to determine the recommended phase II dose (RP2D) of TAS0728 at escalating doses of 50, 75, and 100 mg BID for 21-day cycles. Methods: This multicenter phase I study includes dose escalation and expansion parts. The primary endpoint is to determine the RP2D in the dose escalation part and to evaluate safety at the RP2D in the expansion part. Pharmacokinetics and efficacy are also assessed. In the dose escalation part, the dose level is escalated according to a typical 3 + 3 design, and dose-limiting toxicities are assessed during the first cycle. A total of 30 patients will be enrolled. Inclusion criteria include ≥ 18 years old, unresectable tumors with confirmed HER2 aberration (protein overexpression, gene mutation or amplification), ECOG performance status 0-1, and adequate organ function. Exclusion criteria are significant cardiovascular conditions and active brain metastases. TAS0728 is a substrate for a transporter of breast cancer resistance protein (BCRP). Therefore, the genotypes for ABCG2, which encode a BCRP, are evaluated as a candidate factor for interpatient variability of pharmacokinetics. HER2 gene status is also analyzed from circulating tumor DNA in blood at baseline and disease progression.
Presentation numberPS5-07-26
Ribociclib with fulvestrant versus physician’s choice treatments in patients who recurred after completion of adjuvant cyclin-dependent kinase 4/6 inhibitors as first-line treatment in HR+, HER2- advanced breast cancer
Hee Kyung Ahn, Samsung Medical Center, Seoul, Korea, Republic of
H. Ahn, J. Shin, J. Kim, J. Ahn, Y. Park; Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF.
Background: The NATALEE phase 3 trial demonstrated a significant reduction in the risk of invasive disease recurrence when ribociclib was added to endocrine therapy in patients with early-stage hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer at intermediate to high risk of recurrence, which supports the expanding therapeutic role of ribociclib from the advanced to the adjuvant setting. However, optimal treatment after recurrence following adjuvant cyclin dependent kinase 4/6 (CDK4/6) inhibitor remains a clinical concern. While postMONARCH and EMBER3 showed benefits of maintaining CDK4/6 inhibitors after disease progression in advanced setting, there remains an unmet need to determine the optimal treatment after recurrence after adjuvant CDK4/6 inhibitor therapy. Method: This randomized, phase 2, open-label, multicenter trial aims to compare the efficacy and safety of ribociclib plus fulvestrant versus physician’s choice treatments (PCT) in patients with distant recurrent HR+/HER2- breast cancer after completing adjuvant CDK4/6 inhibitor therapy. Patients will be randomized in a 1:1 ratio to either the treatment arm (ribociclib and fulvestrant) or the control arm (PCT: fulvestrant or exemestane plus everolimus). Leuporelin will be incorporated into peri and premenopausal patients. Eligible participants include men or pre-, peri-, postmenopausal women aged ≥19 with HR+/HER2- advanced breast cancer based on most recently analyzed biopsy that recurred at least one year after the last dose of adjuvant CDK4/6 inhibitor. The patients must have had previously received at least 1 year of adjuvant abemaciclib or ribociclib and a minimum of 2 years of adjuvant endocrine therapy (either alone or in combination with CDK4/6 inhibitors). Additional inclusion criteria include Eastern Cooperative Oncology Group performance status of 0 or 1 and evidence of at least one measurable lesion according to RECIST v1.1. Patients whose cancer recurs 1 year or later after completing adjuvant endocrine therapy or those who have been free from endocrine therapy for at least 2 years will be excluded. The primary objective is to determine whether ribociclib with fulvestrant prolongs progression-free survival (PFS) compared to PCT. PFS will be determined based on local tumor assessment using RECIST v1.1. Assuming a median PFS of 6.75 months in the control arm and 9 months in the treatment arm, the target hazard ratio was 0.75. Using the O’Brien-Fleming boundary with a one-sided type I error rate of 10% and 80% power, the total required sample size was 272 patients (136 per arm). The planned overall study duration is 72 months (36 months for accrual and randomization, 36 months of follow-up for primary endpoint PFS). This trial received local Institutional Review Board approval and is registered in ClinicalTrials.gov (NCT06849947).
Presentation numberPS5-07-27
INAVO123: Phase 3 study of first-line inavolisib / placebo + a cyclin-dependent kinase 4/6 inhibitor + letrozole in participants with <i>PIK3CA</i>-mutated, hormone receptor-positive, HER2-negative, endocrine-sensitive advanced breast cancer
Reva K. Basho, Ellison Medical Institute, Los Angeles, CA
R. Basho1, K. Jhaveri2, J. King3, S. Kümmel4, Y. Park5, V. Krishnan6, P. Gyamfi7, X. Liu8, V. Khor9, A. Mani9, J. Cortés10; 1Department of Medicine, Ellison Medical Institute, Los Angeles, CA, 2Breast and Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 3Oncology Department, Royal Free London NHS Foundation Trust, Royal Free Hospital and Barnet Hospital, London, UNITED KINGDOM, 4Breast Unit, Kliniken Essen-Mitte, Department of Gynecology with Breast Center, Charité – Universitätsmedizin Berlin, Berlin, GERMANY, 5Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 6Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, 7Portfolio Clinical Safety (PCS), Product Development Safety (PDS), Genentech, Inc., South San Francisco, CA, 8Data & Statistical Sciences, Genentech, Inc., South San Francisco, CA, 9Product Development Oncology, Genentech, Inc., South San Francisco, CA, 10Pangaea Oncology, Quiron Group, International Breast Cancer Center (IBCC), Barcelona, SPAIN.
BACKGROUND Standard of care for PIK3CA-mutated (mut), hormone receptor-positive (HR+), HER2-negative (HER2-), endocrine-sensitive advanced breast cancer (aBC) is endocrine therapy + a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i), but progression is expected due to resistance. Inavolisib (INAVO) is a highly potent and selective PI3Kαi that also promotes degradation of mut p110α. INAVO120 (NCT04191499) showed that first-line (1L) treatment with INAVO + palbociclib (PALBO) + fulvestrant (FULV) resulted in a significantly improved progression-free survival (PFS) benefit vs placebo (PBO) + PALBO + FULV (stratified hazard ratio 0.43; 95% confidence interval 0.32-0.59; p < 0.0001) in patients with PIK3CAmut, HR+, HER2-, endocrine-resistant aBC, and is now approved by the Food and Drug Administration for use in such patients. TRIAL DESIGN INAVO123 evaluates 1L INAVO + a CDK4 / 6i + letrozole (LET) vs PBO + a CDK4 / 6i + LET in participants (pts) with PIK3CAmut (determined by central circulating tumor DNA [ctDNA] or existing local tumor tissue / ctDNA testing), HR+, HER2-, endocrine-sensitive (as defined in the protocol) aBC. Pts will be randomized 1:1 to INAVO (9 mg oral daily [QD]) or PBO + a CDK4/6i (e.g. PALBO 125 mg oral QD 21 days on, 7 days off) + LET (2.5 mg oral QD) in 28-day cycles. Pts will receive treatment until disease progression, unacceptable toxicity, withdrawal of pt consent, death, or study termination. ELIGIBILITY Pts must have measurable disease, fasting glucose < 126 mg/dL, and glycated hemoglobin < 6.5%. Pre- and perimenopausal women, and men, must receive luteinizing hormone-releasing hormone agonist. Pts with aBC who progressed while on / within 12 months of ending adjuvant therapy are ineligible. AIMS Primary endpoint: PFS. Secondary endpoints: overall survival, confirmed overall response rate, duration of response, clinical benefit rate, patient-reported outcomes, and safety. STATISTICAL METHODS PFS will be assessed via a stratified log-rank test with a two-sided significance level of 0.05 and Kaplan-Meier methods. Stratification factors: de novo HR+ aBC (yes vs no) and visceral metastases (yes vs no). An independent data monitoring committee will oversee safety and interim efficacy. ACCRUAL Target randomization is 450 pts across > 200 centers, globally.
Presentation numberPS5-07-28
The SURVIVE HERoes study NCT06643585: Targeting molecular relapse in breast cancer.
Kerstin Pfister, University Hospital Ulm, Ulm, Germany
K. Pfister1, T. W. Friedl1, S. T. Huesmann1, S. Heublein1, H. Schäffler1, F. Mergel2, F. Mehmeti1, A. Fink1, T. Braun1, T. N. Fehm3, V. Müller4, L. Wiesmüller1, K. Pantel5, B. Rack1, A. D. Hartkopf6, C. Pipinikas7, A. Chevalier7, P. A. Fasching8, W. Janni1; 1Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, GERMANY, 2Department of Gynecology and Obstetrics, SLK Kliniken Heilbronn, Heilbronn, GERMANY, 3Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Düsseldorf, GERMANY, 4Department of Gynecology, University Hospital Hamburg-Eppendorf, Hamburg, GERMANY, 5Institute of Tumor Biology, University Hospital Hamburg-Eppendorf, Hamburg, GERMANY, 6Department of Women’s Health, University Hospital Tübingen, Tübingen, GERMANY, 7NeoGenomics Inc., NeoGenomics Inc., Fort Myers, FL, 8Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen, GERMANY.
Background: Current research on circulating tumor DNA (ctDNA) in the adjuvant setting of early breast cancer (eBC) highlights its strong prognostic significance. Patients who are ctDNA-positive but show no radiological signs of relapse (i.e., molecular relapse) exhibit reduced disease-free and overall survival. Secondary adjuvant intervention studies with potent therapeutics represent an innovative and promising approach to intercept molecular relapase. Study Design: The SURVIVE HERoes study is a phase III randomized clinical trial comparing the antibody-drug conjugate trastuzumab deruxtecan (T-DXd) to standard of care (SoC) in patients with molecular residual or recurrent disease. Patients tested positive for ctDNA in a tumor-informed approach (RaDaR®) are eligible, if staging examinations do not show any residual or recurrent cancer. Participants must have HER2-positive or HER2-low eBC (hormone receptor (HR) positive or negative) and have completed primary therapy (operation (R0) ± chemotherapy ± radiation).A total of 180 participants are randomized in a 2:1 ratio to receive T-DXd (+ endocrine therapy for HR positive patients) or standard of care for 48 weeks, followed by a 48-week follow-up phase. Stratification factors include hormonal receptor status (positive versus negative) and HER2-status (positive versus low).Staging examinations and ctDNA assessments will be performed every 12 weeks during the 2-year study phase per patient. The study is accompanied by a comprehensive translational research program. Endpoints: The primary endpoint is the ctDNA clearance rate after 48 weeks, comparing participants in the experimental arm (receiving T-DXd) with those in the control arm (receiving standard of care), irrespective of ctDNA test results at other time points.Secondary endpoints include invasive disease-free survival, overall survival, ctDNA clearance at other time points, safety, and quality of life (QoL), as assessed by EORTC QLQ-C30 and PA-F12. Recruitment: Recruitment began in Q2/2025 and is anticipated to continue until 2030. Fifty sites across Germany are planned for participation, and as of July 2025, two sites are actively recruiting. Discussion: Treating ctDNA-positive patients without radiographic evidence of recurrence is a novel therapeutic strategy. If SURVIVE HERoes and similar studies targeting molecular relapse yield positive results, they could pave the way for a new molecularly driven personalized surveillance and treatment approach. Funding: AstraZeneca, NeoGenomics Inc., Menarini Silicon Biosystems, Tosoh Bioscience
Presentation numberPS5-07-29
Trial in progress: a phase 1 dose-escalation study evaluating BGB-21447 (BCL2 inhibitor) in combination with fulvestrant with and without BGB-43395 (CDK4 inhibitor) in patients with previously treated HR+/HER2− metastatic breast cancer
Geoffrey J. Lindeman, Peter MacCallum Cancer Centre and the Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
G. Lindeman1, C. Muttiah2, P. Lau3, N. Hunter4, J. Zhang5, Y. Liu6, Y. Dong7, P. Zhou8, A. Kavi9, K. Keyvanjah9, Z. Shao10; 1Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre and the Eliza Hall Institute of Medical Research, Melbourne, VIC, AUSTRALIA, 2Medical Oncology, Western Health, Melbourne, VIC, AUSTRALIA, 3Breast Cancer Research Center, Linear Clinical Research, Nedlands, WA, AUSTRALIA, 4Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 5Oncology, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 6Clinical Biomarker Science and CDx Development, BeOne Medicines (Shanghai) Co., Ltd, Shanghai, CHINA, 7Global Statistics and Data Science, BeOne Medicines, San Carlos, CA, 8Clinical Development, Solid Tumors, BeOne Medicines (Shanghai) Co., Ltd, Shanghai, CHINA, 9Clinical Development, Solid Tumors, BeOne Medicines, San Carlos, CA, 10Surgery and Breast Surgery, Fudan University Cancer Center, Shanghai, CHINA.
Background: HR+/HER2− tumors account for ~70% of all breast cancers (BCs)1 and are responsible for most BC-related deaths. CDK4/6 inhibitors (CDK4/6i) + endocrine therapy (ET) have improved outcomes for HR+/HER2− BC2; however, patients eventually experience progressive disease and require new therapies. B-cell lymphoma 2 (BCL2) is an anti-apoptotic protein involved in regulation of the intrinsic apoptosis pathway that is often dysregulated in tumors. In BC, estrogen receptor signaling leads to upregulation of BCL2, which is overexpressed in 70-80% of HR+ BC.3-5 When administered in rational combinations with other anticancer agents, BCL2 inhibitors (BCL2i) are likely to enhance potential antitumor effects.5 In HR+ BC mouse xenograft models, the addition of a BCL2i to ET enhanced antitumor activity relative to single agent ET.6 Furthermore, the addition of a CDK4i or CDK4/6i to BCL2i + ET significantly augmented this antitumor activity, suggesting further exploration of the triplet is warranted.7-9 BGB-21447 is a novel, orally bioavailable, next-generation BH3 mimetic that was designed to be a highly potent inhibitor of both wild-type and mutant BCL2, which is also active against BCL-xL and is currently in clinical trials for hematologic cancers and BC. BGB-43395 is a selective CDK4i with improved CDK4 coverage and greater selectivity for CDK4 over CDK6, thus minimizing off-target toxicities. BGB-43395 is currently under development with ET in HR+ BC and other selected solid tumors (NCT06120283, NCT06253195). Here, we describe an ongoing phase 1 study of BGB-21447 + fulvestrant with and without BGB-43395 (NCT06756932). Methods: This is an open-label, international, multicenter, phase 1 dose-escalation study evaluating the safety, tolerability, pharmacokinetics (PK), pharmacodynamics, and preliminary antitumor activity of BGB-21447 in combination with fulvestrant without (Part A) or with BGB-43395 (Part B) in patients with previously treated HR+/HER2− metastatic BC. Sequential cohorts will receive ascending doses of BGB-21447 and fulvestrant. BGB-43395 will be added to the doublet combination dose-escalation component. Key eligibility criteria include patients ≥18 years with histologically or cytologically confirmed HR+HER2− metastatic BC, previous treatment in the advanced/metastatic setting, including prior treatment with CDK4/6i and ET in any setting, Eastern Cooperative Oncology Group Performance Status ≤1, and adequate organ function. Patients with prior BCL2i exposure are excluded. Primary objectives are to assess the safety and tolerability of BGB-21447 in combination with fulvestrant, with or without BGB-43395, and to determine the maximum tolerated dose, maximum administered dose, and recommended dose for expansion of both the doublet and triplet combination. Secondary objectives are preliminary antitumor activity (objective response rate, duration of response, and time to response per Response Evaluation Criteria in Solid Tumors v1.1), characterization of the PK (single dose and steady state), and the preliminary effect of food on PK. This study is currently recruiting patients, with sites open across Australia, China, and the United States (Part A only). References: 1. Burstein HJ. N Engl J Med. 2020;383:2557-2570. 2. Thill M and Schmidt M. Ther Adv Med Oncol. 2018;3:1758835918793326. 3. Kawiak A, Kostecka A. Cancers (Basel). 2022;14:279. 4. Dawson SJ et al. Br J Cancer. 2010;103 :668-675. 5. Lok SW, et al. Cancer Discov. 2019;9:354-369. 6. Vaillant F, et al. Cancer Cell. 2013;24:120-129. 7. Whittle JR, et al. Clin Cancer Res. 2020;26:4120-4134. 8. Lindeman G, et al. Clin Cancer Res. 2025;31(12_Supplement):P2-07-18. 9. BeOne Medicines. Data on file.
Presentation numberPS5-07-30
Open-label, randomized, multicenter, phase 3, ELAINE 3 study of the efficacy and safety of lasofoxifene plus abemaciclib for treating ER+/HER2-, locally advanced or metastatic breast cancer with an ESR1 mutation
Matthew P Goetz, Mayo Clinic, Rochester, MN
M. P. Goetz1, S. A. Wander2, T. Bachelot3, G. Curigliano4, A. de Nonneville5, E. N. Gal-Yam6, K. Jhaveri7, S. L. Sammons8, S. Shen7, C. J. Twelves9, G. Gasper10, P. V. Plourde10, D. J. Portman10, S. Damodaran11; 1Department of Medical Oncology, Mayo Clinic, Rochester, MN, 2Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, 3Department of Medical Oncology, Centre Léon Bérard, France, FRANCE, 4Department of Oncology and Hemato-Oncology, Istituto Europeo di Oncologia IRCCS, Milano, ITALY, 5Department of Medical Oncology, Institut Paoli Calmettes, Marseille, FRANCE, 6Department of Medical Oncology, Sheba Medical Center, Tel-Hashomer Ramat Gan, ISRAEL, 7Department of Medicine, Memorial Sloan Kettering Cancer Center/Evelyn H. Lauder Breast and Imaging Center, New York, NY, 8Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 9Medical Oncology, Univsersity of Leeds and Leeds Teaching Hospitals NHS Trust, Leeds, UNITED KINGDOM, 10Clincal Development, Sermonix Pharmaceuticals, Columbus, OH, 11Department of Medical Oncology, MD Anderson Cancer Center, Houston, TX.
Background: Most patients with estrogen receptor-positive (ER+), metastatic breast cancer (mBC) treated with endocrine therapy (ET) will ultimately develop resistance. A large unmet need exists especially when resistance occurs following treatment with a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i), which is potentially driven by a mutation in the ERα-coding gene, ESR1. Lasofoxifene (LAS), an oral, next-generation ET and ER breast antagonist, was evaluated in two phase 2 studies of women with ER+/HER2- mBC and an ESR1 mutation who had disease progression on previous ET and CDK4/6i. In the ELAINE 1 trial, LAS monotherapy provided numerically greater progression-free survival (PFS, median 5.6 mos vs 3.7 mos; HR: 0.669 [95% CI, 0.445‒1.125], P=0.138), objective response rate (ORR, 13% vs 3%; P=0.124), and clinical benefit rate (CBR, 37% vs 22%; P=0.117) compared with the ER degrader fulvestrant (fulv), with a favorable safety profile (Goetz MP et al. Ann Oncol 2023;34:1141‒51). The single-arm, ELAINE 2 trial showed that LAS combined with abemaciclib (Abema) was well tolerated with a median PFS of ~13 mos, ORR of 56%, and CBR of 66% (Damodaran S et al. Ann Oncol 2023;34:1131‒40). The phase 3, registrational, ELAINE 3 trial was initiated based on these promising earlier-phase data. Recent results from postMONARCH confirmed superior efficacy of fulv/Abema over fulv/placebo (mPFS 6.0 mos vs 5.3 mos, HR 0.73 [0.57‒0.95], nominal P-value 0.017) in non-biomarker‒selected, mBC patients after progression on a prior CDK4/6i and aromatase inhibitor (AI), with similar HR in an ESR1-mutated cohort (Kalinsky K et al. J Clin Oncol 2025;43:1101‒12). Additionally, recent data from EMBER-3 demonstrated meaningful PFS with a novel ET combined with abemaciclib after progression on AI/CDK4/6i (Jhaveri KL et al. N Engl J Med 2025;392:1189‒202). The ELAINE 3, global trial will compare LAS/Abema with fulv/Abema in a post-CDK4/6i, ESR1-mutation‒selected, mBC population. Methods: ELAINE 3 (NCT05696626) is an open-label, phase 3, multicenter study evaluating the efficacy, safety, and tolerability of LAS plus Abema versus fulv plus Abema. Patients are being enrolled at approximately 152 sites in 13 countries (Australia, Belgium, Canada, China, France, Italy, Israel, Poland, Romania, Spain, Taiwan, United Kingdom, United States). Key inclusion criteria are pre- and postmenopausal women and men aged ≥18 yrs; ER+/HER2-, locally advanced and/or mBC (measurable and/or non-measurable disease); ≥1 acquired ESR1 mutation; progression on an AI plus palbociclib or ribociclib as their first hormonal treatment for advanced/mBC; and ≤1 line of chemotherapy in the advanced/metastatic setting. Patients will be randomized 1:1 to receive LAS 5 mg/day plus Abema 150 mg BID, or fulv 500 mg IM (days 1, 15, and 29, then once monthly) plus Abema 150 mg BID. Treatment will continue until progression, death, unacceptable toxicity, or withdrawal from the study. The primary endpoint is PFS per RESIST 1.1 by blinded independent central review (BICR); key secondary endpoints are ORR, overall survival, CBR, duration of response, and time to response. Safety, time to cytotoxic chemotherapy, and quality of life (including vaginal health) will also be evaluated. Blood samples for circulating tumor DNA (ctDNA) will be collected for genomic analyses at screening, at weeks 4 and 8 and every 8 weeks thereafter, and at the final visit. Outcomes with LAS/Abema and fulv/Abema will be compared using a stratified Cox proportional hazards model and stratified logrank test with an expected PFS hazard ratio of 0.68 at final analysis. Enrollment is currently ongoing to meet the target sample size of 500 patients.
Presentation numberPS5-08-01
Hormone receptor (HR)-positive HER2 negative breast cancer patients treated with preoperative Elacestrant and PULSAR adaptive radiotherapy: a phase II study (HELP Trial)
Luca Visani, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
L. Visani1, C. Becherini1, M. Valzano1, V. Salvestrini1, M. Loi2, G. Simontacchi2, D. Greto2, M. Carracino3, M. Pacinico3, A. Duatti3, C. Mattioli3, L. Caprara3, C. Bellini4, J. Nori4, D. Cassetti5, L. Orzalesi5, S. Bianchi6, A. Morandi3, L. Livi3, I. Meattini3; 1Radiation Oncology & Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Firenze, ITALY, 2Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Firenze, ITALY, 3Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, Firenze, ITALY, 4Breast Imaging Unit, Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Firenze, ITALY, 5Breast Surgery Unit, Azienda Ospedaliero-Universitaria Careggi, Firenze, ITALY, 6Division of Pathological Anatomy, Department of Health Sciences, University of Florence, Firenze, ITALY.
Background. Chemotherapy-based neoadjuvant treatment in HR-positive (HR+), HER2-negative (HER2-) breast cancer (BC) patients is associated with the lowest pathologic complete response (pCR) rates among all subtypes. However, despite the exquisite hormone sensitivity of these tumors and a more favorable toxicity profile compared to chemotherapy, pCR rates after neoadjuvant endocrine therapy are even lower. Elacestrant, an oral selective estrogen receptor degrader (SERD), has recently demonstrated its efficacy in the metastatic setting. The use of preoperative radiation therapy (RT) has been questioned due to a potential immunosuppressive effect that may be detrimental for tumor control. This potential negative aspect may be circumvented by the introduction of Personalized Ultrafractionated Stereotactic Adaptive Radiotherapy (PULSAR), a novel concept in with each fraction is administrated at least 10-20 days apart, resulting in excellent tolerability and a potential enhancement of anti-tumor immunity. ELIPSE was a window of opportunity trial evaluating the effect of 4 weeks of Elacestrant in postmenopausal women with HR+ HER2- BC amenable to surgery. It significantly reduced Ki67 expression and was associated with a shift towards a more endocrine sensitive and less proliferative phenotype, higher levels of tumor infiltrating lymphocytes (TILs) and increased expression of immune-response genes. The combination with an adaptive, repeated RT strategy such as PULSAR may further enhance the immune response. In this proof-of-concept trial, the primary objective will be to investigate the safety of this combinatory approach, along with a preliminary evaluation of clinical efficacy.Trial design. This is a proof-of-concept phase II trial to assess the safety (as primary endpoint) and clinical efficacy of neoadjuvant therapy with Elacestrant and PULSAR (Fig.1). The study will enroll postmenopausal patients with early HR+ HER2- node positive BC, clinically staged II-III. Considering an overall incidence of acute skin toxicity of 66.5% with whole breast irradiation (WBI), a number of 21 patients is required to obtain an overall rate of 30% with PULSAR combined with Elacestrant. Patients will receive Elacestrant 4 mg orally once daily for 20 weeks and PULSAR on the MRI-based breast gross tumor volume (GTVt), consisting of 10 Gy “pulses” every 4 weeks for a maximum of 5 or less in case of radiologic complete response. Tumor response will be assessed, as compared to baseline, by monthly breast MRI. Surgery will be planned within 6 weeks from the last Elacestrant administration. Patients will then receive adjuvant endocrine therapy with or without CDK4/6 inhibitors as per standard of care and postoperative RT to the locoregional lymph nodes in case of nodal residual disease, if clinically indicated.Clinical trial identification. EU Clinical Trial Number 2025-520762-22-00; NCT Number NCT07005882
Presentation numberPS5-08-02
Vacuum Assisted Excision (VAE): A single-step approach to the diagnosis and percutaneous treatment of Early Breast Cancer (THE VAE BReast 01 TRIAL)
Henrique Lima Couto, Redimama-Redimasto, Belo Horizonte, Brazil
H. L. Couto1, B. A. Coelho2, T. C. Oliveira3, B. F. Ricardo4, P. H. Toppa5, D. d. Pires3, S. d. Ferreira3, L. B. Oliveira3, A. C. Mendonça3, R. G. Saliba3, T. P. Moraes3, P. C. Soares3, C. A. Padua6, G. F. Cunha Júnior6, M. S. Castilho7, L. L. Dominguez3, J. S. Oliveira3, A. C. Oliveira3, D. R. Siqueira3, B. A. Pires3, A. Mattar8, G. d. Silva Junior9, M. Antonini10, E. C. Pessoa11, F. M. Reis12, BreastMit Collaborative Research Group; 1CEO, Redimama-Redimasto, Belo Horizonte, BRAZIL, 2CEO, Mater Clinic, Montes Claros, Minas Gerais Brazil., Montes Claros, BRAZIL, 3Breast Cancer Center, Redimama-Redimasto, Belo Horizonte, BRAZIL, 4Breast Pathology, Anatomia BH Pathology, Belo Horizonte, BRAZIL, 5Breast Pathology, Analys Pathology, Belo Horizonte, BRAZIL, 6Cetus Oncology, Belo Horizonte, Minas Gerais, Brazil., MIRA – Medical Group, Belo Horizonte, BRAZIL, 7Radio-oncology, Radiocare, Belo Horizonte, Minas Gerais, Brazil., Belo Horizonte, BRAZIL, 8Mastology, Women Hospital, São Paulo, São Paulo, Brazil., Belo Horizonte, BRAZIL, 9Mastology, Orizonti Hospital, Belo Horizonte, BRAZIL, 10Department of Gynecology and Obstetrics, Hospital of the State Public Servant of São Paulo, Sao Paulo, Sao Paulo Brazil., Belo Horizonte, BRAZIL, 11Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil., Botucatu, BRAZIL, 12Department of Gynecology and Obstetrics, Faculty of Medicne of The Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL.
Abstract Introduction Vacuum-assisted excision (VAE) of breast lesions is a technique used for diagnostic and therapeutic purposes and is performed on an outpatient basis, with local anesthesia and image guidance. Currently, VAE is used in the management of benign lesions and lesions of uncertain malignant potential (B3 lesions). More recently, there has been interest in the application of VAE for the percutaneous treatment of small breast cancers with the aim of reducing morbidity and aggressive surgical treatment of breast cancers detected by screening programs. The way conventional VAE is performed, histopathological assessment of resection margins is not possible. Obtaining free margins after a breast cancer resection is a primary objective in the surgical treatment of this disease. If VAE could ensure free margins and absence of residual tumor in the resection cavity, this would also be a method safely used for minimally invasive treatment, providing an effective percutaneous treatment of early breast cancers.Objectives: evaluate the effectiveness of vacuum-assisted excision associated with percutaneous sampling of cavity margins for the complete resection of breast cancers ensuring the absence of residual disease in surgical pathology.Methods: Inclusion criteria are women with unifocal lesions smaller than 1.5 cm, Category 4 or 5 ACR BI-RADS™, identified by screening or clinical alteration. Non-inclusion criteria are multifocal and multicentric breast cancers, breast cancers associated with diffuse and extensive microcalcifications. Sensitivity, specificity, accuracy, positive predictive value, negative predictive value, false negative and false positive rate of vacuum-assisted excision associated with percutaneous sampling of cavity margins for the complete resection of breast cancers will be calculated. The data collected will also encompass demographics, characteristics of the lesion, and information regarding the biopsy, surgical pathology, and surgical procedures, as well as data on side effects, patient acceptance, cosmetic results and patients’ experience during VAE (BreastQ questionnaire).Ethics and dissemination Ethics approval has been obtained by the Brazilian National Research Ethics Commission (CONEP) 61781922.5.0000.5109 and is registered in Brazilian Registry of Clinical Trials (https://ensaiosclinicos.gov.br/), identifier U1111-1301-4235. Participants will provide written informed consent, and researchers will follow institutional guidelines for data collection and management.
Presentation numberPS5-08-03
Denali-1: a seamless phase 1/2 study of A2B395, a logic‑gated, allogeneic, Tmod chimeric antigen receptor T-cell (CAR T) therapy, in patients with triple-negative breast cancer and other EGFR-expressing solid tumors with HLA-A*02 loss of heterozygosity (LOH)
Jennifer Marie Specht, University of Washington/Fred Hutch Cancer Center, Seattle, WA
J. M. Specht1, S. R. Punekar2, M. Ulrickson3, D. J. Wong4, J. R. Hecht5, M. Maus6, P. M. Grierson7, J. B. Sunwoo8, J. R. Molina9, D. G. Maloney10, K. Spencer11, H. E. Fuentes Bayne9, M. Davila12, S. P. Patel13, B. Creelan14, D. B. Zhen15, J. Niu16, D. C. Prasad17, R. L. Marar17, N. A. Bagegni18, G. Palluconi19, J. Lee19, A. Mardiros19, W. Bretzlaff19, W. J. Langeberg19, J. S. Welch19, E. Ng19, W. Y. Go19, K. Kirtane20, R. A. Shatsky13; 1Medicine, Division of Hematology & Oncology; Clinical Research Division, University of Washington/Fred Hutch Cancer Center, Seattle, WA, 2Department of Medicine, NYU Grossman School of Medicine, New York University Langone Health, Perlmutter Cancer Center, New York, NY, 3Hematology & Oncology, Banner MD Anderson Cancer Center, Gilbert, AZ, 4Department of Medicine, Division of Hematology-Oncology, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, 5Department of Medicine, Hematology/Oncology, Gastrointestinal Oncology, UCLA Jonsson Comprehensive Cancer Center, Santa Monica, CA, 6Hematology and Oncology, Harvard Medical School/Massachusetts General Hospital, Boston, MA, 7Division of Oncology, Washington University in St. Louis, St. Louis, MO, 8Division of Head & Neck Surgery, Stanford University School of Medicine, Stanford, CA, 9Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 10Division of Hematology and Oncology/Translational Science and Therapeutics Division, University of Washington/Fred Hutch Cancer Center, Seattle, WA, 11Department of Medicine at NYU Grossman School of Medicine, New York University Langone Health, Perlmutter Cancer Center, New York, NY, 12Department of Medicine Translational Research, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 13Medical Oncology, University of California San Diego Moores Cancer Center, San Diego, CA, 14Medical Oncology, Moffitt Cancer Center, Tampa, FL, 15Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, 16Medical Oncology, Banner MD Anderson Cancer Center, Gilbert, AZ, 17Internal Medicine, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 18Medical Oncology, Washington University in St. Louis, St. Louis, MO, 19Clinical Development, A2 Biotherapeutics, Inc., Agoura Hills, CA, 20Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL.
Background: Despite the success treating hematologic malignancies, chimeric antigen receptor T-cell (CAR T) therapies face challenges in breast cancer and other solid tumors due to lack of targets that distinguish tumor from normal cells. Epithelial growth factor receptor (EGFR) plays a critical role in oncogenesis across several cancers and is often upregulated [1]. While monoclonal antibodies targeting EGFR have demonstrated efficacy, these approaches are often limited by on-target, off-tumor toxicities, such as skin and gastrointestinal toxicity, which constrains dose escalation and efficacy [2]. A2B395 is an allogeneic, logic-gated, EGFR-targeted, TmodTM CAR T therapy designed to address these limitations and provide a convenient and consistent off-the-shelf option. This therapy incorporates 2 CARs: an activator targeting EGFR and a blocker targeting human leukocyte antigen (HLA)-A*02. The activator recognizes EGFR on both tumor and normal cells, whereas the blocker inhibits CAR T activity against normal cells (with preserved HLA expression) while allowing activation against tumor cells (with HLA-A*02 loss of heterozygosity [LOH]). This approach is designed to avoid on-target, off-tumor toxicity, a significant limitation to CAR T treatment of solid tumors [3]. To address potential graft-vs-host response, a short-hairpin RNA expression module targeting beta-2 microglobulin is included in the Tmod construct, which significantly reduces major histocompatibility complex class I levels and subsequent host immune response [4]. Importantly, the Tmod system is modular and adaptable to multiple targets. Initial data on autologous Tmod CAR T therapy suggest reduced off-tumor toxicity and encouraging clinical efficacy [5]. A2B395 represents a novel approach for targeting EGFR-expressing solid tumors with HLA-A*02 loss of heterozygosity.Methods: DENALI-1 is a phase 1/2, open-label, nonrandomized study evaluating the safety and efficacy of A2B395 in adults. Patients are enrolled through BASECAMP-1 (NCT04981119), a master prescreening study that identifies patients with HLA loss of heterozygosity at any time in the course of their disease via next-generation sequencing. Key inclusion criteria include histologically confirmed recurrent, unresectable, locally advanced, or metastatic cancers associated with EGFR expression, including triple-negative breast, colorectal, non-small cell lung, squamous cell head and neck, and renal cell cancers. Patients must have received ≥1 line of prior therapy, such as a checkpoint inhibitor, molecular targeted therapy, or chemotherapy. The primary objective of the phase 1 portion is to evaluate safety, tolerability, and the recommended phase 2 dose (RP2D) using a Bayesian optimal interval design for dose escalation. The dose-expansion phase will confirm the RP2D and collect and analyze biomarker data. The Phase 2 portion will assess overall response rate per RECIST v1.1. As of 03 July 2025, the first patient dosed with A2B395, who has breast cancer, completed the 28-day safety review period. Dose-escalation is ongoing.
Presentation numberPS5-08-04
A phase 1b, multi-centre, open-label, randomized study to evaluate the safety, tolerability, and clinical activity of combining paxalisib with olaparib or pembrolizumab/chemotherapy in patients with advanced breast cancer
Sudha Rao, QIMR Berghofer Medical Research Institute, Herston, Australia
M. Nottage1, M. Melino2, A. Bain2, W. Tu2, S. Goh2, K. Houston3, J. Sanmugarajah4, J. Friend5, S. Rao2; 1Medical Oncology, The Royal Brisbane and Women’s Hospital, Herston, AUSTRALIA, 2Cancer Research, QIMR Berghofer Medical Research Institute, Herston, AUSTRALIA, 3Medical Oncology, Sunshine Coast Hospital and Health Service, Birtinya, AUSTRALIA, 4Medical Oncology, The Gold Coast University Hospital, Southport, AUSTRALIA, 5Kazia Therapeutics Ltd, Kazia Therapeutics Ltd, Sydney, AUSTRALIA.
Background PaxPlus-ABC (ACTRN12624001340527) is a Phase 1b open-label, parallel-group study evaluating the dual PI3K-mTOR inhibitor paxalisib in women with advanced breast cancer. Previously presented preclinical data (SABC 2024 abstract SESS-2122) support the rationale for combining paxalisib with agents such as olaparib or pembrolizumab plus chemotherapy in this setting (Melino, Tu et al 2025). In this presentation, we will share additional preclinical findings from our liquid biopsy biomarker analyses and provide an updated overview of the ongoing clinical trial. Methods Preclinical: Liquid biopsy biomarker analysis was used to evaluate the efficacy of paxalisib in combination with either immunotherapy or olaparib in the 4T1 triple-negative breast cancer (TNBC) mouse model. Circulating tumor cells (CTCs) were characterized through detailed immunofluorescence profiling, while immune cell interactions and spatial dynamics within the tumor microenvironment were assessed using PhenoCycler-Fusion (CODEX®) spatial proteomics technology. Clinical: This Phase 1b open-label, parallel group study has two arms. Arm A (paxalisib + olaparib) will enrol patients with germline BRCA-mutated HER2-negative metastatic breast cancer previously treated with chemotherapy. Patients will receive olaparib (300 mg BID) plus daily paxalisib (15 mg or 30 mg) in 28-day cycles. Arm B (paxalisib + pembrolizumab/chemotherapy) will enrol patients with recurrent, unresectable, or metastatic triple-negative breast cancer with PD-L1 CPS ≥10. Patients will receive daily paxalisib (15 mg or 30 mg) in 21-day cycles, pembrolizumab (200 mg IV every 3 weeks), and standard-of-care chemotherapy (investigator’s choice: nab-paclitaxel or gemcitabine-carboplatin). Primary objectives are to assess the safety, tolerability, and establish a recommended Phase 2 dose of paxalisib in both combinations. Secondary objectives include evaluating liquid biopsy markers (circulating tumour cells, immune cell signatures, reinvigoration/exhaustion scoring) using digital pathology and flow cytometry. Clinical efficacy measures include progression-free survival, overall response rate, clinical benefit rate, time to response, time to progression, and overall survival. Results Preclinical: Dual inhibition of PI3K and mTOR using paxalisib in combination with either anti-PD-1 immunotherapy or the PARP inhibitor olaparib significantly reduced circulating tumor cell (CTC) counts in the 4T1 triple-negative breast cancer (TNBC) mouse model. Further characterization revealed a marked decrease in the aggressive, mesenchymal CTC phenotype (CD45⁻/VIM⁺/SNAIL⁺). Spatial profiling with PhenoCycler-Fusion (CODEX®) uncovered distinct differences in immune cell localization between the paxalisib + anti-PD-1 and paxalisib + olaparib treatment groups, highlighting divergent patterns of immune cell-tumor microenvironment interactions. Clinical: Upcoming data will include updated liquid biopsy biomarker analyses, encompassing CTC enumeration and phenotyping, as well as profiling of immune cell exhaustion and reinvigoration signatures. Conclusions Our preclinical studies demonstrated that combining paxalisib with either immunotherapy or PARP inhibition significantly reduced CTC burden and reshaped the immune landscape, leading to a decrease in metastasis. These promising findings have advanced into a Phase 1b clinical trial in patients with advanced breast cancer, with updated analysis to be presented at this meeting. References Melino, M., Tu, WJ et al. (2025). Molecular Cancer Therapeutics PMID: 40497697
Presentation numberPS5-08-05
Predictive Role of the Gut Microbiome in Abemaciclib-Associated Diarrhea: A Prospective Translational Pilot Study
Soo Jung Lee, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea, Republic of
S. Lee1, H. Park2, I. LEE1, Y. Chae1, J. Lee3, B. Kang3, H. Park3, J. Moon3; 1Department of Oncology/Hematology, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF, 2Division of Allergy and Clinical Immunology, Department of Internal Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF, 3Department of Breast Surgery, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF.
Background: Abemaciclib, a CDK4/6 inhibitor, is approved for the treatment of both early-stage and metastatic HR+/HER2− breast cancer. While it improves survival outcomes, diarrhea remains the most frequent and dose-limiting adverse event, often resulting in treatment interruption, dose reduction, or discontinuation. Maintaining full-dose intensity is particularly crucial in the metastatic setting, where drug exposure is directly correlated with efficacy. Preclinical studies suggest that abemaciclib-induced gastrointestinal (GI) toxicity may result from off-target inhibition of GSK3β, leading to activation of the Wnt/β-catenin pathway and epithelial hyperproliferation. Meanwhile, the gut microbiome has emerged as a key regulator of mucosal integrity, immune homeostasis, and drug response. Inter-individual variability in microbiome composition and function may modulate susceptibility to abemaciclib-related GI toxicity. We hypothesize that baseline gut microbiome characteristics are associated with the incidence, severity, and trajectory of abemaciclib-induced diarrhea. Identification of predictive microbial features may inform microbiome-modulating strategies to improve tolerability and maintain full-dose therapy. Methods: This is a prospective, double-arm pilot translational study enrolling 90 patients with HR+/HER2− breast cancer initiating CDK4/6 inhibitor therapy. Participants will be assigned to two cohorts: one receiving abemaciclib, and the other receiving a comparator CDK4/6 inhibitor with minimal GI toxicity (e.g., palbociclib or ribociclib). The primary objective is to evaluate the association between baseline gut microbiome composition and function with the severity of abemaciclib-induced diarrhea. Stool samples will be collected at baseline (pre-treatment) and one month after treatment initiation. Comprehensive multi-omic analyses will be performed, including shotgun metagenomics for microbial genome reconstruction and functional pathway annotation using tools such as KEGG, MetaCyc, and MAC. Stool metabolomic profiling will be conducted to elucidate potential mechanistic pathways. Systemic biomarkers such as circulating cytokines, intestinal fatty acid-binding protein (I-FABP), and lipopolysaccharide-binding protein (LBP) will also be assessed to evaluate epithelial damage and microbial translocation. The primary endpoint is the association between baseline microbiome features and diarrhea severity, graded using CTCAE v5.0. Secondary endpoints include longitudinal microbiome changes, treatment adherence, dose intensity, and patient-reported GI symptoms. Exploratory analyses will identify microbial taxa and metabolic pathways linked to susceptibility or resilience to GI toxicity. This pilot study aims to elucidate the role of the gut microbiome in modulating abemaciclib-associated diarrhea and to identify predictive biomarkers of toxicity. Findings may support the development of microbiome-informed strategies to enhance drug tolerability and maintain therapeutic intensity in HR+/HER2− breast cancer.
Presentation numberPS5-08-06
A prospective, multicenter comparative pilot study to evaluate targeting axillary lymph node surgery for patients with clinically node-positive breast cancer (LUCAS study, KBCSG36)
Joon Suk Moon, Kyungpook National University, Daegu, Korea, Republic of
J. Lee1, H. Park1, B. Kang1, J. Moon1, Y. Chae2, L. Soo Jung2, L. In Hee2, J. Park3, N. Park3, E. Kim4, S. Jeong4, J. Kang4, J. Yang5, Y. Lim1, T. Jung6; 1Surgery, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 2Department of Oncology/Hematology, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 3Pathology, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 4Breast Cancer Pecision Medicine Institute, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 5Plastic and Recontructive Surgery, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 6Rehabilitation Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF.
Background: Accurate targeting of axillary lymph nodes (ALNs) is important in breast cancer surgery, especially in patients with ALNs metastasis. Conventional methods such as charcoal tattooing, needle localization, clips or skin marking with ultrasound have significant limitations: incorrect targeting, rapid dispersion, irreversible pigmentation, or dislocation, complicating precise localization. LuminoMark, a novel near-infrared fluorescence (NIRF) imaging agent combining indocyanine green (ICG) with a dispersion-limiting polymer, offers real-time visualization of ALNs without staining or dislocation risk. Methods: This multicenter, prospective study aims to evaluate the safety and efficacy of LuminoMark compared to standard ALN-targeting methods (charcoal tattooing, needle localization, and ultrasound-guided skin marking). The study includes 330 breast cancer patients (165 per group), including those undergoing upfront surgery or surgery following neoadjuvant chemotherapy. The experimental group receives a 0.2-mL subcutaneous injection of LuminoMark into pathologically confirmed metastatic ALNs prior to surgery, while the control group follows institutional standard targeting procedures. The primary outcome is the successful detection rate of the targeted lymph node and check-up of adverse events, and secondary outcomes include time from incision to node detection, total axillary surgical time, postoperative complications, and correlation with sentinel lymph nodes. A prior Phase 2 trial showed no adverse drug reactions or significant differences in efficacy between doses of LuminoMark® and charcoal. A preliminary study involving 13 patients demonstrated a 100% detection rate with LuminoMark® compared to 92.3% with charcoal, with superior intraoperative visualization and no adverse events. The inclusion criteria include women aged 20-70 with invasive breast cancer and clinically/pathologically confirmed ALN metastasis. Exclusion criteria include stage IV disease, pregnancy, history of allergic reactions to the study drug, or inability to comply with study protocols. All participants undergo postoperative monitoring for 48 hours for safety evaluation. Randomization will be performed preoperatively in upfront surgery cases, or after neoadjuvant chemotherapy in neoadjuvant treatment cases. All procedures adhere to NCCN guidelines, which recommend excision of targeted ALNs. In a real comparative study, if LuminoMark® (100% detection rate) is set as experimental group and the various conventional methods (approximately 89.7%) is set as control group, approximately 1,480 subjects are required for each group. However, this study is composed as pilot study including only 10% of the actual required sample size, and the dropout rate was set to 10%. Therefore, 165 patients (164.4 patients) are needed in each group. Since the control group is composed of methods currently being implemented in each institution (charcoal dyeing, needle positioning, ultrasound-guided skin marking, etc.), the number of patients for each method is expected to be different. However, to configure the ratio as close to 1:1:1 as possible, the number of cases will be assigned differently for each institution. This study seeks to confirm its utility in a clinical setting and support future large-scale trials to establish LuminoMark as a feasible material in targeting ALNs for breast cancer surgery. The full protocol of this study was registered in clinicaltrial.gov (NCT06903429).
Presentation numberPS5-08-07
Open-label Single-arm Phase 2 Trial of Trastuzumab Deruxtecan in Previously Treated HER2-Immunohistochemistry (IHC) 0 Advanced Breast Cancer – HER2 PARADIGM trial
Adriana Kahn, Yale University, New Heaven, CT
A. Kahn1, J. Du1, N. Casasanta1, S. Schellhorn1, M. Digiovanna1, T. Sanft1, M. Rozenblit1, A. Silber1, L. Pusztai1, Z. Rahman1, D. O’Neil1, R. Legare1, W. Kidwai1, J. Kanowitz1, A. Bulgaru1, N. Fischbach1, S. Hall1, K. Fenn1, K. Blenman1, O. Blaha2, E. Winer1, I. Krop1, D. Rimm3, M. Lustberg1; 1Internal Medicine (Medical Oncology), Yale University, New Heaven, CT, 2Biostatistics, Yale University, New Heaven, CT, 3Pathology, Yale University, New Heaven, CT.
Background: Trastuzumab-deruxtecan (T-DXd) showed impressive efficacy in patients whose breast cancers are either human epidermal growth factor receptor 2 (HER2) immunohistochemistry (IHC) positive or HER2-low/ultralow. In a phase 2 trial, patients with HER2-IHC 0 who received T-DXd had an objective response rate of approximately 30%, revealing activity regardless of HER2-IHC status. In this study, we will treat patients with advanced HER2 IHC 0 breast cancers with T-DXd and assess biomarkers of response and resistance to therapy. We will investigate patients’ tumor biopsies for more precise quantitative HER2 tests, including the High-Sensitivity (HS)-HER2 Troplex testing, that could help determine if T-DXd is active in patients with HER2-IHC 0 tumors regardless of HER2 quantitative expression or if there is a potentially optimal cutoff value of HER2 expression with respect to clinical outcomes for response, improving patient selection and prevention of unnecessary toxicity. Methods: This is a non-randomized, single-arm, open-label, phase 2 study to assess the efficacy and safety of T-DXd in patients whose breast cancer is HER2-IHC 0 (including 0-null and 0-ultralow) in all prior biopsies, with unresectable and/or metastatic disease regardless of hormone receptor (HR) status (NCT06750484). Key eligibility criteria include ECOG-PS 0-2; up to 2 prior lines of systemic cytotoxic therapy for treatment in the metastatic setting; no upper limit of prior endocrine, immunologic or targeted therapy lines. Fifty subjects will be included to achieve 82% power to detect a difference in ORR of 0.15 from historical control (ORR in experimental arm of 0.3 vs. ORR in historical control of 0.15) using a two-sided exact test with a target significance level of 0.1. The study treatment with T-DXd administered intravenously every 3 weeks at a dose of 5.4 mg per kilogram of body weight will be continued in the absence of withdrawal of subject consent, progressive disease (PD), death, or unacceptable toxicity. Patients will be followed and evaluated by RECIST v1.1 criteria. Objective response to T-DXd will be evaluated in the entire study population and separately in the High-Sensitivity (HS)-HER2 Troplex detectable and non-detectable cohorts (defined by the limit of detection [LOD] of the analytic HS-HER2 Troplex assay). In addition, HER2 tissue concentrations (measured by the HS-HER2 Troplex assay in attomole/mm2) will be plotted and analyzed as a function of response to determine levels of tumor expression and potential cut-points associated with benefit from T-DXd. Since the Troplex assay simultaneously provides TROP2 levels in attomole/mm2, the influence of TROP2 levels on response to T-DXd will also be exploratorily investigated. Additional objectives are to assess efficacy in terms of progression-free survival and overall survival, safety, and explore potential biomarkers of response and resistance to therapy with T-DXd. The trial is currently open and enrolling patients.
Presentation numberPS5-08-08
A phase II neoadjuvant clinical trial of the androgen receptor inhibitor darolutamide in early-stage androgen receptor positive (AR+) triple-negative breast cancer (NCT 07016399)
Lila Duke, Vanderbilt University Medical Center, Nashville, TN
L. Duke, J. M. Sharpe, B. Lehmann, C. Brothers, J. Pietenpol, V. G. Abramson; Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN.
Background: Neoadjuvant chemotherapy combined with immunotherapy is the standard of care in the treatment of early-stage triple-negative breast cancer (TNBC) with pathological complete (pCR) rates of approximately 65%. Though TNBC is currently treated as one disease, TNBC is transcriptionally diverse with at least four biological subtypes including basal-like (BL1, BL2), mesenchymal (M) and luminal androgen receptor (LAR) subtypes. The LAR subtype is characterized by androgen receptor (AR)-regulation and luminal gene expression in tumor specimens. Up to 20% of TNBCs are the LAR subtype, and compared to other TNBCs, they are less proliferative, less responsive to chemotherapy, more genomically stable, and are typically diagnosed in older patients. Studies in the metastatic setting have revealed that chemotherapy with/without immunotherapy is not an optimal treatment strategy for LAR tumors. Darolutamide is a potent second-generation AR inhibitor approved for the treatment of prostate cancer. While earlier studies using AR inhibitors in breast cancer showed limited effectiveness, these studies used IHC cut-offs for AR that were below the threshold to capture true LAR tumors and often used less potent AR inhibitors. Ki-67 is a marker of cell proliferation and is validated as a prognostic and predictive marker in estrogen receptor positive breast cancer. Decreases in Ki-67 after two weeks of endocrine therapy correspond with improved outcomes to anti-estrogen therapy. Treatment with the AR inhibitor enzalutamide has been shown to reduce Ki-67 levels in luminal AR tumors. We hypothesize that targeting AR with darolutamide in the neoadjuvant setting and using Ki-67 as a biomarker of response will identify breast cancers responsive to AR inhibitors. Methods: Fifty-one patients with a histologically-confirmed new diagnosis of stage II-IIIa TNBC that is AR positive (>= 80% staining by IHC) will be randomized to receive neoadjuvant standard of care (SOC) chemotherapy and immunotherapy for TNBC. Patients will be randomized 2:1 to receive either the oral androgen inhibitor darolutamide for 14 days followed by darolutamide in combination with SOC treatment for 24 weeks (34 patients) or receive 24 weeks of SOC treatment alone (17 patients). Eligible patients must be appropriate for neoadjuvant treatment, have an ECOG PS of 0-1, and must not have non-resectable or metastatic breast cancer. Biopsies will be taken at baseline, 2 weeks after starting darolutamide or SOC chemotherapy (both arms), 2 weeks after starting darolutamide and SOC chemotherapy (intervention arm only), and at surgery. Blood draws for ctDNA analysis will be taken at the same time points. The primary endpoint is change in Ki-67 level between baseline and post-treatment measurements. This study has 80% power to detect a biologically and clinically significant effect size of 1 with a two-sided type I error of 5%. Secondary endpoints include pCR, EFS, and their correlation to changes in Ki-67 as well as percent nuclear AR positivity. Exploratory objectives include assessing changes in ctDNA levels while undergoing neoadjuvant therapy for TNBC as well as investigating additional AR characteristics and mechanisms of resistance to AR inhibitors. This study is open at Vanderbilt University Medical Center and will be opening at 6 additional sites.
Presentation numberPS5-08-09
A Pilot, Single-Arm, Phase II Trial of Tamoxifen plus Pegylated Liposomal Doxorubicin in Patients with Metastatic Triple Negative Breast Cancer
Zunairah Shah, Roswell Park Cancer Center, Buffalo, NY
Z. Shah1, C. C. Oturkar2, I. Aijaz2, H. Yu3, M. L. Tarquini1, G. M. Das2, S. kabraji1; 1Medicine, Roswell Park Cancer Center, Buffalo, NY, 2Pharmacology & Therapeutics, Roswell Park Cancer Center, Buffalo, NY, 3Biostatistics & Bioinformatics, Roswell Park Cancer Center, Buffalo, NY.
Background: Triple-negative breast cancer (TNBC) accounts for approximately 15-20% of all breast cancers and disproportionately contributes to breast cancer mortality due to its aggressive clinical course, high propensity for visceral and brain metastases, and lack of targeted therapies. While TNBC lacks estrogen receptor alpha (ERα), up to 80% of tumors express estrogen receptor beta (ERβ), and a similar proportion harbor TP53 mutations. Pioneering work by our group demonstrated that ERβ directly interacts with p53, exerting tumor-suppressive effects in the presence of mutant p53 (mut-p53), but potential oncogenic effects in wild-type p53 settings (Mukhopadhyay et al., J Natl Cancer Inst, 2019). We have also showed that tamoxifen (Tam), a selective estrogen receptor modulator, stabilizes ERβ and enhances its binding to mut-p53, sequestering it away from TP73 and restoring TP73’s tumor suppressor activity. In vitro, Tam synergistically enhances the cytotoxicity of doxorubicin in ERβ/mut-p53 positive TNBC cell lines, while in vivo xenograft models demonstrate superior tumor regression with the combination. Therapeutic proof-of-concept was observed in a patient with metastatic Erβ/mut-p53 expressing TNBC who had marked tumor regression following Tam treatment (Scarpetti et al., The Oncologist, 2023). Given the frequent co-expression of ERβ and mut-p53 in TNBC and the favorable safety profile of tamoxifen and pegylated liposomal doxorubicin (PLD), we have initiated a phase II clinical trial evaluating this combination in patients with metastatic(m) or unresectable TNBC. Methods: This is an open-label, single-arm, phase II study (NCT06434064) enrolling patients(pts) ≥18 years with mTNBC characterized by ERα-low (≤10%) expression and progression on ≥2 prior lines of systemic therapy. Pts will receive Tam 20 mg orally once daily and PLD 50mg/m² intravenously every 28 days. Treatment continues until disease progression, unacceptable toxicity, or patient withdrawal. Tam is continued unless dose-limiting toxicity occurs, as no dose reductions are allowed. The primary endpoint is overall response rate (ORR) as assessed by RECIST v1.1. Secondary endpoints include progression-free survival (PFS), overall survival (OS), duration of response (DOR), and safety/tolerability per CTCAE v5.0. Exploratory endpoints include changes in circulating tumor DNA (ctDNA), ERβ-mut-p53 interaction assessed by proximity ligation assay (PLA), differential gene expression analysis by RNA-seq, immune deconvolution via CIBERSORT, and peripheral immune profiling through cytokine analysis and flow cytometry. A safety lead-in will be conducted in the first 6 pts. The study uses a Simon two-stage design to detect an increase in ORR from 16% to 33%, with 80% power and α = 0.10 (n1 = 12, n2 = 18). This design ensures early stopping for futility and supports efficient evaluation of efficacy. Technical and analytic validation of ER-beta expression as a possible biomarker of response is ongoing. Significance: If successful, this study will establish a novel, mechanism-driven treatment option for previously treated mTNBC. Correlative analyses will uncover molecular and immune pathways linked to therapeutic response. Erβ and mut-p53 interaction profiling may support future patient selection for Tam based therapies. Future studies will explore potential synergy between antibody-drug conjugates that induce DNA-damage e.g. Sacituzumab govitecan and Tam as well as adding immune checkpoint inhibition to Tam and Doxil to improve patient outcomes. Status: The study is currently open to enrollment. Clinical trial information: (NCT 06434064)
Presentation numberPS5-08-10
Phase III study to evaluate the efficacy and safety of GLSI-100 (GP2 + GM-CSF) in breast cancer patients with residual disease or high-risk PCR after both neo-adjuvant and postoperative adjuvant anti-HER2 therapy, Flamingo-01
Snehal Patel, Greenwich LifeSciences, Stafford, TX
S. Patel1, J. Thompson1, M. Patel1, J. Daugherty1, M. Rimawi2; 1Clinical Research, Greenwich LifeSciences, Stafford, TX, 2Hematology and Oncology, Baylor College of Medicine, Houston, TX.
Background: GP2 is a biologic nine amino acid peptide of the HER2/neu protein delivered in combination with Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) that stimulates an immune response targeting HER2/neu expressing cancers, the combination known as GLSI-100. Of the 146 patients that have been treated with GLSI-100 over 4 clinical trials, GLSI-100 was well-tolerated and no serious adverse events observed were considered related to the immunotherapy. Methods: This Phase III trial is a prospective, randomized, double-blinded, multi-center study. After 1 year of trastuzumab-based therapy, 6 intradermal injections of GLSI-100 or placebo will be administered over the first 6 months and 5 subsequent boosters will be administered over the next 2.5 years. The participant duration of the trial will be 3 years treatment plus 1 additional year follow-up. Study Size – Interim Analysis: Approximately 498 patients will be enrolled. To detect a hazard ratio of 0.3 in invasive breast cancer free survival (IBCFS), 28 events will be required. An interim analysis for superiority and futility will be conducted when at least 14 events have occurred. This sample size provides 80% power if the annual rate of events in placebo patients is 2.4% or greater. Up to 250 non-HLA-A*02 subjects will be enrolled in an open-label arm. Eligibility Criteria: The patient population is defined by these key eligibility criteria: 1) HER2/neu positive and HLA-A*02, 2) Residual disease or High risk pCR (Stage III at presentation) post neo-adjuvant therapy, 3) Exclude Stage IV, and 4) Completed at least 90% of planned trastuzumab-based therapy. Trial Objectives: The trial objectives are to: 1) Determine if GP2 therapy increases IBCFS, 2) Assess the safety profile of GP2, and 3) Monitor immunologic responses to treatment and assess relationship to efficacy and safety. Study Status: The study is actively recruiting and enrolling patients in the US and Europe at up to 150 sites. Contact Information: Greenwich LifeSciences, Inc. Stafford, TX. Email: Flamingo-01@greenwichlifesciences.com. Website: greenwichlifesciences.com. Funding: This trial is supported by Greenwich LifeSciences.
Presentation numberPS5-08-11
Trial in Progress: Phase II Open-Label Study of ARX788 (anti-HER2 Antibody Drug Conjugate (ADC)) for Patients with HER2-Low Locally Advanced Unresectable or Metastatic Breast Cancer
Laura A Huppert, University of California, San Francisco, San Francisco, CA
L. A. Huppert1, N. D. Pasricha2, K. Nathalie1, C. Galia1, J. Rossi1, A. DeLuca1, K. Natsuhara1, H. Batra-Sharma1, M. Majure1, M. Melisko1, J. Chien1, H. Rugo3; 1Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, 2Ophthalmology, University of California, San Francisco, San Francisco, CA, 3Department of Medical Oncology & Therapeutics Research, Division of Breast Medical Oncology, City of Hope, Duarte, CA.
Background: ARX788 is a next-generation, site-specific ADC composed of a monoclonal antibody targeting human epidermal growth factor receptor 2 (HER2) conjugated via a non-natural amino acid linker to a potent tubulin inhibitor payload AS269. The phase I ACE-Breast-01 and ACE-Pan Tumor-01(NCT03255070) studies previously evaluated the safety and efficacy of ARX788 in patients with HER2-positive breast cancer and pan tumor types respectively, with promising efficacy results and with associated ocular adverse events. The clinical benefit of HER2-targeted ADCs for patients with HER2-low breast cancer (HER2 IHC 1+ or 2+ and FISH/ISH negative) has been established for other HER2-targeted ADCs; novel treatment strategies in this population remain an area of high unmet clinical need. Therefore, we hypothesized that ARX788 will also be safe and efficacious for the treatment of patients with HER2-low locally advanced unresectable or metastatic breast cancer, including in patients who have progressed on prior chemotherapies and ADCs. Methods: This is a single-arm, open-label, Phase II clinical trial of ARX788 monotherapy in patients with HER2-low locally advanced unresectable or metastatic breast cancer (NCT06224673). Key eligibility criteria include patients with locally advanced unresectable or metastatic breast cancer, measurable disease, local confirmation of HER2-low status, and receipt of at least one prior line of chemotherapy or ADC therapy in the metastatic setting (no limit on prior lines of therapy). ARX788 monotherapy will be administered at a dose of 1.5mg/kg intravenously every three weeks until progressive disease or intolerable side effects. A total of 30-36 patients will be enrolled in one of two cohorts: 1) hormone receptor (HR) positive/HER2-low (n=20-24) and 2) HR-negative/HER2-low (n=10-12). The primary endpoint is objective response rate (ORR); The proposed sample size will provide ORR estimates with margin of error of 14% and 18% in each cohort respectively at 90% confidence level with anticipated 25% ORR. Secondary endpoints include duration of response (DOR), best overall response (BOR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and efficacy of an eye toxicity prevention regimen to prevent grade 2 or higher ocular toxicity. Exploratory endpoints include blood- and tumor-based biomarker analyses and patient-reported outcomes (PROs). Enrollment to all cohorts will begin in the third quarter of 2025.
Presentation numberPS5-08-12
Start of SerMa – EUBREAST 5 (Seroma of the Mammary Gland) study (NCT05899387)
Nina Ditsch, University Augsburg, Augsburg, Germany
N. Ditsch1, M. Koepke1, N. Pochert2, U. Jeschke1, M. Schneider1, M. Reiger2, C. Traidl-Hoffmann2, Y. Masannat3, A. Mattmer1, S. Hunstiger1, J. Sagasser1, C. Dannecker1, M. Wild1, C. Hinske4, M. Hauptmann5, S. Hartmann6, H. Kahl7, D. Krug8, N. Krawczyk9, K. Stemmer10, D. Rezek11, A. Raue12, M. Gasparri13, R. Reitsamer14, J. Jelinska15, G. Cakmak16, E. Bonci17, R. Wuerstlein18, V. Fink19, V. Bjelic-Radisic20, M. Thill21, C. Heitmann22, W. Weber23, E. Stickeler24, A. Lebeau25, P. A. Fasching26, H. Kolberg27, T. Reimer6, L. Bauer28, U. Beckmann29, B. Aktas30, S. Hasmueller31, U. Schlembach32, R. Di Micco33, J. De Boniface34, O. Gentilini33, M. Untch35, T. Kühn36, M. Banys-Paluchowski37; 1Clinic for Gynecology and Obstetrics, University Augsburg, Augsburg, GERMANY, 2Institute of Environmental Medicine and Integrative Health, University Augsburg, Augsburg, GERMANY, 3Breast Surgery, The London Clinic, London, GERMANY, 4Digital Medicine, University Augsburg, Augsburg, GERMANY, 5Institute of Biostatistics and Registry Resarch, Brandenburg Medical School Theodor Fontane, Neuruppin, GERMANY, 6Clinic for Gynecology and Obstetrics, University Rostock, Rostock, GERMANY, 7Clinic for Radiotherapy, University Augsburg, Augsburg, GERMANY, 8University Medical Center / Clinic for Radiooncology, University Hamburg-Eppendorf, Hamburg, GERMANY, 9Clinic for Gynecology and Obstetrics, University Düsseldorf, Düsseldorf, GERMANY, 10Molecular Cell Biology / Institute of theoretical Medicine, University Augsburg, Augsburg, GERMANY, 11Clinic for Gynecology and Obstetrics, Gesundheitscampus Wesel, Wesel, GERMANY, 12Modeling and Simulation of Biological Processes, University Augsburg, Augsburg, GERMANY, 13Clinic for Gynecology and Obstetrics, Ospedale Regionale di Lugano, Lugano, SWITZERLAND, 14Clinic for specialized Gynecology, University Salzburg, Salzburg, AUSTRIA, 15Clinic for Surgery, University Zielonogorski, Zielonogorski, POLAND, 16General Surgery Department, Breast and Endocrine Unit, Zonguldak BEUN The School of Medicine, Zonguldak, TURKEY, 17Department of Surgical Oncology, University Cluj-Napoca, Cluj-Napoca, ROMANIA, 18Clinic for Gynecology and Obstetrics / Breast Cancer and CCC Munich, University LMU München, München, GERMANY, 19Clinic for Gynecology and Obstetrics, University Ulm, Ulm, GERMANY, 20Department for Human Medicine, University Witten-Herdecke, Witten-Herdecke, GERMANY, 21Clinic for Gynecology and Obstetrics, Agaplesion Markus Hospital Frankfurt, Frankfurt, GERMANY, 22Operative therapy of the Breast GmbH, SENO MVZ München, München, GERMANY, 23Clinic for Gynecology and Obstetrics, University Basel, Basel, SWITZERLAND, 24Clinic for Gynecology and Obstetrics, University Aachen, Aachen, GERMANY, 25Institute of Pathology, University Hamburg-Eppendorf, Hamburg, GERMANY, 26Clinic for Gynecology and Obstetrics, University Erlangen, Erlangen, GERMANY, 27Clinic for Gynecology and Obstetrics, Marienhospital Bottrop, Bottrop, GERMANY, 28Brustzentrum, Hospital of Weinheim, Weinheim, GERMANY, 29Clinic for Gynecology and Obstetrics, Niels-Stensen-Kliniken Harderberg, Harderberg, GERMANY, 30Clinic for Gynecology and Obstetrics, University Leipzig, Leipzig, GERMANY, 31Clinic for Gynecology and Obstetrics, Hospital Ebersberg, Ebersberg, GERMANY, 32Clinic for Gynecology and Obstetrics, Caritas-Krankenhaus Bad Mergentheim, Bad Mergentheim, GERMANY, 33Breast Surgery Unit, San Raffaele University and Research Hospital, University Milan, Milan, ITALY, 34Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Stockholm, SWEDEN, 35Clinic for Gynecology and Obstetrics, Helios Klinik Berlin-Buch, Berlin, GERMANY, 36Clinic for Gynecology and Obstetrics, Filderklinik, Fildertstadt-Bonlanden, Filderstadt-Bonlanden, GERMANY, 37Clinic for Gynecology and Obstetrics, University Schleswig-Holstein, Lübeck, Lübeck, GERMANY.
Background The standard local therapy for breast cancer is surgery. Postoperative seroma formation is one of the most common and serious complications after breast surgery for primary breast cancer, especially in patients undergoing mastectomy with or without implant-based breast reconstruction. Seromas may lead to infections and wound dehiscence, which can result in loss of reconstruction, ugly scars and poor cosmetic outcome.. To date, the cause of seroma development has not yet been clarified. Trial Design The main objective of the multicenter SerMa study is to identify a subgroup of patients with an increased risk of developing seromas based on immunological or inflammatory markers in a planned cohort of 2200 subjects. The study is designed as international, prospective cohort study in cooperation with the EUBREAST Network (European Breast Cancer Research Association of Surgical Trialists). articipating study groups are AGO-B, AWOgyn and OPBC. Patients with primary breast cancer or ductal carcinoma in situ (DCIS) planned for mastectomy with or without reconstruction can participate in this study (study group 1 and 2). Study group 3 includes patients at high risk for breast cancer planned for risk-reducing subcutaneous mastectomy and implant-based reconstruction. Study group 4 includes healthy women with implant insertion for exclusively cosmetic reasons. Currently, the logistics phase with 50 patients has been completed, which was carried out to optimize the preservation of blood samples and seroma fluid. Primary endpoints: To achieve the primary endpoint – the identification of a patient group with an elevated risk of developing a seroma – it is planned to examine the groups with and without postoperative seroma formation regarding immunological markers in seroma fluid as well as in blood samples. In addition, local microbiome analyses, as well as tumor analyses with focus on microenvironment will be performed to differentiate possible carcinoma-specific immunological processes. Secondary endpoints: To answer the question whether differences are caused by immunological or cancer-related reasons, groups with and without postoperative seroma formation as well as groups with or without breast cancer are compared. Current status of the study: Following the current finalization of the logistics phase the study is open for recruitment, The study is currently expanding worldwide. Target accrual: 2200 participants in total.
Presentation numberPS5-08-13
Reevaluating Follow-Up in Early Breast Cancer, guided by Liquid Biopsy: the SURVIVE Study (NCT05658172)
Kerstin Pfister, University Hospital Ulm, Ulm, Germany
S. T. Huesmann1, T. W. Friedl1, K. Pfister1, H. Schäffler1, A. Fink1, F. Mehmeti1, S. Heublein1, A. Hartkopf2, V. Müller3, L. Wiesmüller1, T. Fehm4, K. Pantel5, P. Fasching6, A. Chevalier7, C. Pipinikas7, B. Rack1, W. Janni1; 1Gynecology, University Hospital Ulm, Ulm, GERMANY, 2Gynecology, University Hospital Tübingen, Tübingen, GERMANY, 3Gynecology, University Hospital Hamburg Eppendorf, Hamburg, GERMANY, 4Gynecology, University Hospital Düsseldorf, Düsseldorf, GERMANY, 5Institute of Tumor Biology, University Hospital Hamburg Eppendorf, Ulm, GERMANY, 6Gynecology, University Hospital Erlangen, Erlangen, GERMANY, 7Clinical Programs, NeoGenomics Inc., Fort Myers, FL.
Background: For decades, international guidelines unanimously recommend to limit routine follow-up in early breast cancer (EBC) to clinical exams and breast imaging, to detect potentially curable local relapse early. Additionally, guidelines explicitly advise against screening for distant recurrence in asymptomatic patients, as there remains no survival benefit in patients with earlier detected metastatic disease, at the cost of quality of life (1). However, as most of this data was published over 30 years ago, a re-evaluation of EBC follow-up strategies is urgently needed, considering numerous novel treatment options for both EBC and metastatic breast cancer patients. Study Design:The SURVIVE study is a prospective, multicenter, randomized controlled trial investigating a liquid-biopsy based follow-up in intermediate- to high-risk EBC patients. 3.500 EBC patients after completion of primary therapy (surgery, chemo- or radiotherapy) are randomized 1:1 to either standard or liquid-biopsy guided (intensified) follow-up. All patients receive guideline-based follow-up procedures. Additionally, blood samples are taken every 3 months (years 1-3), later every 6 months (years 4-5). While samples from patients in the standard-arm are biobanked, samples from the interventional arm are analyzed longitudinally for tumor markers (CA 27.29, CEA, CA 125), CTCs (CellSearch®, Menarini), and ctDNA (RaDaR®, Neogenomics). The detection of abnormalities via liquid biopsy leads to staging examinations. For patients with confirmed recurrence, treatment hereafter follows national guidelines. The detection of true molecular relapse (positive liquid biopsy without clinical evidence of recurrence) leads to further monitoring within the SURVIVE protocol and creates the unique opportunity for treatment-intervention studies, such as SURVIVE HERoes (NCT 06643585). Endpoints: The two co-primary endpoints are overall survival (OS) and the lead time, measured as the time from positive biomarker to positive imaging in the interventional arm. Secondary endpoints include breast cancer-specific survival, quality of life, and biomarker performance. Recruitment: As of July 2025, recruitment is active in 91 study centers in Germany. Currently, nearly half of the patients planned for inclusion have been randomized since December 2022. Conclusion: The SURVIVE study is a long-anticipated study assessing whether intensified follow-up, guided by liquid biopsy, leads to earlier detection of recurrence and improved overall survival in EBC. Funding: German Federal Ministry of Education and Research (BMBF), NeoGenomics, Menarini Silicon Biosystems, Tosoh Bioscience Bibliography: 1. Moschetti I, Cinquini M, Lambertini M, Levaggi A, Liberati A. Follow-up strategies for women treated for early breast cancer. Cochrane Database Syst Rev. 2016;CD001768.
Presentation numberPS5-08-14
Destiny-breast respond her2-low europe: description of first enrolled patients in the non-interventional study of t-dxd in her2-low metastatic breast cancer
Valentina Guarneri, Veneto Institute of Oncology (IOV); Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
V. Guarneri1, J. Passos Coelho2, F. P. Duhoux3, D. Egle4, J. García-Sáenz5, F. Penault-Llorca6, H. Wildiers7, K. Zaman8, P. Laeis9, M. Lucerna9, J. Pierga10; 1Medical Oncology 2, Veneto Institute of Oncology (IOV); Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, ITALY, 2Oncology Department, Hospital da Luz Lisboa, Lisbon, PORTUGAL, 3Department of Medical Oncology, Cliniques universitaires Saint-Luc, Brussels, BELGIUM, 4Medical University of Innsbruck, Department of Gynaecology and Obstetrics, Innsbruck, AUSTRIA, 5Department of Medical Oncology, Hospital Clínico San Carlos, Madrid, SPAIN, 6Centre Jean Perrin, Université Clermont Auvergne, Clermont Ferrand, FRANCE, 7Department of General Medical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 8Department of Oncology, Lausanne University Hospital CHUV, Lausanne, SWITZERLAND, 9Daiichi Sankyo Europe GmbH, Daiichi Sankyo Europe GmbH, Munich, GERMANY, 10Department of Medical Oncology, Université Paris Cité, Paris, FRANCE.
DESTINY-Breast04 showed significantly longer PFS and OS in patients (pts) with HER2-low (IHC 1+ or IHC 2+/ISH-) mBC treated with trastuzumab deruxtecan (T-DXd) vs physician’s choice of chemotherapy.1 As such, there was a shift from the binary HER2 classification towards a HER2 spectrum, including HER2-low as a targetable biomarker for T-DXd. The ongoing DESTINY-Breast Respond HER2-low Europe study (NCT05945732) aims to evaluate real-world effectiveness and safety of T-DXd in pts with HER2-low mBC. We report an initial snapshot of baseline characteristics and prior treatment lines. The design has been published.2 The study is enrolling pts with HER2-low mBC who received ≥1 prior line of chemotherapy in the metastatic setting and are being treated with T-DXd or standard chemotherapy, aiming to enroll 1010 pts from 206 sites across 10 European countries. Primary objective is effectiveness of T-DXd based on real-world time to next treatment, secondary endpoints include baseline demographics, clinical characteristics and other outcome parameters. The first pt was enrolled in Jan 2024. Data cutoff for initial baseline characteristics assessment was 15 April 2025. A total of 234 pts have been enrolled from 8 countries (Austria, Belgium, Denmark, France, Italy, Spain, Sweden and Switzerland). Most pts were female (98.3%). Median age was 62 yrs, 15% aged >75 yrs. Nearly half of pts had an ECOG score ≥1, 8.5% presenting with brain metastasis and 2.1% with lung-related comorbidities. HER2-low pts with HR+ (78.2%) and HR- (18.8%) disease were enrolled and HER2 status was categorized using the most recent biopsy (mostly using newly obtained tissue [61.5%] vs archival tissue [36.3%] from metastases [62.4%] vs primary tumour [35.0%]). Pts previously received a median of 3 prior lines in the metastatic setting. The most common treatments received were CDK4/6 inhibitor-based regimens (52.0%) in the first line, and chemotherapy in the second and third line (47.5% and 56.0%). The most common prior chemotherapy received for mBC was capecitabine (35.6%). An updated analysis will be available for presentation at SABCS, and results are expected to be consistent with the snapshot analysis. This initial snapshot analysis indicates that HER2-low mBC pts treated with T-DXd in the real-world are more diverse versus those in DESTINY-Breast04; the number of prior treatment lines is comparable while age and overall health status differ, highlighting the importance of ongoing recruitment to this large, longitudinal, observational study. Overall, initial data indicate that the DESTINY-Breast Respond HER2-low Europe study will add valuable insights on real-world effectiveness, treatment management, safety and quality of life for HER2-low mBC pts treated with T-DXd in the European region. 1. Modi, et al. NEJM. 2022;3887:9-20. 2. Guarneri, et al. Future Oncol. 2024;20:1237-50.
Presentation numberPS5-08-15
Targeted axillary dissection vs axillary node clearance in patients with positive axillary nodes in early breast cancer: a multicentre, pragmatic, phase 3 randomised controlled trial
Shelley Potter, Bristol Medical School, Bristol, United Kingdom
S. Potter1, K. Avery2, P. Baji1, I. Bhattacharya3, N. Blencowe2, L. Culliford4, R. Cutress5, L. Dabner4, D. Dodwell6, K. Fairhurst1, J. Frost4, J. Harris4, E. Marques1, H. Markham7, A. Morgan8, M. Perkins9, S. Rees4, K. Roberts4, T. Robinson2, N. Sharma10, S. McIntosh11; 1Translational Health Sciences, Bristol Medical School, Bristol, UNITED KINGDOM, 2Population Health Sciences, Bristol Medical School, Bristol, UNITED KINGDOM, 3Department of Clinical Oncology, Cambridge University Hospitals NHS Trust, Cambridge, UNITED KINGDOM, 4Bristol Trials Centre, Bristol Medical School, Bristol, UNITED KINGDOM, 5Medical School, University of Southampton, Southampton, UNITED KINGDOM, 6Oxford Population Health, University of Oxford, Oxford, UNITED KINGDOM, 7Department of Pathology, University Hospitals Southampton NHS Trust, Southampton, UNITED KINGDOM, 8ICPV, Independent Cancer Patient’s Voice, London, UNITED KINGDOM, 9ICPV, Independent Cancer Patients’ Voice, London, UNITED KINGDOM, 10Department of Radiology, St James University Hospital, Leeds, UNITED KINGDOM, 11The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast, UNITED KINGDOM.
BACKGROUND: Approximately 20% of women with breast cancer will be node-positive at presentation. In the UK, all patients with newly diagnosed breast cancer have axillary staging with an axillary USS +/- biopsy of abnormal/equivocal nodes and currently UK NICE guidelines recommend axillary node clearance (ANC) for all patients with biopsy proven node positive breast cancer having primary surgery, irrespective of the number of nodes involved. This highly morbid procedure leads to life-long complications in 1 in 3 patients including lymphoedema and chronic pain which dramatically impact quality of life. ANC aims to reduce locoregional recurrence (LRR) and improve breast cancer survival but there is no evidence to support these benefits for patients with limited nodal involvement (cN0, radiologically detected disease). These patients would meet the criteria for omission of ANC based on eligibility for the ACOSOG Z0011 trial, but this approach has not been adopted in the UK due to concerns regarding false negative sentinel node biopsy (SNB) in node positive patients. Targeted axillary dissection (TAD) which combines removal of the localised biopsy proven involved node(s) in combination with a SNB may offer an alternative to ANC, effectively addressing concerns regarding false negative rates while reducing the risk of life-changing complications The TADPOLE study aims to determine if TAD is a clinically and cost-effective alternative to ANC in patients with low volume node positive breast cancer having primary surgery. METHODS: TADPOLE is a multicentre pragmatic phase 3 randomised controlled trial comparing TAD and ANC in breast cancer patients with low volume nodal disease having primary surgery. All patients with cN0 biopsy proven low volume axillary nodal disease will be eligible to participate. Excluded will be patients with >3 nodes on USS, those who have recurrent disease, previous axillary surgery or neoadjuvant systemic therapy. Participants will be randomised 2:1 to TAD or ANC. Surgical quality assurance (QA) processes will promote standardised introduction of ‘primary’ TAD in the UK and ensure procedure fidelity within the trial. Participants will have adjuvant therapy as per standard of care but axillary radiotherapy (ART) will be prohibited in the TAD group. Robust radiotherapy quality assurance (RTTQA) will be embedded throughout. The co-primary end-points are:i)Patient reported and objective lymphoedema at 12 months ii)Single arm analysis of LRR at 5 years in the TAD cohort. Recruitment of 390 patients in the TAD arm will be required to detect a 50% reduction in lymphoedema at 12 months with 90% power and a type 1 error of 5% and exclude an undesirable LRR of <5% in the TAD cohort at 5 years with one sided 2.5% alpha and 90% power. Inflating for multiplicity and allowing for 5% dropout and 5% crossover a total of 861 patients (574 TAD:287 ANC) will be required for the trial. 40 UK breast units will recruit to the trial. An embedded qualitative study will optimise recruitment and a SWAT (study within a trial) will optimise recruitment of non-English speaking participants. RESULTS: Consensus work to agree how to axillary surgery including primary TAD should be performed has been completed and will underpin the surgical QA within TADPOLE. The trial will commence recruitment September 2025 and include a 9 month internal pilot phase. Recruitment is planned for 28 months with a target of 1 participant/centre/m at 40 sites. CONCLUSION: TADPOLE will address the top UK breast surgery research priority identified in the James Lind Alliance Priority Setting Process. If TAD causes less lymphoedema and is oncologically safe, TADPOLE will change practice, improving outcomes for thousands of patients with node positive breast cancer each year.
Presentation numberPS5-08-16
Intratumoral Injections of INT230-6 Prior to Neoadjuvant Immuno-chemotherapy in Early-Stage Triple Negative Breast Cancer: Early observations from INVINCIBLE-4-SAKK 66/22 (NCT06358573), a Phase II Randomized Controlled Trial
Ursina Zürrer, Cantonal Hospital Winterthur, Winterthur, Switzerland
U. Zürrer1, A. Müller1, O. Tredan2, C. Micheloud3, J. Musilova4, R. Popescu5, T. Schmid6, L. Rossi7, M. Schwitter8, M. Vetter9, M. Niemeyer10, I. Witzel11, A. Patsouris12, S. Ladoire13, J. Martin-Babau14, A. Deleuze15, M. Robert16, L. H. Bender17, M. Joerger18; 1Medical Oncology and Haematology, Cantonal Hospital Winterthur, Winterthur, SWITZERLAND, 2Département de cancérologie médicale, Centre Léon Bérard, Lyon, FRANCE, 3Statistics, Swiss Cancer Institute, Berne, SWITZERLAND, 4Project Managment, Swiss Cancer Institute, Berne, SWITZERLAND, 5Tumor centre, Tumor Zentrum Aarau and Hirslanden Clinic Aarau, Aarau, SWITZERLAND, 6Oncology and Haematology, Claraspital, Basel, Basel, SWITZERLAND, 7Medical Oncology, Ente Ospedaliero Cantonale, Bellinzona, SWITZERLAND, 8Medical Oncology and Haematology, Cantonal Hospital Graubünden, Chur, SWITZERLAND, 9Medical Oncology and Haematology, Cantonal Hospital Baselland, LIestal, SWITZERLAND, 10Senology, Tumor- und Brust Zentrum Ostschweiz, St. Gallen, SWITZERLAND, 11Gynaecology, University Hospital Zurich, Zurich, SWITZERLAND, 12Medical Oncology, Institut de Cancérologie de l’Ouest Site Paul Papin, Angers, FRANCE, 13Medical Oncology, Centre Georges-François Leclerc, Dijon cedex, FRANCE, 14Medical Oncology, Hôpital Privé des Côtes d’Armor – Centre Cario-HPCA, Plérin, FRANCE, 15Medical Oncology, Centre Eugène Marquis, Rrennes Cedex, FRANCE, 16Medical Oncology, Institut de Cancérologie de l’Ouest Site René Gauducheau, Saint Herblain, FRANCE, 17Sponser, Intensity Therapeutics, Shelton, CT, 18Medical Oncology and Haematology, HOCH Health Ostschweiz, St. Gallen, SWITZERLAND.
Background: Triple Negative Breast Cancer (TNBC) poses significant challenges due to its aggressiveness, high relapse rate and mortality. Neoadjuvant immuno-chemotherapy (NAIC) is now a common treatment for early-stage TNBC before surgery. NAIC aims to eliminate viable cancer in the tumor, lymph nodes and possible occult distant metastases, shrink tumors to improve surgical outcomes and prevent disease recurrence. The Keynote-522 study revealed a 84.5% 3-year event-free survival in patients with early-stage TNBC using NAIC and improved pathological complete response (pCR) rates from 51.2% with neoadjuvant Chemotherapy to 64.8% with NAIC. A new potential method to improve clinical outcome and induce immune activation pre-surgery is through a novel local therapy in combination with NAIC that could cause increased apoptotic cell death and create personalized tumor antigens. Arnaout et al. conducted a randomized, phase 2 neoadjuvant window of opportunity trial using intratumoral (IT) INT230-6, a drug comprising vinblastine, cisplatin and a tumor dispersion and cell penetration enhancer molecule (SHAO), evaluating clinical and biological effects in women with early-stage operable BC (NCT04781725). Results in T2 to T4 tumors showed an average of over 30% necrosis in 74% of subjects at the time of surgery, with some patients having >95% tumor necrosis following a single dose. Adding immune-activating and apoptotic induced necrosis caused by INT230-6 dosed prior to NAIC in TNBC patients has the potential to increase pCR. Methods: This is a randomized, open-label multicenter phase 2 clinical study to determine the clinical activity, safety, and tolerability of IT INT230-6 in patients with early-stage, operable TNBC in combination with NAIC (cohort A) or NAIC alone (cohort B). The INT230-6 dose is dependent on tumor size. The primary endpoint is pCR in the primary tumor (ypT0/Tis) and affected lymph nodes (ypN0). Key inclusion criteria include newly diagnosed, previously untreated, locally advanced non-metastatic TNBC stage cT1c N1-3 M0 or cT2-4c N0-3 M0. Multifocal and multicentric primary tumors are allowed. Patients must have measurable disease in the breast with at least one lesion with a diameter ≥1.5 cm visible in ultrasound and injectable. Patients are either male or female, age ≥ 18 years, ECOG performance status <2, adequate bone marrow, hepatic and renal function. STATS: The sample size calculation for both cohorts is determined based on a single-stage phase II single-arm clinical trial design (A’Hern). Null hypothesis (H0): pCR rate ≤ 0.6, Alternative hypothesis (H1): pCR rate ≥ 0.8. Type I error: 10% (one-sided), Power: 80%. The duration for accrual, patient therapy and follow-up is 12, 8 and 36 months respectively. The sample size per cohort is 27 patients. The study is recruiting in Switzerland and France in up to 16 sites. Results: By July 2025 15 of 54 patients could be enrolled, completion of accrual is expected in 2026. Preliminary safety data do not show unexpected or severe INT230-6 related adverse events. pCR results become available 6 months after the last patient starts the SOC and undergoes surgery. An exemplary case of a patient will be shown.
Presentation numberPS5-08-17
Melody: A prospective non-interventional multicenter cohort study to evaluate different imaging-guided methods for localization of malignant breast lesions (Eubreast-4 / iBra-net, Nct 05559411)
Maggie Banys-Paluchowski, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
J. Ryu1, T. Kühn2, N. Ditsch3, J. Harvey4, S. Hartmann5, N. Cabioglu6, N. Canturk7, A. Karakatsanis8, J. de Boniface9, T. Filtenborg Tvedskov10, L. Pankratjevaite11, M. Gasparri12, S. Alran13, E. Schlichting14, A. Lowery15, A. Esgueva16, R. Di Micco17, D. Murawa18, M. Kontos19, M. Muneer Khan20, Y. Masannat21, V. Fabiano22, K. Abdelwahab23, L. Rebaza24, F. Peintinger25, M. Correia26, E. Giblin27, G. Dindelegan28, S. Nietz29, F. Malherbe30, A. Kothari31, R. Dave4, O. Gentilini32, B. Güllüoglu33, M. Hahn34, G. Karadeniz Cakmak35, M. Lux36, S. Potter37, I. Rubio16, M. Smidt38, W. Weber39, N. Krawczyk40, K. Jursik41, A. Kaiser42, A. Körner5, M. Köpke43, N. Tauber42, S. Lukac44, M. Banys-Paluchowski42, MELODY study group; 1Department of Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 2Breast Cancer Center / Department of Gynecology and Obstetrics, Die Filderklinik gGmbH / University of Ulm, Filderstadt / Ulm, GERMANY, 3Department of Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, GERMANY, 4The Nightingale and PREVENT Breast Cancer Center, Manchester University NHS Foundation Trust & Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UNITED KINGDOM, 5Department of Gynecology and Obstetrics, University Hospital Rostock, Rostock, GERMANY, 6Istanbul Faculty of Medicine, Department of General Surgery, Istanbul University, Istanbul, TURKEY, 7Department of General Surgery, Kocaeli University School of Medicine, Kocaeli, TURKEY, 8Department for Surgical Sciences, Faculty of Pharmacy and Medicine / Department of Surgery, Uppsala University / Uppsala University Hospital, Uppsala Municipality / Uppsala, SWEDEN, 9Department of Surgery, Breast Center / Department of Medical Epidemiology and Biostatistics, Capio St. Göran’s Hospital / Karolinska Institutet, Stockholm, SWEDEN, 10Faculty of Health and Medical Sciences / Department of Breast Surgery, University of Copenhagen / Herlev & Gentofte University Hospital, Copenhagen, DENMARK, 11Faculty of Health and Medical Sciences / Department of Breast Surgery, University of Copenhagen / Herlev & Gentofte University Hospital, Lübeck, DENMARK, 12Department of Gynecology and Obstetrics / Faculty of Biomedicine, Ente Ospedaliero Cantonale, Ospedale Regionale di Lugano / University of the Italian Switzerland (USI), Lugano, SWITZERLAND, 13Breast & Gynecologic Surgery Service Groupe, Hospitalier Paris Saint Joseph, Paris, FRANCE, 14Department of Oncology, Oslo University Hospital, Oslo, NORWAY, 15Discipline of Surgery, University of Galway, Galway, IRELAND, 16Breast Surgical Unit, Clinica Universidad de Navarra, Madrid, SPAIN, 17Breast Surgery Unit, IRCCS Ospedale San Raffaele, Milan, ITALY, 18Surgery and Oncology Collegium Medicum / Department of General Surgery and Surgical Oncology, University of Zielona Góra / The Regional Hospital in Poznan – The Greater Poland Specialist Center, Zielona Gora / Poznan, POLAND, 19Department of Surgery, Laiko Hospital, National and Kapodistrian University of Athens, Athens, GREECE, 20Breast Care Service, Department of Surgery, Khyber Medical College & Khyber Teaching Hospital, Peshawar, PAKISTAN, 21Aberdeen Breast Unit, Aberdeen Royal Infirmary, Aberdeen, UNITED KINGDOM, 22Department of Breast Cancer, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 23Mansoura Oncology Center, Mansoura University, Mansoura, EGYPT, 24Breast Surgery, Clinica Oncosalud Auna, Lima, PERU, 25Department of Gynecology and Obstetrics / Institute of Pathology, Medical University of Graz / Medical University of Graz, Graz, AUSTRIA, 26Breast Unit, Champalimaud Clinical Centre, Champalimaud Foundation, Lisboa, PORTUGAL, 27Breast Care Service, Ascension Medical Group, St. Vincent Carmel Hospital, Carmel, IN, 28First Surgical Clinic, Emergency County Hospital Cluj-Napoca / “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, ROMANIA, 29Breast Health Unit, University of the Witwatersrand, Johannesburg, SOUTH AFRICA, 30Breast and Endocrine Surgery Unit, Groote Schuur Hospital, University of Cape Town, Cape Town, SOUTH AFRICA, 31Breast Cancer Unit, Guy’s & St Thomas NHS Foundation Trust, London, UNITED KINGDOM, 32Breast Surgery Unit, IRCCS Ospedale San Raffaele / Vita-Salute San Raffaele University, Milan, ITALY, 33Department of Surgery, Breast Surgery Unit, Marmara University School of Medicine and SENATURK Turkish Academy of Senology, Istanbul, TURKEY, 34Department for Women’s Health, University of Tübingen, Tübingen, GERMANY, 35General Surgery Department, Breast and Endocrine Unit, Zonguldak BEUN The School of Medicine, Zonguldak, TURKEY, 36Department of Gynecology and Obstetrics, St. Louise Frauen- und Kinderklinik, St. Vincenz Krankenhaus, Paderborn, GERMANY, 37Bristol Population Health Science Institute, Bristol Medical School (THS), Bristol, UNITED KINGDOM, 38Department of Surgical Oncology, Maastricht University Medical Center, Maastricht, NETHERLANDS, 39Division of Breast Surgery, Department of Surgery, Basel University Hospital, Basel, SWITZERLAND, 40Department of Gynecology and Obstetrics, Heinrich-Heine-University Düsseldorf, Düsseldorf, GERMANY, 41EUBREAST GmbH, EUBREAST GmbH, Gladenbach, GERMANY, 42Department of Gynecology and Obstetrics, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, GERMANY, 43Department of Gynecology and Obstetrics, University Hospital Augsburg, Lübeck, GERMANY, 44Department of Gynecology and Obstetrics, University of Ulm, Ulm, GERMANY.
Background:In the last decades, the proportion of breast cancer patients receiving breast-conserving surgery has increased, reaching 70-80% in developed countries. In case of non-palpable lesions, surgical excision requires some form of breast localization. While wire-guided localization has long been considered gold standard, it carries several limitations, including logistical difficulties, the potential for displacement and patient discomfort, and re-excision rates reaching 21%. Other techniques (radioactive seed or radio-occult lesion localization, intraoperative ultrasound, magnetic, radiofrequency and radar localization) have been developed with the aim of overcoming these disadvantages. However, comparative data on the rates of successful lesion removal, negative margins and re-operations are limited. In the majority of studies, the patient’s perspective with regard to discomfort and pain level has not been evaluated. The aim of MELODY (MEthods for LOcalization of Different types of breast lesions) is to evaluate different imaging-guided localization methods with regard to oncological safety, patient-reported outcomes, and surgeon and radiologist satisfaction. Methods:The EUBREAST and the iBRA-NET have initiated the MELODY study to assess breast localization techniques and devices from several perspectives (NCT05559411, http://eubreast.org/melody). MELODY is a prospective intergroup cohort study which enrolls female and male pts. requiring breast-conserving surgery and imaging-guided localization for invasive breast cancer or DCIS. Multiple or bilateral lesions and neoadjuvant chemotherapy are allowed. Primary outcomes are: 1) Intended target lesion and/or marker removal, independent of margin status on final histopathology, and 2) Negative resection margin rates at first surgery. Secondary outcomes are, among others: rates of second surgery and secondary mastectomy, Resection Ratio (defined as actual resection volume divided by the calculated optimum specimen volume), duration of surgery, marker dislocation rates, rates of marker placement or localization failure, comparison of patient-reported outcomes, rates of “lost markers” and diagnostician/radiologist’s and surgeon’s satisfaction as well as the health economic evaluation of the different techniques. Target accrual: 7,416 patients. Enrollment started in January 2023. The study will be conducted in 30 countries and is supported by the Oncoplastic Breast Consortium (OPBC), AWOgyn, AGO-B and SENATURK. Financial support was provided by Endomag, Merit Medical, Sirius Medical and Hologic.
Presentation numberPS5-08-18
Trofuse-011: a phase 3, randomized, open-label study of sacituzumab tirumotecan with or without pembrolizumab vs treatment of physician’s choice for previously untreated locally recurrent unresectable or metastatic triple-negative breast cancer
Aditya Bardia, University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA
A. Bardia1, X. Peng2, K. L. Smith2, J. A. Mejia2, H. S. Rugo3; 1Division of Hematology-Oncology, University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, 2Global Clinical Development, Merck & Co., Inc., Rahway, NJ, 3Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA.
Background: The standard first-line therapy for most previously untreated locally recurrent unresectable or metastatic triple-negative breast cancer (TNBC) with PD-L1 combined positive score (CPS) <10 is chemotherapy alone. New therapeutic options with superior efficacy are needed. Sacituzumab tirumotecan (sac-TMT; MK-2870/SKB264) is a novel antibody-drug conjugate (ADC) composed of an anti-trophoblast cell surface antigen 2 (TROP2) monoclonal antibody coupled to a cytotoxic belotecan derivative, topoisomerase I inhibitor payload via a novel linker. Sac-TMT monotherapy has shown promising efficacy with a manageable safety profile in metastatic TNBC. There is strong biologic rationale supporting combination of an ADC with pembrolizumab regardless of PD-L1 expression. The TroFuse-011 study (NCT06841354) evaluates sac-TMT ± pembrolizumab vs treatment of physician’s choice (TPC) in participants with previously untreated locally recurrent unresectable or metastatic TNBC with PD-L1 CPS <10. Trial design: Eligible participants (≥18 y) with centrally confirmed locally recurrent unresectable or metastatic TNBC, measurable disease per RECIST v1.1, ECOG PS 0 or 1, and a tumor tissue sample evaluable for central PD-L1 and TROP2 testing are randomized 2:1:2 to receive sac-TMT (Arm A), sac-TMT + pembrolizumab (Arm B), or TPC (Arm C; Table). Primary endpoints are PFS per RECIST 1.1 as assessed by BICR (Arm A vs C and Arm B vs C) and OS (Arm A vs C). Secondary endpoints are PFS (Arm B vs A), OS (Arm B vs C and Arm B vs A), ORR (Arm A vs C and Arm B vs C) and DOR, patient-reported outcomes, and safety. Tumor imaging occurs at baseline, Q8W after randomization until week 48, and Q12W thereafter. Enrollment is ongoing.
| Intervention | |
| Arm A | sac-TMTa |
| Arm B | sac-TMTa + pembrolizumabb |
| Arm C | Paclitaxelc or nab-paclitaxeld or gemcitabinee + carboplatinf |
| a4 mg/kg Q2W. b400 mg Q6W. c80 mg/m2 on Day 1 QW or 90 mg/m2 on days 1, 8, and 15, Q4W. d100 mg/m2 on days 1, 8, and 15, Q4W. e1000 mg/m2 on days 1 and 8, Q3W. fAUC 2 mg/mL/min on days 1 and 8, Q3W. |
Presentation numberPS5-08-19
Randomized phase 3 trial evaluating KAT6 inhibitor PF-07248144 plus fulvestrant in HR+HER2− advanced/metastatic breast cancer after progression on CDK4/6 inhibitor-based therapy
Kevin M. Kalinsky, Emory University, Atlanta, GA
K. M. Kalinsky1, R. M. Layman2, A. Giordano3, T. Mukohara4, Y. Park5, G. J. Lindeman6, G. Bianchini7, A. Nonneville8, V. Diéras9, S. Kuemmel10, S. Wang11, Z. Shao12, S. Kent13, M. Li13, M. Oliveira14; 1Division of Medical Oncology, Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, 2Department of Breast Medical Oncology, Division of Cancer, MD Anderson Cancer Center, Houston, TX, 3Department of Medical Oncology, Dana–Farber Cancer Institute, Boston, MA, 4Department of Medical Oncology, National Cancer Center Hospital East, Tokyo, JAPAN, 5Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF, 6Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research and Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 7Department of Medical Oncology, IRCCS Ospedale San Raffaele, Milan, ITALY, 8Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, Marseille, FRANCE, 9Department of Medical Oncology, Centre Eugène Marquis, Rennes, FRANCE, 10Breast Unit, Kliniken Essen-Mitte, Essen, GERMANY, 11Department of Medical Oncology, Sun Yat-Sen University Cancer Center, Guanghou, CHINA, 12Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 13., Pfizer Inc., New York, NY, 14Medical Oncology Department / Breast Cancer Group, Hospital Universitari Vall d’Hebron / Vall d’Hebron Institute of Oncology, Barcelona, SPAIN.
Background: Patients with HR+HER2− advanced/metastatic breast cancer (ABC) who have tumors that progressed after CDK4/6 inhibitor and endocrine therapy (ET) represent a high unmet medical need. Histone lysine acetyltransferases KAT6A and KAT6B regulate lineage-specific gene transcription via H3K23 acetylation. PF-07248144 is a selective inhibitor of KAT6A and KAT6B. PF-07248144 (5 mg QD) in combination with fulvestrant (FUL) has demonstrated encouraging and durable antitumor activity with a manageable safety profile in patients with heavily pretreated HR+HER2− ABC in a phase 1/2 study. The phase 3 study is designed to evaluate and confirm the efficacy and safety of PF-07248144 plus FUL in patients with HR+HER2− ABC after tumor progression on CDK4/6i -based therapy, when compared to standard of care therapy. Methods: This open-label, randomized phase 3 trial is comparing PF-07248144 plus FUL versus everolimus (EVE) plus ET [FUL or exemestane (EXE)]) in patients with HR+HER2− ABC. Key inclusion criteria are: age ≥ 18 years; histologically confirmed HR+HER2− ABC; progression after prior CDK4/6i-based therapy; available tumor tissue; measurable disease or non-measurable bone-predominant disease, defined by RECIST v1.1; Adequate organ function; ECOG PS 0 or 1. Key exclusion criteria are: presence of detectable PIK3CA/AKT1/PTEN alterations; prior chemotherapy or therapy targeting PIK3CA/AKT1/PTEN in ABC; >2 prior lines post-CDK4/6i of systemic anticancer therapy in ABC and no more than one line of chemotherapy; systemic anticancer therapy or radiation within 2 weeks of randomization. Up to 400 patients will be randomized (1:1) to receive either PF-07248144 plus FUL versus EVE plus ET (FUL or EXE). Patients will be stratified by ET (FUL vs EXE), prior systemic lines of therapy in metastatic setting (1 vs 2), and disease site (visceral vs non-visceral). The primary endpoint is progression-free survival by blinded independent central review. Overall survival is a key secondary endpoint; other secondary endpoints include objective response, duration of response, clinical benefit rate, safety, and pharmacokinetics of PF-07248144.
Presentation numberPS5-08-20
A phase III trial evaluating De-escalation of Breast Radiation (DEBRA) following breast-conserving surgery of stage 1, HR+, HER2-, RS ≤18 breast cancer: NRG-BR007
Julia R White, University of Kansas Medical Center Comprehensive Cancer Center, Kansas City, KS
J. R. White1, R. S. Cecchini2, E. E. Harris3, E. P. Mamounas4, J. G. Bazan5, D. G. Stover6, P. A. Ganz7, R. Jagsi8, S. J. Anderson2, C. Bergom9, V. Théberge10, M. B. El-Tamer11, R. C. Zellars12, D. A. Shumway13, G. Chen14, T. B. Julian15, N. Wolmark16; 1Department of Radiation Oncology, University of Kansas Medical Center Comprehensive Cancer Center, Kansas City, KS, 2Department of Biostatistics and Health Data Science, University of Pittsburgh; NRG Oncology Statistical and Data Management Center, Pittsburgh, PA, 3Department of Radiation Oncology, St. Luke’s University Health Network, Easton, PA, 4Department of Surgical Oncology, AdventHealth Cancer Institute, Orlando, FL, 5Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, 6Department of Medicine, Ohio State University Comprehensive Cancer Center, Columbus, OH, 7Medicine/Health and Policy Management, David Geffen School of Medicine at UCLA; UCLA Fielding School of Public Health; UCLA Jonsson CCC, Los Angeles, CA, 8Department of Radiation Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, 9Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, 10Department of Radiation Oncology, CHU de Québec – Université Laval, Québec, QC, CANADA, 11Department of Surgical Oncology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical School, New York, NY, 12Department of Radiation Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, 13Department of Radiation Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 14Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 15Department of Surgical Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, 16Department of Surgical Oncology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine; NSABP Foundation, Inc, Pittsburgh, PA.
Background Approximately 50% of newly diagnosed invasive breast cancers are stage 1, with the majority being ER/PR-positive, HER2-negative. Genomic assays such as the Oncotype DX® have identified patients (pts) with reduced risk of distant metastasis and without benefit from chemotherapy added to endocrine therapy (ET), freeing them from excess toxicity. Genomic assays are also recognized as prognostic for in-breast recurrence (IBR) after breast-conserving surgery (BCS) and could similarly allow de-escalation of adjuvant radiotherapy (RT). Reducing overtreatment is of interest to pts, providers, and payers. Methods We hypothesize that BCS alone is non-inferior to BCS plus RT for IBR and breast preservation in women intending ET for stage 1 invasive breast cancer (ER and/or PR-positive, HER2-negative with an Oncotype DX Recurrence Score [RS] of ≤18). Stratification is by age (1-2cm), and RS (≤11, >11-18/MammaPrint Low). Pts are randomized post-BCS to Arm 1 with breast RT using standard methods (moderate or ultra hypo- or conventional-fractionated whole breast RT with/without boost, or APBI) with ≥5 yrs of ET (tamoxifen or AI) or Arm 2 with ≥5 yrs of ET (tamoxifen or AI) alone. The specific regimen of ET in both arms is at the treating physician’s discretion. Eligible pts are stage 1: pT1 (≤2 cm), pN0, age ≥50 to <70 yrs, s/p BCS with negative margins (no ink on tumor), s/p axillary nodal staging (SNB or ALND), ER and/or PR-positive (ASCO/CAP), HER2-negative (ASCO/CAP), and Oncotype DX RS ≤18 (diagnostic core biopsy or resected specimen). A “low risk” MammaPrint is permissible if completed as part of usual care prior to screening. Primary endpoint is IBR (invasive breast cancer or DCIS). Secondary endpoints are breast conservation rate, invasive in-breast recurrence, relapse-free interval, distant disease-free survival, overall survival, patient-reported breast pain, patient-reported worry about recurrence, and adherence to ET. We assume a clinically acceptable difference in IBR of 4% at 10 yrs to judge omission of RT as non-inferior (10-yr event-free survival for RT group is 95.6% v 91.6% for the omission-of-RT group). BR007 is powered to detect non-inferiority with 80% power and a one-sided α=0.025, assuming that there would be a ramp-up in accrual in the first two years (leveling off in Yrs 3-5); 1,670 pts (835 per arm) are required for randomization. Conservative loss to follow-up is 1%/yr. Some T1a pts screened may have Oncotype DX scores >18, making them ineligible for the study. In the accrual process, 1,714 pts will be required to register to ensure that our final randomized cohort is 1,670 pts. As of July 9, 2025, 1,490 pts have been screened and 1,349 randomly assigned. NCT #: NCT04852887 Support: U10 CA180868, -180822, U24 CA196067, UG1 CA189867
Presentation numberPS5-08-21
Prospective breast cancer clinical validation study of an ultrasensitive, tumor-informed, whole genome, circulating tumor DNA assay to detect molecular residual disease and predict recurrence of high-risk early breast cancer treated with standard (neo)adjuvant therapy; NSABP B-64/EXActDNA/003/NCT06401421.
Mark Basik, Jewish General Hospital, Montréal, QC, Canada
M. Basik1, E. Diego2, G. Tang3, S. Puhalla4, M. Balic4, M. Mandadi5, J. Foldi6, S. Seaward7, M. George8, A. Shipstone9, S. Vemulapalli10, J. Jones11, W. J. Irvin Jr12, R. L. Mahtani13, V. Valero14, J. Frederick15, B. Arrick16, G. Young17, J. A. Grigorieva18, M. R. Palomares16, F. Baehner18, P. Rastogi4, E. P. Mamounas19, N. Wolmark20, C. E. Geyer Jr4; 1Department of Oncology, Jewish General Hospital, Montréal, QC, CANADA, 2Division of Surgical Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 3Department of Biostatistics and Health Data Science, University of Pittsburgh; NRG Oncology Statistical and Data Management Center, Pittsburgh, PA, 4Division of Hematology and Oncology, University of Pittsburgh School of Medicine; UPMC Hillman Cancer Center; NSABP Foundation, Inc., Pittsburgh, PA, 5Department of Hematology and Oncology, Baptist Health Cancer Research Network, Louisville, KY, 6Division of Hematology and Oncology, University of Pittsburgh School of Medicine; UPMC Hillman Cancer Center, Pittsburgh, PA, 7Department of Radiation Oncology, Kaiser Permanente Oncology Clinical Trials, Vallejo, CA, 8Department of Breast Medical Oncology, Rutgers Cancer Institute of New Jersey; Robert Wood Johnson Medical School, New Brunswick, NJ, 9Department of Hematology and Oncology, Ballad Health Cancer Center, Johnson City, TN, 10Department of Hematology and Oncology, Aultman Health Foundation, Canton, OH, 11Department of Oncology Hematology, Avera Medical Group – Prairie Center, Sioux Falls, SD, 12Department of Hematology Oncology, Southeast Cancer Research Consortium/Bon Secours Mercy Health, Richmond, VA, 13Department of Medical Oncology, Miami Cancer Institute Baptist Health South Florida, Miami, FL, 14Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 15Precision Oncology, Exact Sciences, Madison, WI, 16Precision Oncology, Exact Sciences, Redwood City, CA, 17Biostatistics, Exact Sciences, Madison, WI, 18Precision Oncology, Exact Sciences, La Jolla, CA, 19Department of Surgical Oncology, AdventHealth, Orlando, FL, 20Division of Surgical Oncology, University of Pittsburgh School of Medicine; UPMC Hillman Cancer Center; NSABP Foundation, Pittsburgh, PA.
Background: Detection of occult micrometastatic cancer—molecular residual disease (MRD)—in patients following treatment for high-risk phenotypes of early breast cancer using circulating tumor DNA (ctDNA) is associated with a high risk of recurrence. An ultra-sensitive, tumor-informed, whole-genome, minor-allele-enriched sequencing through recognition oligonucleotides (MAESTRO) assay has previously demonstrated a strong association with residual cancer burden and disease recurrence in an exploratory study in a subset of patients with triple-negative breast cancer (TNBC) treated in a Phase II study in the neoadjuvant setting.1 Here, we describe a large, prospective registry trial in progress designed to: (Part 1) assess the feasibility of using material from formalin-fixed paraffin-embedded (FFPE) diagnostic breast biopsies to create a bespoke MRD assay using the MAESTRO technology, and (Part 2) validate the association between ctDNA status and distant recurrence-free interval (dRFI) in 3 cohorts of patients with early breast cancer and high-risk phenotypes (TNBC, human epidermal growth factor receptor 2-positive [HER2+] inclusive of hormone-receptor positive and -negative [HR+ and HR−], and HR+/HER2-negative [HER2−] tumors) receiving neoadjuvant chemotherapy co-administered with standard targeted therapies, surgery, and standard adjuvant therapies post-surgery. Methods: Trial eligibility criteria include: age ≥18 years, ECOG performance status 0−1, histologically confirmed invasive breast carcinoma, primary tumor size >2.0 cm, planned neoadjuvant chemotherapy, and either clinically node-positive disease, or if node-negative, one of the following: TNBC or HER2+ subtype, or HR+/HER2− subtype with high-risk features. Target enrollment is 1,800 participants, based on the number needed in each of the high-risk subtypes to observe at least 30 events within 3 years. Analyses will be conducted across the 3 subtypes in Part 1 and by breast cancer subtype in tissue-evaluable participants in Part 2 of the trial, once at least 30 distant recurrences have been reported in each subtype. Patients will receive standard therapies per investigator discretion and will be followed for at least 5 years. Serial blood samples will be collected during treatment and follow-up, and analyzed for ctDNA by Exact Sciences, blinded to clinical data. Results of ctDNA analyses for each collection timepoint will be provided to the National Surgical Adjuvant Breast and Bowel Project (NSABP) Foundation statistician to merge with clinical outcomes data and analyze the endpoints. In Part 1, the primary endpoint is the proportion of participants for whom FFPE diagnostic biopsy material is sufficient to create a patient-specific ctDNA assay. In Part 2, the primary endpoint is dRFI with 2 primary objectives, which will be analyzed sequentially: (1) the association of the ctDNA-positive status at any time in serially collected blood samples (starting from the completion of cytotoxic and targeted therapy) and dRFI, and (2) the association of the ctDNA-positive status as a single timepoint at the end of cytotoxic and targeted therapy with dRFI. Secondary endpoints that will be analyzed for association with ctDNA status include pathologic complete response, recurrence-free interval, invasive breast cancer-free survival, event-free survival, and overall survival. Clearance of ctDNA on therapy will also be assessed and correlated with outcomes. ClinicalTrials.gov Identifier: NCT06401421. References: 1. Parsons HA et al. Ann Oncol. 2023 Oct;34(10):899-906.
Presentation numberPS5-08-22
Patients with breast cancer and axillary involvement undergoing Sentinel LYMph Node Biopsy versus Targeted AXillary Dissection after primary systemic treatment: which axillary staging saves more lymphnodes? A registry-based randomized controlled trial (SLYMT-AXSANA EUBREAST 3R)
Maria Luisa Gasparri, Ente Ospedaliero Cantonale, Lugano, Switzerland
M. Gasparri1, S. Hartmann2, M. Hauptmann3, M. Banys-Paluchowski4, N. Ditsch5, R. Di Micco6, O. Gentilini6, T. Kuehn7; 1Gynecology and Obstetrics, Ente Ospedaliero Cantonale, Lugano, SWITZERLAND, 2Gynecology and Obstetrics, University Hospital of Rostock, Rostock, GERMANY, 3Institute of Biostatistics and Registry Research, Brandenburg Medical School Theodor Fontane, Neuruppin, GERMANY, 4Gynecology and Obstetrics, University Hospital of Schleswig-Holstein, Campus Luebeck, Luebeck, GERMANY, 5Gynecology and Obstetrics, University Hospital Augsburg, Augsburg, GERMANY, 6Breast Surgery Unit, San Raffaele University Hospital, Milan, ITALY, 7Breast Cancer Center, Die Filderklinik and University of Ulm, Filderstadt, GERMANY.
Background: Most patients with breast cancer and axillary nodal involvement achieve a clinical and radiological response after primary systemic treatment (PST). In this case, a surgical axillary de-escalation should be offered to reduce surgical trauma and subsequent morbidity. Sentinel lymph node biopsy (SLNB) (with a dual tracer and/or removal of at least 3 sentinel lymph nodes) or targeted axillary dissection (TAD) can safely replace axillary lymphadenectomy. The more axillary lymph nodes removed, the higher the morbidity. Therefore, SLNB and TAD represent surgical de-escalation procedures with lower body impact and arm injury compared to full axillary lymphadenectomy. Retrospective studies observed no difference in oncological outcome between SLNB and TAD. Follow-up data from the current ongoing prospective registry AXSANA (EUBREAST 3) will provide unique real-world evidence on this topic. In the meantime, surgical axillary staging is characterized by a lack of standardization and a wide heterogeneity between countries, institutions, and guidelines. The choice of treatment is therefore determined by preferences of institutions and/or surgeons rather than benefits for the patients. Currently, there is no consensus on whether SLNB or TAD minimizes the number of lymph nodes removed. No randomized clinical trials have compared SLNB versus TAD in node positive breast cancer patients converting to node negative after PST.The aim of the current study (SLYMT-AXSANA, EUBREAST 3R) is to use a pragmatic design and conduct, to our knowledge, the first registry-based randomized controlled trial in this setting. We will compare the number of lymph nodes removed during SLNB versus TAD and related morbidity in patients with breast cancer who initially present with clinically node-positive status and convert to clinical node-negative after receiving PST. The number of additional nodes included, the failure of techniques, and the need for subsequent axillary lymphadenectomy according to the results of the frozen section during SLNB versus TAD will be assessed in order to better define both techniques. Methodology: Following similar inclusion and exclusion criteria of the AXSANA(EUBREAST 3) study (cT1-4, cN+, M0 breast cancer patients undergoing PST), eligible patients will be randomized to SLNB versus TAD, prior to PST. The biopsy of the metastatic lymph node/s and the marking of the positive lymph node(s) will be required, regardless of the arm of the study. Only patients achieving complete clinical and radiological axillary response will be included in the analysis. Data will be prospectively collected in RedCap. Patient-reported outcome measures (PROMS) will be collected at a predefined time. Sample size calculation: For 80% power to detect whether the number of removed lymph nodes differs between SLNB and TAD (type 1 error=5%), a sample size of 170 patients per group (340 in total) would be required if the number of lymph nodes removed after TAD is 15% lower than after SLNB (ratio=0.85), i.e., assuming 2.72 removed lymph nodes after TAD and 3.2 after SLNB (according to preliminary data from the AXSANA(EUBREAST-3) registry on 4,336 patients from 284 study sites and 26 countries). The calculation is based on a test for the ratio of two Poisson rates, and parameters mean=3.2 and variance=2.7 of the Poisson distribution for SLNB were calculated from the AXSANA (EUBREAST 3).
Presentation numberPS5-08-24
Impact of Intermittent Fasting on Biomarkers of Inflammation and Health-related Quality of Life: A Feasibility Trial for Women with HR+/HER2- Early Breast Cancer
Sailaja Kamaraju, Medical College of Wisconsin, Milwaukee, WI
S. Kamaraju, Y. Cheng, L. N. Chaudhary, S. N. Tarima, H. N. Phuntling, P. Jessica, L. Choppavarapu, H. Alison, D. Ashley, V. Jin; Cancer Center, 4th Fl, Admin Offices, Medical College of Wisconsin, Milwaukee, WI.
Background: Within one year of starting adjuvant endocrine therapy (AET), the majority struggle to return to pre-cancer weight. At least 25-30% of women report a weight gain of 10 pounds or more1,2. Previous reports suggested that weight gain occurred in 14.7% of AET users compared to 6.3% of the controls3 and a weight gain of 10 pounds in at least 27% of the women4. Additionally, 61% of participants kept the weight they had gained, while only 12% lost weight4. The Intermittent Fasting (IF) Study is a Phase II, single-arm, feasibility trial examining the impact of intermittent fasting (IF) on biomarkers of inflammation and the Functional Assessment of Cancer Therapy-Endocrine Symptoms (FACT-ES) in women with ER-positive and HER2-negative early breast cancer who are starting AET. Methods: Patients with stages I-III, ER+, and HER2-negative breast cancer completed definitive surgery, +/- chemotherapy and radiation, and patients who started AET within 12 weeks of trial enrollment are eligible. Other criteria include a body mass index (BMI) of ≥25, an ECOG performance status of 0-1, adequate organ function, and agreement to biomarker testing (IGF, IL-6, Glucose, CRP, adiponectin, fasting lipids, and markers of epigenomics (H3K27ac)5-7. Exclusion criteria included a history of diabetes, other diseases with fluctuations in blood sugar, dizziness, vertigo, pregnancy, breastfeeding, and heavy alcohol use as determined by the treating providers. Following consent, all subjects will receive instructions on the 14-hour IF study for six months (14-hour fasting period after dinner and 10-hour eating period), with monthly nutritional counseling. Anthropometric measures, the FACT-ES surveys8, and biomarkers will be collected at baseline and 6 months. A total of 20 subjects will be accrued over 36 months for this study. The primary objective is to evaluate the proportion of subjects who adhere to at least 80% of the IF instructions, i.e., follow the IF schedule, on average, for at least eight out of ten days throughout the six-month intervention period. Secondary endpoints include anthropometric measures and the FACT-ES surveys, which were administered at baseline and 6 months after the conclusion of the intervention. Biomarkers will be collected at the study entry and conclusion. Mixed-effects models will be used to analyze longitudinal data. The study was approved on May 16, 2024, and enrollment began on July 31, 2024. At the time of this submission, a total of 81 patients had been prescreened, with 13 consenting (8 accrued and 5 withdrawn). Clinical Trial Identification. NCT06106477 Short Title: Intermittent Fasting Study. Sponsor: David Uihlein Foundation
Presentation numberPS5-08-25
ReDiscover-2, a phase 3 study of RLY-2608 + fulvestrant versus capivasertib + fulvestrant as treatment for locally advanced or metastatic PIK3CA-mutant HR+/ HER2- breast cancer following recurrence or progression on or after treatment with a CDK4/6 inhibitor (trial in progress)
Hope S Rugo, City of Hope Comprehensive Cancer Center, Duarte, CA
H. S. Rugo1, C. Saura2, K. Jhaveri3, C. Barrios4, S. Loi5, F. Marmé6, F. Bidard7, Y. Park8, P. Schmid9, S. Sammons10, G. Curigliano11; 1Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, 2Medical Oncology, Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 3Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 4Medical Oncology, Hospital São Lucas, PUCRS, Porto Alegre, BRAZIL, 5Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 6Medical Oncology, University Hospital Mannheim, Mannheim, GERMANY, 7Medical Oncology, Institut Curie & UVSQ/Université Paris-Saclay, Institut Curie, Paris, FRANCE, 8Medical Oncology, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 9Medical Oncology, Barts Cancer Institute, London, UNITED KINGDOM, 10Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 11Medical Oncology, Istituto Europeo di Oncologia, IRCCS, University of Milano,, Milano, ITALY.
Background PIK3CA mutations constitutively activate PI3Kα and drive approximately 40% of HR+/HER2- breast cancer (BC). While the PI3K inhibitors alpelisib and inavolisib and the AKT inhibitor capivasertib have been approved by the FDA to treat this substantial patient population, these therapies cause significant toxicity, notably hyperglycemia, rash, and diarrhea, due to their non-selective targeting of the pathway. RLY-2608 was developed as a pan-mutant-selective allosteric PI3Kα inhibitor designed to optimize dose intensity and target inhibition with reduced toxicity and improved tolerability. The first-in-human ReDiscover study of RLY-2608 demonstrated encouraging antitumor activity with a median progression-free survival of 10.3 mo (95% CI: 7.2, 18.4) across a range of PIK3CA genotypes and a favorable safety profile when given in combination with fulvestrant in patients with PIK3CA-mutated HR+/HER2- advanced BC previously treated with a CDK4/6 inhibitor (Sammons, ASCO 2025). Based on these findings, RLY-2608 in combination with fulvestrant is being studied in this phase 3 study, ReDiscover-2 (NCT06982521), in patients with PIK3CA-mutated HR+/HER2- advanced BC following recurrence or progression on or after a CDK4/6 inhibitor. Methods ReDiscover-2 is a global, multicenter, open-label, randomized phase 3 study comparing the efficacy and safety of RLY-2608 + fulvestrant to capivasertib + fulvestrant in adult patients with HR+/HER2- locally advanced or metastatic BC with PIK3CA mutation. Approximately 540 patients will be enrolled and randomized 1:1 to receive RLY-2608 (400 mg BID with food) + standard-dose fulvestrant or capivasertib (400 mg BID, 4 days on and 3 days off, with or without food) + standard-dose fulvestrant. Randomization will be stratified by PIK3CA mutation type (kinase vs non-kinase), visceral disease (yes vs no), and geographic region (Region 1 [US, Canada, Western Europe, and Australia] vs Region 2 [Latin America and Eastern Europe] vs Region 3 [Asia]). The primary endpoint is PFS assessed by blinded independent central review in patients having tumors with PIK3CA kinase domain mutations and in all patients. Overall survival is a key secondary endpoint within the same populations. Key eligibility criteria: • ≥18 years of age with ECOG performance status of 0-1 • Confirmed diagnosis of HR+/HER2- locally advanced or metastatic BC with radiological or objective evidence of recurrence or progression • Presence of one or more primary oncogenic PIK3CA mutations without evidence of AKT or PTEN alterations • Measurable disease per RECIST v1.1 or evaluable bone-only disease • Previous treatment for HR+/HER2- advanced BC with: – at least 1 and no more than 2 lines of endocrine therapy (ET). Prior fulvestrant is allowed – 1 prior line of CDK4/6 inhibitor therapy • Glycosylated hemoglobin (HbA1c) <7.0% (<53 mmol/mol) and fasting plasma glucose <140 mg/dL. Type 1 diabetes, or Type 2 diabetes requiring antihyperglycemic medication are excluded • No prior PI3K, AKT, or mTOR inhibitors or any agent whose mechanism of action is to inhibit the PI3K/AKT/mTOR pathway ReDiscover-2 (NCT06982521) is open for enrollment. For further information, contact: clinicaltrials@relaytx.com.
Presentation numberPS5-08-26
The PREDICT II Registry: A Prospective Study to Evaluate the Clinical Utility of a 7-Gene Predictive Biosignature on Treatment Decisions in Patients with Ductal Carcinoma In Situ
Anna Daily, PreludeDx, Laguna Hills, CA
P. Borgen1, P. Whitworth2, A. Daily2, K. Mittal2, S. Shivers2, T. Bremer2; 1Surgical Oncology, Maimonides Medical Center, Brooklyn, NY, 2Medical Affairs, PreludeDx, Laguna Hills, CA.
Purpose/Objective(s): A majority of women with ductal carcinoma in situ (DCIS) treated with breast conserving surgery (BCS) do not benefit from adjuvant radiation therapy (RT). However, evidence from randomized clinical trials supports the role of RT in reducing the risk of local recurrence. Given the understanding that more than two-thirds of women with DCIS do not recur after BCS alone, NCCN guidelines recommend utilization of RTOG-9804 criteria (screening detected, grade I/II, size < 2.5 cm, margins > 3 mm) to consider omitting RT after BCS for DCIS. To overcome limitations of clfinicopathologic (CP) risk assessment, a validated biosignature that integrates the protein expression of seven genes and four CP factors (DCISionRT, PreludeDx, Laguna Hills, CA) is currently in use in many breast centers. The 7-gene biosignature has been clinically validated to be both prognostic for in breast recurrence (IBR) risk and predictive for response to RT. Recent evidence demonstrates that approximately half of patients meeting RTOG-9804-like critera wil be identified by the 7-gene biosignature as having elevated IBR risk and significant RT benefit. The study assesses factors impacting shared decision-making, including physician recommendations and patient preferences incorporating the biosignature test results along with CP factors. The main outcome is the impact of test results on individual preferences and treatment recommendations. Materials/Methods: PREDICT II is a multicenter (30 site), prospective, observational registry, open to 3000 women with DCIS. Primary endpoints are changes in treatment recommendations for surgery, RT, and hormonal therapy. Secondary endpoints are identification of key drivers for treatment recommendations, such as age, size, grade, patient preference, biosignature status and IBR risk. The study is open to females age 30-85 who are candidates for BCS and eligible for RT and/or systemic treatment. Subjects must not have been previously treated for DCIS or invasive breast cancer. DCIS patients, breast surgeons, and radiation oncologist participate in informed, shared, decision-making based on standard CP criteria. The preference and strength of preference for mastectomy vs. breast conservation (BCS) and radiation treatment (RT) vs. no RT, following shared decision-making, is recorded before and after biosignature results are available. Recurrence rates at 5 and 10 years will be documented. Changes in treatment recommendations will be analyzed using McNemar’s test (alpha level = 0.05). Multivariable logistic regression will be used to determine odds ratios of CP factors affecting pre- and post-test treatment recommendations. Covariates include hormone receptor status, nuclear grade, tumor necrosis, family history of breast cancer, race, ethnicity, patient preference, physician specialty, and the biosignature score. Differences in recurrence-free and overall survival will be assessed by Kaplan-Meier survival analysis using the log-rank test and/or the Cox Proportional Hazards model. The study has been approved by WCG IRB (Tracking #20172841) and/or local IRBs at each site. ClinicalTrials.gov: NCT03448926.
Presentation numberPS5-08-27
Single arm phase II study with abemaciclib and bicalutamide in locoregionally advanced inoperable or metastatic androgen receptor-positive triple-negative breast cancer (ABBICAR)
Kristien Borremans, KU Leuven, Leuven, Belgium
K. Borremans1, A. Laenen2, M. Maetens1, P. Aftimos3, H. Wildiers4, G. Floris5, H. Denys6, C. Fontaine7, A. Requilé8, S. Altintas9, K. Punie10, C. Desmedt1, P. Neven11; 1Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, BELGIUM, 2Leuven Biostatistics and Statistical Bioinformatics Centre (L-BioStat), KU Leuven, Leuven, BELGIUM, 3Clinical Trials Conduct Unit, Institut Jules Bordet, Brussels, BELGIUM, 4General Medical Oncology Unit, Department of Oncology, University Hospitals Leuven, Leuven, BELGIUM, 5Department of Pathology, University Hospitals Leuven, Leuven, BELGIUM, 6Medical Oncology, University Hospital Ghent, Ghent, BELGIUM, 7Medical Oncology Department, Brussels University Hospital, Jette, BELGIUM, 8Limburg Oncology Center, Jessa Hospital, Hasselt, BELGIUM, 9Department of Medical Oncology, Antwerp University Hospital, Edegem, BELGIUM, 10Department of Medical Oncology, Oncology Center Antwerp, Ziekenhuis aan de Stroom, Antwerp, BELGIUM, 11Department of Gynecological Oncology and Multidisciplinary Breast Center, University Hospitals Leuven, Leuven, BELGIUM.
Background: Triple-negative breast cancer (TNBC) accounts for 15% of all breast cancers and has a poor prognosis, with a 5-year survival rate of only 12% in the metastatic setting. Between 20-40% of these express the androgen receptor (AR). The luminal androgen receptor (LAR) subtype of TNBC is additionally characterized by luminal gene expression. This subtype is typically less responsive to conventional chemotherapy and is mostly seen in elderly patients who more often tolerate chemotherapy poorly. Preclinical studies and early-phase clinical trials suggest that anti-androgen therapies may offer clinical benefit at a higher tolerability. Furthermore, loss-of-function alterations in the Retinoblastoma protein (RB1) are less frequent in LAR, making them more susceptible to CDK4/6 inhibition. This provides a rationale for combining abemaciclib, a CDK4/6 inhibitor, with bicalutamide, an anti-androgen—an approach not yet explored in clinical trials. Methods: ABBICAR (NCT06365788) is a multicenter single arm phase II study evaluating the efficacy and safety of bicalutamide in combination with abemaciclib in inoperable or metastatic AR-positive TNBC. Additionally, inclusion is allowed of estrogen receptor low patients (max 10 patients). A total of 53-59 patients will be enrolled, beginning with a dose-finding safety lead-in (n=6-12), followed by a Simon’s two-stage design (n=25-47). The initial dose in the safety lead-in is abemaciclib 150 mg twice daily plus bicalutamide 150 mg once daily. If after one cycle, no more than one dose-limiting toxicity (DLT) occurs in the first six patients, this dose will be used. Otherwise, safety lead-in will be repeated with abemaciclib 100 mg twice daily with the same dose of bicalutamide. The primary endpoint is disease control rate (DCR) at 16 weeks. Translational objectives include evaluation of stromal tumor-infiltrating lymphocytes, AR expression, PBMC dynamics and ctDNA in association with clinical response. The estimated duration is 48 months. Second stage of Simon’s two stage will start if a DCR of 22.6% (n>=7/31) is observed. DCR and binary endpoints will be estimated as a binomial proportion and reported with a 90% confidence interval. Results: Ethics approval has been obtained (EU CT 2022-502272-23-00; available via the EU Clinical Trials Register). Patients will be included in UZ Leuven, UZ Antwerpen, UZ Brussel, UZ Gent, Jessa Hasselt, and ZAS Antwerpen. The initial safety lead-in with abemaciclib 150 mg twice daily began in April 2024. Two DLTs (grade 3 diarrhea) were observed among the first three patients, leading to early termination of this safety lead-in. A second safety lead-in with abemaciclib 100 mg twice daily was initiated, with no DLTs nor additional safety signals observed in six additional patients. The first stage of Simon’s two-stage design began in January 2025. To date, 18 patients have been enrolled and recruitment is ongoing. Conclusion: This trial explores a novel, chemotherapy-sparing treatment strategy for AR-positive TNBC. If successful, it could offer a more tolerable alternative to conventional chemotherapy. Results from the first stage are expected in 2026. This study is funded by a grant from Kom Op Tegen Kanker.
Presentation numberPS5-08-28
Phase I pilot of pegylated liposomal doxorubicin, CD40 agonist antibody CDX-1140, and Flt3 ligand CDX-301 in advanced HER2-negative breast cancer
Sangeetha M Reddy, University of Texas Southwestern Medical Center, Dallas, TX
S. M. Reddy1, C. A. Santa-Maria2, N. Chen3, V. Kaklamani4, J. O’Shaughnessy5, Y. Abdou6, N. Unni1, B. Santos1, S. Syed1, N. Sadeghi1, J. Gruber1, D. Klemow1, Y. Fang1, I. Chan1, N. Peswani1, I. Patel1, S. Shakeel7, M. Carter7, K. Kyle7, R. Nanda3, H. McArthur1, S. Conzen1, C. L. Arteaga7; 1Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 2Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, 3Internal Medicine, University of Chicago, Chicago, IL, 4Internal Medicine, The University of Texas Health Science Center San Antonio, San Antonio, TX, 5Medical Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX, 6Internal Medicine, University of North Carolina, Chapel Hill, NC, 7Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX.
Background: Immune checkpoint inhibitors deliver durable benefit to only a minority of individuals with metastatic breast cancer, in part due to a paucity of activated, antigen-presenting dendritic cells within the tumor microenvironment. Pre-clinical work from our group showed that combining pegylated liposomal doxorubicin (to release tumor antigens) with a CD40 agonist (to license dendritic cells and repolarize macrophages toward an anti-tumor phenotype) and recombinant Flt3 ligand (to expand dendritic cell precursors) produces markedly superior tumor control compared with chemotherapy alone. These findings underpin the first-in-human clinical evaluation of this triplet regimen. Methods: This is a single arm phase I pilot study of the combination of liposomal doxorubicin, CDX-1140 (CD40 agonist monoclonal antibody), and CDX-301 (recombinant Flt3 ligand) in patients with metastatic or unresectable locally advanced HER2 negative breast cancer (triple negative, TNBC; and hormone receptor positive, HR+). Two lead-in cohorts randomize participants in a 2:1 ratio to receive one cycle of either the triplet combination or pegylated liposomal doxorubicin alone, after which all patients transition to triplet therapy; paired tumor biopsies are obtained at baseline and after the lead-in to characterize early immunological changes. Eligibility originally limited enrollment to triple-negative disease but has been broadened to include HR+ tumors. Key eligibility criteria are unresectable stage III or stage IV HER2 negative breast cancer; for TNBC up to three prior therapies for metastatic disease allowed; for HR+ disease prior cyclin dependent kinase 4/6 inhibitors required and up to 3 prior lines of chemotherapy and/or antibody drug conjugates for metastatic disease allowed; and measurable disease by RECIST 1.1 criteria. Key exclusion criteria are prior exposure to an anti-CD40 antibody or Flt3 ligand, anthracycline treatment in the metastatic setting, progression on or within six months of (neo)adjuvant anthracyclines, and a history of non-infectious pneumonitis. CDX-301 is administered only during cycles 1 and 2, whereas pegylated liposomal-doxorubicin and CDX-1140 is continued until disease progression or clinically limiting toxicities. The primary endpoint is determination of a recommended phase II dose of CDX-1140 based on treatment-related adverse events and dose-limiting toxicities. Secondary endpoints include anti-tumor immune responses measured as CD8 T cell infiltration after triplet therapy and after liposomal doxorubicin alone, median progression-free survival, overall response rate, duration of response, and clinical benefit rate. As of June 24th, 2025 this trial is in dose expansion and has enrolled 23 of 30 evaluable patients (NCT05029999). The trial is currently open at University of Texas Southwestern Simmons Comprehensive Cancer Center, University of Chicago, University of Texas San Antonio Health Science Center, Johns Hopkins, and University of North Carolina.
Presentation numberPS5-08-29
Trend-02 study – a phase ii exploratory de-escalation trial of neoadjuvant sacituzumab govitecan plus tislelizumab (sg/i) in early triple-negative breast cancer
Qiang Zhang, Cancer Hospital of DaLian University of Techology,Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province, China
Y. Xu1, M. Wang1, Y. Wei2, Y. Li2, Q. Jin2, L. Chen2, L. Yao1, M. Cui2, Z. Liao2, S. Li2, Q. Zhang2; 1Department of Breast Surgery, the First Hospital of China Medical University, Shenyang City, Liaoning Province, CHINA, 2Department of Breast Surgery, Cancer Hospital of DaLian University of Techology,Liaoning Cancer Hospital & Institute, Shenyang City, Liaoning Province, CHINA.
Background: Neoadjuvant immune checkpoint inhibitor (ICI) combined with chemotherapy has improved pathological complete response (pCR) in triple-negative breast cancer (TNBC). Sacituzumab govitecan (SG), a TROP-2-targeted antibody-drug conjugate, demonstrates efficacy in metastatic TNBC, while tislelizumab (I), a high-affinity PD-1 inhibitor with 14 approved indications in China, shows survival benefits solid tumors. The TREND trial previously reported that eight cycles of tislelizumab plus low-dose chemotherapy achieved a 68.18% pCR, while NeoSTAR showed four cycles of SG plus pembrolizumab yielded 32% pCR with reduced toxicity versus standard care. These findings highlight the need to explore de-escalated, biomarker-driven regimens to optimize efficacy and safety in TNBC neoadjuvant therapy. Objectives: This phase II trial aims to evaluate the pathological complete response (pCR) rate of neoadjuvant SG/I in early TNBC, stratified by TROP-2 expression and combined positive score (CPS). Secondary objectives include assessing imaging response rate (RECIST v1.1), safety profile (CTCAE v5.0), requirement for additional neoadjuvant chemotherapy (NACT), and 2-year event-free survival (EFS). Exploratory analyses will characterize tumor microenvironment dynamics and identify biomarkers associated with treatment response, aiming to establish a precision de-escalation strategy for TNBC. Methods: This single-center, open-label trial enrolls 30 patients with stage II-III TNBC (ER/PR <5%, HER2- by ASCO/CAP, T2<5cm and/or node-positive, ECOG 0-1). Patients are stratified into three cohorts: A (TROP-2(+), CPS<10%, n=10), B (TROP-2(+), CPS≥10%, n=10), and C (TROP-2(-), CPS≥10%, n=10). Treatment consists of SG 10mg/kg IV d1, d8 q3w plus tislelizumab 200mg IV d1 q3w for six cycles (18 weeks). Patients with progressive disease after clinical evaluation following three cycles of treatment were discontinued from the study. Efficacy evaluation at 18 weeks includes imaging assessment and multi-site core needle biopsy. After six cycles of neoadjuvant treatment, patients with no suspected residual disease (RD) proceed directly to surgery, while those with suspected RD undergo biopsy followed by investigator-selected additional NACT before surgery. After surgery, patients achieving pCR will complete a total of one year of tislelizumab (q3w) treatment; non-pCR patients receive investigator-selected adjuvant therapy. The primary endpoint is pCR rate, with secondary endpoints including NACT rate, clinical response rate, adverse events, and EFS.
Presentation numberPS5-08-30
Cryoablation versus Breast Surgery in the Local Treatment of Early-Stage Breast Cancer: Protocol for the CRYSTAL-SIX Trial (CRYoablation for Small Tumors As Local Treatment)
Vanessa Monteiro Sanvido, Hcor, São Paulo, Brazil
V. M. Sanvido, L. Chinen, M. Nicola, J. O. Gomes, L. G. Barbante, A. B. Cavalcanti, A. P. Nazário; Oncology, Hcor, São Paulo, BRAZIL.
SABCS 2025Cryoablation versus Breast Surgery in the Local Treatment of Early-Stage Breast Cancer: Protocol for the CRYSTAL–SIX Trial (CRYoablation for Small Tumors As Local Treatment)Category: ongoing clinical trial Vanessa Monteiro Sanvido1, Afonso Celso Pinto Nazário11 – Hcor Research Institute, São Paulo, SP, Brazil.Background:Breast-conserving surgery followed by radiotherapy remains the standard of care for early-stage breast cancer. However, interest in minimally invasive alternatives has grown. Cryoablation is a nonsurgical, image-guided technique that induces tumor cell death through freezing and thawing, avoiding general anesthesia and hospitalization. The CRYSTAL–SIX trial (CRYoablation for Small Tumors As Local treatment – SIX) is a phase III study evaluating whether cryoablation is non-inferior to standard breast surgery in terms of locoregional control and cost-effectiveness.Two landmark trials have shaped modern de-escalated care. The SOUND trial showed that omitting axillary surgery in tumors ≤2 cm with negative axillary ultrasound was non-inferior to sentinel lymph node biopsy. The FAST-Forward trial demonstrated that 26 Gy in five fractions over one week was non-inferior to 40 Gy in 15 fractions over three weeks for local control and normal tissue effects over five years.These findings, combined with the advantages of cryoablation, led to the SIX trial design, which integrates cryoablation, omission of axillary surgery, and ultrahypofractionated radiotherapy—completing treatment in just six days.Methods:This phase III, randomized, controlled, multicenter trial compares cryoablation to breast surgery in 750 patients with unifocal, invasive breast carcinoma (≤2.0 cm, T1N0M0), visible on ultrasound and eligible for upfront surgery.Cryoablation will be performed using Cryocare® or ProSense IceCure® under local anesthesia and ultrasound guidance. Surgery includes lumpectomy or mastectomy with sentinel lymph node biopsy. Axillary surgery is omitted in the cryoablation arm. All patients will receive adjuvant treatment according to tumor biology. When indicated, radiotherapy will be delivered as 26 Gy in five fractions over one week.The study incorporates a de-escalated strategy by omitting axillary surgery, applying ultrahypofractionated radiotherapy, and using liquid biopsy for monitoring.Approved by the Ethics Committee, recruitment began in March 2025 and ends in 2030. Registered at ClinicalTrials.gov (NCT06839001).Objectives:Primary: Demonstrate non-inferiority of cryoablation vs. surgery for local treatment over five years, and compare direct treatment costs over one year.Secondary: Assess locoregional recurrence at one year, disease-free and overall survival at five years, and CTCs at baseline, six, and twelve months as prognostic and monitoring tools. Patient satisfaction will be measured via Breast-Q (1 year), and quality of life via EQ-5D.Conclusions:The CRYSTAL–SIX trial investigates cryoablation as a definitive, nonsurgical alternative to breast surgery in early-stage breast cancer. By combining cryoablation, omission of axillary surgery, and ultrahypofractionated radiotherapy, it proposes a modern, patient-centered model.If non-inferiority is confirmed, this strategy may reduce treatment burden, lower costs, and enhance quality of life without compromising oncologic outcomes. The SIX trial reflects and may help redefine current standards in de-escalated breast cancer care.
Presentation numberPS5-09-01
The TeleHealth Resistance Exercise Intervention to Preserve Dose Intensity and Vitality in Elder Breast Cancer Patients (THRIVE-65) Trial
Erica Schleicher, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA
E. Schleicher1, J. Oppenheim2, W. Kemp2, S. Doerksen1, K. Diguglielmo2, S. M. Garrett1, M. Ahmed1, J. Binder1, T. L. Nguyen2, C. Mills3, A. Tanasijevic2, C. Owsusu4, C. Thomson5, K. Basen-Engquist6, J. A. Ligibel2, K. Schmitz1; 1Division of Hematology Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA, 2Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 3Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 4Hematology and Oncology, Case Western Reserve University, Cleveland, OH, 5Health Promotion Sciences, University of Arizonia, Tucson, AZ, 6Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Breast cancer in women over 65 years old is set to increase by 50% over the coming decades. Despite favorable tumor profiles, older women with breast cancer fare worse than younger patients. Age-related declines in functional status, comorbidities, malnutrition, and sarcopenia exacerbate treatment toxicity, contributing to poorer outcomes and lower received dose intensity (RDI). RDI reflects both chemotherapy dose and timing of treatment administration. A RDI below 85% raises recurrence and mortality risk 1.5 to 3-fold. Exercise and higher protein diet interventions targeting physical function may enhance RDI and survival in this vulnerable population. Methods: The TeleHealth Resistance Exercise Intervention to Preserve Dose Intensity and Vitality in Elder Breast Cancer Patients (THRIVE-65) trial is a Phase II randomized controlled trial evaluating a multicomponent resistance and aerobic exercise intervention with protein intake support versus Health Education and Support (HES) control group on RDI (primary aim) in 270 older women with breast cancer while actively undergoing neo/adjuvant chemotherapy. Secondary outcomes include chemotoxicities; physical function, psychological, psychosocial status (via comprehensive geriatric assessment); quality of life, fatigue, sleep; health care utilization; physical activity behaviors; dietary intake; strength; body composition; and treatment dose delays, reductions early termination. We will also explore intervention implementation barriers and facilitators. Eligibility criteria are women age >65 with stage I-III invasive breast cancer, 18-50 kg/m2 BMI, starting at least 10 weeks of neo/adjuvant cytotoxic chemotherapy for curative intent, able to walk for 6 minutes and/or 2 blocks, no current exercise training, no therapeutic diet/weight-loss medication, and English speaking. The HES group receives a tablet with supportive programming (e.g., meditation, yoga, non-tailored nutrition programs) and Fitbit. The virtually-delivered THRIVE-65 intervention includes twice-weekly supervised resistance exercise, 90 minutes/week of unsupervised aerobic exercise, and diet counseling aimed to achieve a protein intake of 1.2 g/kg/day, led by trained exercise physiologist and registered dietitian from a central call center, as well as the HES group tablet and Fitbit. Measurements include anthropometrics, questionnaires, comprehensive geriatric assessment, body composition and objectively measured physical activity. The primary analysis assumes 65-75% of the HES group will achieve an RDI of 85% and hypothesizes that 82-89% of the THRIVE-65 intervention will achieve an RDI of 85% (80% power, two-sided 0.05 significance level). To date, 115 women have been randomized across 3 US medical centers. Support: U01CA271277-03, T32CA186873-11 Clinicaltrials.gov identifier: NCT05535192
| 1Outcomes and measures | Instrument | Baseline | At each cycle | End of Study | ||||||||||
Presentation numberPS5-09-02
Phase 2 study of adjuvant liposomal doxorubicin and carboplatin for early-stage triple negative breast cancer
Mridula George, Rutgers Cancer Institute, New Brunswick, NJ
M. George1, C. Omene1, A. Litvak2, G. Raptis2, A. Cruz3, K. Toomey4, S. Kumar5, N. Ohri6, L. Potdevin5, K. Harper7, E. L. Gramiccioni8, B. Haffty6, H. S. Sloane7, D. Toppmeyer1; 1Medicine, Rutgers Cancer Institute, New Brunswick, NJ, 2Medicine, RWJBarnabas Health – Cooperman Barnabas Medical Center, Livingston, NJ, 3Medicine, RWJBarnabas Health, Jersey City, NJ, 4Medicine, RWJBarnabas Health – Steeplechase Cancer Center, New Brunswick, NJ, 5Surgery, Rutgers Cancer Institute, New Brunswick, NJ, 6Radiation Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 7Medical Affairs, Haystack Oncology, Baltimore, MD, 8Medical Affairs, Hay, Baltimore, MD.
Background: Patients with stage 1 triple negative breast cancer (TNBC) are at high risk of recurrence, given the aggressive nature of the disease. A SEER database review of patients diagnosed with Stage IA TNBC between 2010 and 2019 showed that chemotherapy use increased significantly from 2010-2019 among patients with T1b (p = 0.001) and T1c tumors (p < 0.0001), reaching ≥60% in patients with T1b and ≥70% in patients with T1c tumors across most years. In patients with T1c tumors, chemotherapy was associated with improved BCSS, with a 5-year BCSS of 94.5% for patients receiving chemotherapy vs. 91.2% in the no/unknown chemotherapy group (Adjusted HR = 0.64; 95% CI: 0.48-0.85). Other studies have shown the benefit of adjuvant chemotherapy for stage 1 triple negative breast cancers especially T1c tumors. Given the recurrence free survival and overall survival benefit seen with systemic therapy, guidelines recommend adjuvant chemotherapy for TNBC that are at least 0.6 cm in size. Standard anthracycline-based and taxanes based chemotherapy regimens that have shown benefit in stage II-III breast cancers are routinely used for stage 1 TNBC. However, these regimens are associated with significant risk of neuropathy, alopecia and neutropenia. An investigator-initiated trial at our institution studied the combination of liposomal doxorubicin and carboplatin in the neoadjuvant setting for patients with stage II-III TNBC (ClinicalTrials.gov Identifier: NCT02315196). This study showed that the combination achieved pathologic complete response (pCR) similar to standard regimens with good tolerability. The regimen was well tolerated with minimal risk of alopecia, reduced risk of neuropathy, and lower rates of neutropenia. Selection of liposomal doxorubicin has gained favor due to the lower degree of toxicity, including cardiotoxicity, as compared to conventional doxorubicin while retaining similar efficacy. Based on the results of this study, we are conducting a phase 2 study that will evaluate this regimen in the adjuvant setting for patients with Stage 1 and 2 TNBC. (ClinicalTrials.gov Identifier: NCT05949021) METHODS: The primary objective of this study is to evaluate the efficacy of liposomal doxorubicin and carboplatin in the adjuvant phase for patients with Stage 1 or 2 triple negative breast cancer, as measured by the 5-year disease-free survival (DFS) rate. The current 5-year recurrence-free survival (RFS) is 92.5% compared with 66.5% for patients treated without chemotherapy With 2 years of accrual and 5 years of additional follow-up, we will need 30 patients to achieve 80% power to detect an increase in the five-year survival rate from 66.5% to 84.5% using a 5% level test of survival. Patients diagnosed with early-stage (upto 2.5cm) breast cancer (Estrogen Receptor ≤ 20%; Progesterone receptor ≤ 20% and HER2 negative) who completed primary breast surgery with axillary staging are eligible for the study. Patients must have baseline Left ventricular ejection fraction >50%. Patients will receive liposomal doxorubicin (30mg/m2) and carboplatin (AUC 5) every 4 weeks for 4 cycles. Patients will undergo tumor-informed circulating tumor DNA (ctDNA) monitoring with the Haystack MRD test at baseline, cycles 2 and 4, and every 6 months thereafter for 24 months. After completion of chemotherapy, patient will receive adjuvant breast radiation. Patient will be on follow up for up to 5 years. The study is currently enrolling patients at Rutgers Cancer Institute and Robert Wood Johnson Barnabas Health (RWJBH) affiliate sites in New Jersey.
Presentation numberPS5-09-03
Prospective longitudinal study of circulating serum progranulin (PGRN/GP88) levels and its association with tumor response to therapies in patients with metastatic breast cancer (MBC)
Katherine H R Tkaczuk, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD
K. H. Tkaczuk1, K. Hu1, P. Y. Rosenblatt1, N. Tait1, Y. BinBin2, G. Serrero3; 1Medicine, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, 2NA, A&G Pharmaceutical INC, Columbia, MD, 3NA, A&G Pharmaceutical INC, Baltimore, MD.
The ability to monitor response to therapy and disease progression in patients with metastatic breast cancer (MBC) is a major step in patient management. Sequential imaging remains the method of choice for the assessment of disease status and monitoring of disease response or progression. Serum levels of tumor-associated biomarkers such as CA15-3 and CEA are also measured to follow MBC patients’ disease status. However, it may also be valuable to measure levels of novel biomarkers that have been shown to be overexpressed in breast cancer tumors and play a role in drug resistance and for which therapeutic development is on-going. Progranulin, also called Glycoprotein 88kDa (PGRN/GP88) is an autocrine growth factor overexpressed in several cancers including breast cancer. PGRN plays a significant role in breast tumorigenesis. PGRN/GP88 overexpression in invasive ductal carcinoma (IDC) is associated with malignant phenotype, estrogen (ER) independence, increased proliferation, survival, angiogenesis, and drug resistance. High PGRN/GP88 tumor expression measured by immunohistochemistry in ER receptor positive IDC is an independent prognostic marker associated with increased risk of recurrence and mortality. Clinical studies have demonstrated that serum PGRN levels determined by sandwich enzyme immunoassay are elevated in breast cancer (BC) patients, compared to healthy individuals. In MBC patients, low serum PGRN levels correlate with increased overall survival. Based on these observations, an IRB approved, prospective study was established at the University of Maryland Greenebaum Comprehensive Cancer Center to examine serum PGRN/GP88 levels in association with disease status as measured by RECIST 1.1 criteria in MBC patients receiving standard of care therapies. A total of 103 MBC patients with measurable or evaluable metastatic disease will be consented and enrolled. For inclusion, patients must have been re-staged within 4 weeks of study entry and continue or begin new anticancer therapy. Currently, 50 patients have been enrolled with blood samples collected every 2-3 months or whenever there is a disease event such as clinical or imaging progression. Standard of care (SOC) laboratory assessments and radiographic imaging/staging will be done on study to complement blood collection to measure PGRN/GP88. The samples are stored at -70C until evaluation of PGRN/GP88 using an enzyme linked immunoassay developed by A&G Pharmaceutical. The serum PGRN/GP88 levels will then be correlated with disease response by RECIST 1.1 (progression, stable for =6 months, partial/complete response), progression free survival (PFS), duration of response (DOR) and/or need for change of therapy. In this study the decision about the change of cancer therapy will be based on the SOC approaches and will not be guided by PGRN/GP88 results. This study is supported by grant R44CA210817 from the National Cancer Institute to Ginette Serrero and the University of Maryland Greenebaum Comprehensive Greenebaum Cancer Center.
Presentation numberPS5-09-04
Tbcrc 058: a randomized phase 2 study of enzalutamide, enzalutamide with mifepristone, and treatment of physician’s choice in patients with androgen receptor-positive metastatic triple-negative or estrogen receptor-low breast cancer (nct06099769)
Rita Nanda, The University of Chicago, Chicago, IL
R. Nanda1, Y. Chen2, Y. Abdou3, N. Jahan4, E. L. Mayer5, M. Melisko6, A. Syldor2, C. Friedman2, J. Savoie5, F. Pareja7, B. Weigelt8, S. Chandarlapaty2, N. Turner9, M. Sharifi10, S. Conzen11, T. Traina2; 1Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, 2Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 3Division of Oncology, The University of North Carolina, Chapel Hill, NC, 4Division of Hematology/Oncology, The University of Alabama, Birmingham, AL, 5Division of Breast Oncology, Dana Farber Cancer Institute, Boston, MA, 6Division of Hematology/Oncology, The University of California San Francisco, San Francisco, CA, 7Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 8Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 9Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, UNITED KINGDOM, 10Department of Medicine, Section of Hematology, Oncology and Palliative Care, The University of Wisconsin, Madison, WI, 11Division of Hematology/Oncology, The University of Texas Southwestern, Dallas, TX.
Background: Triple-negative breast cancer (TNBC) refers to a heterogenous group of breast cancers that lack expression of ER, PR, and HER2. Despite recent advances with immunotherapy (IO) and antibody-drug conjugates (ADCs), TNBC remains the most aggressive subtype, characterized by a high risk of recurrence and a short overall survival in the metastatic setting. Breast tumors with low levels of ER and PR expression (1-10%) clinically behave like TNBC, and clinical management follows the TNBC treatment (tx) paradigm. We and others have identified a subset of breast tumors which are ER/PR/HER2 negative and express the androgen receptor (AR). Enzalutamide (enza), an AR-antagonist, has demonstrated activity in AR+ metastatic TNBC (Traina et al, JCO 2018). Activation of glucocorticoid receptor (GR) activity has been implicated as a mechanism of resistance to AR inhibition in prostate and breast cancers (Kach et al, Sci Transl Med 2015). Advanced TNBC remains an area of high unmet need, particularly in chemotherapy and checkpoint inhibitor resistant. This randomized study will evaluate the efficacy of AR antagonism alone (enza monotherapy) or enza plus the GR antagonist mifepristone (mif) as compared to physician’s choice chemotherapy (TPC). Methods: This is a randomized phase II trial; 201 patients (pts) will be randomized in a 1:1:1 fashion to enza, enza plus mif, or TPC (carboplatin, paclitaxel, eribulin, or capecitabine). The primary endpoint (endpt) is progression-free survival (PFS), and the trial is designed to test the hypothesis that PFS in the pooled enzalutamide arms is superior to TPC; there is 80% power to detect a hazard ratio (HR) of 0.70, corresponding to increase in PFS from 3.5 months (mos) with TPC to 5.0 mos with enza-based tx. Secondary endpts include comparisons of PFS among the 3 arms and evaluation of response rate, clinical benefit rate, duration of response, overall survival, safety/toxicity, and patient-reported outcomes by arm. Exploratory endpts include correlation of tumor and circulating markers (AR-V7 in circulating tumor cells and circulating tumor cell DNA) with tx response. Eligible pts must have: ECOG 0-2, metastatic ER/PR/HER2 negative or low breast cancer (BC), tumor AR expression >/= 10% by central testing, measurable or evaluable disease, up to 2 prior lines of chemotx, any # prior endocrine txs, no prior anti-AR tx, no prior mif tx, and concurrent CYP17 inhibitor use prohibited. Pts with PD-L1+ BC must have received prior IO if not contraindicated. Pts must have normal organ function and no history of brain mets. As of 7/9/2025, 19 of 201 pts have begun protocol-specified tx. This trial is supported by the Translational Breast Cancer Research Consortium, The Breast Cancer Research Foundation, The TaTa Sisterhood Foundation, Pfizer/Astellas, and Corcept Therapeutics.
Presentation numberPS5-09-06
A Phase II Adjuvant Trial Evaluating the Impact of Omitting Chemotherapy Based on Patient’s Selection for Moderate to High-Anatomical Risk, Low-Genomic Risk, ER-positive, HER2-negative Breast Cancer with a Combination Regimen of Ribociclib and Optimized Endocrine Therapy – SELECT Trial
Jing Du, Yale Cancer Center, New Haven, CT
J. Du, W. Wei, A. Kahn, M. Rozenblit, L. Pusztai, A. Silber, S. Schellhorn, T. Sanft, M. DiGiovanna, N. Fischbach, R. Legare, D. O’Neil, J. Kanowitz, Z. Rahman, K. Fenn, A. Bulgaru, S. Hall, W. Kidwai, K. Blenman, I. Krop, M. Lustberg, E. Winer; Medical Oncology, Yale Cancer Center, New Haven, CT.
Background: Most patients with ER-positive, HER2-negative, node-positive breast cancer are treated with curative intent, which typically involves some combination of surgery, radiation, endocrine therapy (ET) and often chemotherapy. The recent integration of CDK 4/6 inhibitors into adjuvant ET has complicated treatment decision-making for this subtype of breast cancer. These agents have demonstrated substantial benefits in reducing recurrence risk when combined with ET, prompting a reassessment of the role and necessity of chemotherapy in certain patient populations. However, it remains unclear to what extent prior exposure to a course of adjuvant chemotherapy impacts treatment persistence of CDK 4/6 inhibition. In the NATALEE and monarchE trials, approximately 90% of participants received prior chemotherapy, yet 28 – 36% participants discontinued the CDK 4/6 inhibitors prematurely. These findings underscore the significant barrier in maintaining long-term persistence to CDK 4/6 inhibitor therapy and emphasize the need to develop and implement strategies for optimizing treatment regimens to minimize discontinuation rates, especially in patients for whom the benefit of chemotherapy is uncertain. Methods: This is a Phase II, two-arm, single center, patient preference clinical trial designed to evaluate the impact of omitting adjuvant chemotherapy on ribociclib treatment persistence in moderate to high-anatomical risk, low-genomic risk (Oncotype DX Recurrence Score, RS ≤ 25), ER-positive, HER2-negative breast cancer participants treated with a combination regimen of ribociclib and optimized endocrine therapy. Eligible participants aged ≥18 years, ECOG PS of 0 to 1, with anatomic stage II/III ER-positive (≥10%), HER2-negative breast cancer (men or premenopausal women with T1-3N1-2 or postmenopausal women with T3N1 or T1-3N2) and RS 0-25 will be included. Participants will have the option to decide whether to include adjuvant chemotherapy in their treatment plan. Arm 1 will receive the combination of ribociclib and optimized ET. Arm 2 will be treated with adjuvant chemotherapy followed by a combination of ribociclib and optimized ET. Optimized ET includes an oral aromatase inhibitor (letrozole or anastrozole) for all participants, with the addition of a GnRH agonist (goserelin) to achieve gonadal suppression in all men and premenopausal women. Key exclusion criteria are distant metastases, T4, N3, neoadjuvant chemotherapy and clinically significant uncontrolled heart disease at screening. The primary objective is to evaluate 1-year discontinuation rate of ribociclib in each arm. Secondary endpoints include 3-year invasive disease-free survival, factors impacting treatment decision, quality of life, decisional regret, and fear of recurrence. Estimated enrollment is 140 participants (70 each arm) to ensure we can estimate one-year discontinuation rate with a reasonable level of precision. Recruitment is ongoing. Clinical trial information: NCT06953882.
Presentation numberPS5-09-07
Etic-lm : a multicenter, single-arm, phase 2 study evaluating the efficacy of oral tucatinib, oral capecitabine, and intrathecal trastuzumab in patients with her2-positive metastatic breast cancer with leptomeningeal metastases, a ucbg study
Thomas Bachelot, Centre Léon Bérard, LYON, France
L. Larrouquere1, S. Chabaud2, C. Poisson3, G. Emile4, C. Bailleux5, A. Darlix6, C. Jouannaud7, S. Mijonnet8, T. Roque8, T. Bachelot1; 1Medical Oncology Department, Centre Léon Bérard, LYON, FRANCE, 2Direction de la Recherche clinique et de l’Innovation (DRCI), Centre Léon Bérard, LYON, FRANCE, 3Breast unit, medical oncology department,, Gustave Roussy Cancer Campus, VILLEJUIF, FRANCE, 4Medical Oncology Department, François Baclesse Comprehensive Cancer Center, CAEN, FRANCE, 5Medical Oncology Department, Centre Antoine Lacassagne, NICE, FRANCE, 6Medical Oncology Department, Montpellier Cancer Institute (ICM), MONTPELLIER, FRANCE, 7Medical Oncology Department, Institut Jean Godinot, Reims, FRANCE, 8R&D department, Unicancer, Paris, FRANCE.
Background Leptomeningeal metastases (LM) are a rare but devastating form of disease progression in HER2-positive (HER2+) metastatic breast cancer (MBC), occurring in 5-10% of patients. Median overall survival (OS) historically ranges from 4 to 6 months, with poorer outcomes observed in type 1 LM (characterized by the presence of tumor cells in the cerebrospinal fluid (CSF)), and longer OS reported with newer therapies such as trastuzumab deruxtecan (T-DXd), brain-penetrant HER2 tyrosine kinase inhibitors, or intrathecal (IT) trastuzumab. Tucatinib is a highly selective HER2 tyrosine kinase inhibitor that has demonstrated significant intracranial activity in HER2+ MBC with brain metastases, including in heavily pretreated patients, when combined with intravenous trastuzumab and oral capecitabine. IT trastuzumab has shown encouraging signs of efficacy without safety concerns in two prospective studies in HER2+ MBC with LM, offering one of the few targeted strategies capable of delivering anti-HER2 therapy directly into the CSF, thereby bypassing the blood-CSF barrier. Based on the complementary pharmacological properties and mechanisms of action of these agents, the ETIC-LM trial was designed to evaluate the efficacy and safety of a triplet regimen composed of oral tucatinib, oral capecitabine and IT trastuzumab in patients with HER2+ MBC and LM, including those previously exposed to T-DXd and those presenting with the most severe form of LM: type 1 LM. Trial designETIC-LM is a national, open-label, single-arm phase II study enrolling patients with histologically confirmed HER2+ MBC and LM confirmed by MRI and/or presence of tumor cells in the CSF. Eligible patients must have an ECOG performance status of 0 to 2 and evaluable disease according to RANO-LM and RECIST1.1 criteria. The investigational treatment consists of oral tucatinib (300 mg twice daily, continuously), oral capecitabine (1000 mg/m² twice daily on days 1 to 14 of each 21-day cycle), and intrathecal trastuzumab (150 mg weekly, administered via lumbar puncture or Ommaya reservoir). Treatment is continued until disease progression, unacceptable toxicity, or patient withdrawal. Objectives The primary objective is to evaluate the 12-month overall survival (12m-OS) rate. Secondary objectives include the assessment of neurological symptom improvement using the NANO scale, progression-free survival (PFS), LM-specific and brain-specific PFS using RANO-LM, OS, CSF cytological response at 4 weeks, quality of life (assessed by EORTC QLQ-C30 and BN20 questionnaires), and cognitive function (evaluated using the Montreal Cognitive Assessment (MoCA)).Safety endpoints include the incidence and severity of adverse events according to CTCAE version 5.0, as well as longitudinal cognitive evaluations.Ancillary studies aim to explore pharmacokinetics and potential biomarkers in CSF and plasma. Statistical Methods Using a Fleming single-stage design, the trial aims to detect an increase in 12-month overall survival (12m-OS) from 20% (null hypothesis) to 45% (alternative hypothesis), with 90% power and a one-sided alpha of 5%. A total of 29 evaluable patients are required. If at least 10 patients are alive at 12 months, the regimen will be considered worthy of further investigation. Current Status The trial began enrollment in February 2024. As of June 2025, 11 patients have been enrolled. Recruitment is ongoing (NCT05800275). Funding Pfizer
Presentation numberPS5-09-08
Despatil study: a phase II trial of de-escalated neoadjuvant chemotherapy for early-stage triple-negative breast cancer guided by tumor-infiltrating lymphocytes (TILs) and radiologic response
Renata C Bonadio, Instituto D’Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil
R. C. Bonadio1, R. Barroso-Sousa2, J. Bines3, C. dos Anjos4, D. Assad-Suzuki5, G. Tiburzio6, L. Testa1; 1Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), São Paulo, BRAZIL, 2Oncology, Brasilia Hospital, Oncologia Américas, Brasília, BRAZIL, 3Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, BRAZIL, 4Oncology, Hospital Sírio-Libanês, São Paulo, BRAZIL, 5Oncology, Hospital Sírio-Libanês, Brasília, BRAZIL, 6Patology, Instituto D’Or de Pesquisa e Ensino (IDOR), São Paulo, BRAZIL.
Background:Currently, a four-chemotherapy drug regimen (plus pembrolizumab if stage II-III), is frequently used as neoadjuvant strategy for triple-negative breast cancer (TNBC). Theses regimens are associated with high toxicity rates, and, given the heterogeneity of the disease, some patients may achieve favorable outcomes with less intensive treatment strategies. In this context, high levels of tumor-infiltrating lymphocytes (TILs) have been identified as a marker of excellent prognosis. Additionally, radiologic response can assist in treatment decision-making, allowing escalation only for patients with inadequate response, thereby optimizing therapy. The DespaTIL study investigates a biomarker-driven de-escalation approach to neoadjuvant chemotherapy (NAC) in early-stage TNBC. Methods:DespaTIL is a multicenter, single-arm, investigator-initiated phase II trial evaluating a de-escalated NAC strategy in patients with stage I TNBC with any level of TILs, or stage II TNBC with ≥50% stromal TILs. All patients receive four cycles of carboplatin and a taxane. Radiologic response is assessed via breast magnetic resonance imaging after cycle 4. Patients achieving a radiologic complete response proceed to surgery. Those without a radiologic complete response are escalated to four additional cycles of anthracycline and cyclophosphamide (AC), with or without pembrolizumab at the physician’s discretion.Eligible participants are patients with HER2-negative invasive breast carcinoma, estrogen receptor and progesterone receptor expression <10%, and one of the following: (1) clinical stage T1c N0 M0 with any TIL level; or (2) clinical stage T2 N0 M0 with TILs ≥50%.A total of 40 patients will be enrolled.The primary endpoint is pathologic complete response (pCR) following four cycles of carboplatin and taxane. Secondary endpoints include the correlation between radiologic and pathologic response, residual cancer burden (RCB), event-free survival (EFS), overall survival (OS), and subgroup analyses by disease stage. Patient enrollment is expected to begin in the second half of 2025.
Presentation numberPS5-09-09
Ctc-express: a prospective observational study for ctc biomarker expression and cfdna/cfrna assessment in metastatic breast cancer
Marija Balic, UPMC, Pittsburgh, PA
M. Balic1, D. Riseberg2, T. Pluard3, C. Tweed4, J. Foldi1, F. de Snoo5, L. Stork-Sloots5, F. K. Kuhr6, S. Lazaro7, D. S. Spinner8, R. Prendergast7, C. Dorris6, M. Albitar9, A. M. Brufsky1; 1Division of Hematology/Oncology, UPMC, Pittsburgh, PA, 2Medical Oncology, Mercy Medical Center, Balitimore, MD, 3Medical Oncology, Saint Luke’s Cancer Center, Chesterfield, MO, 4Oncology Hematology, Maryland Oncology Hematology, Annapolis, MD, 5Medical Affairs, Medex15, Amsterdam, NETHERLANDS, 6Medical Affairs, Menarini Silicon Biosystems, Huntingdon Valley, PA, 7Clinical Laboratory, Menarini Silicon Biosystems, Huntingdon Valley, PA, 8Market Access & Reimbursement, Menarini Silicon Biosystems, Huntingdon Valley, PA, 9Medical Affairs, Genomic Testing Cooperative, Lake Forest, CA.
Background Metastatic breast cancer (MBC) remains an incurable disease with a median overall survival (OS) depending on prognostic factors and systemic therapy. As of yet, prospective real-world data on circulating tumor cell (CTC) phenotyping such as expression of HER2, estrogen receptor (ER), and PD-L1 on CTCs and response to therapy is limited. Furthermore, adding cfDNA/cfRNA to CTC phenotyping may provide additional clinical information, for which prospective data are also not yet available. This prospective study in a real-world clinical setting assesses the impact on treatment decisions and response assessment of CellSearch CTC enumeration, CTC biomarker expression, and cfDNA/cfRNA results in subjects with MBC. Trial design CTC EXPRESS is a prospective, multicenter, observational study of CellSearch CTC enumeration, CTC HER2, ER, and PD-L1 expression, and cfDNA/cfRNA results in subjects with histologically proven MBC. Treatment for MBC will be chosen and performed by treating physicians/providers. Subjects will be followed until disease progression. The study will simultaneous enroll two MBC cohorts: Cohort 1 will include HR+/HER2- subjects before starting 1st line therapy and Cohort 2 will enroll triple-negative subjects starting any line of therapy and HR+/HER2- or HER2+ subjects starting 2nd line therapy or beyond. CTCs and cfDNA/cfRNA will be collected minimally at Baseline and before the first re-staging visit. In Cohort 1, additional cfDNA/cfRNA will be collected at week 4, 8 and 12. Physicians will complete a short questionnaire regarding the impact of CTCs and cfDNA/cfRNA on treatment decisions at several timepoints. A total of 187 subjects will be enrolled in up to 10 US centers. The main objectives are: 1. To prospectively assess the impact on treatment decisions and response assessment of CTC enumeration and CTC biomarker expression, and cfDNA/cfRNA results in subjects with MBC. 2. To evaluate the correlation of CTC HER2, ER and PD-L1 expression with response to therapy and PFS. 3. To evaluate the correlation of cfDNA/cfRNA genomic alterations with response to therapy and PFS. 4. To evaluate the correlation of early cfDNA/cfRNA dynamics with response to 1st line therapy and PFS in HR+/HER2- MBC. Clinical trial identification NCT NCT06833853
Presentation numberPS5-09-10
A randomized phase II study comparing monthly versus 3-monthly GnRH agonist for ovarian function suppression in premenopausal patients with HR-positive breast cancer: study design and rationale
Daniella Audi Blotta, Memorial Sloan Kettering Cancer Center, New York, NY
D. Audi Blotta1, Y. Chen2, A. Buzaid3, M. Robson1; 1Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 3Department of Oncology, Beneficência Portuguesa de São Paulo, Sao Paulo, BRAZIL.
Background: The SOFT and TEXT trials have shown substantial survival benefit from adding ovarian function suppression (OFS) with gonadotropin-releasing hormone agonists (GnRHa) to aromatase inhibitors (AI), compared with tamoxifen alone in premenopausal patients with hormone receptor (HR)-positive breast cancer. This benefit was particularly pronounced among women with a higher risk of recurrence, such as younger patients and those who received adjuvant chemotherapy, with an absolute improvement of 15% in disease-free survival. Although most pivotal trials used monthly GnRHa, many clinicians prescribe 3-monthly depot injections for convenience, despite limited prospective data supporting their efficacy. Retrospective data from a Brazilian cohort presented at the 2023 ASCO Annual Meeting [JCO 41, no. 16_suppl (June 1, 2023) 527-527] suggested fewer estradiol (E2) increases with 3-monthly compared to monthly GnRHa, prompting the need for a prospective randomized trial. Methods: This single-institution, randomized, investigator-initiated phase II trial will enroll 50 premenopausal breast cancer patients (age ≤40) who are candidates for adjuvant OFS plus AI at Memorial Sloan Kettering. Eligible participants are women diagnosed with stage I, II, or III HER2-negative or HER2-positive, HR-positive breast cancer (ER >1% and/or PR >1%, according to American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines). Patients will be stratified by prior chemotherapy (yes vs. no) and randomly assigned in a 1:1 ratio to receive either leuprolide at a dose of 22.5 mg every 12 weeks (Cohort A) or leuprolide at a dose of 7.5 mg every 4 weeks (Cohort B), both administered as a single intramuscular injection, in combination with an AI. Leuprolide was selected due to its widespread use in the United States. E2 levels will be measured using high-sensitivity gas chromatography-tandem mass spectrometry (GC/MS/MS) at baseline and at 3, 6, 9, and 12 months. Patients with at least one E2 level above the pre-established threshold of 2.72 pg/mL during the 12-month follow-up will be considered to have suboptimal OFS. Patients in the 3-monthly group will undergo testing at the same frequency as those in the monthly group. The primary endpoint is the difference in the proportion of patients in each treatment arm who experience at least one E2 level > 2.72 pg/mL during the 12-month follow-up. Additionally, E2 levels will be described at each time point, and associations between baseline characteristics and E2 increases will be explored. Conclusions: OFS management remains a major challenge for oncologists worldwide, with limited evidence to guide optimal GnRHa dosing schedules. This study will generate prospective data comparing OFS using monthly versus 3-monthly GnRHa in combination with adjuvant endocrine therapy, addressing an important gap in clinical practice. The findings may contribute to future clinical guidelines for OFS, while also improving convenience, supporting treatment adherence, and enhancing quality of life for premenopausal breast cancer patients globally. Current Status: This investigator-initiated study is supported by the Long-term International Fellowship (LIFe) Award from Conquer Cancer, the ASCO Foundation. IRB approval was obtained in July 2025, and the study is currently in the activation phase. Patient enrollment is expected to begin shortly thereafter.
Presentation numberPS5-09-11
Dose-dense Paclitaxel with Empegfilgrastim vs weekly Paclitaxel in luminal B HER2-negative early breast cancer (PULSE trial)
Petr Krivorotko, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, Russian Federation
P. Krivorotko1, N. Amirov1, V. Mortada2, A. Emelyanov1, R. Pesotskiy1, E. Zhiltsova1, T. Tabagua1; 1Breast Tumor Department, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, RUSSIAN FEDERATION, 2Reconstructive Surgery Department, NMRC of Oncology named after N.N. Petrov, Saint Petersburg, RUSSIAN FEDERATION.
Background. Luminal B HER2-negative breast cancer (BC) is an aggressive subtype with lower chemosensitivity than HER2-positive or triple-negative disease. Standard neoadjuvant chemotherapy (NCT), consisting of doxorubicin-cyclophosphamide (ACq21 or ddACq14) followed by weekly paclitaxel, yields a pathological complete response (pCR) in approximately 20% of patients, which is significantly lower than in other subtypes. Dose-dense (dd) chemotherapy has been shown to improve outcomes in high-risk BC and even in hormone receptor-positive/HER2-negative patients. Paclitaxel can be delivered weekly, biweekly (dose-dense) or every 3 weeks. Evidence suggests that more frequent (weekly or biweekly) paclitaxel dosing improves long term outcomes compared to 3-week schedule. However, no prospective studies have directly compared weekly and biweekly paclitaxel schedules in luminal B early BC. This exploratory trial aims to detect clinically meaningful differences in efficacy of a biweekly paclitaxel schedule supported by empegfilgrastim in comparison to historical weekly paclitaxel and provide early data to inform the planning of a future phase III trial. Aim. To determine whether a biweekly paclitaxel regimen is not inferior in efficacy (as measured by pCR rate) to the standard weekly paclitaxel regimen in patients with luminal B HER2-negative early or locally advanced BC receiving NCT. Study Design. Prospective, single-center, open-label, non-inferiority phase II trial using historical control. Patients in the intervention group will be treated prospectively and compared to a historical cohort treated with weekly paclitaxel. Matching will be performed in a 1:3 ratio using key clinical and pathological variables (age, clinical stage, Ki-67, tumor grade, ER and PR expression). Population. Eligible patients are adult women (≥18 years) with histologically confirmed luminal B breast cancer, defined as estrogen receptor (ER≥1%) positive, HER2-negative tumors with Ki-67 ≥30%. Patients must have early or locally advanced disease (T2-T4 and/or N1-N3, M0) for which NCT is appropriate. Key exclusions include HER2-positive tumors, metastatic disease and any prior chemotherapy for the current cancer. Treatment. All patients in the prospective arm will receive 4 cycles of ddAC (doxorubicin 60 mg/m² + cyclophosphamide 600 mg/m² every 14 days) with empegfilgrastim 7.5 mg as G-CSF support. This is followed by 4 cycles of biweekly paclitaxel (175 mg/m² every 14 days) with empegfilgrastim 7.5 mg support. The historical control arm will include patients previously treated with AC (q21 or q14) followed by weekly paclitaxel (80 mg/m² weekly ×12). Primary endpoint is pCR rate, defined as no residual invasive cancer in breast and lymph nodes (ypT0/is, ypN0) at surgery. Secondary endpoints include RCB 0-I rate, event-free survival, relative dose intensity, breast conservation rate, treatment completion rates, and safety outcomes such as neuropathy, neutropenia, and febrile neutropenia. Statistical Analysis. Analyses will be conducted on an intention-to-treat basis. The primary endpoint will be compared between groups using a one-sided 95% confidence interval to evaluate non-inferiority, with a non-inferiority margin of 10%. Propensity score matching (1:3) will balance baseline characteristics. Logistic regression adjusted for matching strata will estimate odds ratios. Secondary endpoints will be analyzed using Kaplan-Meier estimates, log-rank tests, and Cox proportional hazards models for time-to-event outcomes. Sample Size Justification. Assuming a pCR rate of 20% in the historical weekly paclitaxel group and a non-inferiority margin of 10%, a sample size of 102 patients in the prospective biweekly arm and 307 matched historical controls (1:3 ratio) achieves 80% power with one-sided α=0.05.
Presentation numberPS5-09-12
Small: open surgery versus minimally invasive vacuum-assisted excision for small screen-detected breast cancer – a UK phase III randomised multi-centre trial
Stuart McIntosh, Queen’s University Belfast, Belfast, United Kingdom
S. McIntosh1, C. Coles2, D. Dodwell3, K. Elder4, B. Elsberger5, J. Foster6, C. Gaunt6, J. Henderson7, C. Mabena8, J. Morgan9, Z. Nabi10, I. Lyburn11, S. Paramasivan12, S. Pinder13, S. Pirrie6, S. Potter14, T. Roberts6, N. Sharma15, E. Southgate6, H. Stobart16, A. Talwalkar17, S. Taylor-Phillips18, W. Teh8, E. Turner6, M. Wallis19, D. Rea6; 1Patrick G Johnstone Centre for Cancer Research, Queen’s University Belfast, Belfast, UNITED KINGDOM, 2Department of Oncology, University of Cambridge, Cambridge, UNITED KINGDOM, 3Nuffield Department of Public Health, Oxford University, Oxford, UNITED KINGDOM, 4Western General Hospital, NHS Lothian, Edinburgh, UNITED KINGDOM, 5Breast Unit, NHS Grampian, Aberdeen, UNITED KINGDOM, 6CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UNITED KINGDOM, 7Linda McCartney Breast Unit, Royal Liverpool NHS Trust, Liverpool, UNITED KINGDOM, 8N/A, Edgware Breast Screening UNit, London, UNITED KINGDOM, 9University of Sheffield, School of Medicine and Population Health, Sheffield, UNITED KINGDOM, 10National Radiotherapy Trials QA Group, Mount Vernon Cancer Centre, Northwood, UNITED KINGDOM, 11Breast IMaging Centre, Cheltenham General Hospital, Cheltenham, UNITED KINGDOM, 12Bristol Populatoin Health Science Institute, University of Bristol, Belfast, UNITED KINGDOM, 13Department of Pathology, King’s College London, London, UNITED KINGDOM, 14Bristol Population Health Science Institute, University of Bristol, Bristol, UNITED KINGDOM, 15Breast Imaging Department, Leeds Teaching Hospitals NHS Trust, Leeds, UNITED KINGDOM, 16N/A, Independent Cancer Patients’ Voice, Cambridge, UNITED KINGDOM, 17Breat Unit, Wrightington Wigan and Leigh NHS Foundation Trust,, Wigan, UNITED KINGDOM, 18Warwick Medical School, University of Warwick, Coventry, UNITED KINGDOM, 19Breast Unit, Addenbrookes Hospital, Cambridge, UNITED KINGDOM.
Background: Mammographic screening programmes reduce breast cancer mortality but detect many small good-prognosis tumours which may not progress. Screen-detected cancers are currently treated with standard surgery and adjuvant therapies, with associated morbidities. There is a need to reduce overtreatment of good prognosis tumours and numerous studies are evaluating omission of radiotherapy in low-risk disease. However, there is little evidence for surgical de-escalation, although percutaneous minimally invasive treatment approaches have been described. Vacuum-assisted excision (VAE) is in widespread use for management of lesions of uncertain malignant potential and benign lesions. SMALL (ISRCTN 12240119) aims to determine the feasibility of using this approach to treat small invasive tumours detected within the UK NHS Breast Screening Programme. Methods: SMALL is a phase III multicentre RCT comparing standard surgery with VAE for screen-detected cancers. Main eligibility criteria are age ≥47 years, unifocal grade 1 tumours with maximum diameter 15mm, strongly ER/PR+ve and HER2-ve, with negative axillary staging. Patients are randomised 2:1 to VAE or surgery, with no axillary surgery in the VAE arm. Completeness of excision is assessed radiologically, and if excision is incomplete, patients undergo surgery. Adjuvant radiotherapy and endocrine therapy are mandated in the VAE arm but may be omitted following surgery.Co-primary end-points are1. Non-inferiority comparison of the requirement for a second procedure following excision2. Single arm analysis of local recurrence (LR) at 5 years following VAE Recruitment of 800 patients will permit demonstration of 10% non-inferiority of VAE for requirement of a second procedure. This ensures sufficient patients for single arm analysis of LR rates, where expected LR free survival is 99% at 5 years, with an undesirable survival probability after VAE of 97%. To ensure that the trial as a whole only has 5% alpha, the significance level for each co-primary outcome is set at 2.5% with 90% power. The Data Monitoring Committee will monitor LR events to ensure these do not exceed 3% per year. Secondary outcome measures include time to ipsilateral recurrence, overall survival, complications, quality of life and health economic analysis. A novel feature of SMALL is integration of a QuinteT Recruitment Intervention (QRI), which aims to optimise recruitment to the study. Recruitment challenges are identified by analysing recruiter/patient interviews and audio-recordings of trial discussions, and by review of trial screening logs, eligibility and recruitment data and study documentation. Solutions to address these are developed collaboratively, including individual/group recruiter feedback and recruitment tips documents.Results: SMALL opened in December 2019. Recruitment halted in 2020 for 5 months due to COVID-19. At 9th July 2025, 49 centres are open, with 640 patients randomised. The randomisation rate is approximately 45%, and per site recruitment rate is 0.4-0.5 patients/month. Drawing from QRI findings and insights from patient representatives, a recruitment tips document has been circulated (on providing balanced information about treatments, encouraging recruiters to engage with patient preferences, and explaining randomisation). Individual recruiter feedback is ongoing, and wider feedback is being delivered across sites via recruitment training workshops. Patient interviews are ongoing to explore patient views and experiences of the trial.Conclusion: SMALL continues to have excellent recruitment, is expected to complete recruitment in 2026 and have a global impact on treatment of breast cancer within mammographic screening programmes.SMALL is funded by the UK NIHR HTA programme award 17/42/32
Presentation numberPS5-09-13
Evaluation of the diagnostic accuracy of ultra-fast ConfocAl imaging for rEtroareolar Limit in nIpple-sparing mAstectomy: Camelia project
Angelica Conversano, GUSTAVE ROUSSY, Villejuif, France
A. Conversano1, N. Ben-Romdhane2, N. Joyon2, A. Puchar1, A. Ilenko1, A. Turpin1, A. Alfaro3, M. Mathieu2, M. Abbaci3; 1Department of Breast and Plastic Surgery, GUSTAVE ROUSSY, Villejuif, FRANCE, 2Department of Medical Biology and Pathology, GUSTAVE ROUSSY, Villejuif, FRANCE, 3Surgery and Pathology Photonic Imaging Group, UMS, AMMICa 23/3655,Plateforme Imagerie Et Cytométrie, GUSTAVE ROUSSY, Villejuif, FRANCE.
Background Nipple-sparing mastectomy (NSM) followed by breast reconstruction is now widely recognized as a safe technique, including in patients with a history of radiotherapy or chemotherapy, regardless of tumor size or axillary status, except in cases of inflammatory breast cancer or direct involvement of the nipple-areola complex on imaging or clinical examination. The main oncological concern is the potential persistence of tumor remnants in the residual subareolar breast tissue. At least 10% of patients undergo a second surgery to excise this tissue. Such reinterventions may increase patient anxiety, negatively impact body image, raise healthcare costs, and delay adjuvant treatments. A new generation of ultra-fast confocal microscopes (UFCM) enables high-resolution imaging of fresh tissue in a shorter time than frozen section analysis, potentially guiding intraoperative assessment and avoiding reoperation [1-4]. Methods The CAMELIA project is a prospective, non-interventional, ex vivo clinical study analyzing retroareolar margins after NSM, imaged using UFCM and interpreted by pathologists and surgeons. The examined tissue is preserved for subsequent conventional histology (HES). At this stage, UFCM results do not influence surgical management. The primary objective is to use UFCM to provide an accurate intraoperative diagnosis of retroareolar tissue. Accuracy, sensitivity, and specificity are calculated based on the proportion of images correctly interpreted compared to definitive histology on corresponding HES slides, the reference standard. Results Since October 2024, 40 out of 194 patients who underwent Nipple-Sparing Mastectomy (NSM) have been included in this study. Of these, 8 had prophylactic mastectomies and 32 had therapeutic interventions. There were no bilateral procedures, and 16 patients underwent surgery after neoadjuvant chemotherapy. Importantly, four of these patients had positive margins. Three pathologists participated in the image analysis: two seniors (PT1 and PT2) and one junior (PT3). They analyzed 20 out of 40 images. Both PT1 and PT3 have experience with Ultra-Focalized Cancer Mammography (UFCM). Additionally, an experienced UFCM surgeon interpreted all 40 UFCM images. Images were categorized into four groups: deferred diagnosis, cancer, no cancer, or suspicious. For the first 20 cases, the pathologists’ concordance rate ranged from 59% to 75%. Specifically: PT1 classified 2 out of 3 positive margins as suspicious, PT2 deferred diagnosis for 3 cases and classified 1 out of 3 as suspicious. PT3 classified 1 out of 3 positive margins as suspicious. The surgeon’s concordance rate for the first 40 cases was 72.5%, with one positive margin identified out of four existing positive margins. On average, the intraoperative image acquisition time was 16 minutes. Readers rated the quality of these images at 3.4/5. The average image interpretation time across all specialists was 1 minute and 30 seconds. Conclusions Expected outcomes include high accuracy, sensitivity, and specificity. These results will provide proof of concept for cellular imaging of the retroareolar margin during NSM, with the aim of completing tumor resection if necessary and avoiding reoperation. References: 1. Conversano A. et al. BJS Open. 2023 May 5;7(3):zrad046. 2. Mathieu MC et al. Virchows Arch. 2025 Feb;486(2):299-311. 3. Mathieu MC et al. Life (Basel). 2024 Oct 28;14(11):1384. 4. Spoor J et al. Eur J Surg Oncol. 2024 Jun;50(6):108320.
Presentation numberPS5-09-14
Rossini-platform: a ‘basket factorial multi arm multi stage (mams)’ platform trial in surgical site infection – the breast surgery pillar
Katherine Fairhurst, University of Bristol, Bristol, United Kingdom
K. Fairhurst1, S. Potter1, S. McIntosh2, T. Pinkney3; 1Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UNITED KINGDOM, 2School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UNITED KINGDOM, 3Department of Applied Health Sciences, University of Birmingham, Birmingham, UNITED KINGDOM.
Introduction: Surgical Site Infection (SSI) is the most common postoperative complication costing the UK National Health Service (NHS) more than £700million annually. SSI after breast surgery profoundly impacts patients, for example through readmission for antibiotics, further surgery for washout, and an increased number of hospital visits. SSI may also adversely affect cosmetic outcomes, dramatically impacting on quality of life and long-term wellbeing. Finally, SSI may delay adjuvant treatments such as radiotherapy and chemotherapy, and recent evidence from large cohort studies suggests that complications after breast surgery, may increase the risk of breast cancer recurrence. Few interventions to reduce SSI are underpinned by high quality evidence and consequently there is wide variation in practice across the UK. Methods: The National Institute for Health & Care Excellence (NIHR) Health Technology Assessment (HTA) funded ROSSINI-Platform study (NIHR163832) is a multi-speciality ‘Basket Factorial Multi Arm Multi Stage (MAMS)’ platform trial, in which full-scale RCTs are delivered in parallel within 6 different surgical specialties including breast surgery, cardiothoracics, neurosurgery, obstetrics, vascular limb (amputation) and vascular groin. It is the largest surgical trial ever funded by the NIHR and will utilise active learning and cross-cohort transfer of interventions (and/or combinations of), that suggest benefit. Selected initial interventions in breast surgery include prophylactic antibiotics (vs no antibiotics); Granudacyn® wound irrigation (vs standard of care including no wash or other types of irrigation) and; Dialkylcarbamoyl chloride (DACC)-coated dressings (Leukomed® Sorbact®) (vs standard of care including all other types of dressings/glue/steristrips and no dressings). Negative wound pressure therapy (NWPT) is not allowed in the standard of care arm as these dressings are being trialed in the vascular and cardiothoracic pillars initially. It is likely as the trial progresses that NWPT may be evaluated in higher risk breast surgery wounds. Breast surgery SSI risk will be stratified according to BMI, smoking status, diabetes, immunosuppression, and receipt of neoadjuvant chemotherapy and/or immunotherapy. Results: All adult patients undergoing breast cancer resectional surgery are eligible for recruitment with only patients undergoing primary whole breast reconstruction excluded. Implant reconstruction has a higher associated risk of infection and because of the significant consequence of implant loss, UK surgeons are not in equipoise to randomise these patients to a trial of SSI interventions. A sample size of 4280 breast surgery patients (whole trial 25,924) is required to show an absolute risk reduction of 2% from a baseline SSI rate of 5%. The primary outcome of SSI within 30 days of surgery, will be assessed at hospital discharge and/or remotely through a pre-existing Central Digital Wound Hub (CDWH), augmented by questionnaires and wound photographs collected via a patient-facing App (or via email/telephone if preferred). Secondary outcomes include health related quality of life, a cost-effectiveness analysis and other wound complications. Conclusions: The methodologically unique approach of ROSSINI-Platform employs a highly efficient trial design, to provide practice changing results and improve patient outcomes in a more time and cost-effective manner than traditional trials. The results have the potential to change and standardise SSI prevention practice both nationally and internationally. The trial will open in the UK in September 2025.
Presentation numberPS5-09-15
ERADICATE: A phase Ib/II study of elacestrant plus trastuzumab deruxtecan in patients with CDK4/6 inhibitor and endocrine-resistant HR+/HER2-low or HER2-ultralow metastatic breast cancer
Sarah L. Sammons, Dana Farber Cancer Institute, Boston, MA
S. Sammons1, S. Tolaney1, N. Lin1, L. Anderson1, E. Mayer1, N. Sinclair1, J. Stoeckle1, P. Sanz-Altamira1, N. Graham2, R. Jeselsohn1; 1Breast Oncology Center, Dana Farber Cancer Institute, Boston, MA, 2Department of Statistics, Dana Farber Cancer Institute, Boston, MA.
Background: The oral selective estrogen receptor degrader (SERD) elacestrant is FDA-approved for patients with hormone receptor-positive/HER2-negative metastatic breast cancer (HR+/HER2- MBC) with an ESR1 mutation and disease progression on first-line endocrine therapy (ET) with a CDK4/6 inhibitor. Furthermore, most patients with HR+/HER2- MBC have HER2-low or HER2-ultralow disease, which makes them eligible for treatment with trastuzumab deruxtecan (T-DXd). We recently generated preclinical data showing that the SERD fulvestrant produces synergistic activity with the chemotherapeutic agents fluorouracil and capecitabine in ESR1-mutant HR+ breast cancer cell lines.1 We also showed that in ER+/HER2- tumors, higher HER2 expression is associated with higher ER signaling.2 The phase Ib/II ERADICATE trial will evaluate the safety and efficacy of elacestrant plus T-DXd in patients with endocrine-resistant, HER2-low or HER2-ultralow HR+/HER2- MBC. Methods: Eligible patients include women or men age ≥ 18 years with HR+/HER2-low or HER2-ultralow (by local pathology review) unresectable locally advanced or metastatic breast cancer. HR+ is defined as estrogen receptor ≥ 10% (with any progesterone receptor expression). HER2-low is defined as HER2 IHC 1+ or IHC 2+ and ISH non-amplified. HER2-ultralow is defined as HER2 IHC 0 with any membranous staining on any prior primary or metastatic sample. The study population will be enriched for patients with ESR1 mutations (approximately 50%). Participants must have had disease progression on or within 12 months of adjuvant ET with a CDK4/6 inhibitor, or 1-2 lines of ET and CDK4/6 inhibitor in the MBC setting. Patients with stable or untreated, asymptomatic central nervous system (CNS) metastases are eligible. Participants must have measurable disease per RECIST 1.1 criteria and must not have received any prior chemotherapy or ADC in the metastatic setting. All participants will receive elacestrant 345 mg orally on days 1-21 and T-DXd 5.4 mg/kg IV on day 1 of each 21-day cycle. Treatment will continue until disease progression or unacceptable toxicity. The co-primary endpoints are DLT rates/determination of the RP2D (phase 1b) and objective response rate (ORR, phase II) in the overall study population. There will be a safety lead-in among the first 12 patients to determine the RP2D. Analysis of ORR will follow a Simon’s 2-stage design to distinguish between null and alternative ORRs of 50% and 65%, respectively, with 85% power at a one-sided type I error of 10%. Twenty-eight patients will be enrolled in stage 1, and if there are ≥ 15 responses, an additional 37 patients will be enrolled in stage 2. The combination will be considered worthy of further study if ≥ 38 objective responses are observed among the total 65 patients. Secondary endpoints include ORR in the ESR1-mutant population, as well as clinical benefit rate, duration of response, progression-free survival, and overall survival in both the overall study population and the ESR1-mutant population. Research biopsies will be performed at baseline (before initiation of study therapy) and at cycle 2, with an optional research biopsy at progression/end of treatment. Serial research blood samples will be collected for circulating tumor DNA (ctDNA) analyses. ERADICATE is currently pending the start of accrual. Research Sponsor: Stemline Therapeutics. References1.Grinshpun A, Russo D, Ma W, et al: Pure estrogen receptor antagonists potentiate capecitabine activity in ESR1-mutant breast cancer. NPJ Breast Cancer 10:42, 2024 2.Qiu X, Tarantino P, Li R, et al: Molecular characterization of HER2-negative breast cancers reveals a distinct patient subgroup with 17q12 deletion and heterozygous loss of ERBB2. ESMO Open 10:104111, 2025
Presentation numberPS5-09-16
A phase 1b/2 clinical investigation of invikafusp alfa (STAR0602), a first-in-class dual T-cell agonist, in combination with sacituzumab govitecan in patients with metastatic TNBC or HR+/HER2- MBC (START-002 trial)
Steven J Isakoff, Mass General Cancer Center, Boston, MA
S. Isakoff1, P. Bedard2, A. Martynova3, A. Bardia4, M. Gatti-Mays5, N. LeVasseur6, A. Varkaris7, W. Randolph8, K. Srinivasan9, S. McCue10, A. Bayliffe9, K. Liu10, Z. Su10, K. Chin10, V. Kaklamani11, K. McCann4, E. Hamilton12; 1Center for Breast Cancer, Mass General Cancer Center, Boston, MA, 2Cancer Clinical Research Unit, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 3Breast Medical Oncology, Ph1 Program, USC Norris Comprehensive Cancer Center, Los Angeles, CA, 4Breast Medical Oncology, UCLA Health Jonsson Cancer Center, Los Angeles, CA, 5Breast Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 6Breast Cancer Clinical Trials Unit, BC Cancer Vancouver Centre, Vancouver, BC, CANADA, 7Hematology Oncology, Mass General Cancer Center, Boston, MA, 8Clinical Development, Marengo Therapeutics, Cambridge, MA, 9Research & Development, Marengo Therapeutics, Boston, MA, 10Clinical Development, Marengo Therapeutics, Boston, MA, 11Breast Oncology Program, UT Health San Antonio, San Antonio, TX, 12Breast Cancer Research Program, Sarah Cannon Research Institute, Nashville, TN.
Background: Invikafusp alfa (STAR0602) is a novel, first-in-class dual T cell agonist that selectively targets subsets of T cells expressing the germline-encoded variable Vβ6 and Vβ10 variant TCRs that are enriched in tumor-infiltrating lymphocytes. The completed Ph1 dose escalation of monotherapy invikafusp alfa identified a recommended Ph 2 dose (0.08mg/kg IV Q2W) and demonstrated clinically meaningful single-agent anti-tumor activity in anti-PD(L)-1 resistant tumors, including objective responses in patients with tumor mutation burden-high (TMB-H) colorectal cancer (CRC), non small cell lung cancer, and gastroesophageal junction cancer. Invikafusp alfa promoted potent and selective expansion of mainly CD8+ Vβ6/ Vβ10 T-cells. Based on these initial clinical results, US FDA granted Fast Track Designation for invikafusp alfa in TMB-H CRC. Sacituzumab govitecan (SG), an antibody-drug conjugate (ADC), is FDA approved for patients with pre-treated unresectable locally advanced (LA) or metastatic triple-negative breast cancer (mTNBC) and HR-positive/HER2-negative breast cancer (HR+/HER2- mBC). Preclinical studies indicate that treatment with ADCs enhances tumor immunogenicity by promoting immunogenic cell death, increased antigen presentation and tumor immune infiltration. These effects are likely mediated by cytotoxic tumor cell death and subsequent release of tumor-associated antigens. Recent clinical results from the ASCENT-04 trial demonstrated improved progression-free survival in previously untreated PD-L1+ mTNBC patients who received the combination of SG and pembrolizumab, thus confirming the clinical potential of combining an ADC with an IO agent in metastatic breast cancer. START-002 explores the hypothesis that combining SG’s immunomodulatory potential with invikafusp alfa’s selective activation and expansion of tumor-reactive Vβ6/Vβ10 T-cells, will enhance anti-tumor responses and result in improved clinical outcomes in breast cancer patients. Methods:Study design: START-002 is a phase 1b/2 open label, multi-center study to determine the safety, feasibility, and anti-tumor activity of invikafusp alfa in combination with SG in patients with mTNBC or HR+/HER2- mBC. Patients will receive SG on Days 1 and 8 of a 21-day cycle and STAR0602 only on Day 8. Ph1 enrollment will start with an initial 5 patient Safety Run-in cohort at the 0.04 mg/kg dose level of STAR0602 + SG and may proceed to include 1 of 2 dose groups of STAR0602 (0.08 mg/kg or 0.02 mg/kg) + SG in order to determine the recommended dose of STAR0602 that will be used in the Ph2 cohort expansion phase. Using a Simon’s 2-stage design, Ph2 will enroll 20 participants into 2 separate expansion cohorts of patients who have either mTNBC or HR+/HER2- mBC. Major eligibility criteria: TNBC cohort: LA or mTNBC, ≥2 prior systemic therapies, at least 1 for metastatic disease. HR+/HER2- mBC cohort: LA or metastatic HR+/HER2- BC, prior endocrine based therapy and ≥1 additional systemic therapy in the metastatic setting. Prior topoisomerase 1 inhibitor therapy is excluded. Primary objective and endpoint: The primary objective of this Ph1b/2 study is to characterize the safety and tolerability of invikafusp alfa when given together with sacituzumab govitecan, and to evaluate preliminary anti-tumor activity of the combination treatment. The primary endpoints are safety and overall response rate per RECIST and iRECIST. Enrollment to the Ph1 safety run-in cohorts is ongoing. Clinical Trial Information: NCT06827613
Presentation numberPS5-09-17
Phase 1 results of a Phase I II study of Pharmacokinetics and Safety of Epidiferphane and Taxanes in Breast Cancer Patients
Coy Heldermon, University of Florida, Gainesville, FL
C. Heldermon, K. Daily, D. Ogden, P. Gurjar, D. Lyon, B. Reynolds; Medicine, University of Florida, Gainesville, FL.
Phase 1 results of a Phase I/II study of Pharmacokinetics and Safety of Epidiferphane and Taxanes in Breast Cancer Patients Pre-clinical studies in mouse models given chemotherapy regimens containing paclitaxelhave shown that Epidiferphane (EDP) delays tumor progression and reduces both neuropathy and anemia. In an Investigational New Drug Phase I/II trial investigating Epidiferphane in patients with breast cancer receiving a taxane-containing chemotherapy regimen, phase I has completed enrollment. In phase I, we evaluated the safety and pharmacokinetics of two EDP dose levels in neoadjuvant and metastatic breast cancer patients. No dose limiting toxicities were encountered and pK and adverse events are presented.In phase II, we will assess the safety and efficacy at reducing neuropathy and anemia and improving tumor response and patient quality of life in breast cancer patients of adding EDP to taxane containing chemotherapy regimens.We will be assessing the correlation of neuropathy and anemia with the level of neurofilament light chain, VEGFA, Nrf2, NF-κB and IL18. This correlation will determine if EDP is having a similar effect to that seen in the mice and if these markers may be useful in predicting and targeting toxicity.Our intervention will determine pharmacokinetics of and test a novel therapy for safety and effectiveness at improving response to chemotherapy, improve quality of life, and reducing cytopenias and peripheral neuropathy in patients. If effective, this therapy will: improve treatment regimens by making current chemotherapy regimens more effective, less toxic, and impact survival – ie allowing full dose chemotherapy without the side effects of neuropathy, anemia, and other cytopenias.
Presentation numberPS5-09-19
Flex: from genomic profiling to real-world insights in 30,000 patients with early-stage breast cancer
Linsey P Gold, Comprehensive Breast Care, Troy, MI
L. P. Gold1, L. Samiian2, K. Hoskins3, S. Diab4, L. Lee5, V. K. Gadi6, E. A. Brown7, J. R. Ramadurai8, J. A. Elayoubi9, R. E. Kaczynski10, R. E. Fine11, T. Bah12, L. Matt-Amaral13, B. J. Lieblong14, W. Audeh14, J. O’Shaughnessy15; 1Breast Surgery, Comprehensive Breast Care, Troy, MI, 2Breast Surgery, Baptist MD Anderson Cancer Center, Jacksonville, FL, 3Hematology Oncology, University of Illinois Chicago, Rockford, IL, 4Medical Oncology, Intermountain Health, Lone Tree, CO, 5Breast Surgical Oncology, Comprehensive Cancer Center, Palm Springs, CA, 6Medical Oncology, University of Illinois Cancer Center, Chicago, IL, 7Breast Oncology, Comprehensive Breast Care, Troy, MI, 8Medical Oncology, Affiliated Oncologists, Chicago Ridge, IL, 9Breast Oncology, Karmanos Cancer Institute, Detroit, MI, 10Breast Surgical Oncology, Texas Oncology Surgical Specialists – Northeast, San Antonio, TX, 11Breast Surgical Oncology, West Cancer Center, Germantown, TN, 12Medical Oncology, MetroHealth Medical Center, Cleveland, OH, 13Medical Oncology, Akron General Medical Center, Akron, OH, 14Medical Affairs, Agendia, Irvine, CA, 15Medical Oncology, Sarah Cannon Research Institute, Dallas, TX.
Title: FLEX: From Genomic Profiling to Real-World Insights in 30,000 Patients with Early-Stage Breast Cancer Background: Treatment approaches for early-stage breast cancer (EBC) have advanced significantly in recent decades, with the addition of HER2-targeted therapies, immunotherapy, and personalized chemotherapy. However, despite these advances, many questions related to treatment cannot be practically or ethically addressed in prospective, randomized clinical trials. The required extended follow-up in EBC treatment trials may delay the dissemination of practice-changing results. Additionally, many EBC trials have poor representation of less common tumor subtypes (such as invasive lobular carcinoma, ILC) and limited enrollment of patients of diverse racial/ethnic backgrounds. In this context, observational registry Real-World Data clinical trials, such as FLEX, which analyze clinical and pathologic data, treatment history, outcomes, and other metadata such as whole transcriptomics can provide actionable evidence that can inform clinical practice. The ongoing, multi-center, FLEX study (NCT03053193) seeks to enroll 30,000 EBC patients to create a large-scale, diverse, population-based registry of whole transcriptome data matched with clinical data with 10 years of follow-up to investigate new gene expression signatures of prognostic and/or predictive value in a real-world setting. Efforts are focused on enriching enrollment of diverse racial/ethnic minorities, historically underrepresented groups, and uncommon EBC tumor histologies. An additional objective is supporting investigator-initiated sub-studies to address clinically relevant questions in EBC with up to 10 years of follow-up. Methods: FLEX is a large prospective, observational trial that enrolls patients (male or female) who are ≥ 18 years old with stage I-III breast cancer. Patients who receive standard of care MammaPrint® (70-gene signature risk of recurrence), with or without BluePrint® (80-gene signature molecular subtype) on their primary breast tumor and consent to clinically annotated whole transcriptome data collection are eligible for enrollment. Within 7 years of trial initiation, FLEX has enrolled over 20,000 patients across 102 sites in the US, 3 sites in Canada, and 1 site each in Greece and Israel. Of the total FLEX population, 9495 (47%) have reached 3 years of follow-up, and 4047 (20%) have reached 5 years of follow-up. To address racial/ethnic disparities in clinical trials, a concerted effort has led to the inclusion of 1892 Black/African American, 1722 Latin American/Hispanic, and 490 Asian American/Pacific Islander EBC patients of self-reported racial/genetic ancestry, making FLEX a highly diverse study of EBC patients. Similarly, FLEX represents one of the largest cohorts of ILCs, composed of 2196 ILC and 649 mixed ILC/ductal histology tumors. Studies from FLEX have led to 3 peer-reviewed publications, with 3 submitted in 2025 currently under review. Fourteen FLEX investigator-initiated sub-study abstracts have been presented in 2025. Additionally, over 53 FLEX abstracts have been accepted at congresses internationally (2018-2025), including 11 presentations focused on therapy selection, 12 on differences in tumor biology and clinical outcomes by race/ethnicity, and 2 on ILC, among others. Data generated from FLEX include important new findings in EBC management. Among these findings are the prediction of absolute chemotherapy benefit in genomically high-risk patients, prediction of survival benefit from adjuvant anthracyclines, associations of genomic signatures that predict resistance to CDK4/6 inhibition, and overrepresentation of aggressive ER+ basal-type tumors in Black/African American patients.
Presentation numberPS5-09-20
A global Phase III, randomized, double-blind trial of BNT327 plus chemotherapy (chemo) vs placebo plus chemo in patients (pts) with previously untreated locally recurrent inoperable or metastatic, PD-L1 negative triple negative breast cancer (TNBC) (ROSETTA Breast-01)
Peter Schmid, Queen Mary University of London, London, United Kingdom
P. Schmid1, R. Dent2, P. Razavi3, P. Rietschel4, S. Günther5, L. Li6, Ö. Türeci7, U. Şahin7; 1Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 2Medical Oncology, National Cancer Center Singapore, Singapore, SINGAPORE, 3Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY, 4Global Clinical Development, BioNTech US Inc., Cambridge, MA, 5Global Medical Affairs, BioNTech SE, Mainz, GERMANY, 6Biostatistics, BioNTech US Inc., Cambridge, MA, 7Global Clinical Development, BioNTech SE, Mainz, GERMANY.
Background: TNBC, accounting for 15-25% of breast cancer cases, is an aggressive subtype with poor prognosis, and metastatic TNBC remains challenging to treat with current standard of care (SOC). PD-(L)1 inhibitors combined with chemo have improved outcomes in pts with high PD-L1 expression in the 1L setting, but outcomes remain poor in pts with PD-L1 negative tumors. Novel therapies are needed to provide effective treatment with long-lasting clinical benefits for locally recurrent or metastatic TNBC, particularly for pts ineligible for anti-PD-(L)1 treatment. BNT327 is an investigational bispecific antibody, targeting PD-L1 and VEGF-A. Dual targeting of PD-L1 and VEGF-A combines two complementary modalities, aiming to improve outcomes. PD-L1 is often upregulated in tumor cells and tumor-infiltrating immune cells. By simultaneously binding to PD-L1 and VEGF-A, BNT327 is hypothesized to co-localize immunostimulatory and anti-angiogenic activities to the tumor microenvironment, reversing the negative impact of VEGF signaling on immune cell infiltration and activation, and normalizing tumor vasculature, leading to tumor growth inhibition. A Phase II trial (NCT05918133) of BNT327 plus nab-paclitaxel conducted in China in 1L locally advanced/metastatic (la/m) TNBC showed encouraging efficacy across PD-L1 levels and a manageable safety profile (Wu et al. 2023, Wu et al. 2024). Trial design: ROSETTA Breast-01 is a global, randomized, double-blind Phase III trial evaluating the safety and efficacy of BNT327 plus chemo in pts with PD-L1 negative, previously untreated, locally recurrent inoperable or metastatic TNBC. Pts are stratified by prior treatment with cancer immunotherapy in the neoadjuvant/adjuvant setting, on-trial chemo regimen, and geography, and randomized 1:1 to receive a combination treatment with BNT327 or placebo plus physician´s choice chemo (paclitaxel/nab-paclitaxel, gemcitabine plus carboplatin, or eribulin). Chemo is administered per SOC. Pts will receive treatment until disease progression, intolerable toxicity, death, withdrawal, or trial termination. Eligibility criteria: Eligible pts are aged ≥18 yrs, with locally recurrent inoperable or metastatic PD-L1 negative TNBC in the 1L setting, adequate hematologic and organ function, and ECOG PS of 0 or 1. Endpoints: The primary endpoints are overall survival (OS) and progression-free survival (PFS) by blinded independent central review (BICR) per RECIST v1.1. Secondary endpoints include PFS by investigator, objective response rate, duration of response, disease control rate, OS and PFS rates, occurrence of treatment emergent adverse events (TEAEs), dose interruption, reduction, and discontinuation due to TEAEs, and changes in pt-reported outcomes. Statistical methods: The primary endpoints, PFS per BICR and OS, will be compared between treatment arms using a stratified log-rank test. The hazard ratio for PFS and OS will be estimated via a Cox proportional hazards model, adjusted for the randomization stratification factors. Accrual: The trial aims to randomize ~558 pts across sites in North and South America, Europe, Asia, and Australia. Contact information: For more information or to refer pts, please contact: patients@biontech.com. Acknowledgment: The trial is sponsored by BioNTech. BNT327 is being jointly developed by BioNTech and Bristol Myers Squibb. Writing assistance was provided by Kordula Heinen, of BioNTech SE.
Presentation numberPS5-09-21
Feasibility of Clinical Nurse-Initiated Geriatric Assessments in Breast Medical Oncology Clinic: A Pilot Program
Rima Patel, Icahn School of Medicine at Mount Sinai, New York, NY
R. Patel1, T. Hills2, H. Nunez3, A. Farhadi1, A. Bhardwaj1, J. Fasano1, N. Hall1, K. Facey1, S. Lee1, R. A. Freedman4, F. Ko2; 1Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 2Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 3Hematology/Oncology, Hackensack University Medical Center, Hackensack, NJ, 4Hematology/Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: The incidence of cancer in older adults is substantially rising and oncologists must be prepared to care for this growing vulnerable population. Geriatric assessment (GA)-guided management (GAM) reduces treatment toxicity and improves quality of life in older patients with cancer, leading to its inclusion in national guidelines. However, GA adoption by oncologists remains low, especially in racial and ethnic minority populations. We conducted a pilot program implementing clinical nurse-administered GAs in older adults with breast cancer, with the goal of improving GA completion, particularly among Spanish-speaking patients by using Spanish-translated GAs.Methods: The study was conducted over a 16-week period in 2025 in three breast medical oncology clinics at an academic health system in New York City. Eligible patients were ≥ 65 years, spoke English or Spanish, and had a new or prior breast cancer diagnosis. Medical oncologists in the study received education on the benefits of GAM in older adults with cancer and tools to facilitate GAM in clinic. In the first 8 weeks (pre-intervention), we captured baseline GA completion rates by oncologists and in the second 8 weeks (post-intervention), a clinical nurse intervention was implemented to administer the GAs. A clinical workflow was adapted whereby eligible patients were offered the G8 Geriatric Screening Tool by a medical assistant and a score of ≤ 14 would trigger ASCO’s comprehensive Practical Geriatric Assessment (PGA). The self-reported section of the PGA (in English or Spanish) was completed by the patient, and the provider section was administered by the oncologist (pre-intervention) or nurse (post-intervention). The results of the GAs were reviewed with the patient, and referrals and treatment recommendations, as applicable, discussed by oncologist (pre-intervention) or clinical nurse (post-intervention). Here, we describe demographics for the overall population and baseline GA completion rates, geriatric vulnerabilities identified, and associated referrals made during the pre-intervention phase. Feasibility of the pre-intervention approach was measured by the number of vulnerabilities identified and referrals triggered.Results: As of June 2025, 106 patients have been included, 56% (N=59) pre-intervention and 44% (N=47) post-intervention. The median age of the overall population was 73 years (range 65-94), 42% (N=45) were White, 25% (N=27) Black, 18% (N=19) Hispanic, 8% (N=8) Asian and remaining 7% (N=7) other/unknown. Eleven percent (N=12) were Spanish speaking. Pre-intervention completion rate for the screening G8 tool was 78% (N=46). Based on the results of the G8, 48% (N=22) of patients were recommended the comprehensive PGA, of whom 73% (N=16) completed it. Geriatric vulnerabilities were identified in 12 patients (75%), with an average of 4.8 vulnerabilities per patient assessed, including poor physical function (N=12), low performance status (N=10), high multimorbidity (N=10), and low social support (N=9). Thirteen referrals were made, most frequently to physical and occupational therapy and social work.Conclusions: To our knowledge, this is the first study designed to evaluate feasibility of a clinical nurse intervention to improve GA completion in breast oncology patients, and the first to utilize a Spanish translation of ASCO’s PGA to facilitate delivery in racial/ethnic minority populations. The high baseline GA completion rates suggest that screening and assessment of geriatric vulnerabilities in high-risk oncology patients are feasible when they are effectively integrated into existing clinical workflow and have garnered staff and provider buy-in. Results evaluating the impact of nurse-initiated GAs on changes in G8 and PGA completion, as well as feasibility of the nurse intervention, are forthcoming.
Presentation numberPS5-09-22
Phase 1b study of EZH1/2 inhibitor valemetostat in combination with trastuzumab deruxtecan in subjects with HER2 low/ultra-low/null metastatic breast cancer
Senthil Damodaran, MD Anderson Cancer Center, Houston, TX
S. Damodaran1, A. N. Marx1, T. Iwase2, Y. Yuan3, M. Kai1, J. Lee4, C. H. Barcenas1, N. T. Ueno2; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Translational Clinical Research Program, University of Hawaii Cancer Center, Honolulu, HI, 3Biostatistics, MD Anderson Cancer Center, Houston, TX, 4Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI.
Background: Low HER2-expressing breast cancers have traditionally been classified as HER2-negative and treated as TNBC or hormone receptor (HR)-positive. Trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate composed of an anti-HER2 antibody conjugated to topoisomerase I payload, is approved for clinical use in HER2-low (IHC 1+ or IHC 2+/ ISH negative) metastatic breast cancer (MBC). In the DAISY study (NCT04132960), meaningful clinical response was observed with T-DXd in HER2 IHC 0 MBC. Valemetostat is an oral, selective dual inhibitor of enhancer of zeste homolog 1 and 2 (EZH1/2, methyltransferases that specifically methylate histone H3 lysine 27. It is approved for patients with relapsed or refractory adult T-cell leukemia/lymphoma in Japan. EZH2-mediated PP2A inactivation has been shown to confer resistance to HER2-targeted therapy. Additionally, valemetostat has been shown to upregulate Schlafen11 (SLFN11), a putative DNA/RNA helicase that regulates the sensitivity to DNA-damaging agents such as topoisomerase I inhibitors. Accordingly, this study examines the safety and anti-tumor activity of valemetostat in combination with T-DXd in subjects with HER2 low/ultra-low/null MBC.Trial Design: This is a single-arm, phase-1b study to evaluate the safety and clinical activity of T-DXd in combination with valemetostat in patients with HER2 low/ultra-low/null MBC. The dosing for T-DXd is 5.4 mg/kg Q3W administered intravenously as indicated for current clinical use. Valemetostat will be evaluated in up to five doses (50mg, 75mg, 100 mg, 150 mg, and 200 mg) with a starting dose (level 1) at 100 mg QD. The dose-limiting toxicity (DLT) evaluation period will be the first 2 treatment cycles (42 days). Eligibility criteria: Pathologically confirmed HER2 low/ultra-low/null breast cancer• ECOG performance status ≤1• Measurable disease (for expansion)• Received at least one line of chemo in the metastatic setting• Progressed and would no longer benefit from endocrine therapy (HR-positive).• Normal organ and marrow function• Exclusion: symptomatic brain metastases, interstitial lung disease, cord compression, prior treatment with any anti-HER2 therapy. Specific aims: To evaluate the safety and determine the maximum tolerated dose (MTD)/recommended dose for expansion (RDE) of valemetostat in combination with T-DXd.• To evaluate the objective response rate (ORR) of valemetostat at the RDE in combination with T-DXd• To determine the duration of response, clinical benefit rate, progression-free survival, and overall survival of valemetostat at the RDE in combination with T-DXd.• To evaluate the pharmacokinetics and pharmacodynamic markers of valemetostat and T-DXd combination• To evaluate the immunogenicity of T-DXd when co-administered with valemetostat. Statistical methods: Approximately 15 evaluable patients will be enrolled for the dose-escalation portion based on the Bayesian optimal interval design with a target DLT rate of 25%. Patients enrolled and treated in cohorts of 3. The expansion will be performed at the RDE using the 2-stage Bayesian optimal dose-expansion design. In the first stage, 13 evaluable patients (including those treated at the RDE in the dose-escalation part) will be enrolled. If < 5 patients respond in the first stage, the study will be stopped for futility. If ≥ 5 responses are observed, 13 additional evaluable patients will be enrolled. If 11 or more responses are observed among 26 patients, the treatment will be considered promising. This two-stage design yields 78% power under the alternative hypothesis of ORR=50% (null ORR = 30%) while controlling the one-sided type I error at 10%.
Presentation numberPS5-09-23
A Phase 2 study of zelenectide pevedotin, a Bicycle Drug Conjugate, in patients with NECTIN4 amplified advanced breast cancer (Duravelo-3)
Peter A Kaufman, University of Vermont Cancer Center, Burlington, VT
P. A. Kaufman1, P. Aftimos2, J. Cortes3, G. Curigliano4, F. Dalenc5, M. Danso6, K. Jhaveri7, M. Rimawi8, C. Saura9, P. Savas10, P. Schmid11, J. Ricker12, K. Josephs12, C. Xu13, E. Hamilton14; 1Department of Medicine, University of Vermont Cancer Center, Burlington, VT, 2Clinical Trials Conduct Unit, Institut Jules Bordet-Université Libre de Bruxelles, Brussels, BELGIUM, 3Department of Medicine, Universidad Europea de Madrid, Madrid, SPAIN, 4European Institute of Oncology, IRCCS and University of Milan, Milan, ITALY, 5Department of Medical Oncology, Oncopole Claudius Regaud, Institut Universitaire du Cancer, Toulouse, FRANCE, 6Department of Medical Oncology, Virginia Oncology Associates, Norfolk, VA, 7Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, 8Department of Medicine, Lester and Sue Smith Breast Center and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 9Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, SPAIN, 10Department of Medical Oncology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, AUSTRALIA, 11Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 12Clinical Development, Bicycle Therapeutics, Inc., Cambridge, MA, 13Biostatistics, Bicycle Therapeutics, Inc., Cambridge, MA, 14Medical Oncology, Sarah Cannon Research Institute (SCRI), Nashville, TN.
Background: Zelenectide pevedotin (zele; formerly BT8009) is a Bicycle® Drug Conjugate (BDC®) comprised of a highly selective bicyclic peptide targeting Nectin-4 linked to monomethyl auristatin E (MMAE) via a cleavable linker. With low molecular weight and a short plasma half-life, zele has the potential to rapidly penetrate solid tumours with minimal exposure to healthy tissue. The ongoing Phase 1/2 Duravelo-1 trial (NCT04561362; BT8009-100) has shown preliminary antitumour activity and a manageable safety profile of zele monotherapy in patients (pts) with breast cancer (BC). This open-label, multicentre, Phase 2 Duravelo-3 study (NCT06840483; BT8009-201) aims to further evaluate the efficacy and safety of zele monotherapy in pts with recurrent, unresectable, or metastatic NECTIN4 amplified (amp) BC. Trial design: The trial will enrol pts into 2 cohorts: pts with hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)– BC who have received ≤3 prior lines of nonendocrine-based therapy in the metastatic setting (Cohort A; n≈33) and pts with triple-negative BC who have received 1–3 prior lines of systemic therapy in the metastatic setting (Cohort B; n≈33). NECTIN4 amp is defined as a NECTIN4:CEN1 ratio ≥2.0 or NECTIN4 copy number ≥6, per fluorescence in situ hybridization. All pts must have ECOG performance status 0–1 and adequate organ function. Pts previously treated with MMAE will be excluded. All pts will receive zele 6 mg/m2 IV on Days 1 and 8 of a 21-day cycle. The primary endpoint is objective response rate. Secondary endpoints are duration of response, clinical benefit rate, disease control rate, progression-free survival, overall survival, time to progression, and incidence of adverse events. Efficacy endpoints will be investigator-assessed per RECIST v1.1. Exploratory endpoints will include assessment of baseline tumour biomarkers, including Nectin-4 expression and surrogates of NECTIN4 amp, and peripheral blood biomarkers such as circulating Nectin-4 protein and tumour DNA. Pharmacokinetics, correlation of efficacy and safety with biomarkers of NECTIN4 amp, and measurement of antidrug antibody are additional exploratory endpoints. This study is actively recruiting.
Presentation numberPS5-09-25
Claire: a multicenter, prospective single-arm phase II study, evaluating liquid biopsy guided intensified follow-up surveillance in patients with intermediate to high-risk ER+/HER2-negative early-stage breast cancer
Mitchell J Elliott, Princess Margaret Cancer Centre, Toronto, ON, Canada
M. J. Elliott1, E. Amir1, M. Nadler1, M. Li1, C. Yu2, M. Audoin3, G. Putcha4, W. Levin5, S. Birkeäl5, K. Howarth5, P. L. Bedard1, L. L. Siu1, H. K. Berman1, C. Townsley6, M. Wu6, 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, 3Patient Advocate, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 4SAGA Dx, SAGA Dx, Morrisville, NC, 5SAGA Dx, SAGA Dx, Toronto, NC, 6Department of Family and Community Medicine, Womens College Hospital, Toronto, ON, CANADA.
Background: Estrogen receptor-positive, HER2-negative (ER+/HER2-) breast cancer is the most common subtype, accounting for the majority of diagnoses. Despite favorable overall survival rates, it is responsible for over sixty percent of breast cancer-specific deaths. At present, no imaging or laboratory surveillance is recommended to detect metastatic recurrence in asymptomatic patients with early-stage breast cancer (EBC). Circulating tumor DNA (ctDNA), measured using fit-for-purpose, highly sensitive and specific assays anticipates clinical relapse with high positive predictive value and long lead times, creating the opportunity for targeted secondary adjuvant interventions aimed at reducing mortality in this setting. Eligibility Criteria: Circulating Liquid biomarkers for Assessment and Identification of Recurrence in Early breast cancer (CLAIRE) is a multicenter, single-arm phase II study enrolling adults (≥18 years) with resected stage I-III ER+/HER2- EBC who completed curative-intent therapy [(neo)adjuvant chemotherapy, surgery, radiotherapy] within five years, or between 5-10 years of enrolment with a high CTS5 score. Additional eligibility criteria include no clinical evidence of recurrence, completion of ≥24 months of endocrine therapy (and ≥12 months of CDK4/6 inhibitor, if indicated), and adequate organ function. Methods: Clinical follow up will be performed per the standard of care by the primary general practitioner or medical oncologist. Participants will receive ctDNA testing (Pathlight, SAGA Dx) every three months for up to five years alongside standard clinical surveillance. Patients with ctDNA positivity will undergo reflex restaging imaging to exclude metastatic disease. Participants with ctDNA identified in the absence of radiographic relapse will be eligible for accompanying secondary adjuvant interventional trials. Aims: The two primary objectives of CLAIRE are to determine, among ctDNA-positive participants, the incidence of imaging-negative molecular relapse and, among ctDNA-negative participants, the cumulative rate of local or distant recurrence as a cumulative incidence measure. Secondary objectives include the technical and patient-centered performance of the surveillance program (assay success rate, adherence to testing, patient-reported outcomes, and health-care utilization), as well as key clinical and outcomes: distant recurrence-free survival (DRFS), overall survival (OS), and longitudinal quality-of-life (QoL) assessments. Conclusion: CLAIRE will prospectively evaluate serial ctDNA surveillance using a next-generation ultrasensitive ctDNA assay in ER+/HER2- EBC, and identify patients with MRD for interventional trials. The integration of ctDNA-guided surveillance with standard follow-up has the potential to enable targeted therapeutic interventions for those at imminent risk. Ultimately, CLAIRE seeks to demonstrate that a liquid biopsy surveillance strategy, using this ultrasensitive ctDNA assay, enables molecular recurrence detection, informing confirmatory phase III secondary-adjuvant therapy trial design. Clinical Trial Information: NCT05196087
Presentation numberPS5-09-26
Digital health adherence companion study for tetrathiomolybdate (TM) plus capecitabine in triple-negative breast cancer
Arvind Suresh, University of California San Francisco, San Francisco, CA
A. Suresh1, N. Kornhauser2, R. M. Lavoie2, L. J. Sinks2, C. A. Seymour2, M. V. Cobham2, A. E. Gaughan-Maher2, N. Chan3, L. T. Vahdat2; 1Department of Medicine, University of California San Francisco, San Francisco, CA, 2Section of Hematology/Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH, 3Perlmutter Cancer Center, NYU Langone Health, New York, NY.
Background: Copper depletion therapy with oral tetrathiomolybdate (TM) is designed to complement standard adjuvant therapy for high risk for relapse triple negative breast cancer (TNBC) by overcoming TNBC resistance mechanisms. The dosing of TM requires an induction and maintenance phase with subsequent monitoring of ceruloplasmin and absolute neutrophil counts (ANC). Several studies have reported on adherence rates for oral anticancer medications, with prior breast cancer trials having adherence rates of 78% across all cycles. Diaries and paper logs are subject to manipulation and errors for medications where dosing schedules are frequently modified, while microelectric event monitoring systems are not feasible in real-word practice beyond clinical trials. This trial in progress aims to evaluate whether a digital mobile application can improve adherence to TM for patients with high risk for relapse TNBC. Methods: The mobile application (TM Dosing Logger) is being evaluated as part of an open-label, non-randomized, dose escalation (phase 1b) and randomized phase 2 study in patients with high risk for relapse TNBC after completion of neoadjuvant chemo-immunotherapy and surgery. Patients will receive TM with capecitabine and pembrolizumab in 20 mg capsule increments, with daily doses up to 180 mg during the induction phase (first 6 months) and 100 mg during the maintenance phase (subsequent 30 months) in 28-day cycles. Doses are divided into capsules taken between one and three times daily. The mobile app will be offered to all patients taking TM utilizing an opt out function. Patients without a smartphone or tablet will receive paper logs. The mobile app contains three parts: a study calendar to view a real-time dosing schedule, surveys to log each dose taken, and reminder notifications to improve adherence. The dosing schedule is adjusted remotely in REDCap based on patient-specific ceruloplasmin levels and ANC and syncs with the mobile app. Real-time dose adjustments can also be made. Study participants may select notification times that match when they plan to take each dose, and study coordinators will be notified if a patient misses three consecutive doses. Surveys are locked after each calendar day to prevent data manipulation or incorrect data entry. Mobile app patient enrollment began in December 2024 as part of the Phase 1b study. Usability of the mobile app for daily use and patient satisfaction will be assessed with the mHealth App Usability Questionnaire at two and four weeks after study start date. Adherence will be measured as number of doses logged within 24 hours of scheduled time for each cycle and across entire three-year study period. Adherence rates will be compared with patients using paper logs and prior reported data using linear mixed models. A multivariate regression analysis will be used to evaluate predictors of adherence. Clinical Trial Information: NCT06134375
Presentation numberPS5-09-27
A phase i/ii single-arm trial of azenosertib (zn-c3) combined with carboplatin and pembrolizumab in patients with metastatic triple-negative breast cancer (zap-it)
Filipa Lynce, Dana-Farber Cancer Institute, Boston, MA
F. Lynce1, N. Tayob1, N. Chen2, A. Grinshpun3, R. Davis1, M. K. DeMeo1, A. G. Waks1, R. Nanda2, J. L. Guerriero4, G. Shapiro1, S. M. Tolaney1; 1Breast Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Hematology Oncology, The University of Chicago, Chicago, IL, 3Medical Oncology, Hebrew University-Hadassah Medical School, Jerusalem, ISRAEL, 4Surgery Department, Brigham and Women’s Hospital, Boston, MA.
Background: Despite recent advances in breast cancer treatment, metastatic triple negative breast cancer (mTNBC) remains incurable. WEE1 is a tyrosine kinase that inhibits cyclin dependent kinase 1, thus arresting the cell cycle at the G2 checkpoint and providing an opportunity for DNA damage repair. WEE1 is highly expressed in several cancer types, including breast cancer. A phase II study assessing the efficacy of the WEE1 inhibitor adavosertib with cisplatin in 34 patients with mTNBC treated with 0-1 prior lines of chemotherapy showed an ORR of 26% (Keenan. CCR 2021). Although the combination missed the pre-specified ORR cutoff of > 30%, tumors from patients with clinical benefit demonstrated enriched immune gene expression and T-cell infiltration. Azenosertib is a novel, selective, and orally bioavailable small-molecular inhibitor of the WEE1 tyrosine kinase, with strong evidence of in vitro and in vivo activity in different solid tumor types. Most common side effects (≥G3) of ZN-c3 intermittent dosing with carboplatin AUC 5 were thrombocytopenia (36%), anemia (29%), neutropenia (7%), fatigue (7%), no nausea, vomiting or diarrhea (Liu et al. ASCO 2023). The ZAP-IT trial is evaluating the safety and efficacy of azenosertib with carboplatin and pembrolizumab in patients with mTNBC. Methods: This is an ongoing single-arm, open-label, phase Ib/II study designed to evaluate the safety and efficacy of azenosertib in combination with carboplatin and pembrolizumab in patients with mTNBC, defined as ER < 10% or PR < 10%, and HER2- negative by IHC and/or FISH; patients are eligible irrespective of tumor PD-L1 status. Patients must have received 1-3 prior systemic therapies in the advanced setting, have not had disease progression on prior platinum, and must have received at least one antibody drug conjugate. In the phase I portion of this study, patients will receive a predetermined dose of carboplatin on days 1 and 8 of every 21 days, a predetermined dose of pembrolizumab once every 21 days and a predetermined starting dose level of azenosertib 5 days on 2 days off. Paired research biopsies will be required at baseline and at Cycle 2 of therapy in phase 2 patients with an optional end of treatment biopsy. The primary objective of the phase Ib is to determine the maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D) of azenosertib when used in combination with carboplatin and pembrolizumab in patients with TNBC. The primary endpoint of the phase II is to assess the efficacy, as defined by objective response rate (ORR) per RECIST 1.1 criteria, of the combination of azenosertib, carboplatin, and pembrolizumab in patients with mTNBC. A maximum of 78 patients will be enrolled. To our knowledge this is the first and only ongoing study testing the combination of a WEE1 inhibitor with carboplatin and a checkpoint inhibitor for the treatment of breast cancer.
Presentation numberPS5-09-28
Capecitabine for Targeted Eradication of aRising ctDNA Molecular Residual Disease in ER-positive/HER2-negative Early-stage Breast Cancer
Mitchell J Elliott, Princess Margaret Cancer Centre, Toronto, ON, Canada
M. J. Elliott1, E. Amir1, M. B. Nadler1, M. Li1, C. Yu2, M. Audoin3, G. Putcha4, W. Levin4, S. Birkeäl4, K. Howarth4, P. Bedard1, L. L. Siu1, H. Berman1, C. Townsley5, M. Wu5, 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, 3Patient Advocate, Princess Margaret Cancer Centre, Toronto, ON, CANADA, 4SAGA Dx, SAGA Dx, Morrisville, NC, 5Department of Family and Community Medicine, Womens College Hospital, Toronto, ON, CANADA.
Background: Despite advances in early detection and adjuvant therapy, metastatic recurrences of ER-positive/HER2-negative (ER+/HER2-) breast cancer account for over sixty percent of breast cancer-specific deaths. Circulating tumor DNA (ctDNA) has emerged as a highly sensitive and specific biomarker for identifying individuals at specific risk of metastatic recurrence. ctDNA detection (molecular residual disease, MRD) using a tumor-informed structural variant assay can anticipate clinical relapse with a high positive predictive value and long lead time. Capecitabine has clinical activity in ER+/HER2- disease and, when administered metronomically, exerts anti-angiogenic and immunomodulatory effects with a favorable safety profile. This trial will evaluate whether secondary adjuvant metronomic capecitabine can eradicate ctDNA in early-stage ER+/HER2- breast cancer as a surrogate for recurrence prevention. Eligibility Criteria: Adult patients (≥18 years) with resected stage I-III ER+/HER2- breast cancer with ctDNA+ MRD (Pathlight, SAGA Dx) identified through the in Circulating Liquid biomarkers for Assessment and Identification of Recurrence in Early breast cancer (CLAIRE) study (NCT05196087), ECOG PS 0-1, completion of ≥24 months of endocrine therapy (and ≥12 months of CDK4/6 inhibitor, if indicated), adequate organ function and no clinical or radiographic evidence of recurrence will be enrolled in this single arm interventional trial. Methods: This is an open-label, non-randomized Simon two-stage Phase II study enrolling up to 13 evaluable MRD+ patients (up to 15 enrolled to account for dropouts). The primary endpoint is ctDNA clearance at week 16 (ctDNAwk16=0) as measured by the Pathlight assay. In stage I, eight participants receive metronomic capecitabine 500 mg orally TID for up to 12 months; if ≥2 of 8 have ctDNA clearance at week 16, stage II will enroll five additional patients. Secondary endpoints include distant recurrence-free survival (DRFS), invasive disease-free survival (iDFS), overall survival (OS), and toxicity per CTCAE v5.0. Serial plasma for ctDNA and biobanking will occur with each cycle. Radiographic imaging will be performed every 4 months in year 1 and every 6 months thereafter. Follow-up for survival and ctDNA continues for up to 5 years. The Simon design tests P₀ = 0.10 versus P₁ = 0.40 with α = 0.05 and 80% power; interim analysis follows the first eight patients, with continuous accrual between stages. Aims: The primary aim is to evaluate the rate of ctDNA clearance 16 weeks following treatment initiation. Secondary aims are to describe clinical outcomes (DRFS, iDFS, and OS) and to assess the safety and tolerability of metronomic capecitabine in this setting. Exploratory aims include evaluation of ctDNA dynamics and analysis of tissue and plasma genomic features associated with ctDNA persistence or clearance. Conclusion: This proof-of-concept trial will determine whether ctDNA-guided secondary adjuvant therapy with metronomic capecitabine can result in ctDNA clearance in patients with ER+/HER2- breast cancer MRD, potentially preventing metastatic relapse. Findings will inform larger MRD-directed intervention studies aimed at transforming postoperative management, developing secondary adjuvant therapy strategies, and improving long-term outcomes. Clinical Trial Information: NCT05196087
Presentation numberPS5-09-29
Radiant study: Phase 1b study of pre-op radiation with abemaciclib and letrozole in early-stage hormone-receptor positive breast cancer
Mridula George, Rutgers Cancer Institute, New Brunswick, NJ
M. George1, S. Jabbour2, N. Ohri2, C. Omene1, M. Kowzun3, S. Kumar3, D. Toppmeyer1, B. Haffty2; 1Medicine, Rutgers Cancer Institute, New Brunswick, NJ, 2Radiation Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 3Surgery, Rutgers Cancer Institute, New Brunswick, NJ.
Background: CDK4/6 inhibitors (CDK4/6i) have changed the treatment landscape for patients with metastatic hormone receptor positive (HR+)/HER2- breast cancer. Multiple studies have evaluated the role of neoadjuvant CDK4/6 inhibitors. NEOPAL was a phase II study, which compared neoadjuvant chemotherapy to neoadjuvant endocrine therapy (NET) and CDK4/6i, which showed similar clinical response and breast conserving surgery rates but with more favorable toxicity profile with the combination NET and CDK4/6i. There is increasing pre-clinical data showing synergism between CDK 4/6i and radiation therapy (RT) secondary to radiosensitizing estrogen receptor (ER) positive cells leading to decrease in the surviving fraction of cells. Retrospective studies that have looked at the combination of CDK4/6i with RT in patients with metastatic breast cancer have shown that the combination is well tolerated. The advantages of preoperative RT include accurate tumor site identification, better target volume delineation and tumor downstaging with increased rates of breast conserving surgery. Preoperative RT can overcome treatment planning challenges seen in patients who have had reconstructive surgery such as tissue expander, implant and flap irradiation issues. It is relevant to understand the benefit of concurrent radiation and CDK4/6i in the pre-operative setting in select patient population. RADIANT study is a single-arm, phase 1b study that will evaluate the concurrent use of abemaciclib and letrozole with pre-operative RT in HR+/HER2 negative early-stage breast cancer. METHODS: The study will recruit 15 patients diagnosed with HR+/HER2- breast cancer to evaluate combination of endocrine therapy, CDK4/6i (abemaciclib) and preoperative radiation. (Clinicaltrials.gov Identifier: NCT06139107) The study hypothesize that the combination will be safe and tolerable. The primary objective of the study is to evaluate safety and tolerability of the combination of abemaciclib and radiation as assessed by adverse events (AEs). Secondary objective is to determine the clinical efficacy of the study treatment regimen as determined by Residual Cancer Burden (RCB). Patients with cT1c-T2N0 HR+/HER2- tumors with low-risk or intermediate risk Oncotype DX Breast Recurrence score (≤25) will be eligible for the study. The study is divided into 4 parts- PART A (pre-radiation), PART B (Radiation), PART C (post-radiation) and PART D (breast surgery).All cycles will be based on abemaciclib and will be 28 days. PART A: Patient will take letrozole 2.5mg daily and Abemaciclib 150mg twice a day for 3 cycles . Towards the end of PART A, patients will have on-treatment biopsy. PART B: Patients will then receive breast radiation concurrently with abemaciclib and letrozole. While getting concurrent radiation (Part B), we will use a streamlined Bayesian Optimal Interval (BOIN) design. Enrollment will be in cohorts of 3 patients to 2 dose levels of abemaciclib — 100mg BID and 50mg BID. Fifteen patients will be enrolled to part B of this trial based on streamlined BOIN design. The BOIN procedure targets a toxicity rate of 0.33, and we will enroll patients in cohorts of 3 patients. We will use a posterior probability for overdose control of 0.85.A dose limiting toxicity (DLT) is defined as any grade 4 skin, hematologic (white blood cells only) or grade 2 or greater pneumonitis/Interstitial lung disease resulting from Part B of the study. If no DLTs occur in at 100mg dose during PART B, the study will not enroll to 50mg dose. PART C: After the completion of radiation, patients will continue on to PART C for 2 cycles of abemaciclib and letrozole. PART D: Patients will have breast surgery in Part D along with sentinel lymph node biopsy following completion of systemic therapy. The study is currently enrolling patients at Rutgers Cancer Institute.
Presentation numberPS5-09-30
A phase 1/2 trial evaluating the safety, tolerability, and efficacy of the KAT6 inhibitor, PF-07248144, in combination with vepdegestrant in patients with ER+/HER2− locally advanced or metastatic breast cancer
Fengting Yan, Swedish Cancer Institute, First Hill-True Family Women’s Cancer Center, Seattle, WA
F. Yan1, T. Mukohara2, Y. Park3, S. Im4, G. Kim5, T. Yamashita6, D. Sommerhalder7, R. Layman8, E. Hamilton9, S. Kim10, F. Hara11, S. Wang12, J. Chien13, J. Kim14, T. Clay15, R. Joshi16, Y. Chae17, C. Oakman18, B. Holbrook19, B. Dong20, Y. Cheng21, H. Li22, J. Yang23, S. Kent24, L. Liu25, K. Kowalski26, S. Badhrinarayanan27, A. Skoura28, L. Howie29, P. Lorusso30; 1Breast Medical Oncology, Swedish Cancer Institute, First Hill-True Family Women’s Cancer Center, Seattle, WA, 2Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, JAPAN, 3Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF, 4Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, KOREA, REPUBLIC OF, 5Division of Medical Oncology and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 6Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Yokohama, JAPAN, 7Hematology/Oncology, NEXT Oncology, San Antonio, TX, 8Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 9Director of Breast Cancer Program, Sarah Cannon Research Institute, Nashville, TN, 10Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, KOREA, REPUBLIC OF, 11Department of Breast Oncology, Aichi Cancer Center, Nagoya, JAPAN, 12Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 13Breast Medical Oncology, University of California San Francisco, San Francisco, CA, 14Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 15Department of Oncology, Saint John of God Subiaco Hospital, Perth, Western Australia, AUSTRALIA, 16Medical Oncology, Cancer Research SA, Adelaide, AUSTRALIA, 17Department of Hemato-oncology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 18Department of Cancer, Western Health, Sunshine Hospital, St. Albans, AUSTRALIA, 19Hematology Oncology, St. Elizabeth Healthcare, Edgewood, KY, 20Department of Medicine, University of Louisville Hospital James Brown Cancer Center, Louisville, KY, 21Department of Oncology, Jilin Province Tumor Hospital, Changchun, CHINA, 22Department of Breast Medical Oncology, Beijing Cancer Hospital, Beijing, CHINA, 23Department of Oncology, The First Affiliated Hospital of Xi`an Jiaotong University, Shaanxi, CHINA, 24Biostatistics, Pfizer Inc., Cambridge, MA, 25Biostatistics, Pfizer Inc., San Diego, CA, 26Oncology Late Stage Development, Pfizer Inc., San Diego, CA, 27Oncology Late Stage Development, Pfizer Inc., Bothell, WA, 28Oncology Early Stage Development, Pfizer Inc., Collegeville, PA, 29Division of Oncology, Pfizer Inc., San Diego, CA, 30Department of Medical Oncology and Hematology, Yale School of Medicine, New Haven, CT.
Background: Cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) in combination with endocrine therapy are standard for the first-line (1L) treatment of estrogen receptor-positive/human epidermal growth factor receptor 2-negative (ER+/HER2−) advanced/metastatic breast cancer (ABC). Although CDK4/6 inhibitors significantly prolong progression-free survival (PFS) in patients with ER+/HER2− ABC, resistance ultimately occurs. For patients who develop resistance to CDK4/6i-based therapy, there is no universal standard of care, highlighting a key unmet need for this patient population. PF-07248144 is a novel, oral, selective inhibitor of KAT6A and KAT6B, two histone lysine acetyltransferases that regulate lineage-specific gene transcription via H3K23 acetylation. Vepdegestrant (ARV-471/PF-07850327) is an oral PROteolysis TArgeting Chimera (PROTAC) ER degrader that has exhibited clinical activity as monotherapy in heavily pretreated patients with ER+/HER2− ABC. Here, we describe a phase 1/2 study that aims to evaluate the safety, tolerability, and efficacy of the KAT6 inhibitor, PF-07248144, in combination with vepdegestrant in patients with ER+/HER2− ABC (NCT04606446). Methods: This is an open-label, multicenter, phase 1/2 study of PF-07248144 as monotherapy or in combination for patients with advanced or metastatic solid tumors, including breast cancer. The study is comprised of two parts: dose escalation (Part 1) and dose expansion (Part 2). Parts 1E and 2E are focused herein and are evaluating PF-07248144 plus vepdegestrant in patients with ER+/HER2− ABC. Patients are eligible to participate in Parts 1E and 2E if they are ≥18 years of age (or per local regulatory requirements) and have histologically or cytologically confirmed ER+/HER2− ABC. Other eligibility criteria include disease progression after ≥1 prior line of treatment with CDK4/6i and endocrine therapy. Patients may have received fulvestrant. Other inclusion criteria are measurable disease as defined by Response Evaluation Criteria in Solid Tumor version 1.1, adequate organ function, asymptomatic/stable brain metastasis and Eastern Cooperative Oncology Group performance status of 0 or 1. Females must meet postmenopausal criteria, and premenopausal women with ovarian suppression (via chemical menopause) are allowed. Key exclusion criteria are prior malignancies within 3 years of enrollment; prior major surgery, radiation therapy, or systemic anticancer therapy within 3 weeks of study entry; major ECG abnormalities or cardiac disorders; symptomatic brain metastasis or visceral metastasis; active infection; active inflammatory gastrointestinal diseases or conditions; and pregnancy or breastfeeding. The recommended dose for expansion for PF-07248144 plus vepdegestrant will be determined in Part 1E and evaluated in Part 2E. Primary endpoints are dose-limiting toxicities (Part 1E only), safety, and tolerability. Secondary endpoints include pharmacokinetics of PF-07248144 and vepdegestrant in both Parts 1E and 2E, and best overall response, duration of response, clinical benefit rate, progression-free survival, time to progression, and overall survival in Part 2E.
Presentation numberPS5-12-16
A Phase 2 Trial of (Z)-endoxifen + Goserelin as Neoadjuvant Treatment for Premenopausal Women with ER+, HER2-, Breast Cancer (EVANGELINE)
Matthew P Goetz, Mayo Clinic, Rochester, MN
M. P. Goetz1, V. J. Suman2, C. Lopez3, H. Erickson3, L. Beaulieu3, S. S. Hammer3, L. Mina4, R. Leon-Ferre5, K. N. Hunt6, M. Piltin7, A. Degnim7, J. N. Ingle8, J. C. Boughey8, B. A. Sarah9, J. M. Reid9, M. Schellenberg9, J. R. Hawse9, P. Advani10, K. Giridhar1, F. Batalini11, D. Flora12, J. M. Jones13, N. A. Bagegni14, J. Jakub15, V. Abramson16, V. Dabak17, W. J. Irvin18, E. Sima19, K. Seema20, S. C. Quay3; 1Oncology (Medical), Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN, 2Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 3n/a, Atossa Therapeutics Inc, Seattle, WA, 4n/a, Mayo Clinic, Pheonix, AZ, 5Oncology (Medical) Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN, 6Mayo Clinic Comprehensive Cancer Center Mayo Clinic Children’s Pediatrics Radiology, Mayo Clinic, Rochester, MN, 7Mayo Clinic Comprehensive Cancer Center Surgery, Mayo Clinic, Rochester, MN, 8N/A, Mayo Clinic, Rochester, MN, 9n/a, Mayo Clinic, Rochester, MN, 10n/a, Mayo Clinic, Jacksonville, FL, 11N/A, Mayo Clinic, Phoenix, AZ, 12N/A, St. Elizabeth Healthcare, Edgewood, KY, 13N/A, Avera Health, Sioux Falls, SD, 14N/A, Washington University, Rochester, MN, 15N/A, Mayo Clinic, Jacksonville, FL, 16N/A, Vanderbilt Ingram Cancer Center, Nashville, TN, 17n/a, Henry Ford Cancer Institute, Detroit, MI, 18N/A, 10Bon Secours Cancer Institute, Midlothian, VA, 19n/a, University of Arizona, Tucson `, AZ, 20N/A, Northwestern University, Chicago, IL.
Background: Endocrine therapy (ET) with aromatase inhibitors (AIs) plus ovarian function suppression (OFS) is a standard endocrine strategy for premenopausal women with ER+/HER2- breast cancer. However, poor tolerability to AI+OFS limits its use, leaving tamoxifen as the only alternative. Prospective studies demonstrated that premenopausal women treated with neoadjuvant ET whose tumors exhibit Ki-67 ≤ 10% following 4 weeks of therapy achieve 5-year distant disease-free survival > 96%. Problematically, only 41% of patients reach this threshold with tamoxifen compared to 78% with AI+OFS (Nitz JCO 2022). (Z)-endoxifen (ENDX) is a potent selective estrogen receptor modulator that dually targets ERα and PKCβ1. At plasma concentrations of 3-5 ng/mL, it inhibits ERα; at ≥ 500 ng/mL, it targets PKCβ1, leading to AKT suppression. The EVANGELINE trial aims to examine endocrine sensitivity in terms of Week 4 Ki-67 in premenopausal women whose baseline Ki-67 > 10%, and objective response rate after 24 weeks of ENDX + goserelin in premenopausal women with baseline Ki-67≤ 10%. Methods: EVANGELINE (NCT05607004) is a multicenter, open-label, Phase 2 study comprised of three parts. Eligible patients are premenopausal women with Stage IIA/IIB ER+/HER2- breast cancer. • Part 1 (PK Run-in): assessed 40mg vs 80mg ENDX (+/- OFS) in 22 patients. • Part 2: patients with baseline Ki-67 >10% were randomized to 40 mg ENDX + goserelin or exemestane + goserelin for 6 months with the primary objective being 4-week ESD rate. Patients with a baseline Ki-67 ≤10% were assigned to a single-arm cohort of 40 mg ENDX monotherapy (no OFS) to evaluate the 24-week endocrine sensitivity disease rate. • Part 3 (replaces Part 2): all patients are treated with ENDX 40mg daily + goserelin every 28 days. A two stage Phase II design was chosen to assess outcomes as follows: Part 3a: Up to 45 patients with baseline Ki-67> 10% to assess Week 4 Ki-67≤ 10% rate is at least 65%. If 9 or more of these patients have a Week 4 Ki-67>10%, enrollment will be stopped due to futility. Otherwise, enrollment will continue. Part 3b: Up to 20 patients enrolled with baseline Ki-67≤ 10% to assess Week 24 objective response rate. Ki-67 levels are determined by central laboratory and images are centrally reviewed. Secondary objectives include safety, tolerability, surgical outcomes, and biomarker analysis using paired biopsies (baseline, week 4) and surgical specimens. Results: The PK run-in (completed Fall 2024) enrolled 22 patients and assessed ENDX doses (40 mg daily vs 80 mg daily (+/-OFS)). The Week 4 Ki-67 ≤ 10% rate with ENDX was 86% and did not differ according to dose. Early safety, PK, efficacy and compliance data supported the selection of 40 mg ENDX daily as the optimal dose. Enrollment in Part 2 began in May 2025. Part 3 will replace Part 2 with simplification of the trial design. Conclusions: EVANGELINE is the first trial to evaluate (Z)-endoxifen + OFS as neoadjuvant therapy in premenopausal ER+/HER2- breast cancer. By targeting both ER and PKC pathways, ENDX may offer a well-tolerated alternative to AI-based regimens and expand endocrine therapy options for this population. Clinical Trial Registration: NCT05607004
Presentation numberPS5-12-17
A phase 1/2 study of AVZO-023, a next-generation selective cyclin-dependent kinase 4 inhibitor (CDK4i), as a single agent and in combination with AVZO-021, a selective cyclin-dependent kinase 2 inhibitor (CDK2i), and/or endocrine therapy in patients with advanced solid tumors
Timothy A Yap, The University of Texas MD Anderson Cancer Center, Houston, TX
T. A. Yap1, N. P. McAndrew2, D. K. Marks3, A. I. Spira4, D. Sommerhalder5, M. Hirmand6, S. Thamake6, M. Mehta6, A. Giordano7; 1Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Division of Hematology and Oncology, Department of Medicine, University of California Los Angeles, Santa Monica, CA, 3Perlmutter Cancer Center, NYU Langone Health, Mineola, NY, 4Medical Oncology, Clinical Research, NEXT Oncology-Virginia, Fairfax, VA, 5Medical Oncology, Clinical Research, NEXT Oncology-San Antonio, San Antonio, TX, 6Avenzo Therapeutics, Avenzo Therapeutics, San Diego, CA, 7Dana-Farber Cancer Institute, Department of Medical Oncology, Boston, MA.
Background CDK4/6 inhibitors (CDK4/6i) have revolutionized the treatment of HR+/HER2- breast cancer (BC) but they have dose-limiting CDK6i-related hematologic toxicity, and their efficacy is also limited due to resistance secondary to CDK2 activation. Selective CDK4i may have reduced hematological toxicity and enhanced CDK4 inhibition vs CDK4/6i. In addition, combining selective CDK4i with selective CDK2i can overcome resistance. AVZO-023 is an oral, potent, next-generation CDK4i with high selectivity over CDK6 that may offer improved efficacy and tolerability vs current CDK4/6i for patients with HR+/HER2- BC. AVZO-023 showed strong anti-tumor activity in vivo, which was further enhanced in combination with the selective CDK2i AVZO-021 in palbociclib-sensitive and palbociclib-resistant xenograft models. Trial Design This Phase 1 AVZO-023-1001 trial (NCT06998407) will assess the safety, pharmacokinetics (PK), pharmacodynamics (PD), and anti-tumor activity of AVZO-023 as a single agent and in combination with endocrine therapy and/or AVZO-021. HR+/HER2- BC patients represent the key patient population of interest that will be enrolled in this study. Eligible patients have ECOG PS of ≤1, ≤3 lines of chemotherapy/antibody-drug conjugate therapy, no prior exposure to investigational CDKi, and no unstable CNS metastases. Part A will study AVZO-023 monotherapy. Accelerated titration will be used for the first 3 dose levels of AVZO-023, followed by additional dose levels using a BOIN design. A food effect cohort will also be enrolled. Part B will study the combination of AVZO-023 + AVZO-021. Patients with HR+/HER2- BC enrolled in Parts A or B will have the option to add fulvestrant to their study treatment. Part C will study the combination of AVZO-023 + letrozole ± AVZO-021. Backfill enrollment of patients is planned in this study. Primary objectives are safety/tolerability and secondary objectives include preliminary anti-tumor activity. The trial has activated and will enroll patients at approximately 50 centers across North America, Europe and the Asia-Pacific region.
Presentation numberPS5-12-18
OPERA-02: a phase 3 randomized, double-blind, active-controlled study of palazestrant with ribociclib versus letrozole with ribociclib for the first-line treatment of ER+, HER2- advanced breast cancer
Sara M Tolaney, Dana Farber Cancer Institute, Boston, MA
S. M. Tolaney1, P. Bianchini2, V. F. Borges3, E. Ciruelos4, W. Janni5, K. Jhaveri6, P. Schmid7, J. Tsang8, G. Werutsky9, D. Vecchio10, Y. Luan11, N. Di Santo12, B. Pistilli13; 1Breast Oncology, Dana Farber Cancer Institute, Boston, MA, 2Clinical Oncology, University Vita-Salute San Raffaele, Milan, Italy, Milan, ITALY, 3Medical Oncology, University of Colorado, Aurora, CO, 4Breast Cancer Unit, Universidad Hospital 12 de Octubre, Madrid, SPAIN, 5Obstetrics and Gynecology, University of Ulm, Ulm, GERMANY, 6Breast Medical Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, 7Breast Cancer Center, Barts Cancer Institue, London, UNITED KINGDOM, 8Medical Oncology, The University of Hong Kong, Hong Kong, CHINA, 9Medical Oncology, Futtura Oncologia, Porto Alegre, BRAZIL, 10Clinical Science, Olema Oncology, San Francisco, CA, 11Biostatistics, Olema Oncology, San Francisco, CA, 12Clinical Development, Olema Oncology, San Francisco, CA, 13Breast Cancer Unit, Gustave Roussy, Villejuif, FRANCE.
Background: Approximately 70% of advanced breast cancers (ABC) are estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) tumors. The standard-of-care first-line treatment for ER+, HER2- ABC is an aromatase inhibitor (AI) plus cyclin-dependent kinase 4/6 inhibitor (CDK4/6i). As the first line treatment, ribociclib is the only CDK4/6i that in combination with AIs improved both progression-free survival (PFS) and overall survival (OS) in ER+, HER2- ABC. Despite the improved survival outcomes with AI + CDK4/6i, resistance develops eventually in most patients. The most common mechanism of endocrine resistance involves mutations in the ESR1 gene that encodes the estrogen receptor alpha (ERα). Treatment with AIs induces these mutations in the ligand-binding domain of ERα, thereby determining an estrogen-independent constitutive activation of ER. The ability to suppress the activity of both wild-type and mutated ERs in the first line setting is a potentially effective therapeutic approach. By preventing the appearance of ESR1 mutations, this approach has the potential of maintaining endocrine responsiveness and improving clinical outcomes in patients with ER+, HER2− ABC. Palazestrant (OP-1250) is a novel oral, complete estrogen receptor antagonist (CERAN) and selective estrogen receptor degrader (SERD) that acts by blocking both transcriptional activation function domains, AF1 and AF2. Early phase studies investigating palazestrant as monotherapy (Lin et al. ESMO 2023) and in combination with ribociclib (Borges et al. SABCS 2024) demonstrated a tolerable safety profile, favorable pharmacokinetics with no drug-drug interactions with ribociclib, and encouraging antitumor efficacy in both ESR1-mut and ESR1-wt ER+, HER2- ABC. In addition, palazestrant showed promising anti-tumor efficacy in patients who received prior treatment with CDK4/6i in both ESR1-mut and ESR1-wt ER+, HER2- ABC. When combined, palazestrant and ribociclib safety was consistent with known safety profiles of each drug. Methods: OPERA-02 is an international, multicenter, randomized, double-blind, active-controlled phase 3 clinical trial comparing the efficacy and safety of palazestrant in combination with ribociclib versus letrozole plus ribociclib in patients with ER+, HER2- ABC who have not previously received systemic treatment for ABC. Eligible patients are adult women of any menopausal status or men with de novo or recurrent (after 12 months of completion of adjuvant endocrine therapy) ER+, HER2- ABC who have not received any prior systemic therapy for advanced disease. Additional inclusion criteria include measurable disease per RECIST 1.1 or evaluable bone disease, ECOG performance score of 0-1, adequate hematologic, hepatic, and renal functions. Pre- or perimenopausal women and men must be willing to receive GnRH agonists for gonadal suppression. Patients with a contraindication to ribociclib are excluded. Overall, approximately 1000 participants will be randomized 1:1 to receive 90 mg qd palazestrant plus ribociclib 600 mg qd for 21 consecutive days followed by 7 days off plus letrozole-matching placebo or letrozole 2.5 mg qd plus ribociclib plus palazestrant-matching placebo. Randomization will be stratified by menopausal status, visceral metastasis, and de novo metastatic disease versus recurrent disease. The primary endpoint is PFS assessed by local investigator. Secondary endpoints include PFS by a blinded-independent review committee, OS, overall response rate, clinical benefit rate, duration of response, safety, pharmacokinetics and health-related patient-reported outcomes. The study is conducted globally.
Presentation numberPS5-12-19
Pre-ispy trial: a phase i/ib oncology platform program (pre-i-spy-p1) nct05868226
PAULA R POHLMANN, University of Texas MD Anderson Cancer Center, Houston, TX
P. R. POHLMANN1, D. A. Potter2, N. Chen3, N. Jahan4, J. A. Mouabbi5, H. S. Han6, J. R. Diamond7, M. Bhave8, M. Rampurwala9, S. Rayani10, A. Discacciati11, M. Eklund11, A. G. Chappel12, A. L. Delson13, B. K. LeStage14, C. Hendricks-Kretzer14, S. Venters15, L. Brown-Swigart15, G. L. Hirst16, E. Petricoin III17, A. D. Borowsky18, L. J. van ‘t Veer15, S. M. Asare19, S. Jafari20, P. Henderson20, D. Tripathy5, H. Rugo21, R. Nanda3, E. Stringer-Reasor4, D. Yee22, F. Meric-Bernstam23, L. J. Esserman24; 1Breast Medical Oncology and Investigational Cancer Therapeutics (joint appointment), University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 3Department of Medicine, University of Chicago, Chicago, IL, 4Division of Hematology and Oncology, University of Alabama at Birmingham O’Neal Comprehensive Cancer Center, Birmingham, AL, 5Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 6Breast Oncology, Moffitt Cancer Center, Tampa, FL, 7Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 8Department of Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, 9Hematology and Medical Oncology, UChicago Medicine Center for Advanced Care Orland Park, Orland Park, IL, 10Hematology and Medical Oncology, UChicago Medicine Cancer Care at Silver Cross Hospital, New Lenox, IL, 11Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SWEDEN, 12Statistics, Quantum Leap Healthcare Collaborative, San Francisco, CA, 13Breast Science Advocacy Core, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 14Advocates Working Group, Quantum Leap Healthcare Collaborative, San Francisco, CA, 15Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 16Department of Surgery, University of California San Francisco, San Franciso, CA, 17Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 18Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, 19Program Management, Quantum Leap Healthcare Collaborative, San Francisco, CA, 20Collaborations, Quantum Leap Healthcare Collaborative, San Francisco, CA, 21Breast Medical Oncology, City of Hope Cancer Center, Duarte, CA, 22Departments of Medicine and Pharmacology (joint appointment), Masonic Cancer Center, University of Minnesota, Minneapolis, MN, 23Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 24Department of Surgery, University of California San Francisco, San Francisco, CA.
Background Many promising agents or combination regimens relevant to solid malignancies lack sufficient clinical information for use in the curative setting. To enable acceleration of drug development by reducing time and cost of clinical trial planning and conduct, the Phase I PRE-ISPY trial was created. Methods The PRE-ISPY trial was envisioned as part of the growing I-SPY network of trials, leveraging its efficiencies in streamlined regulatory pathways and trial design processes, including contracting, activation, enrollment, data gathering and analysis. The goals of designing the PRE-ISPY trial were to support investigators and industry partners in meeting safety and preliminary efficacy qualifications for phase II/III studies and gathering translational data to identify the optimal clinical development setting in a timely fashion. The primary aims of the trial are to assess drug toxicity/safety, to determine the Maximum Tolerated Dose (MTD) and Recommended Phase 2 Dose (RP2D), to evaluate preliminary treatment efficacy via Overall Response Rate and Clinical Benefit Rate; and biospecimen acquisition, while gaining experience with the drug/regimen in patients with advanced/recurrent solid malignancies. Arms are independent and may have additional specific aims. Sample size depends on the arm design, number of dose-finding cohorts, and number of expansion cohorts. The minimum sample size for an arm is 15 patients, with no maximum. Study arms include dose finding (Part 1) and dose expansion (Part 2) components. Part 1 is optional if MTD and RP2D have already been established. Each study arm may have specific criteria for transitioning from Part 1 to Part 2. Pre-specified exploratory and qualifying transcriptomic, proteomic and phosphoprotein biomarkers will be analyzed. Results The study is sponsored by Quantum Leap Healthcare Collaborative (QLHC). PRE-ISPY uses a master protocol to evaluate multiple regimens in parallel, enabling seamless addition, modification and removal of study arms. New regimens are introduced through appendices in subsequent amendments. A centralized regulatory process accelerates the activation process. Centralized systems handle cost analysis, budgeting, industry contract management, electronic case report forms and electronic data capture systems creation, and data management. Drug and lab kit shipments are coordinated via a unified logistics system. Research specimen collection utilizes a centralized biobanking infrastructure to ensure consistency in sample handling and storage. General eligibility includes adults with advanced/metastatic or recurrent solid tumors, adequate performance status, and organ function. Arm-specific criteria detail additional requirements for tumor histology, treatment history, and organ function. Graduation of a PRE-ISPY regimen to I-SPY2 (neoadjuvant breast cancer trial) or K-SPY (colorectal trial) is not mandatory. The PRE-ISPY trial opened for enrollment on 2/1/2023. Candidate treatment regimens are selected with advice from the I-SPY2 Agent Selection Working Group and K-SPY consortium. The current arms under study are PRE1 ALX-148/T-DXd; PRE2 tucatinib/zanidatamab; and PRE3 vidutolimod/cemiplimab. As of July 2025, eight main sites and two satellite sites are activated, with fourteen additional site activations planned from Q3 2025 to Q1 2026. All sites are academic centers affiliated with NCI-Designated Comprehensive Cancer Centers. An estimated 18 weeks are required from concept arm development to study arm activation; final protocol version to study arm activation requires 6 weeks. Conclusion The PRE-ISPY phase I/Ib trial has been successfully implemented nationally as part of the Consortium of I-SPY trials. Once safety is established, promising regimens can be rapidly transitioned to I-SPY2.2, K-SPY or other phase II/III trials.
Presentation numberPS5-12-20
Synthesis-breast: a prospective early-phase trial of a synthetic lethality-focused algorithm to identify therapeutic options in metastatic breast cancer
Aanika B Warner, National Institutes of Health, Bethesda, MD
A. B. Warner1, B. B. Solarz1, M. Belyov1, V. Bolanos1, A. McCoy1, T. Fujii1, J. Lee1, K. C. Conlon1, G. Dinstag2, R. Aharonov2, T. Beker2, K. Aldape3, E. Ruppin4, S. Lipkowitz1, P. S. Rajagopal4; 1Women’s Malignancy Branch, National Institutes of Health, Bethesda, MD, 2Pangea Biomed, Pangea Biomed, Tel Aviv, ISRAEL, 3Laboratory and Pathology, Cancer Data Science, National Institutes of Health, Bethesda, MD, 4Women’s Malignancy Branch, Cancer Data Science, National Institutes of Health, Bethesda, MD.
Background: Patients with metastatic triple-negative (TNBC) or endocrine-refractory hormone receptor-positive (HR+) breast cancer have limited therapeutic options and survival benefit once reaching chemotherapy-only options. Existing precision oncology uses actionable mutations, which restrict targeted therapies to patients harboring those mutations. ENLIGHT is a computational algorithm that predicts possible treatment options via tumor transcriptomes using synthetic lethality, validated using 21 retrospective pan-cancer trial datasets with 697 total patients. This exploratory clinical trial seeks to evaluate ENLIGHT’s feasibility to prospectively identify treatment options in this population. Design: SYNTHESIS-Breast (Clinicaltrials.gov NCT number requested) is an exploratory single-institution trial that will enroll participants with advanced metastatic TNBC or endocrine-refractory HR+ breast cancer who were previously treated with approved regimens with known overall survival benefits. Tumor biopsy samples (from within 6 months before enrollment) are mailed in for screening and will undergo genomic and transcriptomic profiling using the TruSight Oncology 500 (TSO500) platform; whole-transcriptome RNA-seq will be analyzed through ENLIGHT. Participants – initially seen virtually – will be assigned to 1 of 3 arms based on TSO500 and ENLIGHT as reviewed by a molecular tumor board: Arm 1 (no treatment recommendation), Arm 2 (recommendation for an FDA on-label treatment option), or Arm 3 (ENLIGHT recommendation from 1 of 22 off-label treatment options). Participants in Arm 3 will be treated at the NIH, and those in Arms 1 and 2 will continue with their community oncologists and be followed virtually. The trial uses a Simon two-stage design with a two-part primary objective. Part A assesses feasibility through the number of participants matched to an ENLIGHT-recommended therapy. If enough participants are matched to an ENLIGHT-recommended therapy within 1 year, the trial moves to Part B, evaluating objective response in Arm 3 participants. Overall success of the trial is defined as a 40% response rate of Arm 3 participants.This study is not designed to validate ENLIGHT for routine clinical use but aims to 1) assess the feasibility of prospective use of this computational algorithm within a reasonable timeframe, and 2) guide future trials to validate clinical use of ENLIGHT. In pilot testing of 10 archival breast cancer tissue samples, all RNA-seq-evaluable samples had ≥ 1 ENLIGHT match; 6/10 were matched to immunotherapy options. Among those, 4/6 would not have been eligible under current guidelines. Implications: SYNTHESIS-Breast is a prospective trial that uses ENLIGHT, a computational algorithm based on transcriptome-based synthetic lethality, to predict off-label treatment options for participants with metastatic breast cancer who have exhausted initial-line therapies. The current mutation-based model of precision oncology leaves patients behind, requiring alternative strategies that can use multi-omic data and predictive approaches. Insights from this trial will not only inform future trials using ENLIGHT, but also early-phase trials assessing other computational algorithms.
Presentation numberPS5-12-21
Herthena-breast03: a phase 2, randomized, open-label study evaluating neoadjuvant patritumab deruxtecan (HER3-DXd) + pembrolizumab before or after pembrolizumab + chemotherapy for early-stage TNBC or HR-low+/HER2− breast cancer
Joyce O’Shaughnessy, Baylor University Medical Center, Texas Oncology, US Oncology and Sarah Cannon Research Institute, Dallas, TX
J. O’Shaughnessy1, J. M. Collins2, L. Yao3, K. L. Smith4, J. A. Mejia4, M. Danso5; 1Oncology, Baylor University Medical Center, Texas Oncology, US Oncology and Sarah Cannon Research Institute, Dallas, TX, 2Oncology, Daiichi Sankyo, Inc., Basking Ridge, NJ, 3Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, 4Global Clinical Development, Merck & Co., Inc., Rahway, NJ, 5Medical Oncology, Virginia Oncology Associates, Norfolk, VA.
Background: The standard of care for patients with high-risk, early-stage triple-negative breast cancer (TNBC) is neoadjuvant pembrolizumab + chemotherapy followed by adjuvant pembrolizumab. Patients with HR-low+/HER2− breast cancer are often managed according to TNBC recommendations. There is a need for improved neoadjuvant therapy to increase the rate of pathological complete response (pCR), as patients who do not achieve pCR have a high risk of recurrence, and to reduce risk of long-term toxicities associated with cyclophosphamide and anthracyclines. HER3 is frequently expressed in breast cancer and overexpression is associated with drug resistance. Patritumab deruxtecan (HER3-DXd) is an antibody-drug conjugate comprising a fully human anti-HER3 IgG1 monoclonal antibody linked to a topoisomerase I inhibitor (DXd) via a stable tetrapeptide-based linker that is selectively cleaved within tumor cells. In a phase 2 study, HER3-DXd monotherapy showed antitumor activity and manageable safety in heavily pretreated participants with HER3-high expressing TNBC. This phase 2 study (NCT06797635) is evaluating neoadjuvant HER3-DXd + pembrolizumab administered before or after carboplatin + paclitaxel + pembrolizumab for early-stage TNBC or HR-low+/HER2− breast cancer. Methods: Eligible participants are adults (≥18 y) with untreated, locally advanced nonmetastatic (AJCC stage cT1c, N1-N2 or cT2-cT4, N0-N2) TNBC or HR-low+/HER2− breast cancer. Participants (N ≥10 and ≤30) in Part 1 of the study (safety lead-in) will receive neoadjuvant HER3-DXd + pembro followed by carboplatin + paclitaxel + pembro (Table) followed by surgery. Dose-limiting toxicity evaluation and dose finding for HER3-DXd (up to three dose levels of 5.6 mg/kg Q3W, 4.8 mg/kg Q3W and 3.2 mg/kg Q3W) during cycle 1 of neoadjuvant HER3-DXd + pembrolizumab will be performed in Part 1 to determine an optimal dose of HER3-DXd for Part 2. Participants (N ~342) in Part 2 will be randomly assigned 1:1:1 to Arm A, B or C (Table) for neoadjuvant treatment. Randomization will be stratified by cancer type (TNBC vs HR-low+/HER2−) and, in the TNBC subgroup, PD-L1 status (combined positive score ≥10 vs <10), overall clinical stage (II vs III) and HER3 expression per IHC (low vs high). After neoadjuvant treatment, participants will undergo surgery (with postoperative radiotherapy if clinically indicated) and receive adjuvant pembrolizumab 400 mg Q6W for 5 cycles. Participants with residual disease may receive additional adjuvant treatment of physician’s choice. Primary endpoints are safety (Part 1 and 2) and pCR (ypT0/Tis ypN0) rate (Part 2). Enrollment is ongoing.
| Neoadjuvant cycles 1-4 | Neoadjuvant cycles 5-8 | |
| Part 1 Arm A | HER3-DXd 5.6 or 4.8 or 3.2 mg/kg Q3W + pembroa | Carboplatinb + paclitaxelc + pembroa |
| Part 2 Arm A | HER3-DXd (selected dose from Part 1) + pembroa | Carboplatinb+ paclitaxelc + pembroa |
| Part 2 Arm B | Carboplatinb + paclitaxelc + pembroa | HER3-DXd (selected dose from Part 1) + pembroa |
| Part 2 Arm C | Carboplatinb + paclitaxelc + pembroa | Doxorubicind OR epirubicine + cyclophosphamidef + pembroa |
|
a200 mg Q3W; bAUC 1.5 mg/mL/min QW; c80 mg/m2 QW; d60 mg/m2 Q3W; e90 mg/m2 Q3W; f600 mg/m2 Q3W |
Presentation numberPS5-12-22
Trofuse-012: a phase 3, randomized study of adjuvant sacituzumab tirumotecan plus pembrolizumab vs treatment of physician’s choice in participants with triple-negative breast cancer who received neoadjuvant therapy and did not achieve a pathological complete response at surgery
Heather L McArthur, UT Southwestern Medical Center, Dallas, TX
H. L. McArthur1, R. Dent2, R. Hui3, Y. Park4, P. Schmid5, J. Wei6, J. A. Mejia6, W. Pan6, J. Cortés7; 1Oncology, UT Southwestern Medical Center, Dallas, TX, 2Oncology, National Cancer Center Singapore and Duke-National University of Singapore (NUS) Medical School, Singapore, SINGAPORE, 3Oncology, Centre of Cancer Medicine, School of Clinical Medicine, University of Hong Kong, Hong Kong, Hong Kong SAR China; and Westmead Breast Cancer Institute, Westmead Hospital and the University of Sydney, Sydney, NSW, AUSTRALIA, 4Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF, 5Oncology, Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 6Global Clinical Development, Merck & Co., Inc., Rahway, NJ, 7Department of Medicine, Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences Madrid, Spain; IOB Madrid, Madrid, SPAIN.
Background: Trophoblast cell surface antigen 2 (TROP2) expression is higher in triple-negative breast cancer (TNBC) vs other breast cancer subtypes, and high expression is associated with poor prognosis. Sacituzumab tirumotecan (sac-TMT; also known as MK-2870/SKB264) is a novel antibody-drug conjugate composed of anti-TROP2 antibody coupled to a cytotoxic belotecan derivative via a novel linker (average drug/antibody ratio, 7.4). In a prior phase 3 study (OptiTROP-Breast01), sac-TMT alone improved PFS (HR, 0.31; 95% CI, 0.22-0.45; P < 0.00001) and OS (HR, 0.53; 95% CI, 0.36-0.78; P = 0.0005) vs chemotherapy in participants with heavily pretreated advanced TNBC. The current standard of care (SOC) for patients with newly diagnosed, high-risk, early-stage TNBC is neoadjuvant pembrolizumab + chemotherapy followed by adjuvant pembrolizumab after surgery. Patients who do not achieve a pathological complete response (pCR) with the current SOC have higher rates of recurrence and mortality vs patients who achieve pCR. This study (NCT06393374) evaluates adjuvant sac-TMT plus pembrolizumab vs treatment of physician’s choice (TPC; pembrolizumab ± capecitabine) in participants with TNBC who received neoadjuvant therapy and did not achieve pCR at surgery. Methods: This phase 3, multicenter, open-label study is enrolling participants ≥18 years old with centrally confirmed TNBC per most recent American Society of Clinical Oncology/College of American Pathologists guidelines. Participants have non-pCR after ≥5 cycles of neoadjuvant pembrolizumab plus chemotherapy, including ≥1 cycle of anthracycline-based neoadjuvant therapy. Participants must provide tissue from the surgical specimen for central TROP2 assessment and be able to continue on adjuvant pembrolizumab. Randomization must be conducted ≤16 weeks from surgical resection (window may be extended in consult with sponsor). Participants are randomized 1:1 to pembrolizumab 400 mg Q6W for 5 doses + sac-TMT 4 mg/kg Q2W for 12 doses or TPC with pembrolizumab 400 mg Q6W for 5 doses ± capecitabine 1000-1250 mg/m2 BID on days 1-14 and days 22-35 every 42 days for 4 cycles until completion of therapy or disease recurrence, unacceptable toxicity, or withdrawal. Primary endpoint is invasive disease-free survival. Secondary endpoints are OS, distant recurrence-free survival, patient-reported outcomes, and safety. Enrollment began Q2 2024.
Presentation numberPS5-12-23
Herthena-breast01: a phase 1b/2, multicenter, open-label, dose-finding study to evaluate the safety and antitumor activity of patritumab deruxtecan (HER3-DXd) in HER2+ unresectable locally advanced or metastatic breast cancer
Sara Tolaney, Dana-Farber Cancer Institute and Harvard Medical School Boston, Boston, MA
S. Tolaney1, C. Pinheiro2, A. Sporchia3, Q. Liu2, K. M. Hirshfield2, H. Iwata4; 1Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School Boston, Boston, MA, 2Oncology, Merck & Co., Inc., Rahway, NJ, 3Oncology, Daiichi Sankyo Italy S.p.A., Milan, ITALY, 4Oncology, Nagoya City University, Nagoya, JAPAN.
Background: Although HER2-targeted antibody-drug conjugates (ADCs) have improved outcomes in HER2+ advanced or metastatic breast cancer, safe and effective later-line therapies are needed for patients who have progressed on trastuzumab deruxtecan (T-DXd). Human epidermal growth factor receptor 3 (HER3) is expressed in many tumor types, including HER2+ breast cancer. Patritumab deruxtecan (HER3-DXd), a novel ADC that selectively binds HER3, is composed of a fully human anti-HER3 IgG1 antibody linked to a cytotoxic topoisomerase I inhibitor via a tetrapeptide-based cleavable linker (drug-to-antibody ratio, ~8). In a phase 1/2 study, HER3-DXd monotherapy showed durable antitumor activity and manageable safety in heavily pretreated participants across breast cancer subtypes, including HER2+, HER3-expressing metastatic breast cancer. This phase 1b/2, multicenter, open-label, dose-finding HERTHENA-Breast01 study (NCT06686394) will evaluate safety, pharmacokinetic exposure parameters, and preliminary antitumor activity of HER3-DXd in combination with anti-HER2 agents in participants with HER2+ unresectable locally advanced or metastatic breast cancer. Methods: Eligible participants are ≥18 y with histologically confirmed HER2+ (per most recent ASCO/CAP guidelines: IHC 3+, IHC 2+/in situ hybridization positive [ISH+], or IHC undetermined/ISH+) unresectable locally advanced or metastatic breast cancer. Tumor tissue will be collected at study entry and assessed locally for HER2+ and hormone receptor status. Participants will have measurable disease per RECIST v1.1, ECOG PS of 0 or 1, and adequate organ function. Up to 27 participants will be enrolled in each arm of the study. Participants in Arm 1 will have received 2−5 lines of anti-HER2 therapy in the locally advanced or metastatic setting, including disease progression on prior T-DXd. Participants in Arm 2 will have received a maximum of 5 lines of anti-HER2 therapy in the locally advanced or metastatic setting, with no minimum prior lines of therapy. Participants in Arm 3 will have received 1-3 lines of anti-HER2 therapy in the locally advanced or metastatic setting, including disease progression on T-DXd as the most recent therapy. Participants in Arm 1 will receive HER3-DXd Q3W plus trastuzumab 8 mg/kg IV followed by 6 mg/kg IV Q3W. Participants in Arms 2 and 3 will receive the same regimen as Arm 1, with either pertuzumab 840 mg IV followed by 420 mg IV Q3W (Arm 2) or tucatinib 300 mg orally BID (Arm 3). Dose level decisions (i.e. escalation and de-escalation) will be performed based on the Bayesian Optimal Interval design. Treatment will continue until radiographic progression, unacceptable toxicity, or participant withdrawal. Primary endpoints are dose-limiting toxicities, safety, and treatment discontinuations due to AEs. Secondary endpoints are pharmacokinetics of HER3-DXd ADC, total HER3-DXd antidrug antibody, and free DXd payload. AEs are graded per National Cancer Institute Common Terminology Criteria for Adverse Events 5.0. Enrollment began in early 2025.
Presentation numberPS5-12-25
Trofuse-032: a phase 3, randomized study of pembrolizumab plus sacituzumab tirumotecan or chemotherapy followed by pembrolizumab plus chemotherapy for early-stage triple-negative breast cancer or hormone receptor-low-positive (HR-low+)/ human epidermal growth factor receptor 2-negative (HER2−) breast cancer
Nadia Harbeck, LMU University Hospital, Munich, Germany
N. Harbeck1, J. Wei2, F. Beca3, J. A. Mejia3, P. Schmid4; 1Breast Center, Dept. Obstetrics and Gynecology, LMU University Hospital, Munich, GERMANY, 2Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, 3Global Clinical Development, Merck & Co., Inc., Rahway, NJ, 4Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University London, London, UNITED KINGDOM.
Background: Trophoblast cell surface antigen 2 (TROP2) is highly expressed in triple-negative breast cancer (TNBC) and is associated with poor prognosis. Current standard of care for high-risk, early-stage TNBC is neoadjuvant pembrolizumab (pembro) plus chemotherapy (chemo) followed by adjuvant pembro after surgery. The same approach may be used for HR-low+/HER2− breast cancer. Patients with TNBC or HR-low+/HER2− breast cancer without a pathological complete response (pCR) after neoadjuvant therapy have higher risks of recurrence and mortality. Sacituzumab tirumotecan or sac-TMT (also known as MK-2870/SKB264), a novel antibody-drug conjugate composed of an anti-TROP2 antibody coupled to a cytotoxic belotecan derivative via a novel linker, has demonstrated significant PFS and OS benefits vs chemo in patients with metastatic TNBC. TroFuse-032 (NCT06966700; EU CT 2024-520190-12) evaluates efficacy and safety of neoadjuvant pembro plus sac-TMT or chemo followed by pembro plus chemo in participants with early-stage TNBC or HR-low+/HER2− breast cancer. Trial Design: This phase 3, randomized, open-label study is enrolling participants aged ≥18 y with centrally confirmed untreated, high-risk, early-stage TNBC or HR-low+/HER2− breast cancer per ASCO/CAP guidelines. Participants are randomized 1:1 to neoadjuvant pembro plus sac-TMT followed by pembro plus paclitaxel plus carboplatin (arm 1) vs pembro plus paclitaxel plus carboplatin followed by pembro plus doxorubicin or epirubicin plus cyclophosphamide (arm 2; Table). Surgery is performed 3-6 wk after last dose of neoadjuvant treatment. Adjuvant therapy (Table) is initiated ≤60 d after surgery. Primary efficacy endpoints are pCR (ypT0/Tis ypN0) at surgery and event-free survival (EFS). Secondary endpoints include OS, pCR-no DCIS (ypT0 ypN0), distant progression- or distant recurrence-free survival, patient-reported outcomes, and safety. Enrollment is ongoing.
| Neoadjuvant | Adjuvant | ||
| Cyclesa 1–2 | Cyclesa 3–4 | Cyclesa 5–9 | |
| Arm 1 | Pembrob + sac-TMT (4 mg/kg Q2W) | Pembrob + paclitaxelc + carboplatind |
Participants with pCR: Pembro (400 mg Q6W or 200 mg Q3W) |
| Arm 2 | Pembrob + paclitaxelc + carboplatind | Pembrob + doxorubicine or epirubicinf + cyclophosphamideg |
Participants with residual disease: Pembro (400 mg Q6W or 200 mg Q3W) + optional treatment of physician’s choice (olaparibh, capecitabinei or doxorubicinj,k or epirubicinl,k + cyclophosphamidem,k) |
|
aCycle length = 6 wks; b200 mg Q3W; c80 mg/m2 QW; dAUC 1.5 QW; e60 mg/m2 Q3W; f90 mg/m2 Q3W; g600 mg/m2 Q3W; h100 or 150 mg every 2 wks; i1000 to 1250 mg/m2 every 2 wks; j60 mg/m2 Q2W; kArm 1; l90 mg/m2 Q2W; m600 mg/m2 Q2W |
Presentation numberPS5-10-01
Misinformation about breast cancer treatment on social media: Implications for engagement with patients and social media
Margaret Gatti-Mays, The Ohio State University Comprehensive Cancer Center, Columbus, OH
M. Gatti-Mays1, K. Koehler2, A. VanLaven3, L. Long1, E. Alvardo1, K. Noce1, D. Stover1, S. Collins4, D. Mays1; 1Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 2Hematology/Oncology Fellowship, Department of Internal Medicine, The Ohio State University, Columbus, OH, 3The College, Denison University, Granville, OH, 4Department of Pharmacy, The Ohio State University Comprehensive Cancer Center, Columbus, OH.
Background: Despite lack of clinical trial data supporting drug efficacy or safety, many patients request alternative treatments, such as ivermectin, fenbendazole, mebendazole, and methylene blue, over standard treatments. Social media (SM) is widely available, highly utilized and a likely source of information about alternative treatments. However, SM content on alternative therapies has not been analyzed to inform engagement by oncology providers with the lay public. Methods: We used Infegy, a SM listening platform, to search for content about alternative treatments for breast cancer across SM platforms (Twitter/X, Reddit, Meta platforms) from June 2021 to June 2025. We conducted an initial search for all content about “breast cancer” to establish the denominator of content and provide a comparison. We then conducted a focused search using Boolean logic combining “breast cancer” with “ivermectin,” “methylene blue,” “fenbendazole,” “mebendazole,” and alternative names for each. Infegy provides descriptive information about content captured (e.g., # of posts, people reached, unique impressions), prominent topics, and prominent sources of posts (i.e., account handles). We manually analyzed the content of a subset of posts from the focused search to determine if they were endorsing alternative treatments, asking questions about them, or opposing them. Results: In the initial search for “breast cancer”, there were >3.1 million (M) posts with a potential reach of >33.2 billion people and generated >421M unique impressions. Prominent breast cancer themes included awareness, support, treatment, and survivorship. Prominent sources of content included cancer research and advocacy organizations (e.g., American Cancer Society, Susan G. Komen Foundation, Breast Cancer Research Foundation). In the focused search for alternative therapies, there were >370,000 posts (12% of all breast cancer posts), potentially reaching >55M people and generating >418,000 unique impressions. Prominent sources of content included health professionals affiliated with companies that promote alternative treatments, websites that sell alternative treatments, and suspected “bot” accounts. Common topics included personal testimonials and discussions about treatments, chemotherapy, and described “cures.” Most posts mentioned (not mutually exclusive) ivermectin (344/410, 84%), methylene blue (56/410, 14%), mebendazole (27/410, 7%), and fenbendazole (11/410, 3%). Overall, 73% of posts analyzed (299/410) endorsed these alternative treatments, whereas only a minority asked questions (30/410, 7%) or opposed them (20/410, 5%), and 30 posts (7%) could not be categorized. Examples of posts endorsing alternative treatments included personal testimonials and online retailers selling the treatments. Of the 410 posts from the focused search, 196 were unique content (48%) and manual review of example posts indicates much of the content may be generated by suspected “bots.” Conclusions: Although it represents a minority of SM content surrounding breast cancer, content promoting and endorsing alternative treatments for breast cancer reached an estimated >55M people. Most content promotes treatments that are (1) not guideline concordant and/or endorsed by oncology experts, and (2) have no supporting scientific evidence of safety or efficacy for breast cancer. Our data suggest the bulk of content promoting such treatments may be from automated accounts or “bots.” Given the wide-reaching impact of these SM posts, these findings highlight a need for information about evidence-based treatments in SM discussions, a need to equip breast cancer advocates and oncology providers to have such discussions with breast cancer patients, and a need for policies to prevent misinformation about breast cancer treatment on SM.
Presentation numberPS5-10-02
Evaluating the impact of educational materials about biomarker testingfor patients with metastatic breast cancer
Theresa Petee, Living Beyond Breast Cancer, Bala Cynwyd, PA
T. Petee1, A. Dedmon1, K. Martin Gonzalez2, C. Koffke1, A. Leader3, J. Sachs4; 1Outreach and Educational Programs, Living Beyond Breast Cancer, Bala Cynwyd, PA, 2Center for Connected Care, Thomas Jefferson University, Philadelphia, PA, 3Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, PA, 4Chief Executive Officer, Living Beyond Breast Cancer, Bala Cynwyd, PA.
Background: As part of the No One Missed Campaign, Living Beyond Breast Cancer created educational materials about the importance of biomarker testing for patients with metastatic breast cancer (MBC). Materials were created in English and culturally adapted to Spanish with input from Spanish-speaking patients. We evaluated the educational materials on whether they increased patient knowledge, attitudes, and the likelihood to speak to their doctor, family members, or other MBC patients about biomarker testing.Methods: Patients living with MBC completed an online survey that consisted of answering questions before (baseline survey) and after (endpoint survey) viewing the newly created LBBC educational materials. The surveys and the materials were available in English and Spanish. Survey data was analyzed using descriptive frequencies and mean scores to compare baseline and endpoint results. Results: Twenty-nine participants living with MBC completed the study. Twenty-three participants (79%) completed the study in English while 6 (21%) completed it in Spanish. In total, 60% had been diagnosed with MBC within the past 3 years and 90% were still in treatment. Most (76%) had heard of biomarker testing and 45% had biomarker testing since being diagnosed with MBC. At baseline, 46% of participants strongly agreed that biomarker testing was important, which increased to 61% of participants at endpoint. At baseline, 36% of participants strongly agreed that biomarker testing will improve their cancer journey, which increased to 54% at endpoint. At baseline, 21% of participants strongly agreed that they would talk with their doctor about biomarker testing, compared to 36% at endpoint. Conclusion: The educational materials helped MBC patients learn about biomarker testing, as well as increase positive attitudes towards biomarker testing and the likelihood that patients will speak with their doctor about biomarker testing.
Presentation numberPS5-10-03
The Influence of Sociodemographic Factors on Barriers to Breast Cancer Screening
Alice Barros Câmara, University of São Paulo, São Paulo, Brazil
A. B. Câmara1, C. Luizaga1, L. Cury2, C. Carlos3, R. López4, A. Carvalho5, B. Partha5, W. Victor2; 1School of Public Health, University of São Paulo, São Paulo, BRAZIL, 2Conecta-SP project, Oncocentro Foundation of São Paulo, São Paulo, BRAZIL, 3Statistics, Federal University of Juiz de Fora, Juiz de Fora, BRAZIL, 4Statistics, Cancer Institute of the State of São Paulo, São Paulo, BRAZIL, 5Early Detection, Prevention & Infections Branch, Internacional Agency for Research on Cancer/World Health Organization, Lyon, FRANCE.
Background: Identifying barriers to cancer screening is essential for developing effective strategies to enhance organized screening programs. Aims: This study aims to investigate the impact of sociodemographic factors on barriers to breast cancer screening in the state of São Paulo, a region marked by significant socioeconomic disparities. Methods: A cross-sectional study was conducted using a representative sample of women aged 50 to 69 years, who were users of the Brazilian Health System. Data were gathered through a semi-structured questionnaire designed to assess sociodemographic factors and barriers to breast cancer screening. The sociodemographic factors analyzed as independent variables included age group (50-59; 60-69); education (illiterate; elementary; high school; college graduate); income relative to the minimum wage (<1 minimum wage; 1-2 minimum wages; ≥2 minimum wages); skin color (white; brown; black; yellow); employment status (employed; unemployed); type of employment (domestic work; private sector; public sector; employer; self-employed; unpaid worker); municipality of residence (Metropolitan Region of São Paulo; countryside 1; countryside 2); presence of chronic disease (yes; no); and health insurance (yes; no). Barriers to breast cancer screening were analyzed as dependent variables, grouped into six personal barriers (fear of diagnosis, embarrassment, other priorities, work schedule, pain, and forgetting to schedule) and four system-related barriers (difficulty scheduling the test, long wait time, distance to the screening facility, and difficulty obtaining results). These barriers were assessed through a 5-point Likert scale and logistic regression models were used to examine the relationships between the sociodemographic factors and cancer screening barriers. Results: The most common personal barrier to breast cancer screening was pain during mammography (59%), while the primary system-related barrier was long waiting times (44%). Regarding individual barriers, women with chronic diseases were more likely to fear discovering a serious issue (OR: 3.01; 95% CI 1.2-7.5). Residing in the metropolis was associated with higher odds of embarrassment (OR: 4.7; 95% CI 3.0-7.2) but showed an inverse relationship with pain (OR: 0.3; 95% CI 0.0-0.5). Women with brown and black skin were more likely to feel embarrassed during examination (OR: 2.5; 95% CI 1.2-3.6), while being employed was linked to forgetting to schedule the exam (OR: 3.3; 95% CI 2.1-5.9). Regarding system-related barriers, residing in the metropolis was associated with greater difficulty in scheduling a mammogram (OR: 2.7; 95% CI 1.3-5.8). Furthermore, employment status, having only elementary education, and being brown or black were linked to longer waiting times for results (OR: 3.0; 95% CI 1.5-7.1, OR: 2.9; 95% CI 1.3-6.3, and OR: 2.2; 95% CI 1.1-3.9). Conversely, employment status showed an inverse association with difficulty in scheduling a mammogram (OR: 0.4; 95% CI 0.1-0.7).Conclusion: Barriers to breast cancer screening differ based on sociodemographic factors. Brown or black skin color, residence in the metropolis, employment status, and chronic diseases were predictors of women-related barriers, while brown or black skin color, lower education levels, employment status, and living in the metropolis were predictors of system-related barriers. To improve mammography coverage and successfully implement an organized breast cancer screening program, it is essential to engage healthcare managers in developing key strategies to overcome these barriers. Collaborative efforts are crucial to ensure equitable access to screening and reduce disparities in breast cancer outcomes. These findings offer valuable insights for guiding the implementation of organized breast cancer screening programs in middle-income settings.
Presentation numberPS5-10-04
Willingness to participate in a trial comparing standard genetic counseling versus genetic counseling with personalized cancer risk estimates in patients with Li-Fraumeni syndrome
Ashley H Woodson, MD Anderson Cancer Center, Houston, TX
A. H. Woodson1, J. L. Corredor1, J. Voller1, J. Casey1, S. Green2, S. Shanker1, S. K. Peterson3, W. Wang4, B. K. Arun1; 1Clinical Cancer Genetics, MD Anderson Cancer Center, Houston, TX, 2Center for Health Promotion and Prevention Research, The University of Texas Health Science Center at Houston, Houston, TX, 3Behavioral Sciences, MD Anderson Cancer Center, Houston, TX, 4Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, TX.
Background: Li-Fraumeni Syndrome (LFS), due to germline pathogenic/likely pathogenic variants (PV/LPV) in TP53, is an inherited cancer syndrome with elevated risks for early-onset breast cancer, sarcomas, brain tumors, leukemia, or a combination of multiple cancers. Unlike other hereditary cancer syndromes with personalized risk assessment tools like BRCAPRO™ or MMRPRO™ (Berry, Iversen et al. 2002, Chen, Wang et al. 2006) that are tailored by variables specific to the individual, no validated models currently exist for Li-Fraumeni Syndrome (LFS). As a result, standard genetic counseling (GC) for individuals at risk for, or diagnosed with, LFS currently include only generalized lifetime risk predictions for cancer risk. This study aims to understand patients’ willingness to participate in a randomized trial comparing standard GC practice to personalized GC via a risk model in development known as LFSPRO (Peng, Bojadzieva et al. 2017, Nguyen, Dodd-Eaton, Corredor et al. 2024, Nguyen, Dodd-Eaton, Peng et al. 2024). LFSPRO intends to estimate the likelihood of a proband to test positive for LFS and provide cancer-specific risks of a first and second primary cancer based on personal and family history details. Methods: English-speaking individuals 15 years of age or older, or the parent/guardian of those under age 15, that underwent GC for TP53 genetic testing, and those that tested positive for a PV/LPV in TP53, were eligible to participate. Those eligible were invited by secure email via their medical record to complete the questionnaire. A hypothetical clinical trial scenario in which patients are randomized to receive one of two types of post-disclosure GC approaches was provided. Approach one was standard GC defined as generic risk predictions associated with TP53 PV/LPV and routine surveillance/management recommendations. Approach two was GC informed by LFSPRO data that requires personal and family history details to predict their specific cancer risks. The questionnaire obtained additional demographic and clinical characteristic details and perceived benefits and barriers to research participation. Results: Our initial analysis shows that the return questionnaire rate for invited participants was 17.9% (65/364). Most of the participants strongly agreed or agreed to participate in the hypothetical trial (81.5%, 53/65), additionally, 76.9% (50/65) would prefer to receive personalized GC. While only 7.7% (5/65) reported having previously participated in a clinical trial, most were interested in participating in research studies (76.9%, 50/65), and even further, 66.2% (43/65) strongly disagreed or disagreed with feeling uncomfortable being randomly assigned as participants to different study groups. A review of open responses to why individuals may want to participate identified four common themes: 43.5% (20/46) had a desire to help others with LFS, 39.1% (18/46) wanted additional information to help their own health management, 34.8% (16/46) wanted to improve research and knowledge about LFS, and 32.6% (15/46) wanted to help their own family. Conclusion: Despite the low response rate, individuals in this study report interest and willingness to participate in a hypothetical randomized trial comparing standard GC and personalized GC for LFS to further the research and understanding of how to use this information in a practical way. Funding: This research is supported by the Cancer Research and Prevention Institute of Texas (RP200383).
Presentation numberPS5-10-05
Implementation of strategies across sectors to reduce breast cancer in Greater Houston
Rosalind S Bello, The University of Texas MD Anderson Cancer Center, Houston, TX
M. E. Brown1, R. S. Bello1, C. L. Roberson2, K. Oestman3, R. Rechis1, L. McNeill1; 1Cancer Prevention and Control Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Principal, Daybreak Consulting, LLC., Las Vegas, NV.
Most recently available data indicates promising improvements in rates of breast cancer screening and diagnosis across all populations of women. However, there is still a persistent disparate outcome in breast cancer mortality affecting Black women in Harris County, Texas. These elevated risks are primarily attributed to non-medical drivers of health, such as racism, living and working conditions, structural barriers to care, complex healthcare systems, and cost. Launched in 2023, the Texas Health Equity Alliance for Breast Cancer (THEAL) is a multi-sector collaboration focused on reducing breast cancer disparities in Greater Houston guided by the principles of cross-sector collaboration, system strengthening, and aligned multilateral impact. The goal of THEAL is to decrease the breast cancer mortality gap by 15% and reduce significant barriers faced by Black women across the cancer care continuum over the next decade in Harris County. THEAL builds upon the expertise of The University of Texas MD Anderson Cancer Center’s Health Disparities Research department in addressing the determinants of cancer disparities while leveraging the Cancer Prevention and Control Platform’s experience in leading cross-sector collaboratives for community impact. The initiative structure includes a backbone team of seven organizations and a Steering Committee of 20+ organizations who foster collaboration, support implementation, evaluation, and sustainability. The first year of the initiative was dedicated to coalition building, aligning around a shared vision, and a facilitated competitive and collaborative request for proposals process. In the second year of the initiative and the first year of implementation (2024-2025), eight collaborating organizations were funded to implement seven evidence-based interventions (e.g., patient education, patient navigation and financial assistance) aimed at improving outcomes for Black women across the cancer care continuum. All collaborating organizations submitted quarterly reports to document progress toward set objectives and specific shared measures. Over the first 9 months of implementation, 2,187 clients have been reached by a patient navigator with 967 receiving direct services; 2,143 breast cancer screenings have been completed; 98 clients have received financial assistance; 1,358 people have attended 38 events or classes and of the 115 participants who have completed post class/event surveys, 100% have indicated self-reported knowledge and confidence gained. All the collaborating organizations have demonstrated a commitment to the THEAL goals and are collectively reaching and impacting women across Houston through programs in the community and quality improvement programs in clinics.
Presentation numberPS5-10-06
Evaluating the Increasing Proportion of Breast Cancer among Patients Aged 21 to 30 Since Year 2016
Brianna Cattelino, Drexel University College of Medicine, Philadelphia, PA
B. Cattelino1, J. Yan1, T. McKee2, S. Yacoub3, M. Shriner4, R. Kashyap4; 1Medicine, Drexel University College of Medicine, Philadelphia, PA, 2Surgical Oncology, Wellspan Health, Gettysburg, PA, 3Radiation Oncology, Wellspan Health, Gettysburg, PA, 4Research, Wellspan Health, York, PA.
Background: Recent reports have demonstrated that breast cancer is increasing in women less than age 50. This univariate analysis looks specifically at patients aged 21-30 to evaluate if the proportion of breast cancer is increasing disproportionately in this age group. Method: We utilized data from TriNetX US Collaborative Network, which collates clinical records across 70 healthcare organizations (HCOs). The medical records of over 52 million patients between January 2016 and December 2024 were queried. Records were included in a year-stratified count of breast cancer cases if they contained either the Oncology C50 code for breast cancer or ICD-10 diagnosis code “malignant neoplasm of breast.” We then determined the proportion of total breast cancer cases among 5 age groups (</=20, 21-30, 31-40, 41-50, >50). Chi-square tests were calculated between each year for patients aged 21-30 to assess if the proportion of breast cancer cases in this group has seen an increase year-over-year. Results: Both the absolute number of breast cancer cases in patients aged 21-30 and the proportion of breast cancer in this age group out of total cases increased between 2016 and 2024 (197,313 to 291,750 cases and 0.04% to 0.30%, respectively). There is a significant increase from the baseline year of 2016 in the proportion of cases amongst 21–30-year-olds beginning in 2019 (Diff = 0.0004, X2 = 33.451, p<0.001), and every year after (2020: Diff = 0.0006, X2 = 58.572, p<0.001; 2021: Diff = 0.001, X2 = 127.267, p<0.001; 2022: Diff = 0.0013, X2 = 209.035 , p<0.001; 2023: Diff = 0.002, X2 = 381.962, p<0.001; 2024: Diff = 0.297, p<0.001). (Table 1.) The difference in proportion positives between subsequent years was not significant until 2022-2023. Conclusion: These results demonstrate that breast cancer cases in patients aged 21-30 comprise a growing proportion of the total population. Providers should also be aware of this trend and consider initiating earlier conversations about breast cancer prevention in younger patients, for whom routine screening is not recommended.
| Year | BCage<20 | BCage21-30 | BCage31-40 | BCage41-50 | BCage>50 | TotalBreastCancer | Group# | %ofBCamong21-30 |
| 2016 | 34 | 76 | 1285 | 7849 | 188069 | 197313 | 1 | 0.04 |
| 2017 | 24 | 100 | 1906 | 10223 | 224438 | 224438 | 2 | 0.04 |
| 2018 | 26 | 140 | 2463 | 12286 | 223796 | 238711 | 3 | 0.06 |
| 2019 | 29 | 203 | 3209 | 14614 | 236500 | 254555 | 4 | 0.08 |
| 2020 | 22 | 241 | 3879 | 16280 | 227767 | 248189 | 5 | 0.10 |
| 2021 | 32 | 356 | 4788 | 19691 | 245601 | 270468 | 6 | 0.13 |
| 2022 | 46 | 482 | 6021 | 23388 | 264322 | 294259 | 7 | 0.16 |
| 2023 | 44 | 689 | 7246 | 27102 | 264014 | 299095 | 8 | 0.23 |
| 2024 | 55 | 867 | 8145 | 28879 | 253804 | 291750 | 9 | 0.30 |
Presentation numberPS5-10-08
Advance-bc: a prospective single-center registry to capture real-world outcomes in advanced breast cancer
Abbey J Kaler, The University of Texas MD Anderson Cancer Center, Houston, TX
A. J. Kaler, A. Singareeka, G. Kirklin, A. Anderson, J. Harris, T. Jacobsen, B. Lim; Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Advanced Breast Cancer (ABC) remains a leading cause of cancer mortality among United States women, yet real world outcomes and quality of life data remain underreported. Population-level studies are often limited by incomplete data capture and lack of quality-of-life variables. We present an updated, comprehensive analysis of real-world clinical outcomes that integrates recent therapeutic advances and key social determinants of health. Methods: Data from 361 MD Anderson patients was summarized as counts (%) for categorical variables. Group differences were assessed using χ²/Fisher’s exact tests and t-tests/ANOVA or Wilcoxon/Kruskal-Wallis, as appropriate. Comparisons between subgroups utilized chi-square tests. Overall and progression-free survival (OS and PFS) was estimated with Kaplan-Meier curves and compared by log-rank tests. Cox proportional-hazards models—adjusting for age, subtype, stage, ECOG status, therapy line, and any variable with univariate P < 0.10—were used to identify independent predictors; proportional-hazards assumptions were tested. Missing data <10 % was handled by complete cases; others with multiple imputation. All two-sided analyses (α = 0.05) were conducted in R 4.4 and independently validated. Results: Of 361 patients, 99% (n = 360) were female, 1% male (n=1). Median age was 62 (range: <49 = 19%, 50-69 = 51%, ≥70 = 27%). Most were married (73%), and 54% lived within 150 miles of the center. The cohort was 81% White, 9% Black, 6% Asian, and 5% Hispanic. Subtype classifications were 70% HR+/HER-, 19% TNBC, and 11% HER2+. At diagnosis, 36% presented with de novo stage IV disease. Metastatic sites included bone (47%), liver (19%), lung (19%), and lymph nodes (15%). At last assessment, 75% had an ECOG of 0 or 1. Genomic data was available for 13%. Advance-care-planning notes were present for 32%, and 40% participated in an interventional clinical trial since their ABC diagnosis. In multivariate Cox analysis, younger age predicted worse survival (HR 0.97 per year, p = 0.003). Genomic testing trended toward improved outcomes (HR 0.50, p = 0.35). ER, PR, and stage were not independently prognostic. Conclusions: This analysis highlights key clinical and demographic features and survival outcomes in a real-world ABC cohort. However, structured quality-of-life, toxicity, and patient-reported outcomes—essential to modern ABC care—remain largely absent from standard EHR fields. Many relevant data elements reside in unstructured text, limiting extraction. Future work will leverage large language models (LLMs) to extract and analyze these data at scale. Additionally, the single-center design and demographic homogeneity limit generalizability. To improve representation and impact, we plan to expand the registry through partnerships with community hospitals.
Presentation numberPS5-10-09
Prognostic factor pregnancy associated breast cancer by assisted technical reproduction
Maxim Izquierdo, Hospital Universitario Dexeus, Barcelona, Spain
M. Izquierdo, S. Baulies, M. Gallardo, C. Ara, F. Fargas, F. Tresserra, R. Fabregas, P. Barri S; Gynecology, Hospital Universitario Dexeus, Barcelona, SPAIN.
Aims Study and assess the differences in the prognostic factors in pregnancy associated breast cancer by assisted technical reproduction and without . Methods Study 164 pregnancy associated breast cancer, 124 without assisted technical reproduction and 40 with assisted technical reproduction, treated in the same hospital. Analyze prognostic factors: estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2 , histological grade and ki67, of pregnancy associated breast cancer with and without assisted technical reproduction. Results Patients with pregnancy associated breast cancer without assisted technical reproductio: estrogen receptor is positive in 91 (73’38 %), negative in 33 (26’61 %) patients with assisted technical reproduction ,estrogen receptor is positive in 34 (94’44 %), negative in 2 (5’55%) patients. (p<0’05), ki67 20 in 57 (53’27%) patients without assisted technical reproduction, and ki67 20 in 9 (32’14%) patients with assisted technical reproduction (p<0’05). Progesterone receptor and HER2 receptor there was no difference between with and without assisted tecnical reproduction. (p=ns). Conclusion. Pregnancy associated breast cancer by assisted technical reproduction influence estrogen receptor and Ki67, and do not influence progesterone receptor and HER2 receptor
Presentation numberPS5-10-10
“You have to work with me”: a thematic analysis of patient experiences in breast cancer treatment decision-making
Molly E Boll, University of Chicago, Chicago, IL
M. E. Boll1, L. J. Schmitt2, L. Rabinovich3, H. M. Earl4, M. E. Peek5, A. Wesevich5; 1Biological Sciences Division, University of Chicago, Chicago, IL, 2Center for Personalized Therapeutics, University of Chicago, Chicago, IL, 3The USC Brain Health Observatory, University of Southern California, Los Angeles, CA, 4Department of Oncology, University of Cambridge, Cambridge, UNITED KINGDOM, 5Department of Medicine, University of Chicago, Chicago, IL.
Background: Shared decision-making has been increasingly incorporated into guidelines for breast cancer care. However, there has been little study on how patients with breast cancer are presented with options in their care and the level of involvement they prefer. When deciding between non-inferior options such as 6 versus 12 months of adjuvant trastuzumab, shared decision-making and elicitation of patient preferences are critical. In this study, we sought to understand patient communication preferences and their prior experiences with oncologists regarding decision-making in their breast cancer treatment. Methods: We recruited 20 people diagnosed with breast cancer in the past year from across the US to participate in semi-structured interviews regarding their cancer journeys, experiences with treatment decision-making, and communication with their oncologist. Participants were also presented with an infographic displaying the efficacy and toxicity of 6 versus 12 months of adjuvant trastuzumab for early-stage HER2-positive breast cancer to elicit feedback on the presentation of data as well as their prior experiences with oncologists sharing data in their treatment process. Interviews were transcribed and deidentified before being independently coded by two study team members using Dedoose. Discordant coding was reconciled for each transcript. Themes were identified via inductive thematic analysis. Results: Participants shared both positive and negative factors that contributed to their experiences of breast cancer treatment decision-making. Positive factors were patient engagement in care, patient-centered communication, support systems, and socioeconomic advantage. Negative factors were insufficient explanations, ineffective oncologist communication, higher degree of mental and physical treatment burdens, and systemic barriers to care. Participants frequently shared that oncologists did not present them with treatment options, with one participant stating, “Not a lot of this is my choice, you know? It’s just, ‘This is the treatment.’” There were various preferences regarding control in their care, ranging from one participant saying, “just give me whatever you need to make me feel better” to another sharing, “I feel like that’s my choice, so I should decide, but I do want the doctor to have ideas about what’s going on.” Participants also appreciated the infographic and wished a similar tool had been used in their own care to help them better engage in treatment decision-making conversations. All patients interviewed expressed a desire for more information in their breast cancer care. Conclusion: We identified factors that support or inhibit patient involvement in shared decision-making for breast cancer treatment. While patients varied in how much they wanted to be involved in final decisions, there was a consensus overall that patients wanted more information-sharing and more deliberation about treatment options, two core components of shared decision-making. Presenting efficacy and toxicity information as an infographic to patients with breast cancer may improve their ability to engage in shared decision-making, especially for non-inferior breast cancer treatment options.
Presentation numberPS5-10-11
Metachrone contralateral breast cancers: Differences in detection mode, Nottingham Prognostic Index, ER, PR and HER2
Elien Bloemen, KU Leuven, Leuven, Belgium
E. Bloemen1, A. Laenen2, S. Han3, C. Remmerie4, M. Van Houdt3, G. Floris5, M. Keupers5, C. Van Ongeval5, V. Celis5, H. De Boodt5, A. Coessens5, R. Prevos5, Y. Van Herck6, F. Derouane6, C. Weltens7, H. Janssen7, A. Baten7, J. Verhoeven7, C. Desmedt8, K. Van Baelen3, A. Smeets9, I. Nevelsteen9, H. Wildiers6, N. Patrick3; 1Biomedical Sciensces, KU Leuven, Leuven, BELGIUM, 2Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, BELGIUM, 3Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, BELGIUM, 4Oncology, University Hospitals Leuven, Leuven, BELGIUM, 5Department of Imaging and Pathology, University Hospitals Leuven, Leuven, BELGIUM, 6Department of General Medical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 7Department of Radiotherapy, University Hospitals Leuven, Leuven, BELGIUM, 8Laboratory for Translational Breast Cancer Research, KU Leuven, Leuven, BELGIUM, 9Department of Surgical Oncology, University Hospitals Leuven, Leuven, BELGIUM.
Background: Risk factors for contralateral (CL) breast cancers are well studied, not the tumor characteristics if metachrone diagnosed. We studied mode of detection, tumor characteristics and similarities in receptor status in consecutive women with a metachrone CL breast cancer. Methods: We performed a retrospective study comparing demographics, tumor characteristics (Nottingham Prognostic Index) and histologic type, ER, PR (≥1% is pos) and HER2 status (amplified is pos) in women with a > 3m diagnosed CL breast cancer in UZ Leuven between 2000-2023. We also evaluated patient-related variables which could influence changes in tumor type between lesion 1 (prior tumor) and lesion 2 (metachrone tumor). The Mann-Whitney U test was used for continuous variables, the Chi² test for categoric variables, the Wilcoxon signed rank test for coupled data and Logistic regression for variables predicting changes between lesions. The analyzed variables were : histological subtype (lobular (ILA) vs non-lobular (non-ILA)), time between two diagnosis, age and BMI at diagnosis, age at menarche, number of pregnancies, age at1st term pregnancy, menopausal status, familial risk, germline carrier status, use of hormones. Statistical analysis were done using SAS System for Windows version 9.4. Significance was defined if p<0.05. Results: We selected 289 cases in accordance to our inclusion criteria. Mean age was 54.9y (SD 10.5y); mean BMI 25.8 kg/m² (SD 5 kg/m²); age menarche 13.3y (SD 1.6y); 1st full term pregnancy 26.2 yrs (SD 5.1y)). There was a mean delay of 8.9 yrs (SD 5.2y) between diagnosis of lesion 1 vs 2. Lesion 2 was more likely screen-detected (82.3% vs 41.5%; P <0.001) with a lower NPI (4.14 vs 3.76; P=0.029). The histologic subtype (Table) of lesion 2 was predicted by that of lesion 1 (P= 0.014). The ER-status of the CL tumor was significantly predicted by the ER-status of the 1st; CL tumor was ER-neg in 32.4% of 68 patients with an initial ER-neg lesion while the ER-status in the CL tumor was ER-pos in 85.5% of 221 patients with an initial ER-pos lesion (P < 0.001); change in ER-status was affected by later age at menarche (OR 1.239; 95% CI 1.030; 1.490). Similar findings were observed for PR-status with a P= 0.034 but none of the tested variables affected these changes. The change in HER-2 status was not significant; 88.9% of 243 cases were HER2 neg if initial HER2 neg and 17.1% of 35 cases were HER2 pos if initial HER2 pos; P= 0.302.[table:Columns refer to histologic subtype in tumor 1; rows in tumor 2 . Reported P-values are two-sided] Conclusion: CL tumors can appear beyond 10 years independently of initial hormonal receptor status. They are more likely screen-detected with a better NPI prognosis. Although histological subtype and receptor status change in a large proportion, lobular type and hormonal receptor status predicted respectively the histological subtype and receptor status of CL lesions.
| Variable | Statistic | non-ILA | ILA | P-Value |
| type_T2 | ||||
| non-ILA | n/N(%) | 213/249(85.54%) | 28/40(70.00%) | 0.014 |
| ILA | n/N(%) | 36/249(14.46%) | 12/40(30.00%) |
Presentation numberPS5-10-13
Exploring the Role of Social Media in Shaping Breast Cancer Treatment Decisions Among Women
Tiare Africa Piñeiro, IAF, Buenos Aires, Argentina
T. A. Piñeiro1, V. Constanzo2, A. Nervo2, S. García1, F. Colo3, M. Daniel1, L. Cosaka1; 1Radiology, IAF, Buenos Aires, ARGENTINA, 2Oncology, IAF, Buenos Aires, ARGENTINA, 3Mastology, IAF, Buenos Aires, ARGENTINA.
Exploring the Role of Social Media in Shaping Breast Cancer Treatment Decisions Among Women Breast cancer is a complex pathology and treatments nowadays is not a one size fits all. Social media has emerged as a prominent and widely used platform for sharing health-related information and also, one of the main sources for bread treatment information in patients. The objective of this study is to evaluate the role of social media in shaping women’s knowledge, trust, and engagement with breast cancer treatment information, and to assess its influence on treatment-related discussions with healthcare providers Methods: To evaluate this, a cross-sectional survey was conducted in July of 2025 through an Instagram breast health account using Instagram survey tools. The survey included questions on demographics, engagement with breast cancer treatment-related content, trust in health information shared on social media, and whether any information encountered on social media influenced discussions or decisions made with their doctor regarding treatment. Results: A total of 1026 respondents participated in the survey, all of whom identified as women, with the majority (43%) aged between 41 and 50 years. From this group of patients 94% participants reported searching for breast cancer-related information online, with Instagram identified as the primary source (82%), followed by other internet sites. Trust in online health information varied, largely depending on the credibility of the source (79%) and the platform on which the information was published. Age differences were observed in levels of trust, with younger (20-30 years old) women more likely to trust online information (25%) compared to older age groups. Women who reported higher trust in social media content demonstrated greater engagement with breast cancer treatment-related information and were more likely to report that such information influenced discussions and decision-making with their physicians (17%). Although the majority of women (73%) reported that online information did not alter their treatment plans or decision-making, a subset (10%)—despite not fully trusting all online content—expressed a willingness to discuss the information they encountered with their healthcare providers. Conclusion:In the current digital age, information is increasingly disseminated through non-traditional channels, with social media platforms significantly enhancing patient access to treatment-related content. However, the prevalence of misinformation on these platforms presents a challenge, as regulating the accuracy of shared content remains difficult. This underscores the critical importance of trust in the source of information. Healthcare providers should be aware of this trend and remain open to discussing online information with their patients to support informed decision-making.
Presentation numberPS5-10-14
How Reliable is YouTube Content Targeting Arabic-Speaking Breast Cancer Patients Regarding Post-Mastectomy Reconstruction?
Marya Alsuhaibani, McGill University, Montreal, QC, Canada
H. Alotaibi1, A. Alsulaimani2, M. Alsuhaibani1, E. Belzile3, N. Morena4, A. Meguerditchian1; 1General Surgery, McGill University, Montreal, QC, CANADA, 2General Surgery, Taif University, Saudi Arabia, SAUDI ARABIA, 3Research Centre, St Mary’s, Montreal, QC, CANADA, 4Department of Art History and Communication Studies, McGill University, Montreal, QC, CANADA.
Background: The dynamic, easily accessible nature of social media platforms, particularly YouTube (YT), has revolutionized the way breast cancer patients seek and process information. This study evaluates the quality of YT videos addressing post-mastectomy reconstruction (PMR) that target Arabic-speaking women (ASW), a heavily digitally connected segment of society that may experience challenges in accessing mainstream medical information in Arabic Language (AL). Methods: YT was searched incognito, using the terms: “post mastectomy reconstruction” in (AL) in July 2023. Data collected included upload date, video length, source, sponsorship, and viewer engagement. Thematic content analysis was performed by physician reviewers. Understandability and actionability were assessed using the Patient Education Materials Assessment Tool (PEMAT A/V), and video quality was evaluated using the DISCERN tool. Cluster analysis used to categorize the videos based on quality. Results: A total of 109 videos were included. The average video length was 7 minutes (SD 10.2). Most originated from North Africa (55%), followed by the Persian Gulf (33%) and the Mediterranean region (11%). Videos were predominantly created by individuals (57.8%) and were natively in Arabic (93.6%). Mean views per video were 8,052. Most videos were information-based (92.6%), aimed at the public (82%), and commercially sponsored (62%). PMBR visuals appeared in 31.1% of videos. Common themes included awareness (82.6%), appearance (68.8%), gender identity (57.8%), and treatment (45.0%); sexuality and fertility were least discussed (4.6%). While 99% encouraged PMR, only 34% offered actionable guidance. Mean PEMAT scores were 61.3% for understandability and 20.2% for actionability. The mean DISCERN score was 2.6/5, with only 43.1% of videos rated as recommendable by Arabic-speaking healthcare professionals. Cluster analysis showed only 19.3% of videos were high quality. High-quality videos were significantly longer (p=0.002), less commercially sponsored (p=0.001), more likely to be information- and advice-based, and more likely to include sensitive themes such as sexuality and fertility. Viewer engagement was not correlated with video quality. Conclusion:Arabic-language PMR content on YT has moderate quality, easily understandable but lacks actionability and patient-centered depth. Videos are highly sponsored and questionably representative of patients’ and women’s perspective. Improving content quality is essential to support informed decision-making among ASW exploring PMR.
Presentation numberPS5-10-15
Comparing User Experience Between AI-powered Thermal Test and Mammography: Results from a Single Center Study
Pavani Chalasani, George Washington Cancer Center, Washington, DC
S. S. Kraus, A. Moreno, A. Carrier, P. Zimemrman, D. Roe, P. Chalasani; Division of Hematology and Oncology, George Washington Cancer Center, Washington, DC.
Background: Mammography remains the gold standard for breast cancer screening but is associated with notable discomfort due to breast compression and concerns over ionizing radiation. Prior studies indicate that 25-46% of women report moderate to severe pain, and anxiety related to mammography as a key reason for avoidance. Additionally, perceived invasiveness and privacy concerns can deter women from regular screening. Thermalytix is a novel, non-contact, radiation-free screening modality combining thermal imaging with artificial intelligence (AI)-based analysis. This privacy-preserving approach was developed to enhance user comfort and reduce barriers to participation. Thermalytix has been shown earlier to have good sensitivity and specificity when compared with standard imaging methods [1,2]. We report a single institution comparative user experience between Thermalytix and standard mammography, as a post hoc analysis of ClinicalTrials.gov Identifier: NCT05650086. Methods: In a single-center prospective study at the University of Arizona Cancer Center, 115 women underwent both mammography and Thermalytix scans. Following both procedures, participants completed a standardized survey evaluating three experience parameters on a 1-5 Likert scale (5 = most favorable), (i) Comfort level during the procedure (ii) Level of anxiety experienced and (iii) Perceived time taken for the screening. Participants were also asked about their overall preference between the two modalities and any reason for preferring one test over the other. Results: (A) Overall Response:A total of 115 women participated. Of these,
- 12 women did not complete the survey
- 25 gave similar scores (within 10%) for both tests
- 78 gave clear preferences, wherein 73 favored Thermalytix, 5 favored mammography.
(B) Quantitative Ratings:Average scores on a 1-5 Likert scale (5 = most favorable) for key experience parameters were:
- Comfort: Thermalytix 4.3, Mammography 2.4
- Anxiety: Thermalytix 4.4, Mammography 3.3
- Time Perception: Thermalytix 4.5, Mammography 4.1
We computed Pareto Average as a sum of individual scores and used that as a consolidated preference, and the Pareto Average of Thermalytix was 10.79 (range: 9-15) while Mammography got 7.6 (range: 4-10) (C) Qualitative Insights: Among 66 participants with text comments in their response, the following were the insights:
- 57 (86.4%) explicitly preferred Thermalytix over Mammography
- 5 preferred mammography, as they were unsure about the results of new AI-based test
- 4 women had equivocal comments about both the tests
- Top reasons from women who elaborated their preference for Thermalytix included:
- No pain (17), Comfort (18), No compression (11), Radiation-free (6), Easy/relaxed (5), Private/no-touch (4)
- 3 Thermalytix-preferring women noted it felt cold
Conclusion:In this small US Based cohort, Thermalytix has shown numerically better ratings in comfort and lower anxiety with the test. It compared similar in terms of time needed to do the testing. Majority of participants seemed to prefer a no-contact, non-ionizing radiation approach to breast screening. References: 1. Bansal R et al, A prospective evaluation of breast thermography enhanced by a novel machine learning technique for screening breast abnormalities in a general population, Frontiers in Artificial Intelligence, 2022 [ClinicalTrials.gov Identifier: NCT04688086] 2. Kakileti, S. T et al, Observational study to evaluate the clinical efficacy of thermalytix for detecting breast cancer in symptomatic and asymptomatic women. JCO Glob Oncol. 2020
Presentation numberPS5-10-16
Physical Therapy and Oncology Rehabilitation Referral and Enrollment Patterns among Breast Cancer Patients
Mary Imboden, Providence St Vincent Medical Center, Portland, OR
M. Imboden1, H. Li1, E. Koltner1, C. Murphy1, M. Layoun2, D. Page3; 1Center for Cardiovascular Analytics, Research, and Data Science, Providence St Vincent Medical Center, Portland, OR, 2Providence Heart Institute, Providence St Vincent Medical Center, Portland, OR, 3Providence Cancer Institute, Providence Portland Medical Center, Portland, OR.
Background: Supervised exercise interventions improve disease-specific and overall survival in colon cancer and may offer similar benefits in breast cancer (BC), with prior research demonstrating improvements in chemotherapy-related side effects, cardiorespiratory fitness and quality of life. Oncology rehabilitation (OR) offers supervised and tailored exercise interventions and are routinely available to BC patients via physical therapy (PT) referral, however these programs may be underutilized. We sought to characterize patterns of PT/OR referral, utilization, and clinical benefit among BC patients. Methods: A retrospective cross-sectional analysis of 53,972 BC patients seen within the Providence St. Joseph Health System between October 2015 and March 2025 was performed. Data were abstracted from patient electronic health records, and descriptive statistics were performed to summarize baseline characteristics of the participants. Random-intercept multilevel logistic regression models, including region/hospital as a random effect, were used to assess predictors of PT/OR referral and enrollment. Baseline and Follow-up exercise tests assessing cardiorespiratory fitness (VO2max) were used to assess the clinical benefit of PT/OR. Results: Comparisons between BC patients that were referred to and enrolled in PT/OR versus not are provided in the table. Forty-one percent of the overall study population was referred (n=22,061) to PT. In multivariate analysis, significant predictors of PT referral included younger age (odds ratio: 0.99 [95% CI: 0.99 – 1.00]), higher BMI (odds ratio: 1.01 [95% CI: 1.01 – 1.02]), hypercholesterolemia (Odds ratio: 1.45 [95% CI: 1.33 – 1.58]), and receipt of chemotherapy (Odds ratio: 1.61 [95%CI: 1.47-1.77]). Among those referred, 13,059 (59%) enrolled and participated. Enrolled patients attended an average of 16 ± 15 appointments, with 25% attending ≥ 20 sessions. 2,051 patients (16%) underwent an exercise test, predicted by older age (Odds ratio: 1.03, 95% CI: 1.02 – 1.03) and higher BMI (Odds ratio: 1.03, 95% CI: 1.02 – 1.04). The average baseline VO2max was 14.1 ± 3.6 ml/kg/min. PT/OR participants experienced improvements in VO2max (0.8 ± 2.8), with those attending ≥ 20 sessions experiencing greater benefit (1.2 ± 3.7 ml/kg/min) compared to those attending <20 sessions (0.7± 2.2 ml/kg/min). Conclusion: OR is routinely available via PT referral but uncommonly utilized by BC patients. However, it may be an effective and feasible strategy to improve physical fitness, quality of life, and potentially clinical outcomes.
Presentation numberPS5-10-17
Barriers to Breast Cancer Screening from the Perspective of Primary Care Professionals
Alice Barros Câmara, University of São Paulo, São Paulo, Brazil
A. B. Câmara1, C. Luizaga1, L. Cury2, C. Contreras3, R. Lopes4, A. Carvalho5, P. Basu5, V. Wünsch Filho2; 1School of Public Health, University of São Paulo, São Paulo, BRAZIL, 2Conecta-SP project, Oncocentro Foundation of São Paulo, São Paulo, BRAZIL, 3Statistics, Federal University of Juiz de Fora, Juiz de Fora, BRAZIL, 4Statistics, Cancer Institute of the State of São Paulo, São Paulo, BRAZIL, 5Early Detection, Prevention & Infections Branch, Internacional Agency for Research on Cancer/World Health Organization, Lyon, FRANCE.
Background: Cancer screening consists of the early detection of breast cancer in asymptomatic populations, with the goal of reducing disease-related mortality.Aims: This study aims to assess the knowledge and barriers to breast cancer screening from the perspective of primary care professionals, investigating associations between professionals characteristics, knowledge, and these barriers, with the goal of developing effective strategies to improve the screening program in the state of São Paulo, Brazil—a region marked by significant socioeconomic disparities. Methods: This cross-sectional study included a sample of 50 Primary Health Centers (PHCs) in the state of São Paulo. A population survey was conducted with a representative sample of 401 primary care professionals. Collected data include professional’s knowledge and characteristics, and barriers to breast cancer screening. These barriers were assessed through a 5-point Likert scale and associated with professionals’ characteristics and knowledge using multinomial logistic regression models. Interviews were conducted using a semi-structured questionnaire designed with RedCap™ software. Analyses were performed using the SPSS v.29 software. Results: Among the professionals interviewed, 82.8% are women and 52.9% are under the age of 44. A majority (88.2%) were employed in the health sector, with 32.6% working in nursing and 18.8% in medicine, while 11.8% held administrative positions. Only 60% were familiar with the guidelines for breast cancer screening and 64% were aware of the law ensuring cancer diagnosis within 30 days. Knowledge levels ranged from 37% to 89%, with higher awareness observed among nurses. From the professionals perspective, the most significant obstacle to breast cancer screening was long waiting times to undergo mammograms, reported by 35% of respondents. Other notable challenges included difficulties in verifying whether the target population was up to date with mammograms (33%) and excessive demand at the PHC (32%). These barriers were mainly reported by nurses, physicians, and professionals working in the Metropolitan Region of São Paulo. Importantly, knowledge on breast cancer guidelines showed an inverse association with the difficulty in informing the target population about the mammographic screening and its importance, while knowledge on cancer diagnosis legislation was associated with perceptions about long waiting times for screening and diagnostic procedures. Notably, 73% of professionals agreed that the absence of a health information system for screen women with abnormal test results compromises the effectiveness of breast cancer screening programs.Conclusion: The implementation of an organized breast cancer screening program requires strategic actions, specially aimed at reducing the long waiting times and ensuring the target population is up to date with the screening exam. These strategies may be tailored according to the professional role and geographic region of primary care. Furthermore, ongoing training to enhance the knowledge of both healthcare and administrative personnel is essential for addressing barriers to breast cancer screening—particularly those related to delays in screening and diagnosis. A robust and well-integrated information system is also critical to support the effective screening and follow-up of the target population. Collectively, these strategies are being undertaken to guide the establishment of an organized, sustainable, and population-based breast cancer screening program in São Paulo, Brazil.
Presentation numberPS5-10-18
Trial in Progress: The Physical Activity Index (PAI) Feasibility Pilot Trial for Breast and Colon Cancer Survivors in North Carolina
Shirley Bluethmann, Wake Forest University School of Medicine, Winston-Salem, NC
S. Bluethmann1, J. Tooze2, J. Evans3, J. Katula4, K. Wood4, L. Hitinariansingh5, C. Crotts5, H. Klepin6, S. Zakrzewski1, R. Paluri6, K. Kadakia7, K. Ansley6; 1Div. Public Health Sciences, Dept of Social Sciences and Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, 2Div. Public Health Sciences, Dept of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, 3Div. Public Health Sciences, Department of Biostatistics and Data Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, 4Department of Health and Exercise Sciences, Wake Forest University, Winston-Salem, NC, 5Div of Geriatrics and Gerontology, Wake Forest University School of Medicine, Winston-Salem, NC, 6Division of Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 7Department of Gastrointestinal Cancer, Atrium Health Levine Cancer Institute, Charlotte, NC.
BACKGROUND The majority of the 18 million U.S. cancer survivors do not meet recommended levels of physical activity (PA) or limit sedentary behavior in daily life. Cancer survivors, especially breast cancer survivors (BCS) and colon cancer survivors (CCS), are particularly at risk of insufficient PA and prolonged sitting, contributing to increased physical impairments, symptom burden, and elevated cancer risk. Completion of cancer treatment may represent a “teachable moment” for lifestyle change, yet many survivors lack behavioral guidance from clinical teams. Validated tools to assess and counsel survivors on PA and sedentary behavior are limited. A simple screener—the Physical Activity Index (PAI)—collects information on PA, strength training, and sedentary behavior and may be useful in clinical settings for monitoring patient behaviors and offering specific, tailored recommendations. This research tests the feasibility of intervening with the PAI at the teachable moment, using the screener, coaching, and self-monitoring tools (e.g., activity tracker and PAI “report card”) to increase PA and reduce sedentary behavior. METHODS The intervention includes standard survivorship follow-up care, a PA assessment using the PAI screener (shared with the provider), and five remote coaching consultations with a certified exercise physiologist. Participants also receive resistance bands and a Garmin activity tracker for home-based self-monitoring. TRIAL DESIGN This is a two-arm, 1:1 randomized pilot trial with waitlist control to assess feasibility. We plan to recruit 20 participants (10 BCS, 10 colon cancer survivors), ages ≥50, stages 0-III, within three years of diagnosis and post-treatment. The pilot spans 12 months, with a six-month active intervention. Standardized study measures (e.g., weekly PA, sedentary time, physical function) will be collected at baseline, 3, 6, 9, and 12 months. Waitlist control participants will begin the intervention at month 6 after two assessments. All participants will complete five total research assessments over one year. STATISTICAL METHODS Feasibility will be assessed through recruitment, retention, acceptability, and adherence benchmarks. Secondary analyses will examine changes in PA and sedentary behavior to inform future trials. Descriptive statistics will summarize behavioral outcomes, participant demographics, and potential confounders (e.g., cancer type, treatment, medications) by group. The primary analytic goal is to estimate standard deviations (SDs) for future power calculations. Additional analyses will include mixed ANCOVA models (baseline-adjusted) to evaluate trajectories of PA and sedentary time over time. Acceptability will be measured via participant satisfaction scores (1-10); scores ≥7 will reflect acceptability. If feasible, this intervention may offer a scalable tool to support lifestyle behavior change and symptom management among cancer survivors—especially BCS—ultimately improving recovery, quality of life, and long-term survivorship outcomes. All study procedures were approved by the IRB at Atrium Health Wake Forest Baptist Comprehensive Cancer Center. This trial is registered at ClinicalTrials.gov (NCT06340503).
Presentation numberPS5-10-19
Breast cancer in young women at a private center in Chile
Maria Trinidad Esperanza Gonzalez, Clinica Alemana de Santiago, Santiago, Chile
M. E. Gonzalez, J. Camacho, C. Barriga, M. Bravo, P. Pincheira, M. Gallegos; Surgery, Clinica Alemana de Santiago, Santiago, CHILE.
IntroductionBreast cancer is the most frequently diagnosed cancer worldwide and the second leading cause of cancer-related death. While it predominantly affects women over 40, 5-7% of cases occur in those under 40 in developed countries. Despite its lower incidence in younger women, it remains the leading cause of cancer death in this group, underscoring the need to better understand its biology, treatment, and outcomes in younger patients. ObjectivesThis study aims to analyze the clinical and pathological characteristics of breast cancer cases treated at Clínica Alemana in Santiago, Chile, over the past decade, and to compare them with global data. Specific objectives include evaluating tumor biology, reason for consultation, type of surgery, recurrence patterns, and mortality. Methods and materialsRetrospective descriptive study of clinical presentation, tumor features, treatments, and fertility in 200 women aged ≤40 with breast cancer over 10 years at Clínica Alemana, Santiago, Chile. ResultsAmong the total cohort, 17.7% underwent fertility preservation prior to treatment. Adjuvant radiotherapy was administered to 58.9%, chemotherapy (neoadjuvant/adjuvant) to 40.6%, and hormone therapy to 79.1%—of whom 96.7% received Tamoxifen and 3.3% aromatase inhibitors. Recurrence occurred in 6.8%, with 38.5% local, 7.7% regional, and 53.8% distant metastases. At follow-up, 97.4% were alive; 5 deaths were due to metastatic disease. DiscussionThe distribution of tumor stages in our cohort is consistent with patterns observed in high-income countries, with 69% of cases diagnosed at stage I or II and 3% at the metastatic stage. Although breast cancer in women < 40 is generally associated with more aggressive subtypes, our cohort exhibited lower rates of HER2 (+) (18%) and triple-negative (8%) tumors compared to reported averages. Additionally, the 5-year cumulative incidence of distant metastases was notably lower in our population (4.7% vs. 18-24%), and the overall 5-year survival rate was higher (97.4% vs. 89-93%).
| 0 | 44 (23%) | ||
| I | 80 (42%) | ||
| II | 51 (27%) | ||
| III | 10 (5%) | ||
| IV | 7 (3%) |
| Luminal A | 56 (36.4%) |
| Luminal B | 48 (31.2%) |
| HER 2 (+) | 34 (22%) |
| Triple negative | 16 (10.4%) |
Presentation numberPS5-10-20
Invasive lobular breast cancer and surveillance: A patient experience survey study.
Mason Mitchell-Daniels, Lobular Breast Cancer Alliance Inc., White Horse Beach, MA
M. Mitchell-Daniels1, J. H. Axelrod2, T. Cushing2, L. Hutcheson3, M. Jochelson4, G. H. Joergensen2, T. Langdon2, J. A. Mouabbi5, R. Mukhtar6, B. F. Neilsen2, S. Paluch-Shimon7, L. Petitti2, D. Romer2; 1Research and Operations, Lobular Breast Cancer Alliance Inc., White Horse Beach, MA, 2Research Advocate, Lobular Breast Cancer Alliance Inc., White Horse Beach, MA, 3Director, Lobular Breast Cancer Alliance Inc., White Horse Beach, MA, 4Breastlink/RADNET, Breastlink/RADNET, Beverly Hills, CA, 5Department of Medical Breast Oncology, MD Anderson, Houston, TX, 6Breast Care Center, Bakar Precision Cancer Medicine Building, University California San Francisco, San Francisco, CA, 7Womens Health Center, Hadassah University Hospital, Jerusalem, ISRAEL.
Background: The Lobular Breast Cancer Alliance (LBCA) is a nonprofit patient advocacy group committed to raising awareness and promoting research on invasive lobular carcinoma (ILC), which is 15% of breast cancers and understudied. The hallmark of ILC, lack of E Cadherin, causes non-cohesive tumor growth. Consequently, imaging studies are less accurate for diagnosis, staging and monitoring for recurrence or progression. Patients’ concern about efficacy of surveillance was the impetus for this study. Methods: LBCA conducted an anonymous online survey in 12/2024 of respondents (RSPs) with ILC who had completed local disease treatment and RSPs living with metastatic ILC (mILC), distributed via LBCA’s newsletter, social media, and partners. The survey addressed surveillance and included a 10-point scaled question about fear of cancer recurrence or progression (FCR) (10 being high). Results: 1706 RSPs had ILC or ILC mixed type. 70% of RSPs were from the US, 11% UK, 6% Australia, 5% Canada, and 8% from 26 other countries. Mean age at diagnosis was 60. 88% of RSPs had had early (stage I-III) breast cancer (EBC) and 12% were living with mILC. 70% of RSPs had dense breasts. 44% of US-based RSPs and 13% of non-US RSPs reported having no breast imaging surveillance because they had double mastectomies. For those with EBC ILC having regular breast imaging, the most common surveillance plans included mammogram (US-based RSPs 92% vs. non-US-RSPs-87%), ultrasound (US-26% vs. non-US-38%), breast MRI (US-50% vs. non-US-26%), and contrast enhanced mammogram (CEM) (US-9% vs. non-US-7%). Surveillance including MRI and CEM was significantly (p = .05) associated with high confidence compared to no confidence. Of 37 (3% of 1496) EBC RSPs who indicated they had a local recurrence, only 28% indicated that their routine imaging test initially detected the recurrence. For those with mILC, the most common surveillance modalities included regular CT scans, bone scans, MRIs, FDG PETs, FES PETs, regular CBC, CMP, tumor marker, and some form of liquid biopsy. The reported use of FDG PET, FES PET, CMP and liquid biopsies were higher in US RSPs compared to non-US RSPs. Notably, 81% of EBC RSPs with high FCRs (8-10) were not sure or had no confidence their surveillance plan would detect recurrence, and 49% of RSPs with mLC with high FCRs lacked confidence their plan would detect a progression. Conclusions: Many women with ILC surveyed worldwide, reported high rates of fear of cancer recurrence/ progression, which is correlated with lack of confidence in surveillance plans. These findings align with our prior work showing low confidence in imaging ILC among breast imagers (Coffey, et al., Journal of Breast Imaging, 2024). More research is needed to identify most effective surveillance for ILC recurrence and progression.
Presentation numberPS5-10-21
Real-world patterns and preferences in the use of diagnostic imaging for breast cancer staging in Hispano-America: A multinational physician survey
William Armando Mantilla, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center, Bogota, Colombia
W. A. Mantilla1, M. A. Bravo1, V. Acosta-Marín2, A. M. Osorio1, C. Villarreal-Garza3, A. W. Mushtaq4, F. E. Petracci5, S. Cervera1, G. Santander6, A. Reyes-Morales7, D. U. Landaverde8, J. C. Samamé-Pérez-Vargas9, J. Moreno-Rios10, L. Gutiérrez11, B. Moreno-Jaime12, J. J. Caicedo1, S. Quintero1, A. M. Mejía1, H. Carranza1, S. X. Franco1; 1Breast cancer Functional Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center, Bogota, COLOMBIA, 2Breast cancer Unit, Centro Clínico de la Mama (CECLIM), Caracas, VENEZUELA, BOLIVARIAN REPUBLIC OF, 3Breast Cancer Center, Hospital Zambrano Hellion, Tecnologico de Monterrey, San Pedro Garza Garcia, MEXICO, 4Oncology Service, Hospital de Especialidades Eugenio Espejo, Quito, ECUADOR, 5Breast Cancer Unit, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 6Oncology department, Hospital Militar Uruguay HCFFAA, Montevideo, URUGUAY, 7Oncology Department, Hospital Roosevelt, Ciudad de Guatemala, GUATEMALA, 8Breast cancer Unit, Hospital México, San Jose, COSTA RICA, 9Oncology Department, Clínica San Felipe, Lima, PERU, 10Oncology Department, Instituto Oncologico Nacional, Panama, PANAMA, 11Direction of Research and Education, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center, Bogota, COLOMBIA, 12Oncology Department, Hospital Regional ISSSTE, Leon, MEXICO.
Accurate staging of breast cancer (BC) is essential for guiding treatment decisions and estimating prognosis. Current guidelines discourage routine diagnostic imaging in asymptomatic patients. However, adherence is often inconsistent in clinical practice. In Latin America (LATAM), this is further challenged by higher rates of advanced-stage presentations and aggressive tumor subtypes, which may lead to more frequent imaging. Yet, there is a lack of data on real-world imaging patterns in the region. Understanding current staging practices is critical to identifying care gaps, informing context-appropriate guidelines, and promoting efficient diagnostic strategies. We conducted a cross-sectional, anonymous online survey among oncologists and breast surgeons from LATAM countries. The survey included 63 items covering sociodemographic characteristics, imaging use by clinical stage, decision-making criteria, access to diagnostic technologies, turnaround times, and familiarity with guidelines. Descriptive statistics were used for analysis. A total of 269 physicians participated. Respondents included physicians from 18 LATAM countries and Spain; 55.4% were oncologists and 44.6% breast surgeons; 57.3% worked primarily in public settings, and 55.4% had over 10 years of experience. While 43.5% managed more than 50 BC patients per month, only 18.2% were exclusively dedicated to BC care. Guideline awareness was high (NCCN: 89%, ESMO: 71.4%, national: 70.6%), and 89.1% stated that guideline recommendations were important for decision making in clinical practice. Despite this, routine use of diagnostic imaging was reported in 34.2% of asymptomatic patients with stage I disease, 48% in stage II without nodal involvement, 77.7% in stage II with nodal involvement, and 91.1% in stage III. Chest CT (65.9%), abdominal CT (63.2%), and bone scintigraphy (62.7%) were reported as the most frequently used studies, though abdominal ultrasound (US) (42.2%), chest X-ray (40.9%), and PET-CT (23.3%) were also reported. Imaging decisions were primarily influenced by tumor size (78.3%) and biological subtype (65.9%). Chest X-ray and abdominal US reported use decreased from 62.8% in stage I to 24.7% in stage III patients, while thoraco-abdominal CT use increased from 36.2% in stage I to 85.7% in stage III patients. The reported use of imaging did not change based on medical specialty or years of clinical practice. Access to advanced imaging modalities such as PET-CT was more restricted; only 43.9% had consistent availability, and 44.6% reported turnaround times >4 weeks, compared to <3% for chest X-ray or abdominal US, 15.9% for thoraco-abdominal CT, and 27.1% for bone scintigraphy. Most respondents (62.5%) indicated that imaging decisions and turnaround times (82.2%) would differ based on patients’ insurance status (public vs private). Overall, the findings highlight a disconnect between guideline-based recommendations and real-world practice, shaped by system-level disparities and resource constraints. Our findings underscore a significant gap between guideline recommendations and real-world diagnostic imaging practices for BC staging across LATAM. Despite high awareness of guidelines, the frequent use of imaging reflects inconsistent adherence to guideline recommendations. Early-stage patients often undergo extensive imaging, with limited access and long delays—particularly for advanced modalities like PET-CT— that may contribute to treatment delays. These findings support the need for regionally adapted staging guidelines and targeted educational strategies to improve guideline-concordant care across LATAM. This work was conducted with the collaboration of LABCA, ACHO, ACM and SVM.
Presentation numberPS5-10-22
Allostatic Load and Risk of Obesity Related and Total Cancer in Postmenopausal Women
Jie Chi, Memorial Sloan Kettering Cancer Center, New York, NY
J. Chi1, F. Wang2, Y. Zong3, M. Pennell4, M. Yu4, V. Nair5, A. Vasbinder6, D. Lane7, A. Shadyab8, C. Valencia9, N. Saquib10, J. Wactawski-Wende11, P. Richey12, M. Skiba13, M. Simon14; 1Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Epidemiology and Biostatistics, Indiana University, Bloomington, IN, 3Internal medicine, Wayne State University, Detroit, MI, 4Division of Biostatistics, Ohio State University, Columbus, OH, 5Division of Hematology and Oncology, University of Washington School of Medicine, Seattle, WA, 6Nursing, University of Washington, Seattle, WA, 7Family, Population and Preventive Medicine, Stony Brook University, Lake Grove, NY, 8Epidemiology, University of California, San Diego, La Jolla, CA, 9Department of Family & Community Medicine, University Arizona, Tucson, AZ, 10Clinical Sciences, Sulaiman AlRajhi University, Al Bukayriah, SAUDI ARABIA, 11Epidemiolgy and Environmental Health, University at Buffalo, buffalo, NY, 12Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 13Advanced Nursing Practice & Science, University of Arizona, Tucson, AZ, 14Oncology, Karmanos Cancer Institute, Detroit, MI.
Allostatic Load and Risk of Obesity-Related and Total Cancer in Postmenopausal Women: Insights from the Women’s Health InitiativeBackground: Allostatic load (AL), a composite biomarker of physiological stress, has been associated with chronic disease, but its relationship with cancer—particularly obesity-related cancers—remains understudied in large, diverse populations. In this analysis, we investigated the association between baseline AL and the incidence of obesity-related cancers and total cancer risk, among postmenopausal women in the Women’s Health Initiative (WHI).Methods: Among 161,805 WHI participants, we identified a final cohort of 28,102 women with information on AL after excluding individuals with delayed biomarker collection, or prior cancer diagnosis. AL was calculated using eight biomarkers (pulse, systolic and diastolic blood pressure, body mass index (BMI), waist circumference, C-reactive protein (CRP), fasting glucose, and total cholesterol). Participants were stratified into tertiles of AL (low: 0–2, medium: 3–5, and high: 6–8). Cox proportional hazards and Fine-Gray models were used to assess associations between AL and incident cancers, separately looking at the risk of 13 obesity related cancers and total cancer risk, and adjusting for sociodemographic, behavioral, and reproductive factors.Results: Over median follow up of 18. 64 years, 14.1% of participants developed obesity-related cancers and 22.3% developed any cancer. In fully adjusted models, women in the highest AL tertile at WHI enrollment had significantly elevated risk of obesity-related cancer compared to the lowest tertile (HR 1.20; 95% CI: 1.01–1.42; p=0.044). A 1-point increase in AL score was associated with a 4% higher risk (HR 1.04; 95% CI: 1.00–1.07; p=0.028). Total cancer risk did not increase significantly with higher AL score (HR for high vs. low: 1.07; 95% CI: 0.92–1.24; p=0.388; per point: HR 1.02; 95% CI: 1.00–1.05; p=0.079). These associations did not differ significantly by race, ethnicity or income. Conclusions: Elevated allostatic load is independently associated with increased risk of obesity-related cancer among postmenopausal women in the WHI. These findings highlight the potential role of cumulative physiological stress in in cancer risk beyond obesity alone and suggest that AL may be a valuable biomarker incorporating social and metabolic stressors for cancer risk stratification in older women.
Presentation numberPS5-10-23
Assessing Clinical and Sociodemographic Trends in Contralateral Prophylactic Mastectomy at an Urban Tertiary Care Center
Ariella Deborah Simoni, Montefiore Einstein, Bronx, NY
A. D. Simoni1, R. Greenbaum1, S. Harbour1, S. L. Jao2, M. P. McEvoy2; 1Albert Einstein College of Medicine, Montefiore Einstein, Bronx, NY, 2Department of Surgery, Division of Breast Surgery, Montefiore Einstein, Bronx, NY.
Background: The rate of patients undergoing contralateral prophylactic mastectomy (CPM) for unilateral breast cancer has been rising dramatically despite emerging guidelines which discourage CPM in low-risk patients due to lack of survival benefit. The decision to undergo CPM is personal and often multi-factorial, and has not been well-studied. This study aimed to identify clinical and sociological factors associated with the decision to pursue CPM in an urban, underserved community. Methods: A retrospective review of 259 patients with unilateral breast cancer who underwent mastectomy from 2012-2025 at a tertiary care hospital system was performed. Variables included sociodemographic and clinical features. Primary outcome was rate of CPM. Statistical analyses were performed in SPSS.Results: Total rate of CPM in our population was 29% (n=75) and total rate of reconstruction was 18% (n=47). No male patients (n=9) underwent CPM, whereas rate of CPM in female patients was 30% (p=0.0627). Among female patients, patients who underwent CPM were on average 13.6 years younger than controls (p < 0.001). Having private insurance vs. public insurance (p=0.084) and being currently employed vs. unemployed (p=0.073) trended towards significance for association with CPM. Among clinical factors, undergoing surgical reconstruction (p=0.002), and undergoing neoadjuvant chemotherapy (p=0.001) were associated with CPM. There was no significant association between CPM and racial or ethnic group, tumor size, histologic subtype, clinical stage, or hormone receptor positivity.Conclusion: In our urban population, social factors such as age, insurance status, and employment status and whether patients additionally underwent surgical reconstruction or neoadjuvant treatment with chemotherapy may play a role in the decision to undergo CPM. In management of patients with unilateral breast cancer, surgeons may consider such nuances in patient decision-making when guiding conversations surrounding CPM.
Presentation numberPS5-10-24
Cardiac Monitoring in Patients Receiving HER2-Directed Therapy: A Meta-Analysis and Cost-Effectiveness Analysis
Ilana Schlam*, Dana Farber Cancer institute, Boston, MA
I. Schlam*1, M. C. Saad Menezes*2, J. N. Upshaw3, J. Rabinowitz4, J. Schwartz5, A. Julian5, V. Mico5, S. Papatheodorou6, N. Kunst7, A. Pandya8, A. Barac9, S. M. Tolaney1; 1Medical Oncology, Dana Farber Cancer institute, Boston, MA, 2Internal Medicine, UT Southwestern, Dallas, TX, 3Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, 4Library, Tufts University, Boston, MA, 5Internal Medicine, Tufts Medical Center, Boston, MA, 6Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, 7Statistics, York University School of Medicine, York, UNITED KINGDOM, 8Health Decision Science, Harvard Chan School of Public Health, Boston, MA, 9Cardiology, Inova Schar Heart and Vascular, Fairfax, VA.
Background: HER2-targeted therapies improve outcomes in HER2-positive breast cancer but carry a risk of cardiotoxicity. Guidelines recommend routine LVEF monitoring every three months. However, the incidence of symptomatic heart failure appears low. We conducted a meta-analysis to quantify the risk of cardiotoxicity associated with HER2-directed agents and developed a cost-effectiveness model to evaluate alternative frequency of cardiac monitoring in patients with a standard cardiovascular risk profile. Methods: We performed a systematic literature search (Ovid MEDLINE, Embase, Cochrane) and included 55 studies (n=39,335) evaluating trastuzumab, pertuzumab, trastuzumab deruxtecan (T-DXd), and trastuzumab emtansine (T-DM1). A random-effects single-arm meta-analysis estimated the cumulative incidence of cardiotoxicity. Using these estimates and additional inputs estimated from the published literature, we constructed a decision-analytic model comparing four LVEF monitoring strategies during HER2-targeted therapy: (1) baseline only, (2) baseline and month 12, (3) baseline, month 6, and month 12, and (4) baseline plus every 3 months. We modeled early-stage breast cancer patients over 12 months of therapy. We used the model to project expected per-person cardiotoxicity events (i.e., heart failure with reduced ejection fraction, HFrEF) events detected and undetected, monitoring (i.e., echocardiogram) costs (including patient time), and incremental cost-effectiveness ratios (ICERs). We varied all model input values through plausible ranges in one-way sensitivity analyses. Results: Among 39,335 patients, the pooled incidence of any cardiotoxicity was 8.0% (95% CI 6.0-10.4) with trastuzumab, 1.4% (95% CI 0.8-2.6) with T-DM1, and 2.0% (95% CI 1.4-2.9) with T-DXd. Symptomatic cardiotoxicity was uncommon, occurring in 1.6% of patients treated with trastuzumab, 0.4% of patients treated with T-DM1, and 0.3% of patients treated with T-DXd. Cardiotoxicity rates were slightly higher in metastatic settings. In our initial cost-effectiveness modeling, more frequent monitoring was associated with more HFrEF events detected but at higher monitoring costs (Table 1), with an ICER of $390,500/HFrEF event detected for the strategy of baseline plus every 3 months compared to the strategy of baseline and months 6 and 12. Conclusions: HER2-directed therapies are associated with low rates of symptomatic cardiotoxicity, particularly T-DM1 and T-DXd. Cost-effectiveness modeling demonstrated diminishing returns with more frequent LVEF monitoring. Future analyses will assess the cost-effectiveness of LVEF monitoring strategies in patients with early-stage or metastatic disease treated with trastuzumab, T-DM1, or T-DXd.
| Strategy | Undetected HFrEF | Detected HFrEF | Monitoring cost | ICER |
| Baseline only | 0.02549 | 0.00402 | $502 | Reference |
| Baseline and month 12 | 0.00848 | 0.02103 | $997 | $29,100/HFrEF detected |
| Baseline, months 6 and 12 | 0.00540 | 0.02411 | $1,488 | $159,700/HFrEF detected |
| Baseline and every 3 months | 0.00288 | 0.02663 | $2,471 | $390,500/HFrEF detected |
Presentation numberPS5-10-25
Comparison of survival and recurrence outcomes among breast cancer patients: breast-conserving surgery versus medically necessary and patient preference mastectomy
Claire Liu, University of British Columbia, Vancouver, BC, Canada
C. Liu, R. Warburton, C. Dingee, J. Pao, A. Bazzarelli, A. Nichol, E. McKevitt; Department of Surgery, University of British Columbia, Vancouver, BC, CANADA.
Background: Global mastectomy rates for breast cancer have risen over the past decade for unclear reasons. Patients may perceive mastectomy to improve survival and reduce recurrence risk, despite trials showing breast conserving surgery (BCS) with radiation offers equivalent or superior survival in early breast cancer. Although international guidelines support both BCS with radiation and mastectomy for treatment of early breast cancer, emerging evidence favoring BCS challenges the routine practice of offering both equally. Currently, there is no comparative data assessing survival and recurrence outcomes across BCS, medically necessary mastectomy (Mast-MN), and patient preference mastectomy (Mast-PP). This study compared breast cancer-specific survival (BCSS), overall survival (OS), local recurrence (LR), and regional recurrence (RR) between these surgical approaches. Methods: Patients who underwent surgery for stage pT1-3pN0-3 breast cancer at our provincial referral center from 2012-2018 were identified from a prospectively maintained database. Exclusions were for stage 4 disease, synchronous breast cancers, neoadjuvant therapy, or BCS without adjuvant radiation. The indication for mastectomy was coded preoperatively by the consulting surgeon, where a mastectomy was classified as Mast-MN in patients with contraindications to BCS, and Mast-PP for patient preference when BCS was otherwise feasible. To account for our provincial cancer centre’s post-mastectomy radiation guidelines, analyses were stratified by nodal status. BCSS was assessed using a cause-specific hazard model, OS with a Cox proportional hazards model, and LR/RR using a Fine-Gray competing risk model, controlling for age, T stage, receptor status, grade, lymphovascular invasion, adjuvant hormone and chemotherapy, and axillary dissection. Results: Among 1468 node negative patients, 986 underwent BCS, 275 Mast-MN, and 207 Mast-PP. There was no significant difference in BCSS or LR between Mast-PP and BCS, however Mast-PP was associated with significantly worse OS (HR = 2.03, 95% CI: 1.29 -3.20, p=0.02) and greater risk of regional recurrence (SHR = 39.7, 95% CI: 4.95-318, p = 0.001) compared to BCS. Among 628 node positive patients, 354 underwent BCS, 207 Mast-MN, and 67 Mast-PP. There was no significant difference in BCSS, OS, LR, or RR between Mast-PP and BCS in these patients. Conclusions: Mast-PP does not appear to confer any survival or recurrence advantage compared to BCS with radiation. In fact, for node negative patients, Mast-PP may be associated with a higher risk of regional recurrence compared to BCS, due to omission of adjuvant radiation.
Presentation numberPS5-10-27
Integrating Virtual Collaborative Care Behavioral Health in Breast Cancer Care: Improving Outcomes in Mental Health, Adherence, and Racial Equity
Nina Balanchivadze, Virginia Oncology Associates, Norfolk, VA
N. Balanchivadze1, K. N. Lavin2, J. L. Yourell3, L. Wolfe4, T. P. Menon5, M. A. Danso6; 1Oncology, Virginia Oncology Associates, Norfolk, VA, 2Psychiatry, Cerula Care, Chapel Hill, NC, 3Behavioral Health, Fit Minded Inc, Chapel Hill, NC, 4Psychology, Cerula Care, Chapel Hill, NC, 5School of Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, 6Oncology, Virginia Oncology Associates and Sarah Cannon Research Institute, Norfolk, VA.
Background: Patients with breast cancer often experience depression, anxiety, and reduced quality of life during treatment, yet mental health support is rarely integrated into oncology care. While collaborative care models have demonstrated efficacy in primary care, their impact in oncology remains under-explored. This study evaluated the impact of Cerula Care, a virtual collaborative care program, on mental health and quality of life among patients with breast cancer. Methods: We conducted a retrospective analysis of 207 patients with breast cancer enrolled in a virtual behavioral health program (Cerula Care) integrated into their oncology care. Patients received coordinated care from a psychiatrist, behavioral health care manager, and behavioral health coach. Depression (PHQ-9), anxiety (GAD-7), and quality of life (FACT-G7) were assessed monthly. Changes in outcomes were analyzed using paired-sample t-tests and correlations. Results: Participants were predominantly female (99%), with a mean age of 55.7 years; 58% identified as White and 32.4% as Black/African American. Medicaid patients had statistically significant higher anxiety and depression at baseline (GAD-7 mean = 10.67 vs. 6.62, p = .005), (PHQ-9 mean = 10.62 vs. 7.63, p = .004), and lower directionally lower functioning (FACT-G7 mean = 11.67 vs. 17.30, p = .073) showing the importance of screening Medicaid members who face greater stressors. Statistically significant improvements across all patients were observed in depression scores by month five (-6.63 points, n=32, p<0.001, Cohen’s d=3.77), and anxiety scores improved by month six (-4.61 points, n=31, p<0.001, Cohen’s d=5.38). Quality of life scores demonstrated a positive trend over time. Notably, identifying as Black was significantly correlated with greater improvements in quality of life (r=0.49, p=0.01), while identifying as White was correlated with less improvement (r=-0.46, p=0.01). Among patients who completed initial surveys, 70% reported that the program helped them consistently make it to their oncology visit and 65% noted improved adherence to their non-chemotherapy medications. Conclusions: Cerula Care’s virtual collaborative care model was associated with meaningful improvements in depression, anxiety, oncology visit and treatment adherence, and quality of life among patients with breast cancer. The greater benefit observed in Black patients highlights the potential to help address racial disparities in supportive oncology care. These findings support broader adoption of collaborative care approaches as scalable, patient-centered, and equity-promoting solutions in cancer treatment settings. Future studies should explore long-term outcomes, cost-effectiveness, and implementation across diverse populations. Table 1. Behavioral Health Outcomes in Breast Cancer Patients (N = 207)
| Outcome Measure | n | Mean Change | p-value | Effect Size (Cohen’s d) | |||||
| Depression (PHQ-9) | 32 | ↓ 6.63 | < .001 | 3.77 | |||||
| Anxiety (GAD-7) | 31 | ↓ 4.61 | < .001 | 5.38 | |||||
| Quality of Life (FACT)* | 62 | ↑ 1.58 | NS | – |
Presentation numberPS5-10-28
A Survivor-Led Patient Navigation Model to Improve Breast Cancer Treatment Adherence in India: Results from a Pilot Project
Soumen Das, Institute of Breast Disease, NCRI Hospital, Kolkata, Kolkata, India
S. Das1, T. Mandal2, A. Mallick3, R. Agarwal1, P. Basu4, R. Jena4; 1Surgical Oncology, Institute of Breast Disease, NCRI Hospital, Kolkata, Kolkata, INDIA, 2Medical Oncology, Institute of Breast Disease, Kolkata, INDIA, 3Surgery, IPGME&R,SSKMH, Kolkata, INDIA, 4Medical Oncology, Institute of Breast Disease, NCRI Hospital, Kolkata, Kolkata, INDIA.
Background:Breast cancer is the leading cancer among Indian women, yet the national 5-year survival rate remains below 65%, largely due to delayed presentation and high rates of treatment non-completion. Historical Indian studies report treatment adherence rates between 55-70%, with dropout rates as high as 30-40%, especially in rural and low-income settings. To address these challenges, we piloted a survivor-led patient navigation model grounded in community participation, digital supervision, and sustainability. Methods:A five-phase pilot program was implemented at two tertiary hospitals in West Bengal, India. Fourteen breast cancer survivors were recruited and trained through a structured 4-week online course. After simulation-based assessment, they began navigating real patients, supported via moderated WhatsApp groups that included the patient, navigator, and treating clinician. Navigators from one hospital were assigned to patients from the other to maintain neutrality. Results:Over a 6-month period, navigators supported 82 newly diagnosed breast cancer patients. Compared to historical Indian data indicating adherence rates of ~65%, the pilot demonstrated a treatment completion rate of 89.0%, with a dropout rate of 11.0%—representing a ~60% relative reduction in dropout. Navigators helped patients overcome key psychosocial and logistical barriers, including fear of surgery, misinformation about chemotherapy, financial uncertainties, and loss to follow-up. Patient-reported feedback indicated increased trust, better communication, and improved emotional resilience throughout treatment. Conclusion:This low-cost, survivor-led patient navigator model significantly outperformed historical national benchmarks in terms of treatment adherence and retention. The model is scalable, culturally adaptive, and resource-appropriate for LMICs. It aligns with the WHO Global Breast Cancer Initiative goals and presents a strong case for integration into India’s National Programme for Prevention and Control of Non-Communicable Diseases (NPCDCS).
Presentation numberPS5-10-29
Factors Associated with Discussion of Contralateral Prophylactic Mastectomy (CPM) During Surgical Consultations
Heather Neuman, University of Wisconsin, Madison, WI
H. Neuman1, F. Dickerson1, M. Saucke1, C. Breuer1, L. Bozzuto1, A. Beck1, J. Schumacher2; 1Department of Surgery, University of Wisconsin, Madison, WI, 2Department of Surgery, University of North Carolina, Chapel Hill, NC.
Introduction While surgeons endorse that CPM should not be routinely performed, they also acknowledge that perceived benefits of CPM can outweigh risks for some women. Little is known about how CPM conversations occur in practice. The objective is to determine the frequency with which CPM is discussed in practice and identify patient factors associated with discussion. Methods: This study was a secondary analysis of Alliance clinical trial A231701CD. We identified women ≤65 years who underwent breast cancer surgery and whose transcript of their audio-recorded surgical consultation was available. We excluded women with indications for bilateral breast surgery (e.g., bilateral cancer, genetic mutation). Transcripts were reviewed to determine whether CPM was discussed and who initiated the discussion (surgeon vs patient/support person). Two breast surgeons reviewed transcripts to determine candidacy for breast conserving surgery (BCS) and family history (FH). We used multivariable logistic regression to identify factors associated with CPM discussion, controlling for age, BCS candidacy, FH, invasive vs in-situ cancer, race, and socioeconomic disadvantage using the area deprivation index (dichotimized at ≥80th percentile). We generated predictive probabilities to highlight model findings. Chi square test was used to assess initiation of CPM discussion by race. Results: Of the 347 women, median age was 55 (range 28-65), 38% were non-white, and 24% socioeconomically disadvantaged. 49% of consults discussed CPM, most commonly initiated by the surgeon (64%). Younger ages compared with age >55-65 (age >45-55 OR 4.2 [95% CI 2.3-7.7]; age ≤45 OR 6.9 [95% CI 3.7-13.0]), FH (OR 2.7, 95% CI 1.5-4.6), and White as compared to Black race (OR 4.1 [95% CI 2.2-7.6]) were associated with greater odds of discussing CPM. No statistically significant association was observed for non-BCS compared to BCS candidates (OR 1.6 [95% CI 0.9-2.9]), invasive compared with in-situ cancer (OR 0.9 [95% CI 0.5-1.7], or socioeconomic disadvantage (OR 1.1 [95% CI 0.6-2.1). Although trends in the association between age and FH, and whether CPM was discussed were similar for Black versus White patients, an absolute difference by race persisted (Table). There was no statistically significant difference in who initiated the CPM discussion by race. Discussion: The likelihood of discussing CPM is greater for younger patients and those with a FH, aligned with the potential benefit of risk-reduction with CPM. Interestingly, there was a notable difference in likelihood of discussing CPM for White versus Black patients. Surgeons initiated the discussion in the majority of consults, making the difference unlikely to be simply related to patient motivation for CPM. Future work will focus on the CPM discussions themselves to understand the nature of the CPM discussion, as this likely contributes to the high rates of CPM for White women.
| Race | Age at Diagnosis | |||
| <=45 | >45-55 | >55-65 | ||
| No Family History, BCS Candidate | White | 76% (64-87%) | 65% (53-77%) | 31% (22-40%) |
| No Family History, BCS Candidate | Black | 43% (26-60%) | 32% (17-46%) | 10% (4-16%) |
|
Family History, not a BCS Candidate |
White | 93% (88-98%) | 89% (81-98%) | 67% (51-83%) |
|
Family History, not a BCS Candidate |
Black | 77% (63-92%) | 67% (48-87%) | 33% (15-51%) |
Presentation numberPS5-10-30
A randomized survey study of structured reflective questions and willingness to participate in window of opportunity trials
Colin Bergstrom, Stanford, Stanford, CA
C. Bergstrom1, D. Parikh1, S. Brain1, D. Heditsian1, V. Lee1, B. Shaw2, C. Thompson3, G. Sledge Jr1, J. Caswell-Jin1; 1Department of Medicine, Stanford, Stanford, CA, 2Quantitative Sciences Unit, Stanford, Stanford, CA, 3Department of Surgery, Stanford, Stanford, CA.
Background: Clinical trials are critical to advancing cancer care but often raise patient concerns regarding treatment delays, additional procedures and visits, and toxicity. Understanding factors that influence trial enrollment decisions is essential for patient-centered trial design. Window of opportunity trials, which introduce investigational therapies between diagnosis and standard treatment initiation and are increasingly important in breast cancer research, exemplify a design where these concerns may be magnified. Limited data exist on patients’ evaluation of participation in such trials.Methods: We conducted a randomized survey study to determine patients’ willingness to participate in a window of opportunity trial. Adults (≥18 years) with a self-reported history of stage 0-III breast cancer were recruited via advocacy organizations, online social media platforms, and IRB-approved outreach at breast oncology clinics. All participants were first presented with a standardized definition and visual representation of a window of opportunity trial. They were then randomized in a 2:1 ratio to receive the primary endpoint question assessing willingness to participate either after exposure to reflective questions about potential barriers (e.g., additional biopsies, clinic visits, treatment delays) and motivators (e.g., desire to contribute to science), or immediately without prior reflective questions. Participants were also asked to answer questions about demographics and clinical characteristics as well as attitudes toward clinical research using the Corbie-Smith Index. The primary endpoint was willingness to participate, assessed on a Likert scale.Results: Of 1,104 total responses, 874 met eligibility criteria and 749 completed the primary endpoint question and were included in the analytic cohort. Respondents were primarily female (99%), non-Hispanic White (76%), and college-educated (64%). Among survey respondents, those Participants exposed to reflective questions before the primary question were significantly more likely to express willingness to enroll in a window of opportunity trial (76% vs 64%, OR 1.88, 95% CI 1.22-2.90; P < 0.001). Most demographic and clinical features did not correlate with willingness to participate, including age, race and ethnicity, income, location of treatment (academic vs community), geographic location (urban vs suburban vs rural), time since breast cancer diagnosis, nodal involvement, patient assessment of risk of recurrence, and initial treatment after diagnosis (surgery vs systemic therapy); an exception was that willingness was higher among those without a college degree (80% vs. 71%; P = 0.027), not significant after multiple hypothesis testing. Research distrust (Corbie-Smith Index) was significantly associated with lower willingness to participate (P < 0.001). Willing participants were more open to additional procedures, with 93% accepting at least one biopsy (vs. 57% of those unwilling) and 48% accepting five or more extra clinic visits (vs. 23% of those unwilling).Conclusions: After exposure to reflective questions addressing potential barriers and motivators, participants had significantly greater willingness to participate in a window of opportunity trial. The effect of reflective question exposure on willingness to participate was substantially greater than that of any demographic or clinical characteristic measured in our survey, emphasizing the potential of simple, scalable behavioral strategies to improve patient engagement in clinical trials. These results have important implications for the development of both digital tools and face-to- face communication strategies to enhance clinical trial enrollment and patient satisfaction in early-stage breast cancer treatment
Presentation numberPS5-11-01
Cost-effectiveness analysis of next-generation sequencing (ngs) panel including brca1 and brca2 genes for women diagnosed with non-metastatic breast cancer in brazil
Henrique Lima Couto, Redimama-Redimasto, Belo Horizonte, Brazil
H. L. Couto1, R. M. Rahal2, A. T. Hassan3, F. Zanghelini4, L. P. Gargano5, T. C. Oliveira6, B. A. Coelho7, M. C. Wender8, A. L. Silva9, E. C. Pessoa10, A. Mattar11, C. A. Padua12, G. F. Cunha Júnior13, A. M. Santos14, L. L. Dominguez6, D. R. Siqueira6, M. S. Castilho15, M. Antonini16, J. S. Oliveira6, B. A. Pires6, D. d. Pires6, S. d. Ferreira6, T. P. Moraes6, L. B. Oliveira6, P. C. Soares6, R. G. Saliba6, A. C. Mendonça6, J. C. Cisneros6, F. M. Reis9, G. Novita17, V. M. Oliveira18, F. M. Bagnoli18, G. Tosello19, D. d. Buttros20, B. P. Carvalho21, W. J. Almeida Junior22, M. d. Ramos23, D. Balabram24, É. M. Carvalho25, BreastMit Collaborative Research Group; 1CEO, Redimama-Redimasto, Belo Horizonte, BRAZIL, 2Faculty of Medicine, Department of Gynecology and Obstetrics, Federal University of Goiás, Brazil., Goiânia, BRAZIL, 3Breast Cancer Center, Oncoclínicas – CAM, Salvador, BRAZIL, 4Postgraduate Program in Therapeutic Innovation, Federal University of Pernambuco, Recife, Brazil, Belo Horizonte, BRAZIL, 5Department of Social Pharmacy, Faculty of Pharmacy,, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil., Belo Horizonte, BRAZIL, 6Breast Cancer Center, Redimama-Redimasto, Belo Horizonte, BRAZIL, 7Breast Cancer Center, Mater Clinic, Montes Claros, Minas Gerais Brazil., Belo Horizonte, BRAZIL, 8Department of Gynecology and Obstetrics, Faculty of Medicine of The Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil., Porto Alegre, BRAZIL, 9Department of Gynecology and Obstetrics, Faculty of Medicne of The Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 10Department of Obstetrics and Gynecology,, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, Sao Paulo, Brazil., Botucatu, BRAZIL, 11Breast Cancer Center, Women Hospital, São Paulo, São Paulo, Brazil., São Paulo, BRAZIL, 12Cetus Oncology, Belo Horizonte, Minas Gerais, Brazil., MIRA – Medical Group, Belo Horizonte, BRAZIL, 13Cetus Oncology, MIRA – Medical Group, Belo Horizonte, BRAZIL, 14Mastology, Minas Gerais State Servants Welfare Institute, Belo Horizonte, Minas Gerias, Brazil, Belo Horizonte, BRAZIL, 15Radio-oncology, Radiocare, Belo Horizonte, Minas Gerais, Brazil., Belo Horizonte, BRAZIL, 16Mastology, Hospital of the State Public Servant of São Paulo, Sao Paulo, Sao Paulo Brazil., São Paulo, BRAZIL, 17President, Sociedade Brasileira de Mastologia, Rio de Janeiro, BRAZIL, 18Department of Gynecology and Obstetrics, Santa Casa de São Paulo School of Medical Sciences, São Paulo, São Paulo, Brazil., São Paulo, BRAZIL, 19Breast Cancer Center, Instituto do Câncer Oeste Paulista (InCOP), Presidente Prudente, São Paulo, Brazil., Presidente Prudente, BRAZIL, 20Department of Gynecology and Obstetrics, Faculty of Medicine of Claretiano University, Rio Claro, São Paulo, Brazil., Belo Horizonte, BRAZIL, 21CEO, Sonar Breast Cancer, Belo Horizonte, BRAZIL, 22Department of Gynecology and Obstetrics, Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 23Breast Surgery, Brazilian Society of Mastology, Rio de Janeiro, BRAZIL, 24Surgery, Faculty of Medicne of The Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Belo Horizonte, BRAZIL, 25Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil., Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil., Rio de Janeiro, BRAZIL.
AbstractBackground: Pathogenic BRCA1/2 mutations in women diagnosed with non-metastatic breast cancer (MBC) correlate with worse survival outcomes and elevated risk of contralateral breast cancer. Detecting pathogenic BRCA1/2 mutations via next-generation sequencing panels (NGS) provides clinical benefits by optimizing treatment strategies.Objective: This study estimates the cost-effectiveness of using NGS with genetic counselling to detect pathogenic BRCA1/2 mutations in women diagnosed with non-MBC, compared to no genetic testing, from the perspective of Brazil’s Public Health System.Methods: A hybrid economic model (decision tree plus Markov model) simulated costs and outcomes over a 10-year horizon of NGS with genetic counselling to identify pathogenic BRCA1/2 mutations in women diagnosed with non-MBC. The eligible population was estimated by using epidemiological methods using national data identified in the literature. Were estimated the incremental cost-effectiveness ratio (ICER) per quality-adjusted life year (QALY) gained of adding NGS plus genetic counselling to identify pathogenic BRCA1/2 mutations in women diagnosed with non-MBC.Results: NGS for pathogenic BRCA1/2 mutations combined with genetic counselling showed an incremental benefit of 0.044 QALYs gained at an additional cost of US$ 590.29 compared to no genetic testing and genetic counselling, resulting in an ICER of US$ 13,526.89 per QALY gained.Conclusion: NGS for pathogenic BRCA1/2 mutations combined with genetic counselling was cost-effective from the Brazilian public health system perspective, with an ICER value below the Brazilian oncological willingness-to-pay threshold of US$21,500/QALY (R$120,000.00/QALY).
Presentation numberPS5-11-02
Exploring Sociodemographic and Structural Barriers to Timely Surgery in ER+ HER2- Breast Cancer: A Single-Institution Study in the Bronx
Susan Laura Jao, Montefiore Medical Center, Bronx, NY
S. Jao1, C. Pansa2, V. Mehta3, M. Scheckley1, M. McEvoy1, A. Gupta1, E. Ravetch1, S. Feldman1; 1Breast Surgery, Montefiore Medical Center, Bronx, NY, 2Breast Surgery, Albert Einstein College of Medicine, Bronx, NY, 3Otorhinolaryngology-Head & Neck Surgery, Montefiore Medical Center, Bronx, NY.
Background: Surgical delays in breast cancer are associated with worse outcomes, prompting a Commission on Cancer 60-day time-to-surgery (TTS) benchmark for patients not receiving neoadjuvant treatment. In ER+ HER2- disease, delays beyond 42 days significantly increase breast cancer-specific mortality. Known risk factors for surgical delay include younger age, Medicaid insurance, lower income, and pre-operative MRI. Racial disparities remain inconsistently reported. This study evaluates risk factors for delayed surgery at an academic center that serves a largely urban, multiethnic and socioeconomically disadvantaged population, with attention to modifiable institutional contributors. Methods: A retrospective review was conducted of ER+ HER2- breast cancer patients diagnosed Jan 2023-Dec 2024. Included were women ≥18 undergoing upfront surgery; excluded were men, in situ disease, neoadjuvant therapy, and TTS >120 days. TTS was defined as the interval from biopsy to surgery. Demographic, clinical, and management variables were analyzed using univariate and multivariate linear regression. Results: Among 227 cases, mean TTS was 55.0 ± 21.0 days; 148 (65.2%) had surgery within 60 days and 70 (30.4%) within 42 days. Mean time from biopsy to first clinic visit was 16.7 ± 9.6 days; from clinic to surgery was 38.3 ± 20.0 days. TTS strongly correlated with time from clinic to surgery (Spearman ρ = 0.863, p 65), 57 vs. 48 days (p = 0.0037). On univariate analysis, longer TTS was associated with Black race (p = 0.042), Medicaid (p = 0.005), and mastectomy (p = 0.0044). On multivariate linear regression analysis controlling for demographic, clinical and management variables, significant delays were associated with Black race, ILC pathology, IORT, plastic surgery involvement, medical clearance, breast MRI, and surgery cancellation/rescheduling (all p < 0.05). Plastic surgery had the greatest impact (+17.2 days), followed by canceled surgery (+13.7), Black race (+10.1), IORT (+9.3), medical clearance (+8.9), ILC (+7.9), and MRI (+6.9). Conclusion: In this study, several modifiable and non-modifiable factors were associated with increased time to surgery among ER+ HER2- breast cancer patients. Notably, delays were significantly associated with Black race, ILC pathology, and logistical elements including plastic surgery coordination, IORT, medical clearance, and pre-operative breast MRI. The largest contributors to delay—plastic surgery involvement and canceled/rescheduled surgeries—highlight actionable targets for quality improvement. These findings underscore the need for institution-specific strategies to streamline surgical workflows and address disparities, particularly as delays beyond 42 days may adversely affect survival in this patient population.
| Frequency(%) | MedianTTS(days) | IQR | UnivariateP-value | MultivariateLinearRegressionP-value | BCoefficient(+days) | |
| Race | 0.0432 | |||||
| White | 118(51.98) | 49 | 36.0-62.8 | Ref | ||
| Black | 84(37.00) | 58 | 42.0-75.2 | 0.0033 | 10.113 | |
| Asian | 9(3.96) | 43 | 30.0-54.0 | 0.8089 | ||
| Unknown/Other | 16(7.04) | 60 | 47.2-66.8 | 0.2702 | ||
| HispanicEthnicity | 104(45.81) | 49 | 38.8-65.0 | 0.5964 | 0.4468 | |
| InsuranceCoverage | 0.005 | |||||
| Private | 69(30.40) | 52 | 38.0-70.0 | Ref | ||
| Medicaid | 49(21.59) | 59 | 49.0-83.0 | 0.0557 | ||
| Medicare | 108(47.58) | 48.5 | 36.0-63.0 | 0.8918 | ||
| Pathology | 0.2108 | |||||
| IDC | 185(81.50) | 50 | 38.0-64.0 | Ref | ||
| ILC | 28(12.33) | 64 | 49.8-75.0 | 0.0404 | 7.867 | |
| SurgeryType | 0.0044 | |||||
| Lumectomy | 197(86.78) | 49 | 37.0-64.0 | 0.3936 | ||
| Mastectomy | 30(13.22) | 62 | 52.8-80.2 | 0.4909 | ||
| IORT | 50(22.03) | 57 | 43.0-69.5 | 0.1522 | 0.0039 | 9.275 |
| PlasticSurgey | 30(13.22) | 66 | 57.0-83.0 | <0.001 | 0.0023 | 17.179 |
| MedicalClearance | 70(30.84) | 58 | 42.0-69.8 | 0.0271 | 0.0026 | 8.894 |
| BreastMRI | 102(44.93) | 57.5 | 43.0-72.8 | 0.0017 | 0.0186 | 6.869 |
| StagingScans | 8(3.52) | 69 | 57.5-93.5 | 0.0079 | 0.0566 | |
| AdditionalBiopsy | 17(7.49) | 70 | 64.0-84.0 | <0.001 | 0.3664 | |
| OutsideReview | 18(7.93) | 60 | 19.2-71.2 | 0.042 | 0.2257 | |
| Canceled/RescheduledSurgery | 11(4.85) | 58 | 57.0-85.0 | 0.006 | 0.0195 | 13.772 |
Presentation numberPS5-11-03
The value of innovation in breast cancer treatment: Real Option Value of Olaparib for First-Line Treatment of gBRCA1,2 Pathogenic Mutation HR+/HER2-negative and Triple Negative Metastatic Breast Cancer
Cynthia L Gong, Curta Inc., Seattle, WA
C. L. Gong1, M. Chavez-MacGregor2, D. Elsea1, X. Xu3, L. Park3, S. Katsandres1, K. Migliaccio-Walle1, D. L. Veenstra1; 1Curta Inc., Curta Inc., Seattle, WA, 2Department of Health Services Research, Division of Cancer Prevention and Population Sciences, University of Texas MD Andersen Cancer Center, Houston, TX, 3Global Oncology Outcomes Research, AstraZeneca, Gaithersburg, MD.
Background: Novel drugs for metastatic breast cancer (mBC) can provide patients not only improved survival but also the opportunity to benefit from future treatment innovations, a concept referred to as real option value (ROV). A recent study indicated that 68% of oncologists consider ROV when making treatment recommendations, but relatively few studies have quantified ROV. Extended survival follow-up from the OlympiAD study suggested a greater survival benefit with olaparib compared to chemotherapy in the subset of patients treated in the first-line (1L) setting. Given the rapid pace of innovation for human epidermal growth factor receptor 2 (HER2)-negative mBC, quantifying the ROV of 1L olaparib may help clinicians and patients better understand the impact of utilizing innovative therapies in earlier lines when appropriate. Objective: To quantify the ROV of olaparib for 1L treatment of hormone receptor-positive (HR+)/HER2-negative and triple negative metastatic breast cancer (TNBC) patients with germline BRCA1,2 pathogenic mutations. Methods: We developed a Markov model consisting of progression-free, progression, and death health states to estimate the quality-adjusted survival gain of 1L olaparib vs. treatment of physician’s choice (TPC, i.e. chemotherapy). To derive ROV, a scenario without future drug innovation, estimated by extrapolating the extended survival follow-up OlympiAD 1L data, was compared to one considering anticipated future innovations as of January 2023 (date of trial publication). Key ROV parameters were derived from literature, previous clinical trial data, ongoing studies as of January 2023, previous oncology clinical trial success and timing, and mBC treatment patterns. ROV parameters and survival curves were input separately for HR+ and TNBC due to inherent clinical differences between the two subpopulations. Parameter assumptions were reviewed by experts and assigned values to generate high-, mid-, and low-ROV scenarios; parameters included future innovation effect size (hazard ratios of 0.65, 0.75, and 0.85 for HR+ and 0.4, 0.6, and 0.7 for TNBC), timing of access to innovative therapies (4, 6, and 8 months for HR+ and 1, 4, and 6 month(s) for TNBC), probability of approval success (40%, 60%, and 80%), and rate of uptake (50%, 70%, and 80%). The primary outcome was aggregated quality-adjusted survival, weighted between HR+ and TNBC according to the OlympiAD trial characteristics. Results: Without future innovation, aggregate (weighted across HR+ and TNBC) quality-adjusted survival for olaparib was 23.3 vs. 12.8 months with TPC (10.5 additional months). With innovation, survival increased to 27.4 and 16.2 months (Δ11.2 months) in the high-scenario with olaparib and TPC, respectively (increase of 0.7 months vs. no innovation); 24.8 and 14.0 months (Δ10.8 months) (increase of 0.3 months vs. no innovation) in the mid-scenario; and 23.8 and 13.1 months (Δ 10.7 months) (increase of 0.2 months vs. no innovation) in the low-scenario. ROV thus yielded a relative increase in quality-adjusted survival with olaparib vs. TPC of 6.1%, 3.2%, and 1.0% in each scenario, respectively. Conclusions: Treatment with 1L olaparib likely improves a patient’s opportunity to benefit from future treatment innovations by extending progression free and overall survival. These findings can inform clinical decision-making by guiding early treatment selection to optimize treatment sequencing and positioning, and support further studies that evaluate the ROV of additional guideline-recommended treatments in mBC.
Presentation numberPS5-11-04
Patient Out-of-Pocket Exposure from Initial Staging Breast MRI: Quantifying Financial Toxicity in Breast Cancer Care
Tomas Dvorak, Orlando Health, Orlando, FL
C. Dvorak1, H. Saunders2, T. Dvorak3, J. Smith1; 1Surgical Oncology, Orlando Health, Orlando, FL, 2Breast Imaging, Orlando Health, Orlando, FL, 3Radiation Oncology, Orlando Health, Orlando, FL.
Background:Breast MRI is routinely considered at initial diagnosis to evaluate extent of disease, multicentricity, and contralateral disease in the non-metastatic setting, despite randomized data (e.g., COMICE & MONET) showing little impact on re-excision rates or long-term outcomes. In addition to time delays, each scan may expose patients to out-of-pocket expenses that compound “financial toxicity,” a quality-of-life domain now tracked by ASCO and NCCN. We audited all first-episode breast MRIs at a large community system during 2024 to clarify the economic burden. Methods:Retrospective billing data of all breast cancer MRI scans (ICD-10 C50.x code present in at least 1 of the first 5 diagnosis fields) performed in calendar-year 2024 at a community hospital system was analyzed in this IRB approved study. Cases were included if they had both Hospital (HB) and Professional Billing (PB) available. Only the “initial episode” MRI for each patient, defined as the earliest service date, was used. Primary endpoint was the posted “Patient Payment Amount”. Descriptive statistics including Spearman, Kruskal-Wallis and chi² were performed using Python 3.11 (pandas, scipy) Results:A total of 630 patients underwent a breast MRI in 2024. There was no difference in MRI distribution by service month. CPT code 77049 was billed in 95.4% of scans. Payer grouping included 53.0% Commercial, 32.7% Medicare, 11.7% ACA Marketplace, and 2.7% Military / Self-Paid / Other. Of all “Net Charges”, patients were liable for 13.4 %, insurers covered 75.4 %, and the hospital system had a gap of 11.2 %. For PB, 98.3 % of patients had a $0 copay, and among the 1.7% who did owe, median out-of-pocket responsibility was $94 (IQR $34-$100). The upper 10% of payers owed >$135, and the highest bill was $177 (skew = 0.46). For HB, 63.2% had $0 copay, and among the 36.8% who did owe, median out-of-pocket responsibility was $327 (IQR $171-$648). Ten percent of payers owed >$1,300, and the highest bill was $4,878 (skew = 2.8). For the combined PB + HB exposure, 64.0% of patients had a net $0 copay (including 1.3 % with small credits). Among the 36.0 % who owed anything, median out-of-pocket responsibility was $326 (IQR $174-$645). The upper 10 % of payers owed >$1,290, and the highest total bill was $4,878 (skew = 2.8). Patient responsibility did not show a consistent deductible effect: the proportion of $0 visits rose from 46% in January to 70% in December (Spearman p = 0.07), yet mean liability among paying patients fluctuated throughout the year (p = 0.32). Patient cost sharing with $0 co-pay by Payer class varied from 33.8% for ACA Marketplace, 49.7% for Commercial, 50% Military, 93.2% Medicare, and 100% for Medicaid, Self-Paid, and Other. Among patients who incurred >$0 liability, median responsibility was highest in Commercial plans ($453.09), followed by ACA Marketplace ($184.67), Medicare ($70.81), and Military ($30.50). Conclusions: Patient-level “financial toxicity” from an initial staging breast MRI is relatively minimal in our system—63 % of patients paid nothing and the remainder faced a median $327, almost entirely from the technical fee. These findings reassure clinicians that ordering a breast MRI seldom imposes a direct out-of-pocket burden on patients, though up-front cost counselling is important to prevent the rare but substantial financial surprises we identified. Cost-sharing burden fell mainly on ACA Marketplace and Commercially insured patients; Medicare and Medicaid beneficiaries were largely shielded. In a value-based oncology landscape, the decision to order a staging breast MRI should weigh its uncertain clinical benefit against not merely the patient’s copay at the point of care, but also system-level costs of both the hospitals that absorb charity-care write-offs and occasional under-collections, and the payers that cover the bulk of charges.
Presentation numberPS5-11-05
Multidisciplinary Team Decision-Making in Breast Cancer: Real-World Insights from the PRISMA Study
Maria Mafalda CPR Nogueira, MSD Portugal, Lisboa, Portugal
G. Sousa1, A. M. Ferreira2, I. Pereira3, C. Abreu4, D. Simão5, F. Machado6, G. Fernandes7, R. A. Leonor8, J. Fougo9, M. C. Nogueira10, P. H. Meireles11, J. Abreu Sousa12, P. F. Cortes13; 1Serviço de Oncologia, IPO Coimbra, Portugal, Coimbra, PORTUGAL, 2Departamento de Oncologia, IPO Porto, Porto, PORTUGAL, 3Serviço de Oncologia, Hospital da Senhora da Oliveira, ULS Alto Ave, Guimarães, PORTUGAL, 4Serviço de Oncologia, Hospital de Santa Maria, ULS Santa Maria, Lisboa, PORTUGAL, 5Serviço de Oncologia, Hospital dos Capuchos, ULS São José, Lisboa, PORTUGAL, 6Direcção, Centro Académico Clínico Egas Moniz Health Alliance (EMHA), Aveiro, PORTUGAL, 7Serviço de Oncologia, Hospital da Luz Lisboa, Lisboa, PORTUGAL, 8Serviço de Oncologia, Hospital CUF Tejo, Lisboa, PORTUGAL, 9Unidade de Mama, Hospital de São João, ULS São João, Porto, PORTUGAL, 10Medical Departement, MSD Portugal, Lisboa, PORTUGAL, 11Serviço de Radioterapia, Hospital de São João, ULS São João, Porto, PORTUGAL, 12Departamento de Cirurgia, IPO Porto, Lisboa, PORTUGAL, 13Departamento de Oncologia, Hospital Lusíadas Lisboa, Lisboa, PORTUGAL.
Background: Breast cancer (BC) treatment is increasingly complex, with a strong need for coordinated decision-making among specialists within multidisciplinary teams (MDTs). Despite their critical role in optimizing patient care, limited data exist on the structure and functioning of BC MDTs in Portugal. To address this gap, the PRISMA study collected both qualitative and quantitative data to characterize the organization, composition, and operational practices of BC MDT meetings across various regions and healthcare sectors nationwide. Methods: A mixed-methods approach was used to analyze multidisciplinary team practices from January 2022 to June 2023. For qualitative data, a Delphi methodology was applied through a questionnaire developed from a systematic literature review. Sixty-four Portuguese specialists involved in BC MDT meetings during this period were invited to participate. Two rounds of anonymous online voting were conducted from October 2024 to December 2024, using a five-point Linkert scale; consensus was defined as ≥ 80% concordance among responses. For the quantitative data, retrospective aggregated information from MDT meetings were collected. Results: Forty-six specialists from 13 Portuguese centers participated in the Delphi panel, including representatives from 3 cancer institutes, 3 university hospitals, and 9 general hospitals, encompassing the private (3 centers) and public (10 centers) healthcare sectors. Ten centers also participated in the quantitative phase of the study, where MDT meetings have been held for an average of 18 years. During the study period, each center held an average of 88 meetings, with each meeting lasting approximately 2.3 hours. Most teams had 5-10 members (70%), including medical oncologists (100%), breast surgeons (100%), radiologists (90%), radiation oncologists (90%), pathologists (70%), and oncology nurses (60%). Additional medical and other specialties represented in at least one center included gynecology, nuclear medicine, social service, geriatrics, and data managers. These findings were validated by the Delphi panel, which underscored the role of specialized MDTs with core and supplementary members. During the study period, most meetings were conducted in a hybrid format (60%), with presential (40%) and virtual (30%) formats also reported. On average, 15 cases were discussed per meeting, totaling approximately 767 annually. Of these, on average 45 cases were revised, mainly due to missing prior information (70%). Experts participating in the Delphi panel considered MDT meetings crucial for delivering evidence-based, personalized treatment and minimizing patient care disparities. Key challenges identified included time constraints, delays in diagnosis and staging procedures, and staff shortages. Conclusions: MDT meetings are well established in Portuguese centers and align with international recommendations. This study, through a mixed-methods approach, identified both strengths and operational challenges in MDT practices. Experts emphasize their critical role in ensuring evidence-based, patient-centered care. Findings support efforts to standardize and strengthen MDT functioning to ensure high-quality breast cancer care nationwide.
Presentation numberPS5-11-06
Regional disparities and temporal trends in breast cancer staging and time to treatment in Brazil: A 20-Year analysis of the hospital-based cancer registry
Anne Dominique Nascimento Lima, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
A. Dominique Nascimento Lima1, L. Novis Leite Pinto1, L. Martins de Brito Moraes2, A. Mattar3, E. Camargo Millen4, F. Pimentel Cavalcante5, F. Pereira Zerwes6, M. Antonini7, F. Palermo Brenelli8, A. Frasson9, A. Coelho de Oliveira1, L. Mesentier da Costa10, J. Bines11; 1Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 2PPGCAN – INCA, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL, 3Breast Surgery, Hospital da Mulher SP, São Paulo, BRAZIL, 4Breast Surgery, Instituto Américas – Ensino, pesquisa e inovação, Rio de Janeiro, BRAZIL, 5Breast Surgery, Hospital Geral de Fortaleza, Fortaleza, BRAZIL, 6Breast Surgery, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 7Breast Surgery, Hospital do Servidor Público Estadual, São Paulo, BRAZIL, 8Breast Surgery, Universidade Estadual de Campinas, Campinas, BRAZIL, 9Breast Surgery, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 10DIDEPRE-INCA, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL, 11Clinical Oncology, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL.
Background: Breast cancer remains a major public health concern in Brazil, where advanced stage at diagnosis and delayed treatment initiation are key determinants of poor outcomes. In an effort to improve care, Brazil implemented the National Oncology Policy in 2005 and the “60-day Law” in 2012, mandating treatment initiation within two months of diagnosis. However, legal frameworks do not always translate into clinical reality. This study aimed to evaluate long-term temporal trends and regional disparities in breast cancer staging at diagnosis and treatment delays using national-level data from the Integrator of Brazilian Hospital Cancer Registries (IRHC). Methods: This was a cross-sectional, observational study utilizing secondary, anonymized data from the IRHC, which compiles standardized information from over 300 cancer hospitals across Brazil. Data spanning from 2000 to 2019 were included and categorized into three time periods: 2000-2005 (pre-National Oncology Policy), 2006-2012 (post-policy, pre-law) and 2013-2019 (post-law). Outcomes of interest were advanced stage at diagnosis (TNM stage ≥IIB) and treatment delay (defined as initiation >60 days from diagnosis). Statistical analyses included crude and adjusted odds ratios. A multiple logistic regression model incorporated confounders such as age, race, education level, and referral pathway. Analyses were performed using R software, version 4.4.2. All data were public and exempt from ethics review. Results: Among 282,419 breast cancer cases, the mean age was 55.1 years (SD 13.2), with 99.1% female patients and 30.6% identifying as non-white. The Southeast Region accounted for 56.3% of cases. Invasive ductal carcinoma was the most common histology (86.5%). More than half of the patients (55.0%) were diagnosed at an advanced clinical stage, and 54.4% underwent surgery as initial treatment. Adjusted analyses showed that patients diagnosed in the North (OR=1.60, p<0.001), Northeast (OR=1.14, p<0.001), and Midwest (OR=1.14, p<0.001) were more likely to present with advanced disease compared to those in the Southeast, while the South showed a protective association (OR=0.84, p<0.001). Advanced stage was also more common among non-white patients (OR=1.21, p<0.001) and those without partners (OR=1.19, p<0.001). Conversely, age over 50 (OR=0.68, p9 years; OR=0.70, p<0.001), and referral from non-SUS services (OR=0.65, p<0.001) were protective factors.Regarding time to treatment, delays were more frequent in the North (OR=1.23, p<0.001), among non-white patients (OR=1.13, p50 years (OR=1.26, p<0.001). The Northeast (OR=0.78, p<0.001) and South (OR=0.85, p<0.001) showed lower odds of delay. Patients referred from non-SUS providers (OR=0.82, p<0.001) or who were self-referred (OR=0.48, p<0.001) also had shorter delays. Temporal analyses revealed that overall cancer staging trends remained stable or fluctuated without consistent decline, while treatment delays exceeding 60 days significantly increased. Conclusion: This 20-year nationwide analysis highlights persistent and substantial regional disparities in breast cancer staging and treatment delays in Brazil. Despite federal regulations such as the 60-day Law, treatment initiation beyond the legal window remains common, particularly in underserved regions. These findings underscore the need for improved implementation of oncology policies, especially in the North and Midwest, to ensure timely access to diagnosis and treatment. Strengthening referral pathways, increasing healthcare infrastructure, and addressing socioeconomic inequities are critical to narrowing Brazil’s regional cancer care gap.
Presentation numberPS5-11-07
The Global Landscape of Practice-Changing Breast Cancer De-Escalation Randomized Trials
Lina M. Adwer, University of Nebraska Medical Center, Omaha, NE
L. M. Adwer1, C. E. Dougherty2, J. I. Orozco3, J. A. Santamaria-Barria2; 1College of Medicine, University of Nebraska Medical Center, Omaha, NE, 2Department of Surgery, Division of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE, 3Department of Surgery, Saint John’s Cancer Institute at Providence Saint John’s Health Center, Santa Monica, CA.
Introduction Breast cancer (BC) treatment de-escalation aims to reduce treatment morbidity without compromising oncologic outcomes. Several trials have evaluated surgery, radiation, and systemic therapy de-escalation, but their global distribution and downstream clinical impact have not been described. We hypothesized that the U.S. lags other regions in leading BC de-escalation trials, and that geography, sponsorship, journal impact factor (IF), and intervention discipline influence clinical adoption. Methods We conducted a systematic review of published randomized prospective BC de-escalation trials from 1975 to 2024 following PRISMA guidelines. PubMed, Embase, Scopus, and major clinical guidelines (NCCN, ASCO, ESMO, ASTRO, St. Gallen) were queried. We collected trial characteristics, authorship geography, funding source, and 2023 journal IF. A practice-changing designation was defined by its adoption in clinical guidelines, explicit statements in high-impact publications, or real-world adoption evidence. Historic guideline citation was assessed via the Internet Archive’s Wayback Machine. Statistical tests, temporal trends, and multivariable regression were utilized to identify predictors of practice-changing status and guideline citation. Covariates included publication year, U.S. versus non-U.S. origin, intervention type, sponsor, journal IF, pharma involvement, and multi-center design. Results We included 127 randomized prospective trials. 82.7% were multi-center, 53.5% focused on radiation de-escalation, 23.6% on surgery, and 22.0% on systemic therapy. Publications span 1985-2024, with a marked rise in the 2010s (47.2%) and 2020s (36.2%), compared to 15.0% in the 2000s and <1% each in the 1980s and 1990s (p < 0.001). The share of U.S.-led de-escalation trials declined steadily over time, as non-U.S. activity rose worldwide. Of all trials, 21.3% were U.S.-based, and this proportion dropped from 100% in the 1980s to 14.0% in the 2020s. Among all studies, 58.3% were classified as practice-changing based on adoption in guidelines or clinical uptake. Guideline citation rates were 33.9% for NCCN, 22.8% for St. Gallen, 15.0% for ESMO, 12.6% for ASTRO, and 6.3% for ASCO. Only NCCN demonstrated a significant regional bias favoring U.S. trials (p = 0.0141); no such preference was observed in other guideline bodies. Geographically, most studies were led by European investigators (66.1%), followed by North America (25.2%) and Asia (8.7%). U.S.-based studies most often focused on surgical de-escalation (44.4%), while non-US trials more frequently addressed radiation (60.0%). US trials were more likely to appear in high-impact journals (mean IF: 54.4 vs. 34.5), which may partially explain their higher but not statistically significant practice-changing rate (74.1% vs. 54.0%, p = 0.0975). Among 74 practice-changing trials, those published in high-impact journals (IF ≥10) were more likely to be cited in guidelines (OR = 3.38, 95% CI: 1.47-8.04, p = 0.005). In contrast, U.S. origin was not a significant predictor of practice-changing status (OR = 1.15, p = 0.795). Publication year was consistently associated with decreased odds of downstream influence: each year after 2010 was linked to lower odds of practice-changing designation (OR = 0.84, p < 0.001) and guideline citation (OR = 0.86, p < 0.001). Conclusion Our study highlights global research on BC de-escalation, with most modern trials led by non-U.S. investigators. Although U.S.-based trials were more often labeled practice-changing, geographic origin did not independently predict inclusion in guidelines. Instead, publication in high-impact journals was the strongest predictor of downstream influence. U.S.-led de-escalation trials declined to 14% in the 2020s, which suggests a shifting center of gravity in BC de-escalation research.
Presentation numberPS5-11-08
Characterization and survival outcomes of young women with breast cancer in Mexico: 5-year results of the Joven & Fuerte prospective cohort
Fernanda Mesa-Chavez, Tecnologico de Monterrey, San Pedro Garza Garcia, Mexico
F. Mesa-Chavez1, A. Platas2, E. A. Lopez-Martinez3, B. F. Vaca-Cartagena1, A. S. Ferrigno4, A. Fonseca3, M. Miaja3, M. Cruz-Ramos3, J. E. Bargallo-Rocha3, P. Cabrera-Galeana5, A. Mohar6, C. Villarreal-Garza1; 1Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, MEXICO, 2Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, MEXICO, 3Joven & Fuerte, Programa para la Atencion e Investigacion para Pacientes Jovenes con Cancer de Mama, MILC, Mexico City, MEXICO, 4Department of Medicine, Yale University School of Medicine, New Haven, CT, 5Breast Medical Oncology Unit, Instituto Nacional de Cancerología, Mexico City, MEXICO, 6Epidemiology Unit, Instituto Nacional de Cancerología, Mexico City, MEXICO.
Background: Young women with breast cancer (YWBC) comprise up to 15% of all breast cancer (BC) cases in Mexico. Joven & Fuerte, established in 2014, was the first 5-year prospective cohort of YWBC in Latin America. This study describes the clinicopathologic characteristics, treatment regimens, and survival outcomes of the complete cohort. Methods: Females aged ≤40 with recently-diagnosed BC (<3 months) were consecutively recruited at 1 private and 2 public referral centers in Mexico. Demographic data were collected from patient surveys; and clinicopathologic, treatment, and survival data from medical files. 5-year overall survival (OS), distant recurrence-free survival (DRFS), and invasive disease-free survival (IDFS) were estimated using the Kaplan-Meier method, specifying date of diagnosis as the time origin. Survival differences between groups were assessed with the log-rank test and Cox proportional hazard model. Results: 588 patients were included. Their median age at diagnosis was 36 years (IQR 32-38) and most had public healthcare (79%). Clinical stage distribution was: 0 (2%), I (9%), II (43%), III (35%), and IV (11%). The most frequent subtype was Luminal B (31%), followed by triple negative (TNBC) (26%), HER2 positive (22%), and Luminal A (17%). Among 513 (87%) patients with stage I-III BC, surgical management included mastectomy in 69% vs breast-conserving surgery in 22%, and axillary dissection in 56% vs sentinel lymph node biopsy in 35%. Overall, 84% received chemotherapy, 68% radiotherapy, 58% endocrine therapy, and 19% anti-HER2 therapy. Of 324 (55%) patients with stage II-III HER2 positive or TNBC, or stage II hormone-sensitive BC, 233 (72%) received neoadjuvant chemotherapy. Of these, 40% had pathological complete response (pCR). After a median follow-up of 5 years, the full cohort did not reach median survival times for any endpoint. Only patients with stage IV BC reached a median OS of 55 months (95%CI 26-not reached). Stage III was associated with worse OS, DRFS, and IDFS (all p<.001), and TNBC with worse OS (p=0.003) and DRFS (p=0.038). Very young patients (≤30 years) also exhibited worse DRFS (p=0.007) and IDFS (p=0.025). In Cox multiple regression (Table), significant associations remained between stage III and worse OS, DRFS, and IDFS, between TNBC and worse OS and DRFS, as well as between age ≤30 and worse DRFS and IDFS. Conclusions: In this cohort of YWBC, 5-year survival outcomes were significantly worse among patients with advanced stage, TNBC, and ≤30 years at diagnosis. These findings underscore the importance of BC downstaging and increased access to treatment options particularly for patients with TNBC, who could experience improved prognosis with complete treatment regimens. Moreover, this study suggests that patients aged ≤30 might comprise a specific subgroup of extremely young patients who face worse outcomes than YWBC aged ≤40.
$$MISSING OR BAD TABLE SPECIFICATION {A87400C1-D502-45B4-AEFA-3070F58E1464}$$
| OS | DRFS | IDFS | ||||
| HR (95% CI) | p | HR (95% CI) | p | HR (95% CI) | p | |
| Clinical stage | ||||||
| I-II | 1.00 (reference) | – | 1.00 (reference) | – | 1.00 (reference) | – |
| III | 4.63 (2.57-8.33) | <.001 | 3.73 (2.33-5.97) | <.001 | 2.46 (1.69-3.58) | <.001 |
| Subtype | ||||||
| Luminal A | 1.00 (reference) | – | 1.00 (reference) | – | 1.00 (reference) | – |
| Luminal B | 1.79 (0.72-4.43) | 0.200 | 1.78 (0.84-3.75) | 0.130 | 1.46 (0.83-2.55) | 0.200 |
| HER2 positive | 0.79 (0.27-2.36) | 0.700 | 1.20 (0.52-2.75) | 0.700 | 0.83 (0.43-1.60) | 0.600 |
| TNBC | 2.90 (1.19-7.08) | 0.020 | 2.29 (1.09-4.85) | 0.029 | 1.35 (0.75-2.43) | 0.300 |
| Age | ||||||
| >30 years | – | – | 1.00 (reference) | – | 1.00 (reference) | – |
| ≤30 years | – | – | 1.77 (1.10-2.87) | 0.020 | 1.56 (1.02-2.39) | 0.042 |
Presentation numberPS5-11-09
Increasing rates of germline genetic testing in Veterans with breast cancer
Akiko Chiba, Durham VA Medical Center, Durham, NC
K. Gera1, S. Bushan2, S. Colonna3, T. Shruti4, H. Moss5, S. Collins5, C. Menendez5, M. J. Kelley6, V. Passero7, G. McWhirter8, C. Hirsch6, A. Chiba5; 1Medicine, James A. Haley Veterans Hospital, Tampa, NC, 2Medicine, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, 3Medicine, George E. Wahlen VA, Salt Lake City, UT, 4Medicine, James A. Haley Veterans Hospital, Tampa, FL, 5Surgery, Durham VA Medical Center, Durham, NC, 6Medicine, Durham VA Medical Center, Durham, NC, 7Medicine, VA Pittsburgh Healthcare System, Pittsburg, NC, 8Medicine, NA National TeleOncology, Durham, NC.
Background: Despite increasing guideline support for germline testing in breast cancer—including universal testing recommendations from the American Society of Breast Surgeons and expanded criteria from NCCN and ASCO—uptake remains low, particularly among minority and male patients. The Veteran Affairs (VA) Breast and Gynecology Oncology System of Excellence (BGSoE) established in 2022, provides sub-specialized, evidence-based, comprehensive breast cancer care to Veterans throughout the United States via telehealth including rural and ethnic minorities. BGSoE is responsible for developing and updating the VA Breast cancer pathway, which supports universal genetic testing among patients diagnosed with breast cancer at any age. BGSoE utilizes Point of Care (POC) genetic testing and refers patients to the Comprehensive Genetic Service (CGS) for additional testing and counseling when needed. Veterans with breast cancer may undergo genetic testing through BGSoE and CGS or local community care providers. We aim to evaluate rates of guideline directed genetic testing among Veterans diagnosed with breast cancer. Methods: We conducted a retrospective cohort analysis of breast cancer patients recorded in the VA national breast cancer dashboard from May 2020 to May 2025. We assessed rates of germline and somatic testing by treatment setting (VA, including BGSoE vs. community care), sex, ethnicity, and year of diagnosis. Results: Among 13,819 patients diagnosed with breast cancer, 1601 (11.6%) were male. Germline testing was performed in 1475 (10.7%) and somatic testing in 713 (5.2%). Among females 1,320 (10.8%) underwent germline testing; among males, 158 (9.9%) were tested. Among patients treated within the VA (n=5107), 810 (15.9%) underwent germline testing (722 females [15.7%], 88 males [17.4%]). Among patients treated in the community (n=3460), 263 (7.6%) received germline testing (235 females [7.5%], 28 males [7.8%]). Uptake increased annually over time: 1.1% (15/1364) in 2020-21, 3.6% (72/1974) in 2021-22, 5.4% (102/1873) in 2022-23, 8.8% (199/2255) in 2023-24, and 16.2% (1087/6713) in 2024-25.Further analysis of the Veterans who underwent genetic testing within the BGSoE revealed that 147 out of a total of 439 (33.49%) underwent genetic testing; 134/147(91%) were females and 13/147(13%) were males. 69/147 (46.94%) identified as Black or African American, and 66/147(44.9%) as White. 22/147(14.9%) identify as rural. 103/169 (60.95%) of tests ordered through BGSoE were POC testing. Conclusion: Despite guideline recommendations, rates of genetic testing among Veterans remains very low. Our data suggests that the highest testing rates of testing were among Veterans treated by the VA and lowest among Veterans treated in Community Care, though our dataset may not include all testing performed by non-VA providers. Males and Veterans treated in the community continue to be under-tested. The utilization of POC testing by BGSoE and CGS has resulted in incremental increases in genetic testing within the VA system each year and allows us to ensure veterans can undergo genetic testing in concordance with national guidelines. Additionally, POC testing and telehealth platform provides access to genetic testing for minority and rural Veterans, who often face barriers. A limitation of our study includes a lack of complete data on testing performed through Care in the Community. These data support the BGSoE’s ongoing effort to promote comprehensive breast cancer care within the VA system.
Presentation numberPS5-11-10
Implementing an Our Practice Advisory (OPA) to provide clinical decision support for adjuvant CDK4/6 inhibitors in early-stage hormone receptor positive breast cancer: The Mayo Clinic Experience
Sarah Premji, Mayo Clinic, Rochester, MN
S. Premji1, P. Advani2, S. Chumsri2, L. Mina3, J. Taraba1, J. Carroll1, J. Hoppenworth1, E. Cathcart-Rake1, T. Haddad1, B. Rudder1, G. Choong1, D. Idossa1, C. O’Sullivan1, S. Yadav1, K. Ruddy1, P. Peethambaram1, T. Hobday1, M. Goetz1, R. Rao2, B. Ernst3, F. Batalini3, R. Leon-Ferre1, A. Tevaarwerk1, K. Giridhar1; 1Department of Medical Oncology, Mayo Clinic, Rochester, MN, 2Department of Medical Oncology, Mayo Clinic, Jacksonville, FL, 3Department of Medical Oncology, Mayo Clinic, Phoenix, AZ.
Introduction: An Our Practice Advisory (OPA), formerly known as Best Practice Advisory (BPA), is an electronic health record (EHR) tool. OPAs identify key parameters to alert providers of potential interventions that could enhance quality of care. Such functionality is increasingly relevant given the rapid pace of new drug approvals. Within the Mayo Clinic system, we previously identified that amongst Mayo Clinic patients (pts) meeting the MonarchE criteria, 28% of pts did not undergo a documented discussion with their provider regarding the role of adjuvant abemaciclib (Premji, SABCS 2024). Further, a Flatiron study identified that only 40% of eligible pts received abemaciclib (Sandoval, Miami Breast 2025). To address this practice gap, in 2024 we began to implement EHR clinical decision support by creating an OPA trigger for pts eligible for an adjuvant CDK 4/6 inhibitor (CDK 4/6i). Here we report on ‘action taken’ versus not for the OPA and the most common reasons for re-prompts. Methods: The Mayo Breast Disease Group designed an OPA including pts who are within 14 months of a clinical or pathologic diagnosis of high-risk early-stage breast cancer. We leveraged structured data captured in the Epic staging forms (Epic Systems) to identify pts with clinical or pathologic T-stage, N-stage, grade, ± Ki-67/ multigene signature to identify ER+/HER2- pts eligible for adjuvant ribociclib or abemaciclib based on FDA approvals. The OPA was triggered by a patient encounter in oncology when endocrine therapy (but not CDK4/6i) was on the active medication list resulting in a hard stop. The OPA gathered limited details about whether adjuvant CDK4/6 inhibitors were discussed and whether the provider followed a link that resulted in prescribing one—considered an indicator of action taken. In this IRB-exempt quality improvement project, we describe the responses captured in the OPA trigger utilizing Epic tools. Results: The OPA went live on 1/10/2025. Over a 5-month period post OPA initiation, we identified 517 OPA alerts among 332 unique pts. Their median age was 62.5 years, and the majority were female (97%). The median time to resolve the OPA was 17.5 seconds (range 2-567 seconds). 12.6% (65/517) of the time there was ‘action taken’ by selecting the hyperlink to potentially start a treatment plan. Providers selected the following OPA pre-set responses: ‘re-prompt in one month’ 248 (48%), ‘patient not a candidate for therapy’ 48 (9%), ‘patient declines’ 69 (13%), ‘outside oncologist will prescribe’ 36 (7%), ‘not discussed’ 44 (9%), and unknown 72 (14%). The most common free text comments among the 517 OPA alerts (summarized) included: ‘revisit in future’ 108 (21%), ‘too early’ 65 (13%), ‘patient declines’ 66 (13%), ‘outside oncologist will start’ 31 (6%), ‘not a candidate’ 26 (5%), and ‘co-morbidities’ 18 (3%). Erroneous OPA prompting occurred in 14 (3%) who previously failed a CDK4/6i due to toxicity and in 18 (3%) for pts currently on a CDK4/6i. 30 (6%) OPA comments noted that they discussed or started therapy that day. Conclusion: The OPA offered clinical decision support and ensured that clinicians considered prescribing CDK 4/6i in all pts identified as eligible, and captured deferrals. Refinements are needed to ensure appropriate timing of OPA and minimize erroneous firing. The OPA remains active and captures the reason for deferral of adjuvant CDK 4/6i prescription. This data pairs real world decision making to better understand clinically meaningful application of improved invasive disease-free survival (iDFS).
Presentation numberPS5-11-11
A common sense oncology values-to-action framework for patient-centered randomized trials in oncology
Michelle Tregear, National Breast Cancer Coalition, Washington, DC
M. Tregear1, H. Verduzco-Aguirre2, B. Shkabari3, V. Venkatesh4, R. Chidebe5, B. Sirohi6, R. Koven7, H. Scowcroft8, B. Gyawali3, M. Brundage9, E. Eisenhauer10, C. Booth3, C. Jackson11; 1Programs, National Breast Cancer Coalition, Washington, DC, 2Department of Hematology and Oncology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, MEXICO, 3Department of Oncology, Queen’s University, Kingston, ON, CANADA, 4Board, The Max Foundation, Seattle, WA, 5Project PINK BLUE, Health & Psychological Trust Centre, Abuja, NIGERIA, 6Balco Medical Center, Vedanta Medical Research Foundation, Chhattisgarh, INDIA, 7Patient and Family Advisory Council, Kingston Health Sciences Centre, Kingston, ON, CANADA, 8Freelance, Independent Communications Consultant, London, UNITED KINGDOM, 9Departments of Oncology and Public Health Sciences, Queen’s University, Kingston, ON, CANADA, 10Department, Queen’s University, Kingston, ON, CANADA, 11Department of Medicine, University of Otago, Dunedin, NEW ZEALAND.
Background: Despite advances in oncology, too many randomized clinical trials (RCTs) fail to answer the most meaningful question: does this intervention improve the lives of people with cancer? Many trials prioritize regulatory approval pathways, rely on surrogate endpoints with limited patient relevance, and impose burdensome protocols that deter participation—particularly from underrepresented populations. This project aimed to develop a globally informed, patient-centered framework to guide the design and conduct of oncology RCTs and explicitly bring the patient voice into trial planning and decision-making. Methods: We conducted a multi-phase, mixed-methods development process that included literature review, thematic analysis, and international stakeholder engagement. We conducted a scoping review of published and grey literature, patient rights charters, and existing frameworks to identify gaps in patient-centered trial guidance. A draft “Values to Action” framework was developed through thematic synthesis by two authors and revised in collaboration with an international working group. After an initial pilot in New Zealand, seven focus groups were held between April 2024 and January 2025 with individuals affected by cancer in India, the United States, Canada, Europe, South Africa, Ghana, and Brazil. Prior to each session, participants received a survey assessing the importance of each value and associated action on a 9-point Likert scale, along with opportunities for open-text comments. Focus groups, conducted virtually, explored areas of consensus and divergence and generated qualitative feedback for refinement. Quantitative analysis included descriptive statistics and non-parametric group comparisons by age, gender, region, and income level. Thematic analysis of qualitative data from survey comments and discussion transcripts was used to iteratively revise the framework. Results: We received 59 eligible survey responses from the pilot and 72 from the seven subsequent focus groups. Participants were geographically diverse: 19 (26.4%) from the U.S., 17 (23.6%) from India, 15 (20.8%) from Canada, 12 (16.7%) from Brazil, and 9 (12.5%) from African and European countries; 52.8% were from high-income countries. Half were cancer survivors, 12.5% were living with metastatic disease, and others were caregivers or had other experiences of cancer. Quantitative ratings showed strong agreement across all values and actions (median=9). The action with the lowest median score (median=8)—concerning restrictive exclusion criteria—also had the highest frequency (18%) of neutral responses (scores 4-6). Very few items were rated “not important” (scores 1-3), primarily those related to diversity, exclusion, burden of trial related procedures, and compensation for costs incurred. Themes from qualitative analysis emphasized the importance of meaningful endpoints, transparent communication, equitable inclusion, fair compensation, and plain-language informed consent. The final framework consists of nine core values with associated actions. Conclusions: Patients strongly support trial endpoints that reflect real-world impact, including overall survival and quality of life, over surrogate markers such as radiographic response. Our framework reflects a broad, international consensus on what constitutes a patient-centered oncology trial, including a strong emphasis on co-design, consent, and broad inclusion criteria. The resulting framework can serve as a practical guide for patient advocates involved in reviewing trials as well as trialists, ethics committees, funders, and regulators. This work is especially relevant to the breast cancer field, where advocates have long influenced research priorities and trial design. The framework can help extend this impact globally.
Presentation numberPS5-11-13
Patient navigation improves treatment compliance in women with breast cancer: A prospective observational audit from India
Nishu S Goel, Tata Memorial Center, Mumbai, India
N. S. Goel1, V. Nakkina2, N. Desai3, V. Noronha4, R. Hawaldar5, U. Mahantshetty6, J. Aggarwal7, R. A. Vadgaonkar6, A. S. Padhy8, V. Kapu9, N. Annamreddi2, S. K. Sahoo2, R. A. Badwe10; 1TMC Kevat Patient Navigation Program, Tata Memorial Center, Mumbai, INDIA, 2Kevat Patient Navigation Program, Homi Bhabha Cancer Hospital and Research Center, Visakhapatnam, INDIA, 3Kevat Patient Navigation Program, Tata Memorial Center, Mumbai, INDIA, 4Medical Oncology, Tata Memorial Hospital, Mumbai, INDIA, 5Tata Memorial Center Research Administration Council, Tata Memorial Center, Mumbai, INDIA, 6Radiation Oncology, Homi Bhabha Cancer Hospital and Research Center, Visakhapatnam, INDIA, 7Medical Superintendant, Homi Bhabha Cancer Hospital and Research Center, Visakhapatnam, INDIA, 8Surgical Oncology, Homi Bhabha Cancer Hospital and Research Center, Visakhapatnam, INDIA, 9Medical Oncology, Homi Bhabha Cancer Hospital and Research Center, Visakhapatnam, INDIA, 10Surgical Oncology, Tata Memorial Center, Mumbai, INDIA.
Patient navigation improves treatment compliance in women with breast cancer: A prospective observational audit from India Background Timely initiation of treatment is essential for optimizing breast cancer outcomes, yet in low- and middle-income countries (LMICs), barriers such as stigma, limited health literacy, and logistical challenges contribute to high treatment default rates. We assessed whether a patient navigation (PN) program could reduce default rates and improve treatment compliance in newly diagnosed breast cancer patients. Material & methods We conducted a prospective cohort audit among 391 consecutive women registered for breast cancer treatment at a tertiary care center between November 2024 and April 2025. During November-December 2024, 122 patients received standard care without navigation (NoPN cohort). From January 2025, 269 patients received structured support from trained patient navigators (PN cohort). The institutional benchmark was to plan treatment within 21 days and initiate treatment within 30 days of registration. The primary outcome was treatment default, defined as failure to initiate or complete planned treatment. Secondary outcomes included treatment planning and initiation within defined timeframes. Multivariable logistic regression was used to identify factors associated with timely treatment initiation. Results Baseline characteristics (age, education, socioeconomic status) were similar between cohorts. Overall default rates were significantly lower in the PN cohort (18.2%, 49/269) compared to NoPN (34.4%, 42/122; p=0.03). Defaults after treatment planning were also reduced (2.6% vs. 7.3%; p=0.05). Treatment initiation within 30 days was achieved in 58.7% of PN patients (158/269) versus 40.2% in NoPN (49/122; p=0.009), representing a 46% relative improvement. No significant difference was observed in achieving treatment planning within 21 days (43.8% vs. 41.8%; p=0.78). In multivariable analysis, PN was the strongest independent predictor of timely treatment initiation (OR 2.6, 95% CI 1.65-4.19), followed by higher education level (OR 1.27, 95% CI 1.02-1.58). Conclusion Patient navigation reduced default rate by 47% and increased treatment initiation within 30 days by 46%, thereby demonstrating improved treatment compliance and timely treatment initiation. Integration of navigation programs can be considered as an effective low cost strategy to enhance care delivery for breast cancer in LMIC’s.
Presentation numberPS5-11-15
The intersection of social determinants of health and healthcare needs among breast cancer patients: A qualitative content analysis
Anjanette Alise Wells, University of Cincinnati, Cincinnati, OH
A. A. Wells1, D. Wu2, W. Hsu3, B. Lartey4, V. Schwegmann1, C. Morrison4; 1Social Work, University of Cincinnati, Cincinnati, OH, 2Carolina Health Informatics Program, University of North Carolina, Chapel Hill, Chapel Hill, NC, 3Biostats, Health Information & Data Science, College of Medicine, University of Cincinnati, Cincinnati, OH, 4College of Nursing, University of Cincinnati, Cincinnati, OH.
Importance: Breast cancer is a significant public health issue in the United States, representing 32% of all cancer cases among women and accounting for 14% of cancer-related deaths. Disparities in outcomes exist, particularly among low-income populations, including higher rates of incidence, delayed diagnosis, and poorer outcomes. These disparities are also linked to socioeconomic factors like limited access to and understanding of health information (collectively known as health literacy). Individuals with low-income and lower health literacy are more likely to experience poorer cancer outcomes, including delayed diagnosis, reduced treatment adherence, and higher mortality rates. Objective: The purpose of this study was to explore the intersection of Social Determinants of Health (SDOH) and healthcare needs documented in the electronic health records (EHR) of women with breast cancer, highlighting factors such as economic stability, neighborhoods, education, food access, community context, and healthcare systems. Design/Setting: The Kaiser Family Foundation (KFF) framework categorizes SDOH and emphasizes their impact on breast cancer care. This study utilized a deductive qualitative content analysis approach to identify SDOH documented in EHR of female breast cancer patients treated at a mid-sized mid-western academic cancer center from 2017 to 2022. Participants: Records for patients with a breast cancer diagnosis who received treatment (anticancer medications or therapies, radiation, surgery), were alive, and had at least one social work note were included. Breast cancer patients 18 years of age or older were included. Male breast cancer patients were excluded as a possible confounder due to the small sample size. Patients who were deceased or in end-of-life/hospice were also excluded. We randomly selected 200 patients, intentionally oversampling racial/ethnic minority patients to ensure there was balanced representation and a diverse final sample. The sample distribution of N=200 was 65.5% White, 28.8% Black, 2.2% Asian, 0.3% Native American, Alaskan, Pacific Islander, 1.7% Hispanic, 0.7% Multiracial, and 0.8% Other. The majority of patients were of non-Hispanic ethnicity (97.8%). Most of the participants were older adults, with an average age of 65 years old. Marital status included 38% who were married, 23% who were single, and 17.5% who were widowed. Almost half never smoked (49%), nor drank (42.5%), or used illicit drugs (70.5%). Results: Findings demonstrate the interconnectedness of various SDOH and their cumulative effects on patient outcomes. Economic strain, lack of transportation, and inadequate social support emerged as critical themes influencing patient experiences. Additionally, mental health was identified as significant, underscoring the necessity for holistic approaches that address both medical and non-medical factors in breast cancer management. Conclusion/Relevance: This study emphasizes the importance of healthcare providers routinely assessing SDOH to identify at-risk patients and connect them with necessary resources. This is especially important for patients who struggle to understand health information, follow treatment plans, and actively participate in their care. By integrating SDOH evaluations into clinical practice, healthcare professionals can enhance treatment adherence, improve health outcomes, and reduce disparities in breast cancer care. Ultimately, addressing SDOH can lead to better patient experiences, improved quality of life, and more equitable healthcare delivery.
Presentation numberPS5-11-16
Impact of GLP-1 Receptor Agonist Exposure on the Long-Term Survival in Breast Cancer Patients
Joanne Mortimer, City of Hope, Duarte, CA
R. Nelson1, Y. Lai2, S. Kil3, H. Lee3, J. Mortimer4, P. H. Wang5; 1Computational and Quantitative Medicine, City of Hope, Duarte, CA, 2Internal Medicine & Graduate Institute of Clinical Medicine, National Taiwan University Hospital, Taipei City, TAIWAN, 3Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA, 4Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, 5Diabetes, Endocrinology and Metabolism, City of Hope, Duarte, CA.
Background: Obesity is a known risk factor for the development of breast cancer, and weight gain after treatment of early-stage disease is associated with worse outcomes, including cancer-specific outcomes. Some of the proposed mechanisms for adverse outcomes in obese patients include the pro-tumorigenic environment from circulating cytokines, impaired adipokine function, and differential efficacy of systemic treatments. Weight reduction strategies in breast cancer patients have largely been unsuccessful. The GLP-1 receptor agonists (GLP-1RAs) have the potential to produce meaningful weight loss; however, the impact of these agents on breast cancer outcomes has not been well characterized. Methods: Diabetic patients with invasive breast cancer diagnosed between 2009 and 2024 were identified using the City of Hope tumor registry through an IRB approved protocol. Patients were divided into those who had been treated with GLP-1RA after breast cancer diagnosis (cases, n = 232) versus those who had not (controls, n = 722). The groups were compared using t-tests for continuous variables and chi-square tests for categorical variables. Multivariable logistic regression models were used to examine differences between cases and controls, while multivariable Cox proportional hazards models were used to examine differences in overall mortality. Included in the multivariable models were the following covariates: diagnosis year, age group (<55 vs. 55+), race, ethnicity, BMI, tumor stage, tumor grade, ER/PR status, HER2 status, receipt of surgical resection, receipt of hormone therapy (AI and/or SERMs), receipt of chemotherapy, and receipt of radiation therapy. Results: The cases were younger (57 years vs. 62 years, p < 0.001) and had higher BMIs (33 vs. 29, p < 0.001) at diagnosis than controls. Patients were similar across case/control status for the following covariates: year at diagnosis, tumor stage, tumor grade, ER/PR status, and Her2 status. The use of hormone therapy was also similar across groups. Using a multivariable logistic regression model, cases were found to have significantly lower risk of death than controls (OR = 0.33, CI 0.18 – 0.62, p < 0.001). Similarly, a multivariate Cox proportional hazards model showed a significantly lower risk of mortality in cases than controls (HR = 0.34, CI 0.19 – 0.60, p < 0.001). After stratifying for age group, tumor stage, ER/PR status, and Her2 status, mortality risk remained lower for cases who were aged 55+, stage II, ER/PR positive, and Her2 negative (all p<0.05) compared to controls. The remaining subgroups also showed lower risk of mortality in cases, however, sample sizes were too small to reach statistical significance. Conclusion: Our institutional tumor registry data suggest an advantage for the use of GLP-1RA in women with breast cancer who also have diabetes. Well-designed controlled trials are needed to validate these findings. Importantly, we did not find any adverse effect from the use of GLP-1RA in our diabetic breast cancer population.
Presentation numberPS5-11-17
An integrative approach to improving outcomes for inflammatory breast cancer (IBC) patients: creating a diagnostic tool, advocating for ICD-10-CM codes, and establishing a task force to address critical research gaps
Lindsey J. Anstine, Susan G. Komen, Dallas, TX
L. J. Anstine1, A. A. Erwin1, M. L. Guthrie2, H. D. Baker3, S. S. Badve4, M. L. Alpaugh5, K. A. Sabelko1, G. M. Zinser1; 1Scientific Strategy & Programs, Susan G. Komen, Dallas, TX, 2Policy and Advocacy, Susan G. Komen, Dallas, TX, 3Information Technology, Susan G. Komen, Dallas, TX, 4Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, GA, 5Biological and Biomedical Sciences, Rowan University, Glassboro, NJ.
Inflammatory Breast Cancer (IBC) is a rare, highly aggressive form of advanced breast cancer that has been historically challenging to diagnose due to its ambiguous clinical nature. A general lack of awareness about IBC within the clinical and patient communities further contributes to delays in diagnosis and treatment. Together, these issues hinder IBC research advancements, leaving patients with limited treatment options and poor prognoses. To address these challenges, Susan G. Komen partnered with IBCRF and the Milburn Foundation to form the IBC Collaborative and launch a multidimensional initiative, aimed at tackling systemic barriers to the advancement of IBC research and care. Early efforts of this initiative prioritized the development of the IBC Scoring System, a quantitative diagnostic scoring rubric for IBC. Available as an easy-to-use online tool (komen.org/ibc-calc), the IBC Scoring System has been steadily gaining global adoption, through the Collaborative’s targeted efforts to raise awareness including dissemination of informational pamphlets for clinician and patient communities. To date, the tool has been used by over 5300 users in more than 100 countries. Additionally, IBC-dedicated clinics, multi-institutional IBC networks and community oncology centers have reported using the tool to support clinical decision making when considering ambiguous patient cases. Recent validation studies have demonstrated that the system can distinguish IBC from non-IBC patients (AUC-ROC 0.84; 95% CI: 0.82 – 0.87) (Lynce et al. JNCI. 2025). Efforts to further enhance the clinical utility of this important diagnostic system are underway and funded by Komen. With a diagnostic system in hand, Komen led advocacy efforts to pursue formal disease recognition through the creation of IBC-specific diagnosis codes. By engaging with the U.S Centers for Disease Control and Prevention (CDC), three unique ICD-10-CM diagnosis codes for IBC (C50.A0, C50.A1, and C50.A2) were approved. Effective October 2025, the widespread utilization of these codes is expected to enhance the ability to track IBC incidence by facilitating inclusion in national health datasets and to improve the coordination of multidisciplinary care for IBC patients. In parallel, a task force was formed to identify current scientific barriers limiting research advancements in IBC and to provide tangible recommendations to address them. Through a series of structured working meetings, three overarching areas emerged as needing significant investigation and investment to drive progress in the biology of IBC: (1) onset and development, (2) detection and monitoring and (3) metastatic dissemination. The group identified a dearth of available IBC tools and resources as key barriers to progress with data silos and the lack of open-access data remaining major obstacles in the field. Additionally, the group prioritized research areas with high potential to improve the understanding and management of IBC. This integrative approach has made significant strides in supporting IBC research, diagnosis and care through the development of a validated diagnostic tool, the establishment of ICD-10-CM codes for IBC, and by defining critical research gaps and prioritizing resolutions. Continued momentum in these areas should ultimately lead to improved outcomes for people with IBC.
Presentation numberPS5-11-18
Racial and Ethnic Representation in Antibody-Drug Conjugate Clinical Trials for Breast Cancer: A 10-Year Review
Zouina Sarfraz, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
M. Jaramillo1, Z. Sarfraz1, C. Gillespie2, S. Milan2, S. Sukie3, V. Andion Camargo1, F. Akkoc Mustafayev1, V. Podder4, K. A. Qidwai1, M. Ganiyani1, M. S. Ahluwalia1, R. Mahtani5; 1Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 2Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 3Research, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 4Gynecologic Oncology, Mount Sinai Medical Center, Miami, FL, 5Breast Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Breast cancer (BC) remains a leading cause of cancer-related morbidity and mortality among women. Despite therapeutic advances, racial and ethnic disparities persist in both clinical outcomes and research participation. Black women have a 40% higher mortality rate than White women despite similar incidence, and BC incidence is rising among Hispanic women. Antibody-drug conjugates (ADCs) have emerged as a transformative therapeutic class for patients with advanced and metastatic BC. As the use of ADCs expands, ensuring equitable representation in the clinical trials that support their approval is critical. This study aimed to evaluate racial and ethnic representation in interventional trials of ADC for BC. We specifically assessed the inclusion of underrepresented populations, the completeness of demographic reporting, and disparities in enrollment relative to the population most affected by the disease. Methods: We analyzed 25 interventional clinical trials investigating ADCs for the treatment of advanced or metastatic breast cancer (Stage III-IV) with publicly available results posted on ClinicalTrials.gov between 2015 and 2025. Trials were included if they investigated ADCs as the primary therapeutic agent and reported demographic data for enrolled patients. Key trial characteristics, including study phase, sponsor type, geographic region, and participant demographic data, were extracted. The primary objective was to assess racial and ethnic representation across trials and evaluate the completeness and consistency of demographic reporting. Descriptive statistics, including mean, median, standard deviation, and 95% confidence intervals, were calculated to summarize participant representation by race and ethnicity. Results: A total of 8,459 participants were included across 25 clinical trials. Most trials (72%, n=18) focused on Stage IV BC; 32% (n=8) also included patients with Stage III disease. All trials (n=25) reported racial demographics, while only 44% included data on ethnicity (n=11). White patients accounted for the highest median representation at 63.9% (n=4,993, 95% CI: 52.2-81.7), followed by Asian individuals at 20.5% (n=2,257, 95% CI: 6.7-40.0). Black patients were markedly underrepresented, with a mean enrollment of 2.2% (n=220, 95% CI: 1.4-3.6). Among the 11 trials that reported ethnicity, Hispanic patients represented a median of 8.1% (n=440, 95% CI: 4.3-17.6). Participants identified as “other” or not categorized represented 5.2% (n=989) of total enrollment. A majority of trials were multiregional (64%, n=16), with North America and Europe hosting most trial locations, 84% (n=21) and 56% (n=14) respectively. South America held only 20% (n=5) of clinical trials, with none in Africa. Most studies (84%, n=21) were industry-sponsored, while 16% (n=4) were led by academic or government institutions. Conclusion: This analysis reveals persistent underrepresentation of Black and Hispanic populations in clinical trials evaluating ADCs for BC, despite their disproportionate disease burden. The limited and inconsistent reporting of ethnicity, alongside broad racial categories that obscure the inclusion of American Indian, Pacific Islander, and multiracial participants further hampers comprehensive equity assessments. As a meta-research study limited to publicly reported data, our findings may underestimate disparities due to incomplete or inconsistent demographic documentation. Nonetheless, these results underscore the urgent need to prioritize representative enrollment and standardized demographic reporting in future ADC trials. Doing so is critical not only for ensuring equitable access to novel therapies, but also for enhancing generalizability of efficacy and safety outcomes across diverse patient populations in oncology.
Presentation numberPS5-11-19
Insurance Type Predicts Time-to-Approval for Proton Therapy in Breast Cancer
Aren S Saini, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
A. S. Saini, M. T. Gompels, N. F. Khosrowzadeh, K. R. Chambers, K. Samimi, A. P. Dhawan, C. G. Washington, J. J. Meshman; Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL.
Background: Proton therapy (PT) is an advanced form of radiation therapy that offers dosimetric advantages over conventional photon therapy, particularly in sparing cardiac and pulmonary structures in patients with breast cancer. While PT may reduce long-term toxicity and improve quality of life, access remains limited due to cost, availability, and especially insurance-related hurdles. Prior authorization requirements, peer-to-peer reviews, and lengthy appeals can delay care, with disparities in access often correlating with insurance type. This study investigates whether insurance structure influences time-to-approval and denial rates for breast cancer patients receiving PT. Methods: A retrospective analysis was conducted on 111 breast cancer patients treated with PT at a National Cancer Institute-designated Comprehensive Cancer Center between August 2020 and November 2023. Patients were categorized by insurance type: public (Medicare/Medicaid), private (commercial PPO/HMO), and marketplace (Affordable Care Act plans). The primary endpoint was time from initial insurance submission to formal approval. Secondary endpoints included denial rates and success at each appeal stage. Statistical comparisons were performed using Mann-Whitney U tests for approval times and Fisher’s exact tests for denial rates, with significance defined as p < 0.05. Results: Significant disparities in time-to-approval were observed across insurance groups. Public insurance had the shortest mean approval time (16.6 days), followed by private (34.0 days), and marketplace plans (55.6 days). Median approval times mirrored this trend at 8, 27, and 47 days, respectively. All pairwise comparisons were statistically significant (p < 0.01). Marketplace insurance was associated with the greatest variability and highest rate of initial denial (59%), compared to 27% for private and 14% for public plans. Among denied marketplace patients, approval was eventually obtained in 100% of cases, but typically only after undergoing multiple appeal stages, including external review. First-level and peer-to-peer appeal success rates for this group were low at 33% and 31%, respectively, contributing to prolonged authorization timelines. Conclusions: Insurance structure significantly impacts access to PT for breast cancer patients. Marketplace insurance is associated with longer approval times and higher denial rates despite equivalent clinical indications. Public insurance demonstrated the most efficient and consistent approval process. These findings highlight insurance type as a structural determinant of care, independent of medical necessity, and underscore the need for payer reform to ensure equitable access to advanced radiation technologies. Streamlined authorization protocols, insurer-provider collaboration, and standardized clinical criteria may reduce delays and improve equity in cancer treatment delivery.
Presentation numberPS5-11-20
Long Term Impact of Medicaid Expansion on Breast Cancer Mortality
Ishani Dwivedy, Providence Day School, Charlotte, NC
I. Dwivedy1, A. Jordan1, K. J. Carnes2; 1Science, Providence Day School, Charlotte, NC, 2Department of Health Management and Policy, University of North Carolina Charlotte, Charlotte, NC.
Background: Breast cancer remains one of the most commonly diagnosed cancers among women in the United States and is the leading cause of cancer-related deaths in women under 40. In 2010, the Affordable Care Act introduced Medicaid Expansion (ME) to improve healthcare access for low-income individuals, yet only some states adopted the policy. While earlier studies have shown short-term benefits of ME on cancer outcomes, there remains a need for long-term evaluation, especially in age-specific subgroups and across differing levels of healthcare access. Objective: This study investigates the effect of Medicaid Expansion on breast cancer mortality rates in the United States by: (1) comparing states that did and did not expand Medicaid before and after 2015, (2) analyzing differences in breast cancer mortality outcomes between patients under and over age 40, and (3) re-evaluating these trends after excluding states with the fewest oncologists per capita (Wyoming and Alaska). Methods: Using the CDC WONDER database, we analyzed breast cancer mortality data from 1999-2021 across all U.S. states and the District of Columbia. States were categorized based on Medicaid expansion status as of 2015. Breast cancer crude mortality rates (CMR) per 100,000 population were calculated and compared using t-tests and F-tests. Subgroup analyses were conducted for individuals under 40 and those aged 40-64. Additional analyses excluded states with minimal oncology resources. Results: Medicaid expansion was associated with a significantly larger decline in breast cancer CMR in expansion states (6.53 to 5.64, p < .0005) compared to non-expansion states (6.75 to 6.02, p = 0.02). Among individuals aged 40-64, post-expansion breast cancer mortality was significantly lower in ME states compared to NME states (13.50 vs. 14.78, p = 0.01). For those under 40, breast cancer mortality declined in both ME and NME states, but no significant difference was found between the two after 2015 (p = 0.20). Excluding Wyoming and Alaska further strengthened the post-2015 difference between ME and NME states (p = 0.04). Conclusion: Medicaid expansion is associated with a significant long-term reduction in breast cancer mortality, especially among women aged 40-64 who benefit from routine screenings. While younger women also experienced improvements, the impact was less pronounced, suggesting that insurance alone may not eliminate barriers to care in this age group. These findings support the role of healthcare policy in improving cancer outcomes and highlight the need for targeted interventions for high-risk, underserved populations.
Presentation numberPS5-11-21
Evaluation of patient experience from participation in a randomised trial
Phoebe Mila Coco Collins, University of Cambridge, Cambridge, United Kingdom
P. M. Collins1, N. Khitab2, V. Pitsinis2, J. Benson3; 1Clinical School, University of Cambridge, Cambridge, UNITED KINGDOM, 2Department of Breast Surgery, NHS Tayside, Dundee, UNITED KINGDOM, 3Cambridge Breast Unit, Addenbrooke’s Hospital, Cambridge, UNITED KINGDOM.
Background: There are persistent barriers to recruitment of patients into surgical trials, especially those involving the process of randomization whereby there is uncertainty about allocation to an experimental intervention or standard treatment. A key challenge to recruitment relates to discrepant surgeon and patient preferences with lack of equipoise and a belief that outcomes for treatment options may differ in a predictable manner1. Trials involving novel methodology with limited data on efficacy compound these problems of recruitment. Minimal research has been undertaken on the process of randomization from a patient’s perspective and how this removes an element of management choice with a perceived obligation to accept the allocated treatment and not withdraw from the trial after randomization. This study aimed to explore patient experience from participation in a randomised trial evaluating fluorescent navigation with indocyanine green (ICG) for sentinel node localization in early breast cancer2. Methods: A questionnaire was issued retrospectively to all trial patients (n=100) with key themes that explored motivation for trial participation, attitudes to randomization, adequacy of information provision and concerns about standard tracer agents. Almost half (45%) of patients responded with data quantified using a 10 point Likert scale (strongly agree [1], strongly disagree [10]. A one-sample T-test determined significance of deviation from the hypothesized neutral value of six. A mixed-methods approach provided holistic assessment of patients’ experience with open-ended questions generating qualitative data for context. Employing a comprehensive post-study analysis, several positive messages were identified from this INFLUENCE trial that might inform the design and conduct of future surgical trials Results: A principle motivation for trial participation was a desire to help future patients (1.22; p<0.001). Patients appeared comfortable with random allocation to treatment groups (2.32; p<0.001) and were unconcerned about any potential negative impact from being part of the experimental arm (7.5; p<0.01). Pre-trial information was deemed accessible and comprehensive (1.93; p<0.001) and patients had sufficient time to process this before making any final decision on trial participation (1.75; p<0.001). Although patients were unconcerned about radioactive tracer usage (8.05; p<-.001), avoiding exposure to a source of radioactivity was considered an advantage of ICG alone for detection of sentinel nodes (3.67; p<0.001). Conclusion: Results of this questionnaire are reassuring and confirm that any barriers to trial participation are not primarily related to concerns about the randomization process or allocation to an experimental treatment arm. Furthermore, this data suggests that any pre-existing concerns can be mitigated through effective pre-trial engagement and communication with the research team. A strong sense of altruism prevails amongst patients and highlights the importance of emphasizing the benefits of clinical research to prospective trial participants. Optimal and well-written pre-trial information is essential and patients should be allowed adequate time to process this information and reflect upon trial participation. Well-designed consent processes will positively influence enrolment along with transparent dialogue and trust in the healthcare professionals conducting the study. 1) Ingram J, Beasant L, Benson J, et al. Pilot and Feasibility Studies 2022; 8: (46) 2) Pitsinis V, ⋯⋯⋯Benson J Ann Surg Oncol 2024; 31: 8845 – 55
Presentation numberPS5-11-22
Disparities in Mammography Screening in a Welfare-State Healthcare System: Data from 100,000 Women in Israel
Hadas Kadar Sfarad, Kaplan Medical Center, Rehovot, Israel
H. Kadar Sfarad1, R. Magnezi2, O. Eldar Friedman1, O. Weinstein3; 1Breast Surgical oncology, Kaplan Medical Center, Rehovot, ISRAEL, 2Department of Management, Bar-Ilan University, Ramat Gan, ISRAEL, 3Department of Management, Ariel University, Ariel, ISRAEL.
Background: Breast cancer is the most common malignancy among women in Israel and globally. Although early detection significantly improves prognosis, 30% of women in Israel are diagnosed at a locally advanced or metastatic stage. In the early 1990s, Israel implemented a national breast cancer screening program, offering biennial mammograms at no cost to women aged 50 and older at average risk. Such organized programs, which provide free and accessible mammography, have been shown to improve adherence rates. Despite this, a substantial proportion of women do not participate in regular screening. Objective: To evaluate adherence to national mammography screening guidelines among Israeli women and identify sociodemographic and clinical factors associated with adherence. Methods: This population-based retrospective study included 100,000 women aged 35-75, using data collected between 2006 and 2020 from the centralized healthcare registry of Clalit, the largest health maintenance organization (HMO) in Israel. Women with a personal history of breast cancer, prior breast surgery, or known genetic predisposition were excluded. Adherence was defined as the proportion of recommended mammograms completed, categorized as: <0.2, <0.5, <0.8, and ≥0.8. The ≥0.8 group was considered fully adherent. Results: A total of 95,603 women met inclusion criteria. Only 17,533 (18%) demonstrated full adherence (≥80%). Among the adherent group, 4% were Ultra-Orthodox Jewish and 13% were Arab. Smoking status was associated with lower adherence; 80% of adherent women were non-smokers. In diabetic women (n=39513), only 5,929 women (15%) demonstrated full adherence. Hormone replacement therapy (HRT) users were more likely to adhere, with 8,943 (51%) meeting the adherence threshold. Women engaged in other preventive health measures were significantly more likely to adhere to mammography screening. Among the adherent group, the median index for Pap smears was 0.18 (IQR: 0.07-0.30), for occult blood testing 0.33 (IQR: 0.17-0.56), and for colonoscopy 0.35 (IQR: 0.00-0.59). Adherence to pneumococcal vaccination showed high adherence to screening mammography with a median index of 1.0 (IQR: 0.60-1.0). In contrast, receipt of influenza vaccination was not associated with mammogram adherence. Conclusion: Despite the availability of a national, no-cost mammography screening program, overall adherence remains low. Sociodemographic characteristics and engagement in other preventive health behaviours significantly influence adherence. Targeted interventions are needed to improve screening rates, particularly among underserved populations.
Presentation numberPS5-11-23
Timing of Oncotype DX Testing and Its Impact on Treatment Initiation in ER+/HER2-/N0 Breast Cancer Patients
Eleni Kohilakis, Montefiore Medical Center, Bronx, NY
E. Kohilakis1, J. Taylor2, S. Jao3, E. Hakim4, M. Rony4, L. Coe4, S. Harbour4, N. Degrezia4, A. Brooks2, S. Feldman3, A. Gupta3, E. Ravetch3, M. Sheckley3, M. McEvoy3; 1Internal Medicine, Montefiore Medical Center, Bronx, NY, 2General Surgery, Montefiore Medical Center, Bronx, NY, 3Breast Surgery, Montefiore Medical Center, Bronx, NY, 4Breast Surgery, Albert Einstein College of Medicine, Bronx, NY.
Background:Oncotype DX testing plays a central role in guiding adjuvant treatment decisions for patients with early-stage estrogen receptor-positive (ER+), HER2-negative, node-negative (N0) breast cancer. While the assay informs chemotherapy decisions and may help avoid overtreatment, delays in ordering or resulting may contribute to postponed therapy initiation. This study characterizes the timing of Oncotype DX testing in relation to surgery and evaluates its association with initiation of chemotherapy, endocrine therapy (ET), and radiation therapy (RT). Methods:We conducted a retrospective review of 373 ER+/HER2–/N0 breast cancer patients treated at a single academic center between January 2018 and November 2024. Inclusion required an Oncotype DX order and available treatment dates. Patients treated externally or with missing key dates were excluded. Data collected included demographics, tumor characteristics, and dates of definitive surgery, Oncotype DX order/result, and treatment initiation. The primary outcome was time from surgery to first treatment. We also evaluated intervals from surgery to Oncotype order/result and from result to treatment. Results:The cohort had a mean age of 58.9 years (range 30–88) and was predominantly female (98.9%). Racial/ethnic distribution was 57.7% White, 31.3% Black, and 43.3% Hispanic. Most tumors were stage IA (90.1%) with low (55.6%) or intermediate (31.8%) recurrence scores. Oncotype DX was ordered before surgery in 40 patients (8.6%), on the day of surgery in 105 (22.5%), and a mean of 30.2 days postoperatively (SE 2.59) in the remainder. Results became available a mean of 41.1 days after surgery (SE 1.33), excluding 21 patients with preoperative results and one with same-day result. Chemotherapy was the first treatment in 65 patients, initiated a mean of 64.2 days after surgery and 33.7 days after the Oncotype result. ET was first in 135 patients; 13 began therapy preoperatively and 29 started before Oncotype results. Among those who initiated ET postoperatively and post-result, the mean time to treatment was 61.3 days after surgery and 36.4 days after result. RT was the first treatment in 173 patients, excluding 70 who received intraoperative RT. Among these, RT began a mean of 63.8 days after surgery and 28.8 days after Oncotype result. Two patients began ET and RT on the same day, averaging 41 days post-surgery and 25.5 days post-result. Conclusion:Delays in Oncotype DX ordering and resulting were associated with prolonged time to treatment. Patients with earlier testing had shorter delays. When compared to national guidelines, many patients initiated therapy later than recommended. Adjuvant chemotherapy is ideally started within 4–6 weeks and no later than 12 weeks post-surgery; our cohort averaged over 9 weeks, with some beyond 12 weeks. RT is recommended within 6–8 weeks of surgery (or 4–6 weeks post-chemotherapy), but was frequently delayed. While ET is more flexible, most patients started beyond 6 weeks. Workflow adaptations during the COVID-19 pandemic, including use of core-biopsy Oncotype testing and neoadjuvant ET to bridge surgical delays, may have contributed to these patterns. Early integration of Oncotype testing into the surgical workflow may reduce treatment delays and improve adherence to care timelines.
Presentation numberPS5-11-24
Does Early Awareness of Clinical Trials Translate to Increased Adoption of Results? Factors Associated with Intent to Omit Axillary Surgery Based on the SOUND Trial
Ko Un Park, Dana-Faber Cancer Institute/Brigham and Women’s Hospital, Boston, MA
K. Park1, M. Delisle2, K. Eum3, M. J. Hassett4, C. A. Minami1, R. S. Punglia5, M. Brindle3, E. A. Mittendorf1, T. A. King1; 1Surgery, Dana-Faber Cancer Institute/Brigham and Women’s Hospital, Boston, MA, 2Surgery, University of Manitoba, Winnipeg, MB, CANADA, 3Surgery, Harvard T.H.Chan School of Public Health and Brigham and Women’s Hospital, Boston, MA, 4Medical Oncology, Dana-Faber Cancer Institute/Brigham and Women’s Hospital, Boston, MA, 5Radiation Oncology, Dana-Faber Cancer Institute/Brigham and Women’s Hospital, Boston, MA.
Background: Oncology trials typically take a decade to mature. We hypothesized that anticipating results may facilitate early implementation. The SOUND trial, published November 2023, demonstrated that sentinel lymph node biopsy (SNB) omission is oncologically safe for select early-stage breast cancer (EBC) patients. Here we evaluate if timing of awareness of the SOUND trial translates to increased intent to implement the SOUND trial results. Methods: This multicenter, cross-sectional study surveyed a purposive sample of North American physicians from August to December 2024 (US) and March to May 2025 (Canada) on current SNB use, when they became aware of the SOUND trial and intent to implement the trial results. Two sets of questions assessed intent on a 5-point scale: 1) Validated measure by Moulin (Innovation-Specific Implementation Intentions); 2) case questions on likelihood of omitting SNB for a 1.5cm grade 2, luminal A EBC with different patient ages and radiation plans (whole vs partial breast irradiation; WBI, PBI). The outcome of interest was intent to omit SNB to a “high extent”, defined as survey answers of ‘moderate’ to ‘very great extent’. Multiple logistic regression was performed to identify factors significantly associated with intent. Results: The overall response rate was 21% (208/1004), including 145 (72%) surgeons, 32 (16%) medical and 27 (13%) radiation oncologists. Most respondents (73.6%) were from the US; 26.4% were from Canada. The mean year respondents became aware of the SOUND trial was 2020 (range 2011-2024). At the time of the survey, 47.8% (n=99) of respondents had already omitted SNB in select patients < 70 years with EBC and 63.6% had high intent to use SOUND trial results. Among those who hadn’t omitted SNB (n=108), 59.3% had high intent to do so. Each additional year of awareness of the SOUND trial was associated with a 13% increase in the intent to omit SNB in patients < 70 years (OR 1.13, p = 0.01). Compared to physicians practicing in Western US, those in the Northeast US had a significantly higher intent to omit SNB (OR 3.87, p=0.02) but Midwest, South, and Canada did not. In the case-based question of a 60-year-old patient undergoing lumpectomy and WBI, 50.5% highly recommended omitting SNB but this decreased when the scenario changed to PBI or age 50 (Table 1); intent to omit SNB was not impacted by length of awareness of SOUND trial, years in practice, type of practice, proportion of clinical practice in breast or specialty. However, regional differences were noted with those in the Southern region and Canada being less likely to omit SNB compared to the West (OR 0.3, p=0.04; OR 0.27, p=0.03; respectively). Conclusions: Early awareness of the SOUND trial and regional differences were associated with higher intent to omit SNB. Further research is needed on dissemination strategies of trial concepts while studies are underway.
| Low (n,%) | High (n,%) | |
| Age 60, WBI | 103 (49.5) | 105 (50.5) |
| Age 60, PBI | 154 (74.0) | 54 (26.0) |
| Age 50, WBI | 160 (76.9) | 48 (23.1) |
| Age 50, PBI | 183 (88.0) | 25 (12.0) |
Presentation numberPS5-11-25
Oncology patient engagement to improve outcomes: the power of patient navigation
SANDRA MARQUES SILVA GIOIA, PinkMapp, Rio de Janeiro, Brazil
S. M. GIOIA, L. Fonseca, I. Santos, M. Rocha, L. Brigagão, R. Costa, M. Moraes, P. Martins, A. Andrade; Medical, PinkMapp, Rio de Janeiro, BRAZIL.
Background: In 2017, a Breast Cancer (BC) Patient Navigation Program was implemented in diagnostic centers of the public health system in Rio de Janeiro, Brazil. This program achieved excellent results, such as reduced time to treatment initiation, reduced mortality from breast cancer, increased overall survival, and increased patient satisfaction with the use of health services. However, one of the greatest benefits of the PNP Rio was promoting patient engagement, which made them proactive in their health care and participated in changes to public policies in the country. Aim: To describe the success of the implementation of a breast cancer patient navigation program in the public health network, from the perspective of patients. Methods: We conducted a longitudinal observational and retrospective study with women over 18 years of age diagnosed with BC. Clinical and sociodemographic data were collected from medical records of the Rio Imagem Diagnostic Center and the Heloneida Studart State Hospital, in Rio de Janeiro, from 2017 to 2024. Interviews were conducted with patients to learn about their participation and that of their friends and family in the trajectory of political achievements on the topic of patient navigation in Brazil. Results: There were 840 BC patients with a mean age of 57 years (26 to 93 years) and 40% with initial staging (0-IIA). The increase in the mean rate for initiation of treatment within 60 days was 9% to 52%. 79 patients died of BC. The 5-year specific survival rate was 92.6%. Survival was higher for patients initially treated with surgery (97.9%, p < 0.0001), histological grade 1 (94.5%, p = 0.002), luminal biological profiles (98.4%, p < 0.0001); and treatment within 60 days (95.3%, p = 0.005). Stratification of the 5-year mortality risk by advanced stage revealed higher mortality among women who were not treated within 60 days (HR = 2.00 [1.23; 3.24]). 100% of patients are satisfied with patient navigation services. Patients participated in the maintenance of the PNP, approaching local politicians and medical managers. Patients helped to draft the first law in the national territory on patient navigation in oncology, Municipal Law No. 7,197/21, which inspired Federal Law No. 14,758/23 on the subject. Conclusion: A Breast Cancer Patient Navigation Program has contributed to increased adherence to the federal law that requires treatment to be initiated within 60 days, contributing to reducing mortality from this disease in patients in the public health system. One of the greatest contributions of the introduction of the patient navigator was the promotion of patient engagement, who became co-responsible for their treatment and allowed the social participation of these patients in the construction of effective public policies for BC control in Brazil.
Presentation numberPS5-11-27
Impact of Protein Energy Malnutrition on Clinical Outcomes in Hospitalized Breast Cancer Patients
Kalaivani Babu, Allegheny General Hospital, Pittsburgh, PA
K. Babu1, S. Rajarajan1, M. Jin1, R. Dileo1, G. Gorecki1, S. Arivazhagan2, C. J. Hilton2; 1Internal Medicine, Allegheny General Hospital, Pittsburgh, PA, 2Medical Oncology, Allegheny General Hospital Cancer Center, Pittsburgh, PA.
Background Protein Energy Malnutrition (PEM) is associated with adverse outcomes in hospitalized patients, yet its impact among those with breast cancer remains poorly characterized. This study evaluates the burden of PEM and its effect on inpatient mortality, length of stay, and hospitalization charges in breast cancer hospitalizations. Methods We analyzed the 2021 National Inpatient Sample to identify breast cancer hospitalizations and cases of PEM using appropriate ICD10 codes. Survey-weighted logistic and linear regressions assessed the association between PEM and clinical outcomes. Models adjusted for age, sex, race, Charlson Comorbidity Index, ZIP code income quartile, hospital location (urban or rural), teaching status, region, and bed size. Results A total of 491634 hospitalizations were identified as breast cancer related. Among these, 98327 patients (20 percent) had a diagnosis of Protein Energy Malnutrition. PEM prevalence was highest in the South (24 percent), followed by the Midwest (18 percent), Northeast (17 percent), and West (15 percent). The cohort had a mean age of 65.2 years, and 63.4 percent were female. Racial distribution included 67.1 percent White, 16.9 percent Black, 9.8 percent Hispanic, and 6.2 percent from other racial groups. More than 60 percent of patients had a Charlson Comorbidity Index score of three or greater. The majority of hospitalizations occurred in teaching hospitals (75.4 percent) and urban facilities (92.2 percent). In adjusted analyses, PEM was independently associated with significantly worse clinical and economic outcomes. Patients with PEM had more than twice the odds of in-hospital mortality compared to those without PEM (adjusted odds ratio 2.41, 95 percent confidence interval 2.25 to 2.59, p less than 0.001). Additionally, PEM was associated with an average increase of 5.3 days in length of stay (beta equals 5.33, 95 percent confidence interval 5.09 to 5.58, p less than 0.001) and 65294 dollars higher total hospitalization costs (beta equals 65294, 95 percent confidence interval 57863 to 72726, p less than 0.001). Conclusion In this national cohort of hospitalized breast cancer patients, one in five had documented Protein Energy Malnutrition. The presence of PEM was strongly associated with increased in-hospital mortality, prolonged hospital stay, and substantially higher hospitalization costs. The disproportionately high burden in the Southern United States underscores potential regional disparities in nutritional screening, access to care, and social determinants of health. These findings highlight the critical importance of early nutritional screening and intervention to improve outcomes in this vulnerable population.
Presentation numberPS5-11-29
Implementation of a Rapid Access Referral Program for Breast Cancer: A Quality Improvement Project
Devin Johnson, University of North Carolina, Chapel Hill, NC
D. Johnson, J. Thomason, J. Vionito, S. Javvaji, K. Reeder-Hayes, J. Aldrich; Oncology, University of North Carolina, Chapel Hill, NC.
Background:Timely access to cancer care is a barrier to many newly diagnosed breast cancer patients, and treatment delays may negatively affect outcomes. This quality improvement project aimed to reduce time from referral to initial provider visit to <72 hours through the implementation of a Rapid Access (RA) Program. Methods:A multidisciplinary working group utilized Lean methodology to develop a RA process map. Referrals for newly diagnosed stage I-III breast cancer as well as high risk breast lesions were considered RA-eligible. Other types of new referrals (e.g metastatic disease, transfer of care) were RA-excluded for the initial roll-out. A dedicated referral nurse navigator (RA NN) had first contact with patients. Surgical oncology advanced practice providers (RA APP) conducted RA intake visits, with subsequent multi-disciplinary consultation. The following metrics were assessed at pre-intervention baseline and analyzed on a monthly basis after the implementation of the RA program in January 2024: navigation time (time from referral to RA NN phone call), turnaround time (time from referral to scheduling RA APP visit), schedule to service (time from scheduling to RA APP visit), and cycle time (time from referral to RA APP visit). Results:The pre-intervention referral turnaround time, schedule to service, and cycle time was 2.9, 11.7, and 14.5 days, respectively for all new breast referrals. Pre-intervention baseline data was collected from July-June 2023. The intervention evaluation period spanned January-December 2024. Table 1 shows the post intervention metrics at the 1-month and 6 through 12-month timepoints for the RA-eligible cohort as well as post intervention metrics for the RA-ineligible cohort along with the percentage change from pre-intervention. The navigation time, turnaround time, schedule to service, and cycle time at 1 month was .5, .7, 2.0 and 2.70 days respectively. Each of the reported metrics showed sustained improvements at the 6 through 12-month time periods except navigation time. RA-ineligible patients saw worsening in turnaround time and cycle time. Conclusion:Participants in the RA program had improved access to timely oncologic care that was durable over the study period compared to pre-intervention programmatic baseline; however, timeliness of care for RA-excluded new referrals lengthened over the same period. Whether resource concentration on RA patients, or other temporal factors such as workforce changes or increased referral volume, were the drivers of decline in timeliness for RA-excluded patients will be further evaluated. Expansion of RA to a wider variety of referral types, is a potential solution to extend RA benefits.
Presentation numberPS5-11-30
Racial and ethnic differences in site of metastasis at presentation and treatment in de novo metastatic triple-negative breast cancer
Jincong Q. Freeman, UChicago Medicine Comprehensive Cancer Center, Chicago, IL
J. Q. Freeman1, W. Guo2, S. Poland2, A. Ravichandran3, M. Hennessy2, R. Nanda2; 1Department of Public Health Sciences; Cancer Prevention and Control Research Program, UChicago Medicine Comprehensive Cancer Center, Chicago, IL, 2Section of Hematology & Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, 3Internal Medicine Residency Program, University of Chicago Medicine, Chicago, IL.
Background: Triple-negative breast cancer (TNBC) is an aggressive disease that disproportionately affects Black patients in the U.S. Racial/ethnic minoritized patients are diagnosed at younger ages, later stages, face barriers in accessing oncology care, and overall experience worse outcomes, despite improvements in early detection and treatment. There remains limited data on racial/ethnic and socioeconomic disparities in disease presentation, treatment receipt, and initiation in metastatic TNBC nationally. Methods: Using the National Cancer Database (NCDB), we analyzed a retrospective cohort of patients diagnosed with de novo metastatic TNBC between 2010-2022. We evaluated 3 metastatic sites, including brain, bone only, and visceral (liver and/or lung). Three treatment modalities were assessed: immunotherapy, chemotherapy, and radiation. Immunotherapy use was restricted to patients treated with chemotherapy from 2019-2022, following the initial accelerated approvals for atezolizumab in 2019 and pembrolizumab in 2020 for metastatic TNBC. Treatment initiation delay (i.e., number of days) was defined as the time from initial diagnosis to treatment start and compared by metastatic disease presentation and across racial/ethnic groups. Multiple linear regression was used to examine racial/ethnic differences in treatment initiation, adjusting for clinical and socioeconomic factors. Regression coefficients (β) and 95% CIs were calculated. Results: Of 16,897 patients included, the mean age was 61.6 years; 62.9% were White, 26.3% Black, 6.9% Hispanic, 2.9% Asian or Pacific Islander (API), and 1.0% Other (per self-report); 12.0%, 24.5%, and 57.4% had brain, bone only, and visceral metastases at presentation, respectively; 55.8% received chemotherapy alone, 44.2% chemoimmunotherapy, and 31.8% radiotherapy as their first line of treatment. Higher percentages of White (12.6%) patients were diagnosed with brain metastasis at presentation compared to 9.2% API, 10.8% Black, and 11.4% Hispanic patients (P = .009). A higher proportion of API patients (27.5%) presented with metastatic bone only disease than White (25.7%), Hispanic (23.5%), or Black (21.8%) patients (P <.001). Among patients with visceral metastases, a higher proportion were Black (59.1%) vs White (57.2%), API (56.4%) and Hispanic (53.3%) (P = .008). There was a shorter time to first-line radiotherapy initiation among patients with brain metastases compared to those without (median days: 29.0 vs 140.0; P <.001). Treatment receipt and initiation varied across racial/ethnic groups. In the adjusted models, Black (β = 3.0; 95% CI, 0.6-5.3) and Hispanic (β = 6.8; 95% CI, 2.6-10.9) patients experienced a longer time to chemotherapy initiation than White patients. Immunotherapy was delayed, on average, for 7.8 days (95% CI, 0.6-15.1) in Black and for 16.6 days (95% CI, 6.0-27.2) in Hispanic as compared to White patients; however, these differences were not significant when further controlling for socioeconomic factors. API, Black, and Hispanic patients also had a longer time to radiotherapy initiation. Compared to private insurance, Medicare was associated with chemotherapy initiation delay (β = 3.0; 95% CI, 0.3-5.7) while Medicaid was associated with immunotherapy initiation delay (β = 12.9; 95% CI, 2.0-23.7). A shorter time to immunotherapy initiation was observed in areas with higher levels of education. Conclusions: This NCDB study found notable racial/ethnic and socioeconomic disparities in metastases at presentation and treatment initiation delays among patients diagnosed with de novo metastatic TNBC the U.S. More effort is needed to alleviate these disparities and to improve care equity and treatment outcomes for all patients with metastatic TNBC.
Presentation numberPS5-12-01
Breast cancer in chile: incidence rising, young women dying, why?
Claudio Salas, Clinica Alemana de Santiago, Santiago, Chile
C. Salas1, Y. Bernal2, I. Delgado3; 1Oncology, Clinica Alemana de Santiago, Santiago, CHILE, 2 Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, CHILE, 3Centro de Epidemiología y Políticas Públicas, Instituto de Ciencias, Universidad del Desarrollo, Santiago, CHILE.
Introduction In Chile, official databases on mortality and hospitalizations systematically record all events, identifying causes using the ICD-10 coding system. Since 2005, the Explicit Health Guarantees (GES) program has provided time-bound treatment and financial coverage for specific conditions, including breast cancer (BC). In 2024, we conducted a descriptive-analytical study using official secondary databases from Chile, covering mortality from 1997 to 2023. We found that the BC mortality rate increased by 19.2% among women under the age of 40, while it decreased by 21% among women aged 40 and older. Objective: To describe the incidence of BC in Chile, with a specific focus on women under 40 years of age. Materials and methods: This is a descriptive study. For incidence, data from the GES program were used as a proxy. We analyzed the period between 2007 and 2023. All data sources had near-complete national coverage. The study variables included: Temporal trends (year of event) through Annual Percentage Change (APC), individual characteristics (age), and disease coding using ICD-10 code “C50” for BC. Due to the case distribution, incidence trends were examined annually. Crude and age-adjusted rates were calculated. Temporal trends were assessed using Joinpoint regression models (Version 5.2). Data were processed using R software (2023). Results According to GES data, from 2007 to 2023:
- The number of new BC cases in women under 40 increased with an APC of 7.42% between 2014 and 2023.
- Among women aged ≥40, incidence rise of BC by an APC of 9.94% between 2020 and 2023.
- The crude incidence rate of BC per 100,000 women aged 20-39 increased from 8.7 in 2007 to 19.75 in 2023 (+227%), indicating a concerning upward trend among younger women.
- For women ≥40, the crude incidence rate of BC rose from 104.82 in 2007 to 162.00 in 2023 (+154%).
- Among women under 40 years old, the APC was 4.82, indicating a faster rise in incidence rates between 2007 and 2023. In contrast, women over 40 years old experienced a more gradual increase, with an APC of 2.15
Conclusions At SABCS 2024, we reported a 19.2% increase in BC mortality among women under 40, with this trend accelerating after 2015, while mortality decreased by 21% in women over 40. In this report, we present incidence data from 2007 to 2023, revealing a faster rise among younger women. It is important to note that the increase in mortality among women <40 is not solely explained by rising incidence. While both age groups have experienced increasing incidence, the magnitude and associated mortality differ. Further research is needed to identify other contributing factors to mortality in women under 40, including lifestyle changes, comorbidities, healthcare access, and educational disparities. Although breast cancer policies in Chile have led to favorable outcomes for women over 40, similar progress has not been observed in younger women, emphasizing the need for age-specific strategies targeting this demographic.
Presentation numberPS5-12-02
Quantifying the value of progression-free survival in breast cancer treatment among a general population of US women
Jaehong Kim, FTI Consulting, Los Angeles, CA
J. Kim1, K. Spurrier1, M. Jakobsson2, B. Hauber3, J. Doan4, B. Korytowsky5, J. Shafrin1; 1ECON, FTI Consulting, Los Angeles, CA, 2Department of Medical Affairs, Pfizer AB, Stockholm, SWEDEN, 3Patient Preference Integration, Pfizer Inc., New York, NY, 4Breast Cancer and Precision Medicine, Pfizer Inc., New York, NY, 5Healthcare Innovation Center, Pfizer Inc., New York, NY.
Title: Quantifying the value of progression-free survival in breast cancer treatment among a general population of US women. Background: While progression-free survival (PFS) often serves as a surrogate endpoint in metastatic breast cancer (mBC) treatment approvals, it is unclear whether patients prefer PFS to overall survival (OS), or whether patients are willing to accept greater uncertainty in OS in exchange for more immediate PFS gains. Methods: A nationwide survey was administered to US women aged ≥ 21 years. The survey asked respondents to imagine they were a mBC patient choosing between two hypothetical mBC treatment options (Table): a “certain OS” option (PFS=5.5 months, OS=24 months) versus an “uncertain OS” option defined as having higher PFS (8 months) and uncertain OS with the possibility of both higher OS (80% chance of OS=28 months) as well as lower OS (20% chance of OS=22 months). A multiple random staircase design was used to iteratively adjust OS probabilities in the “uncertain OS” option until the respondent was indifferent between the “certain OS” treatment option with 5.5-month PFS and the “uncertain OS” treatment option with 8-month PFS. The point of indifference was estimated by taking the midpoint between the maximum and minimum expected OS values for the “uncertain OS” treatment option, as observed in the final question of the multiple random staircase. The value of the increase in PFS was the difference between the expected OS of the “uncertain OS” option at the indifference point and the risk-neutral expected OS (26.8 months). Results: The 306 respondents had an average age of 52 years, 94% were insured, 76% were white, and 52% had a bachelor’s degree or higher. Among these respondents, 53% preferred the “uncertain OS” option with greater PFS over the “certain OS” option with lower PFS at baseline. However, preferences were highly heterogeneous; 19.6% preferred the “uncertain OS” treatment even when the probability of lower OS (22 months) was 90% whereas 24.8% did not choose to take any risk and always preferred the “certain OS” option, even if the probability of greater OS (28 months) was 95%. On average, respondents valued the “uncertain OS” treatment profile more as they were willing to forego 0.96 months of certain OS in exchange for a 2.5-month increase in PFS and the possibility for extended survival. Conclusion: On average, women attributed more value to increases in PFS even if this meant there was some possibility of a lower OS. However, preferences varied significantly across individuals in the sample. Because, on average, patients are willing to trade off OS for increases in PFS, shared decision-making processes should be considered to ensure mBC patient care aligns with individual preferences.
| Indifference Point for Expected OS | N | % | |
| Risk Averse (n=144, 47.1%; without considering respondents’ choice due to PFS duration) | 27.85 | 76 | 24.8% |
| 27.55 | 23 | 7.5% | |
| 27.25 | 24 | 7.8% | |
| 26.95 | 21 | 6.9% | |
| Risk Neutral (Baseline) | 26.80 | – | – |
| Risk Accepting (n=162, 52.9%; without considering respondents’ choice due to PFS duration) | 26.20 | 43 | 14.1% |
| 25.00 | 41 | 13.4% | |
| 23.80 | 18 | 5.9% | |
| 22.60 | 60 | 19.6% |
Presentation numberPS5-12-03
Improving Clinical Trial Awareness Through Community Education: A Retrospective Survey of Diverse South Florida Participants
Ana Sandoval-Leon, Miami Cancer Institute, Miami, FL
A. Sandoval-Leon1, L. Dumeny2, L. Carcas1, C. Thiry1, Y. Chamorro1, M. Roy1, M. Rubens1, N. Dempsey1, R. Mahtani1; 1Medical Oncology, Miami Cancer Institute, Miami, FL, 2Family Medicine, Florida International University, Miami, FL.
Background: Clinical trials (CTs) are essential for advancing breast cancer (BC) treatment, yet participation remains low, especially among underrepresented groups due to barriers such as mistrust, limited awareness, and perceived burdens. Community-based educational interventions have proven effective in overcoming these barriers. This study retrospectively evaluated changes in group-level knowledge and willingness to participate in CTs before and after a series of community presentations conducted in diverse South Florida communities. Methods: Adults aged 18 or older residing in Palm Beach (PB), Broward (B), or Miami-Dade (MD) counties attended one of three community-based educational presentations on CTs. Presentations were conducted in English (E) in PB and B, and in Spanish (S) in MD and included patient testimonials. Educational sessions were promoted through grassroots community outreach efforts and were funded by the V Foundation for Cancer Research. Anonymous pre- and post-surveys were distributed to assess self-reported understanding of CTs and willingness to participate. Surveys were unlinked and not coded to track individual responses over time, therefore only group level comparisons were possible. One identical question assessing willingness to participate in a clinical trial was included in both pre- and post- surveys at all 3 events. The MD (Spanish-language) event included a second repeat question. Although data collection was originally intended for educational purposes, de-identified responses were retrospectively analyzed after obtaining IRB exemption for secondary use. Chi-square tests were used to compare pre- and post- event responses and to assess differences between E and S-language events. Results: A total of 115 individuals participated across the three CT educational events: 29 in PB, 42 in B, and 44 in MD. Overall, 84% of participants reported understanding the importance of CTs prior to the event, which increased to 100% post-event (p<0.05). Following the sessions, 77% indicated an increased willingness to participate in a CT and 96% reported improved understanding of the types and significance of CT. Among those who were initially hesitant to participate, the most frequently cited concern (33%) was fear of side effects or doubts about treatment efficacy. Additionally, 25% of respondents reported they had never been offered the opportunity to participate in a CT, despite expressing interest. In the S-language MD event, willingness to participate increased from 45% pre-event to 74% post-event; however, this change was not statistically significant (p=0.23). Minimal differences were observed between E and S-language events in terms of overall attitudes and response patterns. Conclusion: This study demonstrates that community-based education can enhance group-level awareness and interest in CTs. The use of patient testimonials and culturally tailored outreach likely contributed to the observed improvements in understanding and willingness to participate. However, the unlinked survey design, small sample size, retrospective analysis, and limited number of repeat questions restrict interpretation of individual-level change and consistency across groups. The gap between interest and opportunity highlights the need for more inclusive recruitment strategies. Future efforts should focus on sustained education, proactive outreach, and trust-building to address misconceptions and improve access. Targeting these efforts at the point of recruitment may help increase CT diversity and participation.
Presentation numberPS5-12-04
Initial factors affecting recruitment of Black women in the U.S. Mid-South region diagnosed with or at elevated risk for de novo metastatic breast cancer
Janeane N. Anderson, University of Tennessee Health Science Center, Memphis, TN
A. N. Curry1, Y. Hull1, B. Johnson2, F. Mzayek3, C. Hogea4, S. Meyers4, G. A. Vidal1, J. N. Anderson2; 1West Cancer Center and Research Institute, West Cancer Center and Research Institute, Germantown, TN, 2Department of Community and Population Health, College of Nursing, University of Tennessee Health Science Center, Memphis, TN, 3School of Public Health, University of Memphis, Memphis, TN, 4Gilead Sciences, Inc., Gilead Sciences, Inc., Foster City, CA.
Background: Data from the Surveillance, Epidemiology, and End Results program, supported by internal findings from the West Cancer Center and Research Institute (WCCRI), show that Black women are nearly twice as likely as White women to be diagnosed with advanced-stage or de novo metastatic breast cancer (dnMBC). Disparities in dnMBC incidence and mortality remain particularly pronounced among Black women living in the southern United States, contributing to poorer clinical outcomes and higher mortality. The multilevel, culturally specific factors that shape these disparities remain inadequately understood. We designed this mixed methods study, utilizing clinical and self-report quantitative data, patient interviews, and shadowing, to explore how socioecological factors influence healthcare-seeking behaviors and potential challenges to accessing appropriate healthcare. Methods: The observational study employs a multimodal research design informed by the socio-ecological model and critical race theory to achieve three objectives: a) explore barriers that influence screening behaviors and healthcare utilization among Mid-Southern Black women with or at high risk for dnMBC; b) identify unreported structural barriers in social and clinical environments; and c) explore clinician perceptions of healthcare quality received by Black patients. A total of 150 participants will be enrolled—75 with dnMBC and 75 identified as high-risk (e.g., self-report of known mutations or known family history of breast cancer via blood relative or self-report of more than two lifetime breast biopsies). Data collection methods include semistructured interviews with photo elicitation, field observations, and ethnographic “go-alongs” in participants’ homes, neighborhoods, and clinical settings. Eligible patients are identified through the WCCRI electronic medical record (EMR) and screened against inclusion criteria. Race- and gender-concordant study staff contact eligible individuals in Arkansas, Tennessee, and Mississippi via phone and/or email using an IRB-approved script. Results: As of June 20, 2025, 219 WCCRI patients have been screened, with 30 of the planned 150 participants enrolled (dnMBC: n = 19; high-risk: n = 11). Participants averaged 21 days between the date of consent and first study activity (dnMBC=21.54; high risk=20.29). Recruitment challenges include structural and contextual issues (e.g., EMR screening tool, social determinants of health (SDoH), and engagement approaches). Specifically, the EMR screening tool failed to distinguish between eligible living and deceased patients, resulting in 31 ineligible cases (five high-risk, 26 dnMBC). Patients were later identified as deceased after exhaustive verification with hospice care teams, funeral homes, and obituary searches. SDoH challenges were participant work obligations, caregiver responsibilities, and social support needs. Additionally, high-risk patients who attend clinic visits less frequently were less reachable via the EMR, which resulted in the addition of community-centric recruitment strategies. Early recruitment successes include sample diversity (e.g., participant age, income), geographic dispersion, and low study attrition. Conclusions: The ACCESS study seeks to uncover the underlying drivers of dnMBC disparities among Black women in the U.S. Mid-South region. Preliminary findings highlight the willingness of this population to engage in research when approached by race- and gender-concordant researchers and the complex, often unseen challenges that justify the study’s focus and innovative study design. Early challenges affirm the critical need for tailored, community-informed approaches in disparity-focused cancer research.
Presentation numberPS5-12-06
Supporting language access for persons with a breast cancer diagnosis
Ana María López, Thomas Jefferson University, Philadelphia, PA
J. Kim1, J. Witkowski2, N. Nikita2, O. Trachtenberg3, A. López2; 1Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 2Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 3JMG, Thomas Jefferson University, Philadelphia, PA.
Background: Limited English Proficiency (LEP) serves as a barrier to cancer healthcare. LEP may result in miscommunication and misunderstanding around critical cancer care information and decreased engagement around digital health tools which are ubiquitous in cancer care. These factors contribute to treatment non-adherence and reduced participation in clinical trials. LEP is generally addressed by engaging certified medical interpreters or clinicians proficient in the patient’s language, either in-person or virtually as per the CLAS Standards. The Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Thomas Jefferson University serves a diverse patient population across five practice sites. At its central Philadelphia location, approximately 13% of patients document a preference for communication with a language other than English. Methods: To assess our efforts at addressing LEP, we conducted a retrospective chart review to evaluate how often patient language services are documented in the Electronic Medical Record (EMR). Persons with a breast cancer diagnosis who self-identified their language preference as a language other than English and who had a clinic visit in central Philadelphia between January 1 and March 31, 2025 were evaluated. Clinical notes from the most recent clinic visit were reviewed for documentation of interpreter services utilization. Results: Out of 5,054 clinical visits during this period, 326 (6.5%) involved a language preference other than English. Of these, 40 visits (12.3%) were for patients diagnosed with breast cancer. Among these breast cancer patients, the most common preferred languages were Cantonese (30.0%), Spanish (25.0%), Mandarin (22.5%), and Indonesian (5.0%). Interpreter usage was recorded in 18 (45.0%) of the breast cancer patient visits, which aligns closely with the overall clinic rate of 47.9%. For these breast cancer visits:
- GLOBO, the telecom translation service used at SKCCC, was used 5 times (27.8%)-36.5% for the entire clinic population.
- In-person interpreters were used 6 times (33.3%)-26.9% for the entire clinic population.
- A physician fluent in the patient’s preferred language conducted the breast cancer visits on 2 occasions (5.0%)-8.3% for the entire clinic population.
- The interpreter type was unspecified 7 times (38.9%)-22.4% for the entire clinic population.
Overall, the breast cancer data are not significantly different from those of the entire clinic population suggesting data reliability. Conclusion: These data may indicate incomplete documentation of interpreter usage, instances where patients declined interpreter services, or situations where interpreters were not utilized as per CLAS standards. Our next steps include a quality improvement initiative aimed at enhancing language access for patients with a preferred language other than English who are receiving breast cancer care at the Sidney Kimmel Comprehensive Cancer Center by proactively engaging interpreter services at appointment initiation and maintaining that connection virtually via the EMR.
Presentation numberPS5-12-07
Breast Cancer Treatment in Brazil: The Gap Between Incorporation and Real-World Access
Luciana Holtz de Camargo Barros, Oncoguia Institute, São Paulo, Brazil
L. H. Barros1, I. C. de Lima1, P. S. Kindi1, P. F. da silva2, H. N. Esteves1; 1Advocacy, Oncoguia Institute, São Paulo, BRAZIL, 2Oncology, Hospital Israelita Albert Einstein, São Paulo, BRAZIL.
Introduction: Breast cancer is the second most commonly diagnosed cancer worldwide, after lung cancer, and remains the leading cause ofcancer-related deaths among women globally (1). In Brazil, it is the most common cancer, with an estimated 66,280 new cases annually (2). Brazil has a universal healthcare system(SUS), which serves approximately 160 million people, or 75% of the population (3). Ensuring equitable access to breast cancer treatment within such a complex system is a significant challenge.The incorporation of new technologies into SUS is determined by the National Commission for the Incorporation of Technologies into the Unified Health System (Conitec), which conducts Health Technology Assessments (HTA) to decide on their adoption. Theoretically, once incorporated by Conitec, these technologies should be available to all patients cared for by SUS (4). However, Patient Advocacy Organizations frequently support patients who face barriers in accessing approved treatments. To strengthen advocacy efforts and generate evidence regarding the real-world availability of incorporated treatments, a cancer advocacy group in Brazil conducted a study to assess the accessibility of breast cancer medications incorporated by Conitec. Methods: The study focused on all medications incorporated by Conitec for breast cancer until September 2023, namely: trastuzumab and pertuzumab for HER-2 positive metastatic breast cancer (incorporated in 2018) (5), and abemaciclib, palbociclib, and ribociclib succinate for advanced or metastatic HR+ and HER2+ breast cancer (incorporated in 2021) (6). To investigate the availability of these treatments, the Brazilian Freedom of Information Law (7) was used, which allows citizens to request data from public institutions. Between September 2023 and January 2024, requests were made to 268 accredited public cancer centers, asking fortheir treatment protocols for five types of cancer (breast, prostate, colorectal, lung, and melanoma). This publication focuses on the responses related to breast cancer. Results: The total of 42 oncology centers submitted their breast cancer treatment protocols specifying the medications available. Each protocol was analyzed to determine the inclusion of the most recently incorporated Conitec-approved treatments. Of the protocols received, 42 (100%) listed trastuzumab; and 40 (95.2%) listed pertuzumab; but only 2 (4.8%) listed abemaciclib, palbociclib, and ribociclib succinate. The incorporation of trastuzumab and pertuzumab was accompanied by a centralized purchasing strategy by the Ministry of Health. The remaining medications listed were incorporated without a defined purchasing strategy. Conclusion: This study highlights significant disparities in the availability of breast cancer treatments within Brazil’s public healthcare system. Despite their official incorporation, access to these medications remains inconsistent across hospitals. The higher presence of pertuzumab and trastuzumab in the protocols may indicate that the centralized purchasing strategy, implemented at the time of incorporation, ensures broader dissemination of Ministry of Health decisions across hospital protocols. Even with two medications being acquired through centralized purchasing, their availability for patients remains unequal. Moreover, the inclusion of a medication in a hospital’s protocol does not guarantee its consistent availability or use, as factors such as stock shortages and implementation barriers may affect real patient access. The findings emphasize the need for improved mechanisms to ensure that the incorporation of new treatments translates into real-world access for patients. Strengthening monitoring and enforcement strategies is essential to bridge the gap between policy decisions and patient care.
Presentation numberPS5-12-08
Survival Outcomes and Tumor Site Distribution by Molecular Subtype in Breast Cancer Patients Under 40
Muhammad Noorani, University of Texas Rio Grande Valley School of Medicine, McAllen, TX
M. Noorani1, S. Goyal1, S. Viduarri1, A. Calderon1, C. Pham1, S. Singh1, A. Dhasmana1, S. Dhasmana1, M. Yallapu1, S. C. Chauhan1, D. Nguyen1, S. Fofana2, S. Khan1; 1Cancer & Immunology, University of Texas Rio Grande Valley School of Medicine, McAllen, TX, 2School of Mathematical and Statistical Sciences, University of Texas Rio Grande Valley, Edinburg, TX.
Survival Outcomes and Tumor Site Distribution by Molecular Subtype in Breast Cancer Patients Under 40BackgroundScreening guidelines for breast cancer typically begin at age 40, leaving younger women at risk for delayed detection. The survival outcomes and tumor site patterns by subtype in this younger population remain underexplored. Our analysis leverages comprehensive national data to fill this critical knowledge gap, offering one of the most detailed evaluations to date of molecular subtype distribution, prognostic disparities, and anatomical presentation in women under 40. MethodsUsing the SEER 18 registry, we identified 5,026 female patients diagnosed under age 40 with invasive breast cancer in 2018. This year was selected for the most recent data available on SEER with its complete molecular subtype, staging, and anatomical site information. Breast cancer subtypes were categorized as Luminal A (HR+/HER2−), Luminal B (HR+/HER2+), HER2-Enriched (HR−/HER2+), and Triple-Negative (HR−/HER2−). We stratified patients into four age groups (15-29, 30-39, 40-44, 45-49) and used Chi-square tests to assess distributional differences. Kaplan-Meier curves estimated 5-year cancer-specific survival by subtype, with log-rank tests for comparison. Multivariable Cox models, random survival forests, and Mahalanobis distance matching estimated prognostic outcome and treatment effects. ICD-O-3 topography codes (C50.0-C50.9) were used to analyze tumor primary site distributions across subtypes. ResultsAmong women under 40 with breast cancer, Luminal A was the most common subtype (52.6%), followed by Triple-Negative (18.5%), Luminal B (17.0%), HER2-Enriched (6.1%), and unclassified (5.9%). Younger women presented more often with metastatic disease and had lower rates of localized tumors than women over 40 (5.0% and 60.2%, respectively). Luminal B demonstrated the highest 5-year survival probability in the case of young women, followed by Luminal A, HER2-Enriched, URNA, and Triple-Negative, which had the poorest prognosis (log-rank p < 0.0001), whereas Luminal A showed better survival in patients above 40. Black women under 40 had the highest rate of Triple-Negative cancers (22.6%) compared to 20.6% for Hispanic, 18% for Non-Hispanic White, and 12% for other. White/non-Hispanic and other (including Asian) women have a higher percentage of cases diagnosed as Luminal A (53.3% and 58.4%) to 49.3% for Black and 50.2% for Hispanic. Additionally, lower-income women under 40 had more aggressive subtypes and poorer survival than higher-income groups. Those living in rural areas also face significantly worse outcomes than those living in urban metros. The most common tumor site was the upper-outer quadrant (33.0%), especially in Luminal subtypes, followed by overlapping lesions (C50.8, 22.6%) and breast, NOS (C50.9, 14.8%). While Triple-Negative and HER2-Enriched tumors were more often in overlapping or non-specific locations (p < 0.0001). ConclusionsBreast cancer in women under 40 presents distinct biological behavior, survival patterns, and tumor site distributions by subtype. The favorable prognosis of Luminal B tumors highlights the impact of targeted therapies and supports aggressive, subtype-specific treatment in young patients. In contrast, poorer outcomes among young women with Luminal A tumors suggest even traditionally low-risk subtypes require close monitoring. Triple-Negative disease remains the most lethal, emphasizing the urgent need for novel therapies and clinical trial inclusion. Subtype-related differences in tumor localization may inform imaging and surgical strategies. Our findings highlight the need for earlier detection strategies, personalized management approaches that consider tumor subtype biology, genetic risk, and fertility concerns in this high-risk population.
Presentation numberPS5-12-09
Breast cancer patient attitudes and experiences of health care in the post-COVID era: Landscape of recovery and trust at an urban US cancer center
Alyson Moadel-Robblee, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
A. Moadel-Robblee1, C. Garrett2, D. Makower3; 1Albert Einstein College of Medicine, Dept Epidemiology & Population Health, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, 2Dept Epidemiology & Population Health, Albert Einstein College of Medicine,, Bronx, NY, 3Albert Einstein College of Medicine, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY.
Background: The fall-out of COVID-19 on breast cancer care delivery is yet to be fully understood and measured from the vantage point of all stakeholders, particularly the most vulnerable. At a U.S. comprehensive cancer center serving a very diverse, urban, and low-income community that was at the epicenter of the pandemic, identifying the impact on health care trust among these breast cancer patients is an important step in developing a roadmap for full recovery and responsiveness in the backdrop of a changing health care model. Methods: This descriptive analysis of an IRB-approved psychosocial needs assessment study examines attitudes and experiences around COVID-19, cancer care delivery, and medical mistrust from March 2024 to June 2025 among an ethnically diverse and underserved breast cancer patient cohort. Patients (n=135) were referred to a psycho-oncology program by their providers based on interest in counseling or support services. Patients identified as Hispanic (56%) or Black (38%) and were a mean age of 57.15 (25-86 yrs), with a median time since diagnosis of one year. Results: A COVID-19 history assessment found that 52% of patients had a past diagnosis and 28% lost a loved one to the virus. Almost all (94%) received the COVID-19 vaccine for reasons of: protecting self (63%), family (56%), or community (45%), chronic health condition (31%), MD recommendation (41%), reducing worry (38%), and job requirement (39%). While 65% felt getting vaccinated was the right decision and 42% report they are likely to get boosters, 27% regret getting the vaccine and 19% felt it harmed them. Next, patients were queried about their impressions of their most recent health care visit (80% oncology-related) using the Health Service Utilization Scale. Positive to negative ratings were as follows: wait time (85% to 5%), treated respectfully (93% to 3%), clear provider explanations (90% to 4.5%), shared treatment decisions (89% to 4%), private discussion time with providers (95% to 2%), happy with provider (88% to 5%), and facility cleanliness (96% to 1%). Lastly, based on items from the Group-based Medical Mistrust Scale in which “people of my ethnic group” is the reference perspective, 26% of patients reported disagreement with the statement that they receive the same medical care as other groups with 20% endorsing that doctors don’t take their medical complaints seriously and 14% felt they should be suspicious of information from health care workers and modern medicine. Conclusions: This psychosocial assessment of underserved breast cancer patients in the immediate post-COVID period sheds light on a wide range of health care perceptions from high satisfaction with care to substantial levels of medical mistrust and decisional regret. The contrast in these held beliefs may hold an important guidepost in how both the patient and the cancer care system can mutually heal and rebuild its relationship, with dialogue around values, goals, and shared decision making at the center.
Presentation numberPS5-12-10
Impact of KEYNOTE-522 on BneoCT: An Institutional Quality Measure Improvement Analysis
Karishma Vijay Rupani, Icahn School of Medicine at Mount Sinai, New York, NY
K. Rupani1, C. Williams2, P. Klein1; 1Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 2Clinical Data-Quality Operations, Cancer Registry, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: Early stage HER2-positive breast cancer (HER2+) and triple-negative breast cancer (TNBC) are associated with a high recurrence risk. In 2022, BneoCT was formally introduced as a Commission on Cancer (CoC) breast quality measure. The metric focuses on the timely initiation of neoadjuvant therapy in eligible patients. More specifically, BneoCT is a measure of patients 75 years of age or younger, with early stage HER2-positive breast cancer or TNBC, for whom neoadjuvant chemotherapy, targeted therapy, and/or immunotherapy is initiated within 60 days of diagnosis. Methods: We evaluated annual BneoCT performance rates from 2021 to June 2025 at 4 Icahn School of Medicine/Mount Sinai campuses: Mount Sinai Downtown, Mount Sinai Brooklyn, Mount Sinai West, and Mount Sinai Morningside, as reported in institutional quality dashboards. The benchmark for BneoCT is 95%. Results: At our institutions, performance on the BneoCT measure improved steadily over 4 consecutive years (Table 1). This upward trend reflects a 6.25% absolute increase from 2021 to 2024, and a 10.31% absolute increase from 2021 to 30th June 2025. While multiple factors likely contributed, we believe that results from the KEYNOTE-522 trial, which demonstrated that the addition of pembrolizumab to neoadjuvant chemotherapy in high-risk early-stage TNBC significantly improved pathologic complete response (pCR) rates and event-free survival, followed by the National Comprehensive Cancer Network’s (NCCN) endorsement of KEYNOTE-522 in May 2021 (NCCN Version 4.2021), was a critical driver for earlier multidisciplinary coordination and greater utilization of neoadjuvant therapy pathways. The inclusion of pembrolizumab in NCCN Version 4.2021 marked a pivotal shift in practice standards for TNBC, prompting widespread institutional adoption of this immunotherapy-based neoadjuvant regimen. This facilitated earlier referrals and systemic therapy initiation, directly improving adherence to the BneoCT benchmark. Although HER2+ breast cancer cases were not directly affected by KEYNOTE-522, the broader trend toward aggressive, guideline-aligned neoadjuvant therapy likely enhanced care timeliness across both TNBC and HER2+ populations included in BneoCT. Conclusion: The integration of KEYNOTE-522 into the NCCN Guidelines in May 2021 played a substantial role in driving improvements in the BneoCT quality measure over the subsequent years. This quality measure improvement analysis illustrates how high-impact clinical trial data, when rapidly translated into national guidelines, can meaningfully influence patient care outcomes and institutional quality metrics. Sustained performance improvement at or above benchmark levels will require continued investment in multidisciplinary coordination, treatment pathway optimization, and early diagnosis-to-therapy transitions.
| Measure | Rolling Year 2025 Performance Rate [95% CI] | 2024 Performance Rate [95% CI] | 2023 Performance Rate [95% CI] | 2022 Performance Rate [95% CI] | 2021 Performance Rate [95% CI] |
| BneoCT | 95.24% | 91.18% [81.64% – 100.00%] | 86.67% [78.07% – 95.27%] | 81.82% [72.51% – 91.12%] | 84.93% [76.72% – 93.14%] |
Presentation numberPS5-12-11
Clinicopathological Characteristics and Treatment Patterns in Young Women with Breast Cancer at the Brazilian National Cancer Institute
Matheus Feres de Melo, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
M. F. de Melo, A. Boukai; Oncology, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL.
Background: Breast cancer in women aged 40 years or younger is typically associated with more aggressive biological behavior and unfavorable prognosis. In low- and middle-income countries, healthcare system limitations may further impact outcomes. In Brazil, data specifically characterizing this population remain scarce. Objectives: To describe the epidemiological and clinicopathological features, and treatment patterns, among young women with breast cancer treated at a public cancer center. Methods: This retrospective observational study included young women (aged 18-40 years) with breast cancer who were registered at the Brazilian National Cancer Institute (INCA) between October 2022 and October 2023. Data were extracted from medical records. Results: Among 1,321 patients registered at the Breast Cancer Department during the study period, 75 (5.7%) were aged ≤40 years and included in the analysis. The median age at diagnosis was 37 years, and 68% of patients were non-white. A family history of breast cancer was reported in 45.3%, and 5.3% had a family history of ovarian cancer. At the time of diagnosis, 5.3% of patients were pregnant. Invasive carcinoma of no special type was the most common histologic subtype (82.4%), followed by other subtypes (12.2%) and lobular carcinoma (5.4%). Regarding histologic grade, 4.2% of tumors were grade 1, 68.1% grade 2, and 27.8% grade 3. Clinical stage (AJCC 8th edition) at diagnosis was: stage I: 6.7%, stage II: 38.7%, stage III: 34.7%, and stage IV: 20%. T classification: T1: 8.1%, T2: 31.1%, T3: 25.7%, and T4: 35.1%. Nodal status: N0: 41.3%, N1: 37.3%, N2: 16%, and N3: 5.3%. High Ki-67 (≥20%) was observed in 87.5% of tumors. Estrogen and progesterone receptor positivity were 74.3% and 66.2%, respectively; HER2 positivity (IHC 3+ or FISH positive) was observed in 27.4%, and 17% of tumors were triple-negative. Among the 60 patients with stage I-III disease, 78.3% received neoadjuvant chemotherapy. Surgery was performed in 86.7% of patients, with 67.3% undergoing mastectomy and 32.7% breast-conserving surgery. Among those who underwent mastectomy, more than half (54.2%) did not undergo breast reconstruction. Sentinel lymph node biopsy was performed in 61.6%, and axillary dissection in 38.4%. Conclusions: Young women with breast cancer treated at the Brazilian National Cancer Institute appear to present with advanced-stage disease, high-grade tumors, elevated Ki-67, and nodal involvement. Limited access to breast reconstruction was also observed. These findings highlight the need for targeted strategies to improve care delivery and outcomes for this population.
Presentation numberPS5-06-01
Tattoo Ink Fluorescence Interfering with Axillary Reverse Mapping Using Indocyanine Green in Breast Conservation Surgery
Aishwarya Vijay, Marshall University Joan C. Edwards School of Medicine, Huntington, WV
A. Vijay1, B. Andrew1, J. Anderson1, M. Legenza2, D. Krutzler-Berry2; 1Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 2Oncology and Surgery, Edwards Comprehensive Cancer Breast Center, Huntington, WV.
Introduction: Breast cancer-related lymphedema (BCRL) is a significant postoperative complication, affecting approximately 1 in 5 women undergoing surgical treatment for breast cancer. Axillary reverse mapping (ARM) aims to reduce BCRL by differentiating lymphatic drainage paths in the arm versus the breast. Sentinel lymph node biopsy (SLNB) using dual tracers such as radioisotope (RI) and/or blue dye (BD) has been considered the standard of care for early breast cancer surgical management. Recently, indocyanine green (ICG) has been utilized as an alternative for blue dye due to safety and limitations with blue dye utilization. However, ICG and certain tattoo inks can demonstrate fluorescence and therefore limit ICG’s utility in certain cases. We present a case of a 55-year-old woman whose tattoo ink interfered with the ARM procedure. Case Presentation: The patient is a 55-year-old postmenopausal woman diagnosed with ER/PR positive, HER2 negative, Grade 1 Invasive Ductal Carcinoma (IDC) following abnormal findings in the right breast on screening mammography. She underwent right breast lumpectomy with SLNB, using a radioactive tracer and ICG for lymphatic mapping. Intraoperatively, green fluorescence was unexpectedly observed in axillary lymph nodes containing pigment from previous tattoos, raising concerns about false-positive identification of sentinel lymph nodes. Discussion: Tattoo ink-stained lymph nodes are indicated in the literature as a source of interference during SLNB. In this case, the fluorescence of tattoo pigment mimicked that of ICG, complicating intraoperative differentiation between nodes draining the arm lymphatics and those stained by ink. This may lead to over-excision of axillary nodes, under-staging of cancer due to misidentification, or increase the risk of lymphedema. Literature suggests that lipid-bound ICG utilizes the hydrophobic suspension in black tattoo ink to bind to and travel to the lymph node, potentially contributing to the false-positive node findings in this patient. Conclusion: This case highlights a clinically relevant interaction between tattoo ink and ICG during SLNB, which may influence the accuracy of lymphatic mapping and surgical outcomes. It demonstrates the need to expand current understanding of tattoo ink interference in breast cancer surgery guided by ICG fluorescence. Further exploration of the biochemical interactions between ICG and tattoo ink can refine surgical approaches and improve patient outcomes in the context of BCRL prevention.
Presentation numberPS5-06-02
Cdk4/6 inhibitor-induced interstitial lung disease: a rare but severe complication in breast cancer treatment
Stephanie Adel Haddad, UT Health San Antonio/Mays Cancer Center, San Antonio, TX
S. A. Haddad1, D. U. Portillo1, A. M. Nambiar2, M. J. Kim3, V. Kaklamani1, K. I. Lathrop1, R. Pathapati1; 1Hematology & Medical Oncology, UT Health San Antonio/Mays Cancer Center, San Antonio, TX, 2Pulmonary Diseases & Critical Care Medicine, UT Health San Antonio, San Antonio, TX, 3Pharmacotherapy and Pharmacy Services, University Hospital, San Antonio, TX.
Introduction:Cyclin-dependent kinases 4 and 6 (CDK4/6) inhibitors are a class of targeted therapies that inhibit two enzymes involved in regulating the cell cycle. CDK4/6 inhibitors are widely used in patients with hormone receptor-positive (HR), human epidermal growth factor receptor-2 (HER2) negative breast cancer for early-stage and metastatic breast cancer. While these therapies have improved clinical outcomes, they carry a risk of rare but serious adverse events, including interstitial lung disease (ILD)/pneumonitis. We present a case of a patient with CDK4/6 inhibitor-induced grade 4 pneumonitis and ILD, requiring consideration for lung transplantation. Clinical Case: A 61-year-old female with a medical history of hypothyroidism, hypertension, and hormone-receptor-positive breast cancer was found to have a 3 cm irregular spiculated right breast mass and enlarged right axillary lymph nodes. A core needle biopsy revealed invasive ductal carcinoma with confirmed involvement of the right axillary lymph node, estrogen-receptor positive (ER, 90%), progesterone-receptor positive (PR, 90%), HER-2 negative (1+), and Ki-67 20%. She was clinically staged as IIB (T2N1Mx), Oncotype DX score of 28, and negative genetic testing aside from an ATM VUS. She received four cycles of neoadjuvant docetaxel/cyclophosphamide followed by bilateral mastectomy with reconstruction in October 2023. Pathology from the right total mastectomy revealed pT2N2a disease with 6/10 positive lymph nodes. Postoperative course was complicated by a tissue expander infection requiring removal in December 2023. The patient completed adjuvant radiation therapy in March 2024 and began aromatase inhibitor, anastrozole in November 2023, and CDK 4/6 inhibitor, abemaciclib in April 2024. After six months of treatment, she developed acute hypoxic respiratory failure, requiring hospitalization in November 2024. Bronchoscopy with BAL and lung biopsy indicated focal fibrosis with acute and chronic inflammation without infection, granuloma, or malignancy. She was treated with oxygen, high-dose corticosteroids (methylprednisolone 1mg/kg daily), and three doses of infliximab for abemaciclib-induced pneumonitis. Abemaciclib was permanently discontinued, and she was discharged on a prednisone taper in December 2024. The patient has no history of smoking or prior lung injury. She was evaluated by the pulmonology team given her severe, chronic pulmonary fibrosis and advanced interstitial lung disease with worsening hypoxia and dry cough despite prolonged high doses of prednisone. Mycophenolate (MMF) was initiated as a steroid-sparing agent with both anti-inflammatory and anti-fibrotic effects. The patient is referred for lung transplantation evaluation in the setting of a serious, chronic, and irreversible lung disease with poor prognosis and increased risk for sudden, unpredictable, acute, and life-threatening exacerbations. Discussion: CDK4/6 inhibitors are used for HR-positive, HER2-negative breast cancer, with expanded use in high-risk early-stage disease following the NATALEE and monarchE trials. This drug class carries a rare yet life-threatening adverse event that requires early recognition and monitoring. A meta-analysis of 12 randomized trials (>16,000 patients) reported an increased incidence of ILD/pneumonitis in treated patients (1.6% vs. 0.7%; OR 2.12, 95% CI [1.57-2.86], P < 0.00001). The exact mechanisms underlying CDK4/6 inhibitor-induced pneumonitis and ILD remain unclear, and retrospective studies have found no significant correlation between ILD and risk factors such as smoking history, lung metastases, or prior thoracic radiotherapy. This emphasizes the need for early recognition and multidisciplinary management involving medical oncology, pulmonology, and infectious disease teams.
Presentation numberPS5-06-03
Early-stage hormone receptor-positive metaplastic breast cancer treated with chemo-immunotherapy: a case report
Dali Edwards, Medical University of South Carolina, Charleston, SC
D. Edwards1, C. Coleman1, A. Jamil2; 1Hematology and Oncology, Medical University of South Carolina, Charleston, SC, 2Internal Medicine, Samaritan Medical Center, Watertown, NY.
Background: Metaplastic breast cancer (MpBC) is a rare and aggressive histologic subtype, accounting for less than 1% of all breast cancers. It is characterized by two or more histological types, usually mixed epithelial and mesenchymal differentiation. MpBC typically presents with rapidly growing tumors and poor response to standard therapies. The majority of MpBCs are triple-negative. Hormone receptor-positive MpBC is exceptionally rare. Due to the rarity of this subtype, no MpBC-specific treatment guidelines exist. According to current NCCN recommendations, MpBC should be managed similarly to invasive ductal carcinoma of corresponding receptor status. However, outcomes remain significantly worse, with higher rates of chemoresistance, recurrence and distant metastasis. Emerging evidence suggests that PD-L1 expression is present in up to 95% of metaplastic breast cancers, supporting the rationale for incorporating immune checkpoint blockade into treatment. Case: A 54-year-old Hispanic woman presented with a painful, enlarging right breast mass. Diagnostic breast imaging revealed a 7.3 × 4.6 × 5.6 cm irregular, heterogeneously enhancing mass in the superior right breast, along with enlarged level 1 right axillary lymph nodes. Biopsy of the mass confirmed invasive metaplastic carcinoma with focal squamous differentiation, Grade 3, ER positive 84%, PR negative, HER2 IHC 1+, and Ki-67: 67%. Right axillary lymph node biopsy demonstrated benign lymph node tissue. Staging scans showed no evidence of distant metastases. The disease was staged as clinical stage IIIA (cT3, cN0, cM0). MammaPrint testing revealed MammaPrint Ultra High risk (MP2), qualifying her for enrollment in SWOG S2206, a randomized phase III trial evaluating neoadjuvant Durvalumab plus chemotherapy versus chemotherapy alone for adults with MammaPrint ultrahigh (MP2) hormone receptor (HR) positive / human epidermal growth factor receptor (HER2) negative stage II-III breast cancer. She was randomized to the standard chemo-immunotherapy arm, receiving 12 weeks of weekly paclitaxel, followed by dose-dense doxorubicin and cyclophosphamide every 2 weeks for 4 cycles, in combination with durvalumab every 2 weeks. To date, she has reported good treatment tolerance. Further clinical, radiographic, and pathological response assessment is pending and will be presented at the time of the symposium. Conclusion: This case highlights a rare histological subtype of hormone receptor-positive breast cancer and explores the potential role of chemo-immunotherapy in this biologically aggressive disease within the context of a clinical trial. A small prospective study from Memorial Sloan Kettering Cancer Center (MSKCC) demonstrated higher pathologic complete response rates in patients with triple-negative metaplastic breast cancer treated on the KEYNOTE-522 regimen. However, data on the role of immunotherapy in hormone receptor-positive MpBC remain limited. This case may contribute to a better understanding of the potential role of adding immunotherapy to chemotherapy in the treatment of early-stage hormone receptor-positive metaplastic breast cancer and may help inform future directions in managing this rare and understudied subgroup.
Presentation numberPS5-06-04
Cdk4/6 inhibitor in extensive cutaneous metastasis breast carcinoma: a case report
Ricardo Fernández-Ferreira, HOSPITAL CENTRAL SUR DE ALTA ESPECIALIDAD PEMEX, CIUDAD DE MEXICO, Mexico
R. Fernández-Ferreira1, V. Bautista-Piña2, M. Barragán-Dessavre3, G. Cruz-Morales4, R. Vicuña-González2, R. Anaya Jimenez,5, R. Moreno López1; 1ONCOLOGIA MEDICA, HOSPITAL CENTRAL SUR DE ALTA ESPECIALIDAD PEMEX, CIUDAD DE MEXICO, MEXICO, 2PATOLOGIA, HOSPITAL CENTRAL SUR DE ALTA ESPECIALIDAD PEMEX, CIUDAD DE MEXICO, MEXICO, 3DERMATOLOGIA, HOSPITAL CENTRAL SUR DE ALTA ESPECIALIDAD PEMEX, CIUDAD DE MEXICO, MEXICO, 4GINECO-ONCOLOGIA, HOSPITAL CENTRAL SUR DE ALTA ESPECIALIDAD PEMEX, CIUDAD DE MEXICO, MEXICO, 5ONCOLOGIA MEDICA, HOSPPITAL MEDICA SUR, CIUDAD DE MEXICO, MEXICO.
Introduction: Breast cancer is one of the most common malignant tumors affecting women worldwide and Mexico. Cutaneous metastases (CM) occur in 0.7 to 10.4% of all patients diagnosed with cancer although they represent only 2% of all skin tumors. Case report: A 68-year-old female with a positive smoking history. Comorbidities: Chronic obstructive pulmonary disease. Major depressive disorder.History of unspecified right breast cancer, Clinical stage IIB, in 2000, treated with right radical mastectomy and hormonal therapy for 5 years. In 2023, the patient presented with disseminated dermatosis, predominantly on the anterior and posterior thorax and skull, bilateral, characterized by multiple subcutaneous neoplasms measuring 1-3 cm, as well as multiple lymphadenopathy in the cervical and retro auricular regions. In November 2023, an excisional biopsy of a subcutaneous nodule was taken, reporting moderately differentiated infiltrating ductal carcinoma, as well as immunohistochemistry with Estrogen Receptor positive 95%, Progesterone receptor positive 90%, negative HER2, and Ki67 positive in 30%. A Computed Tomography scan of the head-neck-chest-abdomen-pelvis also showed evidence of metastatic disease to the lung, liver, and blastic lesions in the skull, lytic lesions in spine, sacrum, and iliac bones. Examination patient was found with an ECOG score of 2. Her scalp showed multiple subcutaneous nodular lesions with alopecia, multiple cervical and retroauricular lymphadenopathy, and a right mastectomy. She had multiple subcutaneous nodular lesions measuring approximately 1-3 cm, fixed, stony, and painless, in the anterior and posterior regions of the chest. She had bilateral axillary lymphadenopathy, as well as multiple lesions in the abdomen and back. In April 2024, treatment was started with ribociclib 600 mg orally every 24 hours and letrozole 2.5 mg orally every 24 hours plus denosumab. The treatment was well tolerated, with no reported toxicity. During follow-up, all subcutaneous nodular lesions decreased significantly. The lesions were flat and brownish pigmented at that site within the first 3-4 months after treatment. Currently, the skin has returned to normal color at the sites where the lesions were located, and most of the nodules are absent. The scalp has completely disappeared, and hair has reappeared at that site. The latest imaging study (CT scan of the head, chest, abdomen, and pelvis with contrast) reports a partial response with a significant reduction in lesions in the lungs, liver, and bones. Discussion: Extensive CMBC hormone receptor (HR)-positive subtypes are generally present late in the course of the disease and treated with hormonal therapy. CMBC are usually solitary or multiple nodular pinkish lesions (ranging between 1 and 3 cm). The spectrum of presentation of cutaneous metastasis in breast cancer varies in frequency from the common papulonodular variant to much rarer presentations of dermatitis-like metastases. Breast cancer scalp metastases generally present as one or more inflammatory or nodular lesions, telangiectasias, or irregularly shaped skin lesions that are flesh or reddish, making them difficult to identify from other skin carcinomas. The use of aromatase inhibitors (anastrozole and letrozole) in CMBC has been reported; however, treatment with modern CDK4/6 inhibitor-based therapy has not been reported, nor is there any experience with this presentation of the disease. Our case demonstrates that this type of treatment not only acts on visceral and bone disease but also offers adequate penetration at the subcutaneous level, achieving an adequate response in extensive disease. Conclusion: Our case demonstrates that treatment with CDK4/6 inhibitor, achieving an adequate response in CMBC-HR-positive extensive disease and scalp.
Presentation numberPS5-06-05
A Rare Case of Breast Cancer Recurrence Presenting as Laryngeal Metastasis
Divya Samat, Tower Health-Reading Hospital, Reading, PA
D. Samat1, S. Iqbal2, M. Haas3, N. Leasure2, S. Singh1; 1Internal Medicine, Tower Health-Reading Hospital, Reading, PA, 2Hematology/Oncology, Tower Health-Reading Hospital, Reading, PA, 3Radiation Oncology, Tower Health-Reading Hospital, Reading, PA.
Background: Breast cancer is the most commonly diagnosed malignancy in women and the second leading cause of cancer death. While bone, lung, liver, and regional lymph nodes are frequent sites of metastasis, laryngeal involvement is exceedingly rare, accounting for <1% of all laryngeal tumors, with breast cancer representing an uncommon primary source. Diagnosis can be challenging when laryngeal lesions present as the sole clinical manifestation of recurrence. Case Presentation: A 79-year-old woman with history of hormone receptor-positive, HER2-negative, high-risk luminal breast cancer (stage IIB) diagnosed in 2015, previously treated with mastectomy, adjuvant chemotherapy (docetaxel/cyclophosphamide), and seven years of endocrine therapy, presented with progressive hoarseness and a 10-pound unintentional weight loss over four months. She denied dysphagia, decreased appetite, recent infections, neck trauma, surgery, or tobacco and alcohol use. On physical examination, the larynx was midline with mild left-sided tenderness, but no palpable cervical or supraclavicular lymphadenopathy. Flexible laryngoscopy revealed left vocal cord swelling, reduced mobility and fullness of the left aryepiglottic fold. Computed tomography (CT) revealed a left laryngeal mass destroying the thyroid cartilage, associated left level II lymphadenopathy, and lytic bone lesions in the right sixth rib and T11 vertebra, concerning for metastatic disease. Biopsy of the laryngeal lesion confirmed poorly differentiated carcinoma, which was strongly estrogen receptor-positive (100%), moderately progesterone receptor-positive (30%), HER2-negative, high Ki-67 (30%). Immunohistochemistry supported metastatic breast carcinoma. Positron emission tomography-CT showed intense FDG uptake in the laryngeal mass (SUVmax 15.1) and multiple skeletal metastases, without visceral involvement. Intensity-modulated radiation therapy was administered to the larynx for local control and symptom palliation. Antiestrogen therapy with letrozole was initiated, with plans to add a CDK 4/6 inhibitor after the completion of radiation. Given her diffuse bony disease, she was started on monthly denosumab, with radiation planned for symptomatic bony lesions. Discussion: Laryngeal metastasis from breast cancer is exceedingly rare and carries a poor prognosis, typically signaling disseminated disease. Laryngeal involvement usually presents with nonspecific symptoms such as dysphonia, chronic cough, dysphagia, or stridor, which are often misattributed to benign conditions or primary laryngeal cancer, leading to diagnostic delays. Hormone receptor status is a key determinant of prognosis and mortality in metastatic breast cancer, including rare sites like the larynx. ER-positive metastatic disease is associated with longer median survival, whereas triple-negative breast cancer has the poorest prognosis and highest mortality. Notably, receptor status can change between the primary tumor and metastatic sites; thus, biopsy and reassessment of receptor status in metastatic lesions is recommended, as it may impact both prognosis and therapeutic options. Management remains multidisciplinary, with systemic endocrine therapy preferred for hormone receptor-positive disease and localized radiotherapy for symptom control. Conclusion: Laryngeal metastasis from breast cancer, though rare, should be considered in patients presenting with persistent hoarseness or other unexplained laryngeal symptoms. Improved survival among breast cancer patients has increased the risk of metastasis to uncommon sites, underscoring the importance of vigilant follow-up and a high index of suspicion for atypical presentations. Timely diagnosis and individualized therapy are essential to optimize outcomes and maintain quality of life.
Presentation numberPS5-06-06
Case Series on Early Methotrexate Use in Reducing Morbidity in Granulomatous Mastitis
Agnes M Otieno, Howard University, Washington, DC
A. M. Otieno1, D. Dewar1, T. Haroun2, Q. Chu1, S. Nagel1, M. Tee1, R. Williams1; 1Division of Surgical Oncology/Department of Surgery, Howard University, Washington, DC, 2Division of Rheumatology/Department of Internal Medicine, Howard University, Washington, DC.
Background: Granulomatous mastitis (GM) is a rare, benign inflammatory breast condition primarily affecting parous, reproductive-aged women. However, Hispanic and African American women are disproportionately affected. As GM can mimic malignant breast disease, it remains a diagnostic and therapeutic challenge, especially in the absence of standardized treatment guidelines. Methods: A single-institution, retrospective review of all cases of GM sequentially captured from January 2024 to March 2025 at a minority-serving breast clinic. A total of 4 patients were included in the study. Results: All four patients initially presented with breast pain, swelling, and masses concerning for malignancy based on clinical findings and imaging. Biopsies in each case confirmed granulomatous mastitis without evidence of malignancy. Antibiotics and steroids provided partial or temporary relief in three patients, but recurrent or persistent disease led to the application of disease-modifying antirheumatic drug therapy with methotrexate (MTX). Notably, all patients had a negative rheumatologic workup.
| Patient | Age/Sex | Ethnicity | Presentation | Treatment(s) before MTX | Outcome after MTX initiation |
| 1 | 23/F | Hispanic | Right indurated breast mass, with recurrent multiloculated right breast abscess |
– 3 drainage procedures – Antibiotics for 4 months |
Mass significantly reduced in size, with no recurrences of abscess |
| 2 | 42/F | Hispanic | Enlarging painful lump on left breast, development of abscess after biopsy |
– Antibiotics and NSAIDs for 4 months
– Steroids for 1 month
|
Resolution of breast mass, without recurrence of abscess |
| 3 | 59/F | African-American | Two-year history of a progressively enlarging mass, recurrent abscess after biopsy |
– 1 drainage procedure
– ~1-week course of antibiotics
|
Resolution of breast mass, without recurrence of abscess |
| 4 | 54/F | African-American | Indurated lump in the left breast associated with nipple retraction and dimpling | – ~-1-week course of antibiotics | Pending |
Conclusions: This series illustrates the varied clinical spectrum of GM and underscores the importance of a timely biopsy to differentiate it from carcinoma, particularly in women of color. Notably, our case series suggests that early MTX initiation can be valuable, particularly in patients with persistent or recurrent disease despite antibiotics and corticosteroids, which could reduce morbidity and avoid prolonged surgical interventions. Prospective studies are needed to further clarify the role of early MTX in GM management and to establish standardized treatment protocols.
Presentation numberPS5-06-07
A case report of veno-occlusive disease (VOD) in a patient with metastatic triple-negative breast cancer receiving trastuzumab deruxtecan.
Kenna Noel Koehler, The Ohio State University James Comprehensive Cancer Center, Columbus, OH
K. N. Koehler1, K. Noce1, S. Kelly2, G. Bader1, M. Cherian1, A. Davenport1, K. C. Johnson1, N. Lopetegui-Lia1, A. Roy1, D. Quiroga1, S. Sardesai1, D. Stover1, R. Wesolowski1, N. Williams1, S. Vasu3, M. E. Gatti-Mays1; 1Department of Internal Medicine, Division of Medical Oncology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 2Department of Internal Medicine, Division of Gastroenterology, The Ohio State University Wexner Medical Center, Columbus, OH, 3Department of Internal Medicine, Division of Hematology, The Ohio State University James Comprehensive Cancer Center, Columbus, OH.
Background: Antibody-drug conjugates (ADC) are increasingly utilized to treat a variety of solid tumor and hematologic malignancies. Fam-trastuzumab deruxtecan-nxki (T-DXd) is an ADC that combines the anti-HER2 monoclonal antibody, trastuzumab, to a highly potent topoisomerase I inhibitor payload. It is indicated in the treatment of metastatic HER2-positive and HER2-low and ultralow breast cancer. While veno-occlusive disease (VOD) is commonly described in patients with hematologic malignancies, it is rarely seen in patients with solid tumors but has been documented with other ADCs. We present a rare case of VOD that occurred in a patient with breast cancer receiving T-DXd. Case Presentation: A 45-year-old female with triple-negative breast cancer developed a rapid metastatic recurrence following neoadjuvant chemo-immunotherapy, then rapid progression while on first-line sacituzumab govitecan. Given the tumor was HER2-low (IHC 2+, FISH negative), she received second-line T-DXd. After cycle 4 of T-DXd, she developed rapid elevations of total and direct bilirubin as well as transaminases. Abdominal imaging was negative for progression in the liver. Due to further elevations in bilirubin, a liver biopsy was conducted and revealed venous outflow obstruction consistent with VOD. Defibrotide was considered for treatment, however the patient ultimately pursued hospice care. Discussion: VOD results from damage to the small blood vessels in the liver, leading to obstruction of blood flow, volume overload, and potentially liver failure. It can rapidly progress to multiorgan failure if untreated. VOD presents with tender hepatomegaly, rapid weight gain, ascites, and jaundice, often accompanied by elevations in total and direct bilirubin, as well as transaminases. VOD occurs most commonly after hematopoietic stem cell transplantation, and more rarely can occur secondary to treatment with ADCs. The two most implicated ADCs are gemtuzumab ozogamicin and inotuzumab ozogamicin, utilized in hematologic malignancies. Management of VOD includes defibrotide, a thrombolytic agent that stabilizes the endothelium. On review of the literature, there are reports of VOD occurring after treatment with trastuzumab emtansine, another ADC used in breast cancer. However, to our knowledge, there are no reports of VOD occurring after treatment with T-DXd in breast cancer patients. The most commonly reported adverse effects of T-DXd include edema, cytopenias, GI upset, peripheral neuropathy, alopecia, and elevated liver function tests. Rarely, T-DXd can cause decreased ventricular function, pneumonitis, and febrile neutropenia. Our patient received prior chemotherapy in the neoadjuvant and adjuvant setting, which is a well-described risk factor in the development of VOD. VOD is likely underrecognized in the breast cancer population given overlapping symptoms with the metastatic disease itself, such as elevations in serum liver function tests, ascites, jaundice, pain, and weight fluctuations. There is limited data regarding the optimal management and prognosis of patients with breast cancer who develop VOD after treatment with an ADC. Conclusion: We present a rare case of VOD after receipt of T-DXd in a heavily pretreated patient with metastatic breast cancer.
Presentation numberPS5-06-08
Pseudocirrhosis Following Great Response to CDK4/6 Inhibitor and Endocrine Therapy in a Premenopausal Patient With HR+/HER2- Metastatic Breast Cancer
Bruna Migliavacca Zucchetti, Hospital 9 de Julho – Américas Oncologia, São Paulo, Brazil
M. C. Gouveia1, R. Borges1, P. Zanuncio2, B. Zucchetti1; 1Clinical Oncology, Hospital 9 de Julho – Américas Oncologia, São Paulo, BRAZIL, 2Radiotherapy, Hospital BP Mirante, São Paulo, BRAZIL.
Background: CDK4/6 inhibitors revolutionized the treatment of hormone receptor-positive, HER2-negative (HR+/HER2-) metastatic breast cancer (MBC), significantly improving outcomes. In premenopausal patients, their combination with ovarian suppression and endocrine therapy is the standard of care in the first-line. In patients with extensive liver metastases with a great treatment response, rare but clinically relevant complications such as pseudocirrhosis can occur. Recognizing this radiological mimic of cirrhosis is essential to avoid misdiagnosis with disease progression and ensure appropriate management. Methods: We describe a case of pseudocirrhosis in a 43-year-old premenopausal woman with HR+/HER2- MBC and high hepatic tumor burden at diagnosis, who experienced a robust response to first-line treatment. Results: This patient was diagnosed in August 2022 with grade 3 invasive breast carcinoma of no special type, ER 98%, PR 2%, HER2-negative, Ki-67 85%, and extensive liver involvement. Treatment was initiated with tamoxifen plus abemaciclib, transitioning to letrozole following GnRH agonist. Molecular testing revealed no somatic or germline mutations. She experienced radiologic complete response, with only a solitary liver lesion progression in February 2025, which was treated with stereotatic body radiotion therapy.In July 2025, routine labs revealed elevated liver enzymes without clinical symptoms. Laboratory tests showed: ALT 60 (Reference value – RV: 13-35); AST 43 (RV:15-37), GGT 510 (RV<55), ALP 232 (RV: 46-116). Comprehensive evaluation excluded viral, autoimmune, or drug-induced hepatitis, and imaging suggested morphologic features of cirrhosis, without any evidence of disease progression. Despite imaging findings, the patient had no signs of portal hypertension or liver failure. The diagnosis of pseudocirrhosis has been done and started with ursodeoxycholic acid 300 mg BID and vitamin E 800 mg daily to avoid its progression. She continues on first-line treatment, with stable disease and some improvement in liver enzymes.Discussion: Pseudocirrhosis refers to radiologic changes in the liver—such as capsular retraction, nodularity, and caudate lobe hypertrophy—mimicking true cirrhosis, but without histologic confirmation of fibrosis. It has been reported most frequently in breast cancer, particularly in patients with hepatic metastases undergoing chemotherapy. While the exact mechanism is unclear, proposed explanations include extensive tumor regression leading to capsular retraction, nodular regenerative hyperplasia, desmoplastic reaction, and hepatocellular injury due to oxidative stress from rapid tumor lysis. Despite resembling chronic liver disease radiographically, pseudocirrhosis often lacks clinical features of decompensated cirrhosis, making the correct diagnosis crucial. Importantly, up to 75% of women with metastatic breast cancer may exhibit some degree of liver contour irregularity on imaging, underscoring the need for oncologists to correlate radiologic changes with treatment history and clinical context.Conclusion: This case highlights pseudocirrhosis as a rare but important differential diagnosis in patients with extensive liver metastases responding to systemic therapy. Awareness of this entity can prevent misinterpretation of imaging as disease progression or cirrhosis and help guide appropriate management without unnecessary treatment changes.
Presentation numberPS5-06-09
Immunotherapy induced Kikuchi Fujimoto (Histiocytic Necrotizing Lymphadenitis): A Rare Immune Related Adverse Event
Briana To, The Ohio State University Comprehensive Cancer Center, Columbus, OH
B. To, V. Prasath, L. Adorni, G. Bader, M. Cherian, A. Davenport, K. C. Johnson, N. Lopetegui-Lia, D. Quiroga, S. Sardesai, D. Stover, R. Wesolowski, N. Williams, M. E. Gatti-Mays, A. Meara, A. M. Roy; Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH.
IntroductionImmunotherapy (IO) agents can cause a range of rare immune-related adverse events (irAEs) due to immune system activation. Kikuchi-Fujimoto disease (KFD), a benign inflammatory condition characterized by self-limiting lymphadenopathy, fever, and occasional night sweats, is extremely uncommon and often associated with autoimmune diseases. Its atypical presentation can mimic infection or malignancy, making diagnosis challenging in oncology patients. We report a case of pembrolizumab-induced KFD in a patient with early-stage triple-negative breast cancer (TNBC).Case reportA 28-year-old woman with early-stage TNBC undergoing treatment with chemoimmunotherapy presented to the hospital with fever, left ear pain, left neck tenderness, and swelling. Magnetic resonance imaging (MRI) brain revealed edema of the left external auditory canal, mastoid and middle ear effusion, and soft tissue thickening, raising concern for necrotizing infection. Laboratory studies showed elevated erythrocyte sedimentation rate and aminotransferases. She was empirically started on broad spectrum antibiotics. Computed tomography (CT) neck showed rapidly progressing left neck/face inflammatory edema and possible early subperiosteal abscess. Given disease progression despite antibiotics, additional coverage was initiated for possible herpes zoster, atypical organisms, and fungal infection. Despite broadening infectious disease coverage, the patient continued to develop high grade fevers. With an unrevealing infectious workup, non-infectious etiologies including macrophage activation syndrome/HLH, autoimmune conditions, and KFD were considered. A left supraclavicular lymph node biopsy revealed benign reactive changes with expanded paracortical T lymphocytes, histiocytes, and polytypic plasma cells. Due to suspected inflammatory etiology, she was started on 250 mg methylprednisolone daily, resulting in prompt resolution of symptoms. The clinical presentation, disease course, and pathological findings were most consistent with a Kikuchi-Fujimoto reaction, a rare inflammatory condition likely triggered by pembrolizumab. She was able to safely resume pembrolizumab following a slow steroid taper without recurrence of symptoms.DiscussionAlthough IO has transformed the treatment landscape across many cancer types, it has also introduced a range of rare irAEs. IO-induced Kikuchi-Fujimoto is a very rare phenomenon, with only one previously reported case involving sintilimab. To our knowledge, there have been no reported cases of pembrolizumab-induced Kikuchi-Fujimoto like reactions. Given its nonspecific presentation and overlap with infectious and malignant etiologies, KFD can pose a diagnostic challenge. Clinicians should consider this rare entity in the irAE differential diagnosis when patients on pembrolizumab present with unexplained fever and lymphadenopathy. Notably, patients with IO-induced KFD often respond well to steroids and, may, in selected cases, be safely re-challenged with IO under close surveillance allowing oncologic treatment optimization. ConclusionKFD is a rare inflammatory condition that may occur as an irAE in patients with breast cancer receiving IO. A high index of suspicion is essential for early recognition and timely treatment.
Presentation numberPS5-06-10
A Rare Metastasis of Renal Cell Carcinoma to the Breast: A Case Report
Madelyn G Moulton, Rosalind Franklin University, North Chicago, IL
M. G. Moulton1, E. A. Marcus2, J. S. Wecsler2; 1Chicago Medical School, Rosalind Franklin University, North Chicago, IL, 2Surgical Breast Oncology, Cook County Health and Hospital System, Chicago, IL.
Renal Cell Carcinoma (RCC) has a metastasis rate of approximately 33%, but metastasis to the breast is extremely rare1. The common sites of metastasis include lung, bone, lymph nodes, and liver2. Here we present a case of RCC that metastasized to the breast three months after radical nephrectomy. In September 2019, a 65 year old female was diagnosed with cT2bN0M0 RCC. She was treated with Cabozantinib due to her past history of heart failure. Five months after the initial diagnosis she underwent left radical nephrectomy and left adrenal gland resection due to metastasis. This surgery resulted in negative margins and no lymph node involvement. Three months later a 1.6cm breast mass was discovered and a biopsy confirmed RCC. After it was confirmed that she had no other sites of metastasis and after multidisciplinary discussion, a lumpectomy was performed. All margins were negative apart from a focally positive margin, however fascia was removed without any evidence of muscular invasion. Although it was discussed at a multidisciplinary conference, adjuvant radiation was not started due to a lack of data to support its use in RCC. Four months later a surveillance CT revealed recurrence with right adrenal gland metastases and multiple subcentimeter nodules in the left breast. She was then started on Axitinib with pembrolizumab. After nine cycles of this immunotherapy regimen a CT found equivocal standardized uptake value (SUV) on the right adrenal metastasis and a 6th rib lytic lesion, but the breast mass did not have an elevated SUV. She underwent repeat breast biopsy which showed RCC, and she continued on her immunotherapy regimen for 2 more cycles due to the presence of other metastatic disease. She underwent repeat breast imaging which showed known masses, 2 of which had increased in size (one from 11 mm to 23 mm and one from 7 mm to 10 mm) and osseous destruction of the sixth rib which was causing increased rib pain. She underwent palliative left mastectomy and radiation therapy to the affected rib and chest wall. The final pathology showed 3 foci of metastatic clear cell carcinoma (1.8cm, 2cm, and 0.3cm) with negative margins in the breast and the patient received adjuvant radiation therapy of the left chest wall and was restarted on pembrolizumab. A later CT showed soft tissue invasion on the left chest wall. The chest wall lesion demonstrated metastatic RCC, but the pembrolizumab was stopped due to lack of response and arthritis. Three months later she presented to the ED after a fall and was found to have metastases in the left anterior frontal lobe. After brain lesion resection she was started on Everolimus. Five months later there were no new lesions seen on imaging, and the brain lesion had decreased in size. This case demonstrates a rare breast metastasis of RCC to add to the small body of case reports that describe this phenomenon. Documenting these cases is important in order to direct treatment options for future cases of RCC metastasis to the breast. Although resection of oligometastatic disease has been described, data is insufficient to indicate that it has an effect on survival. 1. Flanigan, R. C., Campbell, S. C., Clark, J. I. & Picken, M. M. Metastatic renal cell carcinoma. Curr. Treat. Options Oncol. 4, 385-390 (2003).2. Bianchi, M. et al. Distribution of metastatic sites in renal cell carcinoma: a population-based analysis. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 23, 973-980 (2012).
Presentation numberPS5-06-11
Exploring Characteristics and Outcomes of Metachronous Primary Breast and Lung Cancer in A Diverse Cohort
Keerthana Sureshkumar, University of Miami, Miami, FL
K. Sureshkumar1, R. Dawar2, S. Kareff3; 1Miller School of Medicine, University of Miami, Miami, FL, 2Breast Medical Oncology, University of Miami, Miami, FL, 3Lynn Cancer Institute, Baptist Health, Boca Raton, FL.
Introduction: Despite breast and lung cancer being the most common malignancies in females, the occurrence of double primary cancers has been rarely reported in literature. It is well-established that at an earlier age of diagnosis in patients with breast cancer is linked to more aggressive phenotypes, highlighting the importance of early screening in this demographic group. There is a deficiency in the literature related to secondary thoracic malignancies among this high-risk group. Methods: This retrospective case series reviewed a cohort of eight patients with double metachronous, primary breast and lung cancers at the Sylvester Cancer Center. Data on patient demographics, diagnoses, treatment modalities, and outcomes were obtained from electronic health records. Results: All 8 patients were biologically female with a median age of 58.5 years. Racial distribution was 25% Asian, 12.5% Black and 62.5% White. 80% of White identified as the Hispanic/Latinx ethnicity. 62.5% of patients were never-smokers and 12.5 % were light, former smokers. The median age of diagnosis of breast and lung cancer was 60 years and 62.5 years, respectively. The average time between both diagnoses was 1.9 years (range 0.33 – 4 years). The majority (87.5%) of genetic profiles indicated a hormone receptor-positive mutation, with 33% of this genetic subset also showing an EGFR mutation. 75% of lung tumors were PDL-1 negative. Our review also indicates a decrease in concern for smoking. Despite no statistical significance between ethnicity and age of diagnosis of breast or lung cancer (p= .157), there was a trend demonstrating Hispanic/Latinx patients being diagnosed at an earlier age for both. There was no statistical significance between ethnicity and smoking status (p= .465), or ethnicity and time between diagnoses (p = 1). Conclusions: In this diverse, real-world cohort of patients with metachronous and synchronous lung and breast cancers, we noted interesting, hypothesis-generating observations. Trends indicated an earlier diagnosis of both breast and lung cancer in Hispanic/Latinx patients. 75% of our cohort had a dual diagnosis of breast ductal carcinoma and lung adenocarcinoma. 87.5% of individuals in our cohort displayed a hormone receptor-positive profile and 75% a PDL-1 negative prevalence. This case series confirms the importance of screening breast cancer patients with low-dose CT for early detection of lung cancer. Since double primary breast and lung cancer is rare, molecular profiling with next-generation sequencing could also provide insightful information.
Presentation numberPS5-06-12
Atypical Presentation of Invasive Lobular Carcinoma Presenting as Periorbital Swelling Despite Negative Mammogram
Sarah A Shaker, UT Health Houston, Houston, TX
S. A. Shaker1, D. Ramirez2; 1Internal Medicine, UT Health Houston, Houston, TX, 2Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Invasive lobular carcinoma (ILC) accounts for approximately 10-15% of invasive breast cancers and is characterized by a unique histologic growth pattern that often results in subtle imaging findings. Notably, up to 30% of cases may be mammographically occult. ILC is also characterized by an atypical metastatic pattern, with a tendency to spread to sites such as the gastrointestinal tract, peritoneum, and orbit. Presentation as isolated periorbital swelling is exceedingly rare and presents a significant diagnostic challenge, particularly in the context of a negative screening mammogram. Case Presentation: A 56-year-old woman initially presented to her primary care physician with left periorbital swelling, redness, and discomfort. Symptoms were recurrent for over a year, and during this time she was evaluated by optometrists, ophthalmologists, and plastic surgeons without a definitive diagnosis. Empiric treatment for presumed allergic conjunctivitis and sinusitis provided little improvement before symptoms would recur. Screening mammogram during this time was negative for malignancy. She eventually underwent a brain MRI for persistent symptoms which demonstrated nonspecific enhancement of the left orbit. Given ongoing symptoms, she underwent surgical resection of the infraorbital mass, and pathology revealed metastatic ILC. The patient had no breast-related symptoms, and repeat diagnostic mammography and ultrasound failed to identify a primary tumor. Subsequent breast MRI detected a subtle right breast lesion, which was confirmed as ILC on biopsy. Discussion: This case highlights the diagnostic limitations of mammography in detecting ILC and underscores the importance of maintaining clinical suspicion when evaluating persistent orbital symptoms. Orbital metastases are a rare and often an overlooked manifestation of ILC, especially when it is the initial presentation. This case also highlights the value of MRI in the diagnostic workup of ILC, even with negative mammography. Conclusion: Periorbital swelling may represent the initial and sole manifestation of ILC. Given the potential for ILC to spread to atypical sites, providers should be aware of this rare presentation and include breast cancer in the differential diagnosis for unexplained orbital symptoms. Further imaging with MRI should be considered, even in the setting of a negative mammogram, if there is high clinical suspicion for ILC to help avoid delays in diagnosis.
Presentation numberPS5-06-13
Carcinoma en Cuirasse
Karishma Vijay Rupani, Icahn School of Medicine at Mount Sinai, New York, NY
K. Rupani, P. Klein; Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY.
Carcinoma en Cuirasse We present the case of a 49 year old postmenopausal female with right breast stage IIIA (mpT2N2aM0) hormone receptor-positive and HER2-negative classic-type multifocal invasive lobular carcinoma who underwent a lumpectomy. She was treated with adjuvant dose-dense doxorubicin and cyclophosphamide, followed by paclitaxel (ddAC-T). She then received adjuvant radiation therapy which was followed by anastrozole. She discontinued anastrozole after losing her health insurance and had a local recurrence with biopsy-proven involvement of the dermis, carcinoma en cuirasse (Figure 1a). The name, carcinoma en cuirasse, derives from its resemblance to a breastplate of armor. Following this, she underwent repeat radiation therapy to the skin, and was started on treatment with exemestane and palbociclib. She later switched from palbociclib to abemaciclib because of insurance issues, and continued exemestane and abemaciclib until progression of disease locally. Due to aversion to parenteral treatment, she was started on everolimus with exemestane instead. Unfortunately, she had further progression of disease locally at which time she agreed to treatment with fulvestrant and everolimus but had worsening of her carcinoma en cuirasse (Figure 1b). It is likely that she had poor disease control because of intermittent compliance with all of her treatment. She most recently received repeat palliative radiation followed by capecitabine with improvement of her skin lesions (Figure 1c).
Presentation numberPS5-06-14
Unexpected B-hCG elevation in metastatic inflammatory breast cancer: A diagnostic dilemma
Katherine Klein, MD Anderson Cancer Center, Houston, TX
K. Klein1, D. Ramirez2; 1Internal Medicine, MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX.
Introduction: The beta subunit of human chorionic gonadotropin (B-hCG) is often used as a marker of pregnancy, but can be produced by several different sources, including the placenta, trophoblastic tumors, nontrophoblastic tumors, and the pituitary gland. While not used as a routine tumor marker for breast malignancies, studies have shown that hCG from a placental source appears to be protective against breast cancer, while ectopic production of B-hCG appears to be associated with poor prognosis. As part of routine testing during chemotherapy at our institution, all patients undergo a pregnancy test due to the risks of chemotherapy on the developing fetus. Here, we present a case of incidentally found false positive pregnancy test in a post-menopausal patient with metastatic inflammatory breast cancer. Case Description: We present a case of a 49 year old female who was initially diagnosed in May 2024 with stage IV HER2+ inflammatory breast cancer, with metastases to multiple lymph nodes and pulmonary nodules. She progressed through several lines of therapy, including docetaxel, trastuzumab, and pertuzumab, dose dense doxorubicin and cyclophosphamide, and fam-trastuzumab deruxtecan. She is currently on therapy with tucatinib, capecitabine, and trastuzumab. In May 2025, she had routine prechemotherapy labs done including a urine pregnancy test which resulted positive. She had two prior urine pregnancy tests which were negative in July and September 2024. We subsequently performed a serum B-hCG quantitative test in May 2025 showed a B-hCG of 82.4 mIU/mL. The patient had reported abstinence and amenorrhea for the past year. It was determined to be a false pregnancy test and her therapy was continued. The clinical case was discussed with our OB/GYN colleagues with consensus to verify menopausal status by hormone levels. Labwork showed an estradiol of <11 pg/mL and FSH of 94.8 IU/L, consistent with a post-menopausal state. She had a repeat set of labs in June 2025 which showed a serum B-hCG of 87.4 mIU/mL, estradiol of 16 pg/mL, and FSH of 97.5 IU/L, again consistent with a post-menopausal state. Discussion: Patients undergoing chemotherapy are often screened for pregnancy, but quantitative measures of B-hCG as a tumor marker are not typically done for breast malignancies. Some case reports have described breast malignancies presenting with elevated B-hCG which was described as having choriocarcinomatous features. There have been case studies evaluating the prognostic implication of elevated B-hCG in breast malignancies however levels in these studies were only very mildly elevated (e.g. <5 mIU/mL). For this patient, it was thought that the elevated B-hCG could be due to a pituitary source, however typically in these cases the B-hCG is only mildly elevated (e.g. in one population of women the mean level of hCG was 9.5 mIU/mL with a range of 2.1-32 mIU/mL). Our patient in this case had a B-hCG value of 82.4-87.4 mIU/mL. However, her low levels of estradiol and elevated FSH confirmed a post-menopausal state. Patients with trophoblastic tumors typically have significantly elevated B-hCG, greatly above the level seen in this patient. Conclusion: In summary, this case highlights an instance of moderately elevated B-hCG in a patient with metastatic inflammatory breast cancer, which has not been well described in the literature to our knowledge. We sought to describe this case to alert providers to the possibility of false pregnancy tests in patients with breast cancer with no other known gynecological cancer. As with this patient, next steps include measurement of estradiol and FSH to confirm a post-menopausal state.
Presentation numberPS5-06-15
Dermatofibrosarcoma protuberans (DFSP) with fibrosarcomatous transformation involving the male breast: A rare case report and comprehensive review of clinicopathologic features
Anne Dominique Nascimento Lima, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
G. Neri Ferreira1, L. Novis Leite Pinto1, L. Martins de Brito Moraes2, D. Franco Vieira de Oliveira3, A. Coelho de Oliveira4, A. Pereira Correia Carneiro5, A. Dominique Nascimento Lima4; 1Gynecology, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 2PPGCAN-INCA, Instituto Nacional de Câncer, Rio de Janeiro, BRAZIL, 3Plastic Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 4Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 5Pathology, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL.
Background: DFSP is a rare, low-grade soft tissue sarcoma characterized by locally aggressive growth and low metastatic potential. The fibrosarcomatous (FS) variant, which occurs in approximately 10-15% of cases, is associated with increased biological aggressiveness, higher rates of local recurrence, and a greater risk of distant metastasis, particularly to the lungs. DFSP involving the breast is exceptionally rare, especially in male patients, making diagnosis and treatment more challenging due to its clinical similarity to breast carcinomas. Case Report: A 65-year-old male, ex-smoker, presented with a progressively enlarging left breast mass over a four-month period. On physical examination, the lesion appeared as an irregular, indurated, mobile mass, measuring 7×6 cm, with hyperemia and cutaneous infiltration. Breast ultrasonography demonstrated a solid, hypoechoic, heterogeneous, lobulated nodule (BI-RADS 4C). Chest CT revealed a 7.1×5.7×4.5 cm mass with no clear cleavage plane with the pectoralis major muscle. Core biopsy demonstrated a spindle cell neoplasm, CD34-positive and Factor XIIIa-negative, consistent with DFSP. Clinical staging was cT4b N0 M0. The patient underwent a wide local excision via simple mastectomy with partial resection of the pectoralis major muscle to achieve adequate deep margins, and immediate reconstruction was performed using an abdominal fasciocutaneous flap. Histopathological analysis confirmed DFSP with FS transformation (DFSP-FS), high mitotic rate, and all surgical margins were negative, with the closest deep margin exceeding 0.5 cm. Immunohistochemistry (IHC) showed diffuse CD34 positivity, negative S-100, p63 and CK, and Ki67 >30% in fibrosarcomatous areas. Discussion: DFSP involving the breast is extremely rare, particularly in males, with few reported cases. It contributes to diagnostic delays and frequent misclassification as more common breast malignancies. Clinical presentation can be insidious. Imaging exams, although useful for anatomical delimitation, are not specific for diagnosis. Ultrasonography and mammography may suggest solid lesions, but magnetic resonance imaging is superior in assessing the extent of local invasion and involvement of adjacent structures. Definitive diagnosis relies on histopathology and IHC, with CD34 positivity and absence of epithelial markers. The presence of fibrosarcomatous areas, with high mitotic activity and an elevated Ki67, indicates aggressive transformation and a worse prognosis. Wide surgical excision with negative margins remains the cornerstone of treatment to minimize recurrence. Simple mastectomy with wide margins, including resection of adjacent muscle tissue when necessary, is recommended in cases of extensive involvement or fibrosarcomatous transformation, as in the present report. Adjuvant radiotherapy is recommended in situations of positive margins, unresectable disease, or locoregional recurrence, contributing to the reduction of the local recurrence rate, as evidenced by retrospective studies and systematic reviews. Radiotherapy should also be considered in cases of DFSP-FS, due to its potential aggressiveness. The use of imatinib, a tyrosine kinase inhibitor that acts on the characteristic COL1A1-PDGFB gene fusion of DFSP, has proven effective in advanced, unresectable, or metastatic cases, offering an important therapeutic option for patients with refractory disease. This rare case of DFSP-FS infiltrating the male breast underscores the diagnostic challenges and therapeutic considerations associated with this entity. It highlights the critical importance of a multidisciplinary approach for timely diagnosis and optimal surgical management, ultimately improving patient outcomes.
Presentation numberPS5-06-16
A case of triple negative breast cancer in Lynch Syndrome
Harshil Bhatt, Maine Health Cancer Care, South Portland, ME
H. Bhatt, D. A. Rabinovich, M. E. Auster; Hematology and Oncology, Maine Health Cancer Care, South Portland, ME.
Background: Although there are numerous inherited syndromes that predispose individuals to developing breast cancer, it is inconclusive whether Lynch syndrome (LS) increases the risk of breast cancer. We present a case of a triple-negative breast cancer in a woman with LS. Case Presentation: Our patient is a 73-year-old woman with a past medical history of LS (MSH2 deletion), endometrial cancer status-post TAH/BSO, rectal cancer status-post abdominoperineal resection and end ileostomy, and colon cancer status-post right hemicolectomy/appendectomy and chemotherapy with 5-fluorouracil and leucovorin. A 2.7 cm mass was found in her right breast during a screening mammogram. MRI showed a 3.7 cm right breast mass. CT of the chest, abdomen, and pelvis showed no lymphadenopathy or evidence of metastatic disease. Biopsy of the right breast mass indicated grade 3 invasive ductal carcinoma with ER 10%, PR 2%, HER2 1+. She began neoadjuvant chemoimmunotherapy based on the KEYNOTE-522 trial. Chemoimmunotherapy was discontinued after five cycles due to intolerable side effects, including neuropathy, rash, and musculoskeletal pain. She then underwent a bilateral mastectomy with sentinel lymph node biopsy, which revealed residual invasive ductal carcinoma measuring 6 mm without lymph node involvement, consistent with stage ypT1b pN0. Pembrolizumab was resumed. Adjuvant treatment with dose-reduced capecitabine was initiated following the CREATE-X trial. Discussion: Lynch syndrome, also known as hereditary nonpolyposis colorectal cancer, is an inherited condition caused by germline mutations in DNA mismatch repair genes, including MLH1, MSH2, MSH6, and PMS2. LS is well known to increase the risk of multiple cancers, such as colorectal, endometrial, ovarian, and stomach cancers. Breast cancer has not been considered a part of the LS spectrum. Despite several studies, a clear link between LS and breast cancer has not been established. A few studies have also looked at the possible association between pathogenic variants of LS and breast cancer risks; however, the data have not been consistent. A cohort study by Sheehan et al. found a significantly higher prevalence of breast cancer in women with PMS2 mutations compared to other genetic alterations, while a study by Scott et al. reported overrepresentation of breast cancer in MLH1 mutations compared to MSH2 mutations. Currently, LS screening guidelines recommend general population-based screening for breast cancer. Immunotherapy with PD-1 blockade has demonstrated profound results with mismatch repair-deficient colorectal cancer as well as other cancers such as endometrial, gastric, pancreatic, ovarian, and cholangiocarcinoma, but whether this translates to breast cancer is unknown. In this case, the patient with triple-negative breast cancer and LS received five cycles of neoadjuvant chemoimmunotherapy, and her mass decreased from 3.7 cm before treatment to 6 mm pathologically. There are no treatment recommendations specific to LS patients with breast cancer. Larger prospective studies are warranted to establish a potential need for intensified breast cancer screening in patients with LS, as well as to study outcomes of breast cancer in LS patients. References: Sheehan, M. et al. Eur J Breast Health 2020; 16(2): 106-109. Schmid, Peter et al. “Pembrolizumab for Early Triple-Negative Breast Cancer.” NEJM vol. 382,9 (2020): 810-821. Masuda, Norikazu et al. “Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy.” NEJM vol. 376,22 (2017): 2147-2159. Scott, R J et al. “Hereditary nonpolyposis colorectal cancer in 95 families: differences and similarities between mutation-positive and mutation-negative kindreds.” American journal of human genetics vol. 68,1 (2001): 118-127. Cercek, A et al. “Nonoperative Management of Mismatch Repair-Deficient Tumors.” NEJM vol. 392, 23 (2025): 2297-2308.
Presentation numberPS5-06-18
Paraneoplastic Encephalitis in Breast Cancer
Chinyere Onyeukwu, Rush University, chicago, IL
C. Onyeukwu1, M. Boamah1, J. Steffes2, N. Dhliwayo2, S. Lo3, T. Christ1; 1Internal Medicine, Rush University, chicago, IL, 2Division of Hematology and Oncology, Rush University, chicago, IL, 3Division of Hematology and Oncology, Rush University, Chicago, IL.
IntroductionParaneoplastic Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is one of the most common types of immune-mediated encephalitis characterized by the collection of immune-mediated nervous system symptoms that arise due to malignancy1,2. However, NMDAR encephalitis is a rare manifestation of breast cancer. We present the case of a 31-year-old female with progressive neurological decline due to paraneoplastic encephalitis with a subsequent breast cancer diagnosis. Treatment with tamoxifen led to significant improvement, emphasizing the importance of identifying underlying etiology and treatment of NMDA encephalitis for improved outcomes. Patient PresentationA 31-year-old female presented with an 11-month history of progressive neurological decline characterized by confusion, psychosis, dysarthria, severe ataxia requiring a walker, amnesia, aggression, violence, and insomnia. Subsequently, she experienced progressive dysphagia and dysarthria resulting in an inability to speak beyond grunts. She was admitted to the psychiatric unit 3 times during this 11-month period. Clinical CourseThe patient was admitted to the Neurology floor and then transferred to Neuro ICU on day 2 of hospitalization due to hypoxic respiratory failure due to fluctuating mental status with associated bradytachycardia, diffuse rigidity, and agitation She underwent tracheostomy and PEG placement.Diagnostic testing demonstrated leukocytosis, anemia, thrombocytopenia, hyperglycemia, +ANA, elevated CRP 21.5 and ESR 40, +SSA, elevated CK, low TSH 0.123, SPEP with diffuse hypergammaglobulinemia. Encephalitis antibody panel was positive for AMPA receptor Ab. Lumbar puncture (LP) was unremarkable, and EEG showed activity consistent with mild encephalopathy. MRI brain demonstrated hyperintense areas in bilateral frontal, parietal, and right temporal lobe suggestive of autoimmune encephalitis. Treatment for neurologic symptoms included methylprednisolone daily and IVIG for 5 days, followed by 7 sessions of therapeutic plasmapheresis. Psychiatry was consulted, and she underwent six sessions of ECT, initiated on Ativan, and transitioned to Thorazine twice daily.CT chest showed right axillary adenopathy without evidence of distant disease. Right axillary biopsy confirmed invasive carcinoma, consistent with breast primary, ER+, PR+. HER2 2+, FISH not amplified, Ki-67 20-30%. She was started on Tamoxifen with gradual improvement in her symptoms. Patient was discharged from hospital and regained complete neuromechanical function after about three months of single agent Tamoxifen. She was transitioned to neoadjuvant dose-dense Adriamycin and Cyclophosphamide followed by Paclitaxel with plan to undergo surgical resection. Conclusion Paraneoplastic encephalitis is a rare diagnosis in breast cancer, reported in less than 1% of patient’s with breast cancer. AMPA-R encephalitis is a paraneoplastic syndrome associated with breast cancer caused by onconeural antibodies directed against cancer cells, resulting in inflammation and injury to limbic strictures in the brain.This case highlights the importance of a thorough assessment in the evaluation of unexplained neurologic and psychological decline which includes imaging, LP, antibody testing, EEG, and biopsy. It also stresses the importance of identifying and treating the underlying malignancy which may result in resolution of symptoms.
Presentation numberPS5-06-19
Sacituzumab Govitecan Combined with Anti-angiogenic Therapy and Radiotherapy in a BRCA1-Mutated Triple-Negative Breast Cancer Patient with Multiple Recurrences: A Case Report
Chunfang Hao, Tianjin Cancer Hospital Airport Hospital, Tianjin, China
C. Hao, Z. Jie; Department of Breast Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin, CHINA.
Introduction: Triple-negative breast cancer (TNBC), accounting for 10%-15% of breast cancers and defined by ER⁻/PR⁻/HER2⁻ status, exhibits aggressive biological behavior with high proliferative indices, genomic instability, and limited targeted therapies. BRCA1-mutated TNBC may respond to PARP inhibitors and DNA-damaging agents. However, a significant proportion of BRCA1-mutated TNBC patients experience disease progression, underscoring the urgent need for novel treatment strategies. This case report details a young female with BRCA1-mutated TNBC presenting with extensive systemic and brain metastases. The treatment strategy combining Sacituzumab Govitecan (SG), Anlotinib, and concurrent whole-brain radiotherapy (WBRT) provides unique insights into managing complex, relapsed TNBC. Case Report: In March 2011, a 28-year-old female was diagnosed with left breast invasive ductal carcinoma (ER⁻/PR⁻/HER2⁻, pT2N0M0). She underwent left modified radical mastectomy, followed by 8 cycles of adjuvant chemotherapy (epirubicin + cyclophosphamide, followed by paclitaxel). In March 2017, a 2×2 cm invasive carcinoma was detected (ER⁻/PR⁻/HER2 1+) in her right breast. She received 6 cycles of neoadjuvant TAC chemotherapy (docetaxel + liposomal doxorubicin + cyclophosphamide), and then underwent right nipple-areolar-sparing mastectomy, axillary dissection, and breast reconstruction on August 7, 2017, followed by radiotherapy. In December 2020, right chest wall recurrence was found, treated with local extended resection in January 2021, and followed by one-year oral capecitabine.In June 2023, she developed lung metastases (germline BRCA1 mutation),then enrolled in an IIT trial (NCT05085626) where she received Fluzoparib + Chidamide, achieving partial response (PR) with a progression-free survival (PFS) of 14 months. In August 2024, the patient developed widespread metastases (brain, lung, soft tissues, liver, spleen, bone, ileum). She declined brain surgery and received Bevacizumab + Trastuzumab Deruxtecan. After two cycles, symptoms of intracranial hypertension improved, and the disease was assessed as stable disease (SD), but grade 3 or 4 nausea/vomiting/fatigue led to discontinuation. In December 2024, original lesions (brain, lung, liver, ileum) progressed with new metastases in retroperitoneum, kidneys, left 4th metacarpal, and phalanges. Treatment with SG in combination with Anlotinib was initiated, accompanied by concurrent WBRT during the first cycle. After two cycles, the disease achieved a PR. Grade 3 anemia occurred during treatment but was managed symptomatically. The PFS has exceeded 7 months to date, and the patient remains under continuous treatment. Discussion: Preclinical studies have shown that SG delivers SN-38 to induce DNA damage in homologous recombination deficiency (HRD) tumors (e.g., BRCA-mutated) with impaired repair. Anti-angiogenics enhance SG’s efficacy via VEGF-targeted vascular normalization and blockade of survival signaling pathways. Additionally, SG can penetrate into intracranial tumors and exhibit promising activity. Combining SG with WBRT can further induce DNA damage and enhance treatment efficacy. The patient’s positive response to this combination regimen suggests potential benefit for metastatic TNBC, especially in those with brain metastases, though grade 3 anemia highlights the need for close toxicity monitoring. Conclusion: This case report highlights the potential efficacy of SG in combination with anti-angiogenic therapy and radiotherapy in treating BRCA1-mutated TNBC patients with multiple recurrences and brain metastases. However, further studies are needed to confirm these findings and to optimize the use of these agents in this subset of patients.
Presentation numberPS5-06-20
Autoimmune Hemolytic Anemia Triggered by Neoadjuvant Pembrolizumab in Triple Negative Breast Cancer: A Case Report
Angela Rita Slaybe, Baptist Health Miami Cancer Institute, Miami, FL
A. R. Slaybe, A. C. Sandoval Leon; Hematology Oncology, Baptist Health Miami Cancer Institute, Miami, FL.
Introduction: Pembrolizumab has improved outcomes in triple negative breast cancer (TNBC), but its use can be associated with serious immune-related adverse events. Autoimmune hemolytic anemia (AIHA) has been reported in patients treated with immune-checkpoint inhibitors for other malignancies, but to our knowledge, it has not been previously described in patients with breast cancer (BC). Herein, we report a case of pembrolizumab-associated AIHA during neoadjuvant therapy for locally advanced TNBC. Case Presentation: A 72-year-old postmenopausal woman with a cT3N3 TNBC started treatment with chemotherapy in combination with pembrolizumab per the KEYNOTE-522 protocol. She completed four cycles of paclitaxel, carboplatin, and pembrolizumab with a good clinical response. The second cycle of doxorubicin and cyclophosphamide (AC) plus pembrolizumab was delayed due to neutropenic fever and symptomatic anemia (hemoglobin 7.1 g/dL). She received one unit of packed red blood cells. At that time, the indirect bilirubin was normal, and her hemoglobin rose appropriately to 11g/dL. However, two weeks later she developed worsening macrocytic anemia (hemoglobin 9.8 g/dL) and evidence of hemolysis: indirect bilirubin elevation (2.3 mg/dL), low haptoglobin (< 10 mg/dL), reticulocytosis (5.8%), and LDH elevation (840 U/L). The patient had no prior history of a blood transfusion, making a delayed hemolytic transfusion reaction less likely. A direct antiglobulin test (DAT) was drawn after steroid initiation and was negative. Due to these findings, she started prednisone 1mg/kg. Pembrolizumab was discontinued and prednisone was tapered over four weeks. Hemolysis resolved (Hemoglobin stabilized at 11g/dL, LDH, indirect bilirubin, and haptoglobin normalized). She completed the remaining 3 cycles of AC uneventfully. Although her hemoglobin declined again during subsequent AC chemotherapy it remained stable, consistent with expected chemotherapy-induced anemia. Importantly hemolysis markers remained normal during this period, supporting resolution of the autoimmune process. Discussion: AIHA is an uncommon but potentially life-threatening immune related adverse event associated with checkpoint inhibitors such as pembrolizumab. In this patient, laboratory diagnostic clues included anemia with reticulocytosis, elevated LDH/indirect bilirubin, and low haptoglobin. While the DAT was negative, it was done after corticosteroid treatment started. The temporal association with pembrolizumab exposure and clinical resolution following its withdrawal and steroid therapy supports a diagnosis of pembrolizumab-induced AIHA. Clinicians should maintain a high index of suspicion since prompt recognition and treatment with corticosteroids is critical.
Presentation numberPS5-06-21
When Signet Ring Cells Mislead: A Case of Metastatic Breast Cancer Presenting as Gastric Malignancy
Jared Eckman, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
J. Eckman1, H. Jeon2, S. Oh3; 1Department of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, 2Georgetown Cancer Institute, Medstar Washington Hospital Center, Washington, DC, 3Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY.
Background Breast cancer is the most frequently diagnosed malignancy among women, with invasive lobular carcinoma (ILC) comprising 15% of cases. ILC frequently harbors CDH1 mutations, resulting in the loss of E-cadherin as well as a dyscohesive growth pattern that hinders detection. Signet ring cell carcinoma of the breast, a rare but diagnostically significant variant with poor prognosis, can closely mimic primary gastric adenocarcinoma. Shared histomorphology, particularly in metastatic setting at diagnosis, predisposes to misclassification and consequent therapeutic misdirection. Case Presentation A 61-year-old woman presented to the emergency room with one-month of dysphagia, nausea, vomiting, and unintentional weight loss. Radiographic studies revealed abdominopelvic ascites, periportal lymphadenopathy, right adnexal enlargement, osseous lesions, and omental carcinomatosis. A biopsy of the gastric fundus was positive for invasive adenocarcinoma, diffuse type with signet ring features, supported by positive pan-cytokeratin AE1/AE3 staining. Further biopsy of an omental mass showed CK7 positivity and negativity for PAX8, CDX2, and CD20. Metastatic gastric adenocarcinoma was presumed, and capecitabine-oxaliplatin was commenced. Additional molecular profiling was completed, which revealed breast markers including ESR1, GATA 3, cytokeratin, and mammaglobin. A pathogenic loss of function variant in CDH1 was also detected. Further immunohistochemistry (IHC) on the omental specimen revealed ER 90%, PR 90%, and HER2 IHC 0. IHC of the omentum was also positive for GATA3, mammaglobin, and E-cadherin. These findings supported a diagnosis of metastatic lobular breast carcinoma with signet ring features. MRI breast did not identify a primary breast mass and there were no pathologic axillary lymph nodes. Following bilateral salpingo-oophorectomy, pathology for the right adnexal mass showed carcinoma of breast origin (ER 95%, PR 70% and HER2 IHC2+ and FISH negative). The treatment was switched to a first line aromatase inhibitor and CDK 4/6 inhibitor and the patient has since undergone multiple lines of therapy. Discussion Although breast cancer metastasizing to the stomach is rare—with an estimated incidence of just 0.04%—accurate diagnosis of primary origin of adenocarcinoma is vital due to the stark differences in management and outcomes. The clinical and morphological overlap between metastatic ILC with signet ring features and primary gastric carcinoma necessitates a high index of suspicion and thorough immunohistochemical analysis. ILC may metastasize in a diffuse pattern mimicking gastrointestinal malignancies and can present without an identifiable breast lesion. Literature has shown that ER, GCDFP-15, mammaglobin, and GATA3 are highly sensitive and specific for identifying breast origin, whereas CK20 and CDX2 are associated with gastrointestinal differentiation. Although not unique to breast tumors, CDH1 and PIK3CA alterations are detected in roughly 63-65% and 46% of invasive lobular carcinomas, respectively, versus only about 9.7% and 12% of gastric adenocarcinomas. Further studies are warranted to refine diagnostic algorithms that couple IHC with next-generation sequencing, clarifying when the addition of breast-specific markers is essential for definitive tumor origin assignment. Conclusion This case emphasizes the need for comprehensive diagnostic workup in atypical presentations and supports broader use of breast-specific IHC markers and molecular profiling in gastrointestinal biopsies with ambiguous or signet ring histology. Integrating markers such as ER, PR, GATA3, mammaglobin, and CDH1/PIK3CA profiling into a tiered diagnostic algorithm can prevent misclassification, facilitate timely therapy, and ultimately improve patient outcomes.
Presentation numberPS5-06-22
Atypical recurrence of invasive lobular breast carcinoma with hormone receptor loss: a Case Report
Rita Quaresma Ferreira, Local Health Unit of São José, Lisbon, Portugal
R. Q. Ferreira, A. Montenegro, L. Fernandes, D. C. Simão; Medical Oncology, Local Health Unit of São José, Lisbon, PORTUGAL.
Introduction: Invasive lobular carcinoma (ILC) is the second most common type of breast cancer and typically shows hormone receptor (HR) positivity. Unlike ductal carcinoma, it tends to metastasize later and to unusual sites such as the peritoneum, gastrointestinal tract, and ovaries. These atypical patterns can mimic primary tumors of other organs, complicating diagnosis. Late recurrences with loss of HR expression are rare and present additional challenges. We report a case of peritoneal recurrence of ILC, years after initial diagnosis, with loss of HR expression. Case Description: This report describes a 51-year-old premenopausal woman with no significant family history of cancer, diagnosed in 2019 with mixed invasive carcinoma of the right breast: invasive lobular carcinoma (ER 95%, PR 75%, HER2 2+ without ISH amplification, Ki67 10%) and invasive carcinoma of no special type (ER 10%, weak staining; PR 90%, HER2 0; Ki67 10%). The disease was staged as cT2N0M0. The patient underwent breast-conserving surgery and sentinel lymph node biopsy. Final pathology revealed pT2N2a disease, with 7 metastatic lymph nodes out of 12 examined. She subsequently underwent axillary lymph node dissection, confirming persistent HR positivity. Adjuvant treatment included chemotherapy with taxanes and anthracyclines, radiotherapy, and endocrine therapy planned for ten years—initially tamoxifen for 15 months, followed by an aromatase inhibitor after menopause onset. Long-term follow-up imaging remained negative for recurrence or metastasis. In 2023, the patient developed pelvic pain, prompting a pelvic MRI that revealed suspicious bone lesions in the left iliac and L5 vertebra, a solid hypogastric mass (30×28 mm), and moderate peritoneal effusion. However, subsequent imaging, including FDG PET-CT and lumbar spine MRI, showed no clear evidence of malignancy, and the patient remained under surveillance. In April 2025, a screening colonoscopy revealed a sigmoid lesion; biopsy showed poorly cohesive carcinoma cells, and immunohistochemistry (CK7+, GATA3++, ER-, HER2-, PD-L1-) favored metastasis from prior lobular breast carcinoma despite complete loss of ER expression. Pelvic MRI confirmed extensive pelvic-peritoneal disease involving the cervix, adnexa, and bilateral ureters, with associated hydronephrosis. Additional findings included suspected ovarian involvement and uterine leiomyomas. Tumor markers showed elevated CA 15.3 and CEA. A multidisciplinary tumor board concluded this was most consistent with metastatic recurrence of the original ILC. A PET-CT in June 2025 showed no FDG-avid disease but confirmed mass effect on the ureters. The patient was referred to urology, and first-line palliative chemotherapy with paclitaxel was proposed for HR-negative, HER2-negative, and PD-L1-negative disease. Discussion: This case shows a late recurrence of ILC with peritoneal involvement and loss of ER expression. Although this metastatic pattern is consistent with ILC, loss of HR expression is rare. This loss may result from clonal evolution due to long-term endocrine therapy, leading to resistance and poorer prognosis. Peritoneal carcinomatosis mimicking gynecologic cancer delayed diagnosis, highlighting the need for thorough histopathological and immunohistochemical evaluation. FDG PET-CT may miss low-metabolic lesions, emphasizing the role of biopsy in unclear cases. Conclusions: This case highlights the need for long-term vigilance in ILC patients and the consideration of metastasis years after treatment. It supports re-biopsy and pathological reassessment in atypical cases, especially when receptor status changes, underscoring the evolving biology of breast cancer and the importance of personalized management.
Presentation numberPS5-06-23
A Functional Reversal of Leptomeningeal Disease without CNS Directed Therapy
Emily Sargent, UTHealth Houston, Houston, TX
E. Sargent1, A. Buzdar2, D. Ramirez2; 1Internal Medicine, UTHealth Houston, Houston, TX, 2Breast Medical Oncology, MD Anderson, Houston, TX.
Background: Leptomeningeal disease (LMD) is a rare complication occurring in up to 5% of breast cancer patients and is associated with a poor prognosis. In a large institutional study of 233 patients with LMD, the median overall survival for those with HR+/HER2− disease was 3.7 months, despite multimodal therapy. Standard treatments include intrathecal chemotherapy and CNS radiation; however, no consensus guidelines or randomized trials exist as most patients are excluded from clinical trials due to poor performance status. Systemic therapy has been associated with modest survival benefits, though responses remain unpredictable. Here, we report a case of LMD in a HR+/HER2- metastatic breast cancer patient with initial poor performance status who has achieved sustained functional recovery with endocrine and CDK4/6 inhibitor therapy alone. Case Presentation: A 76-year-old woman with hypertension and hyperlipidemia presented with fatigue, constipation, and dizziness. CT imaging revealed a soft tissue mass involving the uterus, adnexa, bladder, and left ureter with resulting hydroureteronephrosis. Further workup showed extensive metastatic disease, including osseous metastases, omental/peritoneal carcinomatosis, nodal involvement, and soft tissue metastases to the gluteal and paraspinal musculature, as well as the left breast. MRI brain and spine performed for evaluation of altered mental status demonstrated leptomeningeal disease. Right inguinal lymph node biopsy revealed metastatic carcinoma consistent with breast primary (ER 99%, PR 80-90%, HER2 IHC 0, Ki-67 >50%). A left breast biopsy confirmed invasive lobular carcinoma. Subsequently, the patient presented to the MD Anderson emergency room for dyspnea on exertion and right shoulder pain. The patient was noted to be bed bound consistent with an ECOG performance status of 4 and had an early-stage sacral wound. Further evaluation revealed left pulmonary emboli and a large right sided pleural effusion. She underwent thoracentesis with pleural studies revealing malignant cells consistent with adenocarcinoma favoring breast primary. Based on the right choice trial, the patient was started on an aromatase inhibitor and CDK 4/6 inhibitor in August of 2023. When considering a CDK inhibitor, the decision was made to utilize palbociclib based on the side effect profile. Additionally, zoledronic acid was initiated for her diffuse osseous disease. After 22 months, the patient remains on first-line therapy with anastrozole and palbociclib. MRI Brain, Bone Scan, and CT Abdomen and Pelvis show stable disease. She has demonstrated remarkable functional recovery, evidenced by her transition to an ECOG of 0 with resolution of her prior sacral wound- all without CNS-directed therapy. Discussion: This case highlights an unusual response of LMD in HR+/HER- breast cancer to systemic therapy with an aromatase inhibitor and CDK 4/6 inhibitor. Emerging evidence suggests that CDK inhibitors may have meaningful central nervous system activity in patients with HR+/HER2− breast cancer. In a prospective phase II trial, abemaciclib achieved CSF and brain metastasis concentrations well above the IC50 for CDK4/6 inhibition and was associated with clinical benefit in a subset of patients with brain and leptomeningeal metastases. Our patient’s recovery challenges the conventional guidelines regarding recommendations of intrathecal and/or radiation therapy for LMD and supports further investigation into systemic CDK4/6 inhibition for select patients.
Presentation numberPS5-06-24
When Therapy Leaves a Mark: A Case of Ribociclib-Induced Vitiligo Reversed with Abemaciclib
Daniela Urueta Portillo, UT health Mays Cancer Center, San Antonio, TX
D. Urueta Portillo, S. Haddad, M. Mazo Canola; Hematology Oncology, UT health Mays Cancer Center, San Antonio, TX.
Introduction: CDK4/6 inhibitors are essential in managing HR-positive, HER2-negative metastatic breast cancer, but can cause cutaneous side effects. While alopecia and rash are common, pigmentary disorders like vitiligo are rare and often underrecognized, occurring in fewer than 1% of patients. Emerging reports suggest that vitiligo-like lesions may occur more frequently with ribociclib compared to other CDK4/6 inhibitors. We present a case of ribociclib-induced vitiligo that improved significantly after switching to abemaciclib.Case Presentation: A 44-year-old premenopausal woman was initially diagnosed with multifocal disease in the right breast, consisting of two primary lesions: ductal carcinoma in situ (DCIS), ER/PR positive, grade 3, and invasive ductal carcinoma (IDC), ER/PR positive, HER2 negative. Sentinel lymph node biopsy was negative. She underwent adjuvant therapy with Lupron and tamoxifen but self-discontinued treatment after three years. Ten years later, she presented with new-onset persistent left hip pain, prompting further evaluation that revealed metastatic breast cancer involving the bones. Treatment was initiated with ribociclib, letrozole, and zoladex; genomic profiling (NGS) showed no targetable mutations.After two years of therapy, the patient developed depigmented patches localized predominantly on the dorsal aspect of her hands, anterior neck, and face, along with ill-defined hypopigmented to near-depigmented patches on her forearms and upper arms, involving approximately 20% of her body surface area. Notably, her back and feet were unaffected. Dermatology consultation led to initiation of ruxolitinib cream, applied daily to twice daily on affected areas, along with narrowband UVB phototherapy administered three times weekly starting at 350 mJ/cm², with incremental increases of 50 mJ/cm² per session, up to a maximum dose of 4000 mJ/cm². Due to suspicion of ribociclib-induced vitiligo, ribociclib was discontinued and replaced with abemaciclib, resulting in significant repigmentation of previously affected areas.Discussion: Vitiligo-like lesions are rare cutaneous toxicities associated with CDK4/6 inhibitors and may be underreported due to delayed recognition or asymptomatic presentation. In our patient, the lesions primarily affected sun-exposed areas, consistent with prior reports. Initial treatment with topical ruxolitinib and NB-UVB phototherapy yielded limited benefit, but discontinuation of ribociclib and transition to abemaciclib led to significant repigmentation. This suggests a potential drug-specific effect and highlights the importance of considering medication changes when managing CDK4/6i-induced pigmentary changes. Prompt recognition and tailored dermatologic management, including coordination with oncology for treatment modification, can improve outcomes and patient quality of life.Conclusion: This case contributes to the limited literature on CDK4/6 inhibitor-induced pigmentary changes. It underscores the need for clinical awareness of this rare toxicity and demonstrates that switching agents within the same drug class may offer symptom resolution without compromising cancer treatment.
Presentation numberPS5-06-26
Androgen Receptor Positive Breast Cancer in a Female-to-Male Transgender Individual
Kendra Wilson, UT Health Science Center Tyler, Tyler, TX
K. Wilson1, F. Raza2, K. Donthireddy1; 1Hematology/Medical Oncology, UT Health Science Center Tyler, Tyler, TX, 2Internal Medicine, UT Health Science Center Tyler, Tyler, TX.
Background: Limited guidelines exist regarding breast health in the transgender population. The method of gender-affirming transition undergone by a transgender individual has been shown to influence the risk of developing breast cancer. The role between exogenous testosterone and the pathogenesis of breast cancer remains unclear. Here, we report a case of invasive ductal carcinoma of the breast in a female-to-male transgender patient after receiving gender-affirming hormonal therapy. Case Presentation: A 36-year-old female-to-male transgender individual presented with a palpable left breast mass for evaluation. The patient began testosterone and aromatase inhibitor for transitioning six years prior. They reported menarche at age 12 with continued monthly menstrual cycles since initiation of gender-affirming hormonal therapy. Family history was significant for multiple family members with breast cancer and colon cancer. Clinical exam was significant for 3cm mobile mass at 6 o’clock position and matted level one tender lymph node. The patient underwent mammogram and biopsy that confirmed a 3.8cm mass in the lower inner left breast. Histopathological exam of the biopsy revealed invasive ductal carcinoma, grade III, estrogen receptor positive (39%), progesterone receptor negative (0%), human epidermal growth factor receptor 2 negative (0%), with Ki67 85%. Androgen receptor was positive (34%). The patient then underwent PET scan that confirmed locally advanced disease with 4.1cm left breast mass with left axillary adenopathy. Genetic testing was obtained that was negative for BRCA 1/2. The patient was recommended neoadjuvant chemotherapy and has begun treatment with dose-dense doxorubicin, cyclophosphamide and paclitaxel regime. The patient was advised to discontinue testosterone therapy. They are planned for hysterectomy with bilateral salpingo-oophorectomy following treatment for breast cancer. Conclusion: Our case highlights the importance of identifying personal risk factors for breast cancer such as family history, genetic predispositions, and surgical procedures performed for this patient population. Most notably, our case identifies the need for further research into the role of gender-affirming hormonal therapy on breast cancer risk specifically in patients receiving testosterone therapy without luteinizing hormone-releasing hormone or oophorectomy. Discussion: From review of literature, female-to-male individuals who have undergone chest reconstruction surgery to remove breast tissue have significantly reduced risk of breast cancer. These individuals face higher breast cancer risk compared to cisgender men but lower than cisgender women. The use of testosterone therapy and effects on breast cancer are controversial. One theory is protective by decreasing glandular tissues, ultimately reducing the amount of tissue susceptible to cancer. The counter theory is causative in that testosterone is aromatized to estradiol leading to increased proliferation and breast cancer risk. Our case highlights the risk of inadequate estrogen suppression and prolonged androgen stimulation leading to breast cancer in our female-to-male transgender patient. More research with randomized controlled trials is necessary to determine the overall effect on each patient’s risk to ensure clinicians that are prescribing these hormonal therapies for medical transition can have open risk-versus-benefit discussions with these patients. It will also be necessary to gain surveillance data on gender affirming hormones effect on breast cancer to aid clinicians in screening and clinical recommendations, as current screening guidelines vary among organizations.
Presentation numberPS5-06-27
Reframing Breast Cancer Therapies in a Rare Context – A Case of Metastatic Mammary-Like Vulvar Carcinoma Treated with Breast Cancer Regimens
Dali Edwards, Medical University of South Carolina, Charleston, SC
D. Edwards1, C. Coleman1, A. Jamil2; 1Hematology and Oncology, Medical University of South Carolina, Charleston, SC, 2Internal Medicine, Samaritan Medical Center, Watertown, NY.
Background: Mammary-like adenocarcinoma of the vulva (MLVA) is a rare subtype of vulvar adenocarcinoma that exhibits morphological and immunohistochemical features similar to breast tissue. Arising from anogenital mammary-like glands, this entity poses unique diagnostic and therapeutic challenges due to its extreme rarity and overlapping features with both breast and gynecologic malignancies. Currently, there are no standardized treatment guidelines for MLVA, and clinical management is often extrapolated from therapeutic strategies used in breast cancer, particularly for tumors with hormone receptor or HER2 expression. Case: We report a case of a 60-year-old woman initially diagnosed with stage IV mammary-like adenocarcinoma of the vulva in August 2022. She underwent a modified radical vulvectomy with bilateral inguinal lymphadenectomy. Pathology confirmed adenocarcinoma arising from anogenital mammary-like gland, ER-negative and PR-negative, and HER2-positive. She received adjuvant radiation with concurrent cisplatin. Given HER2 positivity, she was treated with six cycles of docetaxel, trastuzumab, and pertuzumab, followed by maintenance trastuzumab and pertuzumab. In August 2021, staging scans revealed new lung metastasis. A lung biopsy confirmed moderately differentiated adenocarcinoma consistent with metastasis from the patient’s known vulvar primary, HER2 2+ (FISH negative) and ER/PR negative, immunohistochemical stains – GATA3-positive, and PAX8-negative, SOX10-negative. Molecular testing revealed a germline BRCA2 mutation. She was treated with olaparib from February to November 2023. Following disease progression, she transitioned to trastuzumab deruxtecan for HER2-low disease, which resulted in temporary disease control between December 2022 to May 2024. Upon further progression, she received carboplatin, paclitaxel, and bevacizumab, completing six cycles in May 2024, followed by maintenance bevacizumab and pembrolizumab. With continued progression, she was re-challenged with paclitaxel. Staging scans in February 2025 showed disease progression including new brain metastasis. Repeat liver biopsy demonstrated moderately differentiated adenocarcinoma, HER2 1+. For brain metastases, she received local treatment with gamma knife radiosurgery. She was re-referred to the breast oncology clinic for co-management and discussion of next-line therapy. She was initiated on Sacituzumab Govitecan. Initial follow-up showed disease stability and good tolerance to therapy. Notably, the patient continues to work full-time as a dump truck driver, underscoring her preserved functional status and overall quality of life while on treatment. Conclusion: MLVA is an extremely rare malignancy with no established standards of care. Its histologic resemblance to breast cancer suggests it may respond to breast cancer-based therapies. In the absence of defined treatment pathways, applying breast cancer therapeutic strategies—particularly in the metastatic setting—may offer a rational and effective approach for managing this rare tumor type. While Sacituzumab Govitecan is not currently approved for vulvar malignancies, Sacituzumab govitecan targets the TROP2 protein, which is found on the surface of many cancer cells, including some mammary-like adenocarcinomas of the vulva. Trop-2-targeted mechanism of Sacituzumab Govitecan may offer a rational therapeutic approach in rare tumors with overlapping molecular features.
Presentation numberPS5-06-28
Carcinoma en Cuirasse and Radiation-Induced Morphea
Holly Grace, UT Southwestern Medical Center, Dallas, TX
H. Grace, R. Assadi, N. Wandrey, M. Arbab, A. Rahimi; Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX.
Carcinoma en Cuirasse (CeC) and Radiation-Induced Morphea (RIM) are two discrete and rare diagnoses, most commonly found among patients treated for breast cancer. Due to their similar clinical presentation and extremely low incidence rates, it can be challenging for providers to differentiate the pathologies promptly and accurately. Both pathologic processes result in sclerotic skin plaques with fibrotic changes to the dermis and subcutaneous tissue. Timely diagnosis is critical due to the high mortality rate of CeC and the potential reversibility of RIM if treated promptly. For this reason, improved patient care is dependent on increased awareness of how to distinguish these pathologies. In CeC, metastatic cells invade and disseminate via the superficial lymphatic vessels. The tumor cells are visualized in pattern of single-file lines, poorly differentiated, with atypical nuclei. There are hyperpigmented, indurated, and sclerotic plaques as well as firm, erythematous papules. The surrounding tissue is fibrotic with reduced vascularity. This fibrosis inhibits penetration of chemotherapy and can reduce efficacy of systemic therapies. Management is dependent on the cancer morphology and prompt initiation of the next line of therapy is recommended. RIM presents with erythema which progresses to sclerotic plaques and ultimately irreversible fibrosis of the dermis and subcutaneous tissue. Treatment consists of topical corticosteroids and calcineurin inhibitors for early stages, progressing to systemic treatment of immunosuppressants such as mycophenolate mofetil and methotrexate if needed. Retrospective analysis has shown that methotrexate or UVB phototherapy have achieved the best results.
| Characteristic | Case 1: CeC | Case 2: RIM |
| Age at diagnosis (years) | 63 | 65 |
| Sex | Female | Female |
| Past Medical History | Hypothyroidism, seasonal allergies | Hypertension, type 2 diabetes mellitus, stage 3 chronic kidney disease |
| Past dermatologic conditions | None | None |
| Oncologic condition | Invasive ductal carcinoma of the Right breast, ypT1a ypN2a, grade 3, 7/8 lymph nodes positive, negative margins, positive lymphovascular space invasion | Invasive ducal carcinoma of the Left breast, ypT1cN1a, grade 2, 11/15 lymph nodes positive, negative margins, positive lymphovascular space invasion |
| Prior treatments for oncologic condition | Neoadjuvant carboplatin, paclitaxel, and pembrolizumab, bilateral modified radical mastectomy with right axillary lymph node dissection, and adjuvant whole chest wall and regional lymph node irradiation of 50.4 Gy in 28 fractions with a sequential boost to the right mastectomy scar of 10 Gy in 5 fractions | Partial mastectomy with axillary lymph node dissection and adjuvant whole breast and regional lymph node irradiation of 50.4 Gy in 28 fractions |
| Time between RT and onset of condition | 0 months | 8 years |
| Investigation to confirm diagnosis | Punch biopsy | Punch biopsy |
| Treatments | Sacituzumab govitecan, palliative RT 26 Gy in 5 fx to the left chest wall and 20 Gy in 5 fx to the right back | Topical clobetasol and UVB phototherapy |
| Toxicity risks | Reirradiation dermatitis, skin necrosis | Recurrence associated with immunosuppression |
Presentation numberPS5-06-29
Phantom limb pain patients with Breast cancer: Updates in rehabilitation paradigms
Breanne Nguyen, UT Health San Antonio, San antonio, TX
B. Nguyen, S. Anholt, J. Birch; Physical Therapy, UT Health San Antonio, San antonio, TX.
Introduction: By using current evidence-based practice mechanisms of pain or pain phenotypes, we can better select candidates for appropriate interventions within oncology rehabilitation. Using subjective and objective assessment criteria for identifying candidates for nocioplastic treatment can result in better treatment outcomes for persistent pain in this traditionally recalcitrant patient population. This has been explored in the context of populations with amputation and CRPS and has yet to be approached within physical therapy in oncology. Purpose: To demonstrate application of nocioplastic assessment and treatment within the realm of physical therapy, oncology specialized practice. Methodology: Case Study of patient with breast cancer and subsequent upper quarter impairments including pain, limitations in shoulder AROM, and trunk lymphedema after mastectomy, axillary radiation, and chemotherapeutic interventions. Results: Noted improved laterality, and shoulder AROM and functional subjective outcomes noted as measured by the Quick-DASH after nocioplastic specific interventions. Conclusions: By treating the central component of pain regarding mastectomy and subsequent current standard of practice for breast cancer, we mitigate impairments more effectively. The prospective surveillance model to impact known impairments, functional activity limitations, and participation limitations could adopt this assessment in at risk populations within the oncology sector.
Presentation numberPS5-04-30
A Rare Case of Fibroadenoma After Long-Term Estrogen Therapy in a Transgender Woman
Joseph Sahagun, University of Illinois, Chicago, IL
J. Sahagun1, E. A. Marcus2; 1General Surgery, University of Illinois, Chicago, IL, 2Breast Surgery, John H. Stroger Hospital of Cook County, Chicago, IL.
Fibroadenomas are benign breast masses typically present as painless, mobile, well-circumscribed lesions commonly encountered in young women. They are well characterized in this population but not routinely studied in other groups. Fibroadenomas are rare in cisgender men and even less frequently documented in transgender women. Estrogen-based hormone therapy is a fundamental component of gender-affirming treatment for transfeminine individuals, promoting the development of secondary breast tissue. Prolonged exposure to exogenous estrogen may predispose this population to breast pathology not typically observed in cisgender males. We describe the case of a 30-year-old transgender woman who presented with a palpable, mobile breast mass following ten years of continuous oral estrogen therapy. Surgical excision of the breast mass confirmed the diagnosis of a fibroadenoma. With the increasing accessibility of hormonal replacement therapy (HRT) and expanded transgender healthcare services, it is reasonable to expect a rise in breast pathology within this population. HRT-induced breast development facilitates the emergence of breast conditions typically seen in cisgender women, including fibroadenomas and potential malignancy. Despite their rarity in cisgender men and the limited number of reported cases in transgender women, thorough evaluation of any breast lesion in this population remains essential to ensure accurate diagnosis and appropriate management.
Presentation numberPS5-10-26
Breast Cancer Screening Patient Navigation Decreases Time to Screening and Diagnosis for FQHC Patients
Martha Welman, Neighborhood Health, Alexandria, VA
M. Welman, J. Henry, N. Hojvat-Gallin; Neighborhood Health, Alexandria, VA
Background Early breast cancer detection is critical for preventing mortality, yet many patients – especially those that are racial/ethnic minorities, low-income, and underserved – face barriers that prevent them from progressing through the screening and diagnostic pathway in a timely manner. Neighborhood Health (NH) is a Federally Qualified Health Center located in Northern Virginia that serves more than 42,700 patients annually, a majority who identify as racial/ethnic minorities and speak languages other than English. 52% of adult patients are uninsured and 30% are enrolled in Medicaid. Approximately 9,000 patients are women 40-75 years old. In 2024, NH implemented a breast cancer screening and linkage to care program to address low breast cancer screening rates through patient navigation. To assess the impact of our system-level process improvements and pre-diagnostic patient navigation on patient progress through the screening and diagnostic pathway, we compared time between screening and diagnostic exams pre- and post-intervention. Methods To address existing barriers to breast cancer screening for the patient population, two bilingual patient navigators were hired to work one on one with patients, a standing order was implemented to facilitate scheduling by clinical staff and navigators, access to imaging was improved through partnerships with local radiology providers and a mobile mammovan, provider communication was streamlined to ensure timely follow-up, and bi-directional texting was utilized for patient appointment reminders. We analyzed baseline median days between screening referral and completion, screening completion and first diagnostic exam (if recalled), diagnostic referral and completion, and diagnostic exam to biopsy completion to the post-implementation period. We also tracked late-stage cancer diagnoses by year. Results Patient data reflects project period April 2024 to August 2025 and includes completion of 6,602 screening mammograms, 2,072 diagnostic exams, and 325 biopsies. Between 2023 and 2025, the time between screening mammogram referral and completed decreased 48% (79 days to 41 days), time between screening completion and first diagnostic exam (if recalled) decreased 51% (59 days to 29 days), time between diagnostic referral and completion decreased 47% (62 days to 33 days), and time from first diagnostic exam to biopsy completion decreased 48% (40 days to 21 days). Late-stage diagnoses also decreased from 7 (23.3%) patients in the baseline period to 2 (7.4%) patients in 2024. Conclusions System-wide process improvements and implementation of pre-diagnostic patient navigation decreased linkage intervals throughout the breast cancer screening and diagnostic pathway. More work is needed to identify and address remaining barriers to timely completion of outstanding diagnostics and biopsies and to ensure sustainability of patient navigators.
| Days from screening mammogram order to completion | If recalled, days from screening completion to first dx exam | Days from diagnostic order to completion | Days from first dx exam to biopsy completion | |
| 2023 (baseline) | 79 (N=2396) | 59 (N=106) | 62 (N=615) | Data not available |
| 2024 | 55 (N=2719) | 40 (N=439) | 56 (N=1219) | 40 (N=208) |
| 2025 (8 months) | 41 (N=1487) | 29 (N=180) | 33 (N=685) | 21 (N=80) |
Presentation numberPS5-11-12
Economic Impact of Adverse Event Management in HR+/HER2− Metastatic Breast Cancer: A Comparative Analysis of Datopotamab deruxtecan and standard of care in the Brazilian Private Healthcare System
Thiago Belchior de Oliveira, Daiichi Sankyo, São Paulo, Brazil
H. T. dos Santos1, T. B. de Oliveira1, A. B. Villa1, C. d. Huerta1, L. dos Santos1, F. d. Costa2, G. L. V. Araujo1; 1Daiichi Sankyo, São Paulo, BRAZIL, 2COE Ensino e Pesquisa, São José dos Campos, BRAZIL.
Background: TROPION-Breast011 (TB01), a phase III clinical trial evaluated the safety and efficacy of datopotamab deruxtecan (Dato-DXd) in comparison to the investigator’s choice of standard single-agent chemotherapy (ICC). Dato-DXd demonstrated a manageable safety profile, characterized by a significantly lower incidence of grade ≥3 adverse events (AEs) compared to ICC (21% vs. 45%)1. Objective: This study aims to evaluate and compare the economic burden of managing grade ≥3 AEs in patients with HR+/HER2− metastatic breast cancer who have progressed on endocrine therapy and received at least one line of systemic. Methods: The therapeutic regimens analyzed in this study were derived from two pivotal clinical trials: TB01, which compared Dato-DXd with ICC: (capecitabine, eribulin, gemcitabine, vinorelbine), and TROPICS-022, which assessed sacituzumab govitecan (SG) versus ICC. AEs frequencies were extracted from the afore-mentioned clinical trials. To identify clinically and economically relevant events, a threshold of ≥10% frequency for any-grade AEs was applied, considering occurrences in each arm of the studies analyzed. Additionally, all grade ≥3 AEs were included regardless of frequency. Cost estimation was performed using a micro-costing approach3, incorporating direct medical expenses specifically associated with the management of grade ≥3 AEs. The total AE-related cost per treatment arm was calculated by multiplying the unit cost of each selected AE by its respective frequency and summing the resulting values. Results: The AEs selected for analysis are summarized in Table 1. Treatment with Dato-DXd resulted in an estimated AE-related cost of US$ 155.45, reflecting a 41.9% decrease in toxicity-related expenditures relative to its respective ICC arm comparator (US$ 267.36), while SG was associated with a total AE-related cost of US$ 535.66, compared to US$ 304.02 for its respective ICC arm comparator, indicating a 76.2% increase in costs related to adverse event management. Conclusion: This analysis identified differences in AE-related costs across treatment modalities, emphasizing the economic relevance of toxicity management in patients with HR+/HER2− metastatic breast cancer who have progressed on endocrine therapy and received at least one line of systemic treatment. Dato-DXd demonstrated lower expenditures associated with adverse event management compared to its respective ICC arm comparator. SG was associated with higher AE-related costs relative to its respective ICC arm comparator. These findings align with previous evidence, including the Ryczek (2024)4 study. The data highlights the importance of incorporating toxicity-related costs into treatment decision-making within the Brazilian Private Healthcare System.
| Adverse Events | TROPION-Breast01 1 %($) | TROPiCS-02 2 %($) | ||
| Dato-DXd | ICC | SG | ICC | |
| Neutropenia | 1.1 (1.94) | 30.8 (53.72) | 50.7 (88.60) | 38.2 (66.61) |
| Anaemia | 1.1 (24.7) | 2.0 (44.33) | 6.3 (140.98) | 3.2 (71.41) |
| Leukopenia | 0.6 (6.76) | 6.8 (83.23) | 8.6 (104.46) | 5.2 (63.55) |
| Lymphopenia | – | – | 3.7 (2.63) | 3.2 (2.26) |
| Thrombocytopenia | – | – | 0,4 (1.13) | 3.6 (10.95) |
| Diarrhea | – | 1.1 (11.29) | 9.3 (92.38) | 1.2 (11.93) |
| Nausea | 1.4 (5.83) | 0.6 (2.39) | 1.1 (4.70) | 2.8 (11.80) |
| Vomiting | 1.1 (4.67) | 0.6 (2.39) | 0.4 (1.57) | 1.6 (6.75) |
| Abdominal pain | – | – | 0.7 (13.73) | – |
| AST increased | 0.6 (1.56) | 0.6 (1.56) | – | 1.2 (3.38) |
| ALT increased | – | – | – | 2.4 (6.77( |
| Fadigue | 1.7 (18.07) | 2.0 (21.62) | 5.6 (60.69) | 2.8 (30.48) |
| Asthenia | 0.8 (9.04) | 1.1 (12.36) | 1.9 (20.23) | 0.8 (8.71) |
| Decreased appetite | 0.8 (0.74) | 0.6 (0.51) | 0.4 (0.33) | 0.4 (0.33) |
| Stomatitis | 6.4 (81.45) | 2.6 (32.69) | – | – |
| Dry eye | 0.6 (0.34) | – | – | – |
| Keratitis | 0.6 (0.36) | – | – | – |
| Palmar-plantar eythrodyses | – | 2.0 (1.27) | – | – |
| Neuropathy | – | – | 1.1 (4.22) | 2.4 (9.09) |
| Constipation | – | – | – | – |
| Alopecia | – | – | – | – |
| Total | 155.45 | 267.36 | 535.66 | 304.02 |
Presentation numberPS5-05-02
Real world data (RWD) outcome analysis of ESR1 mutation emergence in HR+/HER2- metastatic breast cancer through the continuum of standard of care hormonal therapy
Malvika Pillai, Tempus AI, Chicago, IL
M. Pillai1, N. Vidula2, Y. Hsieh1, Z. T. Rivers1, V. A. Rhodes1, S. Hyun1, V. L. Chiou1, C. Sangli1, H. Nimeiri1, A. M. Brufsky3, B. H. Park4; 1Tempus AI, Chicago, IL, 2Massachusetts General Hospital, Harvard Medical School, Boston, MA, 3Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, 4Vanderbilt University Medical Center, Nashville, TN.
Background Molecular surveillance adoption in hormone receptor-positive, HER2-negative metastatic breast cancer (HR+/HER2- mBC) has potential to inform critical treatment decisions in patients (pts) treated with aromatase inhibitor (AI) plus CDK4/6 inhibitor (CDK4/6i) therapy with detected ESR1 mutations (ESR1m). ESR1m patterns remain poorly characterized in real-world practice. We characterized ESR1m emergence and clinical impact in a large RWD cohort of HR+/HER2- mBC pts monitored with longitudinal testing. Methods Study cohort included pts in Tempus AI’s RWD, multimodal database with HR+/HER2- mBC diagnosis prior to January 2023 who received AI+CDK4/6i therapy, had reported results of comprehensive genomic profiling from tissue (Tempus xT) or liquid (Tempus xF) biopsy within 24 months (mo) of receiving AI+CDK4/6i therapy, and ≥1 on-treatment xF tests. Pts were stratified by ESR1m and ESR1-wildtype (ESR1wt) status during AI+CDK4/6i. Real-world overall survival (rwOS) was estimated using Kaplan-Meier survival curves. Survival differences between groups were assessed with a log-rank test. Followup was measured from AI+CDK4/6i therapy initiation. Delayed entry, due to first biopsy sequencing occurring after therapy initiation, was accounted for using risk set adjustment in rwOS estimates. Pts were censored at 60 mos after AI+CDK4/6i initiation or at last known followup. Pts who switched to elacestrant, ESR1m targeted therapy, were censored at therapy initiation. Incidence and timing of ESR1m emergence and treatment patterns post ESR1m detection were also assessed. Results Among 301 HR+/HER2-mBC pts treated with AI+CDK4/6i and undergoing longitudinal molecular testing, median time to on-treatment xF was 23.1 mo. Median time from AI+CDK4/6i initiation to ESR1m detection was 19.1 months overall, 21.0 months for patients with ESR1m detected during 1L therapy, and 12.3 months for those detected in 2L+ settings. Baseline characteristics were similar between ESR1m and ESR1wt groups (median age 64/63 years; 55%/ 57% White, 16%/12% Hispanic, 7%/7% Black, and 4%/4% Asian). ESR1m status was associated with reduced survival (median rwOS 44.0 mo vs. not reached for ESR1wt; log-rank test p=0.02). Median follow-up time overall was 33.8 mo. From on-treatment xF, median time to therapy switch was 41 days overall in ESR1m (41 days in 1L, 36 days in 2L+) and 107.5 days overall in ESR1wt (137.5 days in 1L, 81 days in 2L+). 18 pts went on elacestrant and 46 pts went on fulvestrant after ESR1m detection. ESR1m incidence was 33.9% overall (102/301 pts), 34.0% (69/203) in 1L and 33.7% (33/98) in 2L+ settings. From on-treatment xF, most common ESR1 point mutations were D538G (64.4%), Y537S (24.4%), Y537N (21.1%), Y537C (10.0%), and E380Q (5.6%). Multiple(M) ESR1 missense mutations were observed within individual tumor samples, with higher death rates in pts with multiple(M) than single(S) mutations(D538G(S): 38.5%; Y537S(S): 40.0%; Triple(M): 68.8%). Rare mutations (e.g., S463P, V422del) were also observed. PIK3CA co-mutations occurred in 35.9% (108 pts), more frequently in ESR1m pts (42.2%) than ESR1wt pts (32.7%) (chi-square test, p=0.13). Conclusion This large multimodal RWD outcome analysis from longitudinal molecular surveillance testing in HR+/HER2- mBC pts treated with AI+CDK4/6i sheds light on the continuum of ESR1m emergence and pt outcomes from 1L and beyond outside of clinical trial data. Our analyses show higher ESR1m incidence is associated with reduced survival regardless of line of therapy. RWD analyses also indicate the complexity of resistance within ESR1 gene including multiple mutations or co-mutations with PIK3CA and occurrence of rare ESR1m variants. The impact of these variants on therapeutic decision-making will be investigated in future research.
Presentation numberPS5-11-14
Systemic Barriers to Breast Cancer Care in Conflict-Affected Syria
Ahmad Al-Bitar, Damascus University, Damascus, Syrian Arab Republic
A. Al-Bitar1, A. Kouli1, F. Nahhat2, M. Saifo1; 1Damascus University, Damascus, Syrian Arab Republic, 2Mayo Clinic, Phoenix, AZ
Background: In conflict zones like Syria, accessing cancer care presents immense challenges. However, there is a critical lack of data on the specific barriers patients face. This study aimed to document these obstacles for breast cancer patients in Syria following 14 years of civil war. We surveyed patients presenting for cancer care to better understand the challenges they faced. Methods: This cross-sectional study included adult patients with a confirmed breast cancer diagnosis presenting to Al-Bairouni University Hospital for care. Data was collected through face-to-face interviews using a structured questionnaire adapted for the Syrian context. The survey covered six key areas: Demographic & Clinical, Healthcare System Barriers, Financial Barriers, Geographic & Security barriers, Reasons for Diagnostic Delays, and Psychosocial Impact. Multivariate analysis via logistic regression was conducted to determine which responses were associated with a higher odds ratio (OR) of having a late-stage (III/IV) cancer diagnosis. Results: A total of 109 patients diagnosed with breast cancer were enrolled. The mean age was 51.4 years (±15.2 SD). Advanced diagnostics (CT/MRI/biopsy) and radiation therapy were inaccessible for 58% (n=63) and 63% (n=69) of patients. Systemic therapy drugs were “rarely” or “never” available for 33% (n=36) of patients, and 14% (n=15) reported waiting over three months for an appointment. The costs of medications “greatly” or “very greatly” impacted 86% (n=94) of patients. Over two-thirds of patients reported that essential pain control medications (e.g., morphine) were “rarely” or “never” available (66%, n=72). Nearly two-thirds (67%, n =73) reported that security concerns “greatly” or “somewhat” affected their ability to access treatment. An overwhelming 89% (n=97) of patients reported a negative psychological impact from their treatment course. Patient responses associated with a higher OR of late-stage cancer diagnosis are present in Table 1. Conclusions: Breast cancer patients in Syria confront overwhelming systemic, financial, and security barriers that compromise their care and contribute to advanced diagnoses. The findings highlight a profound humanitarian crisis and underscore the urgent need for targeted public health and aid interventions to support cancer care in conflict settings. Table 1: Factors Associated with Late-Stage (III/IV) Diagnosis of Breast Cancer (Multivariate Logistic Regression)
| Variable | Adjusted Odds Ratio (aOR) | 95% Confidence Interval | p-value | ||||
| Waiting Time First Oncology appointment >1 month | 3.11 | 1.47 – 6.65 | 0.008* | ||||
| Unemployed due to cancer | 2.83 | 1.21 – 6.73 | 0.011* | ||||
| Sold Assets to Finance Care | 2.42 | 1.14 – 5.21 | 0.002* |
Presentation numberPS5-05-21
Breast Cancer in Syria: An Epidemiological Analysis from the Country’s Main Cancer Center
Ahmad Al-Bitar, Damascus University, Damascus, Syrian Arab Republic
A. Al-Bitar, F. Al-Jojo, A. Kouli, R. Al-Khouli, R. Al-Ahmad; Damascus University, Damascus, Syrian Arab Republic
Background Epidemiological data on breast cancer in conflict-affected regions such as Syria are severely limited. Our study aims to fill in the gap in data by evaluating the demographics of Syrian breast cancer patients over a three-year period. Methods This is a retrospective epidemiological study conducted at Al-Bairouni University Hospital – Syria’s national cancer center serving 60-70% of the country’s cancer patients. We included all patients diagnosed with breast cancer (BC) between January 2022 and December 2024. Data extracted from hospital records included sex, age at diagnosis, cancer stage, smoking status, and the governorate of residence. Patients were grouped by disease group and year to assess annual trends. Institutional ethical approval was obtained to perform the study. Results A total of 5630 were diagnosed with breast cancer during the study period: 1513 in 2022, 1964 in 2023, and 2153 in 2024 (Table 1). Across all years, a consistent female predominance was observed (99.3%). The average age at diagnosis was 50.5 years, 20.75% of patients were smokers, and 59.02%. had advanced/metastatic disease at diagnosis. IDC (4912) was the most common breast cancer. The highest patient loads originated from Damascus (16.1%) and Rural Damascus (17.1%), followed by central governorates such as Homs (10.78%) and Hama (9.15%). Notably, patients from underserved governorates such as Deir ez-Zor (6.47%), Aleppo (8.76%), and Al-Hasakeh (6.77%) represented a significant proportion of the cohort. Conclusions To our knowledge, this study provides the first comprehensive multi-year dataset of breast cancer in Syria, with representation from all 14 governorates in the country. More than 20% of our cohort were smokers, and nearly 59%. were diagnosed with advanced disease, demonstrating a need for public health efforts to improve screening & early detection methods. This dataset offers a foundation for future public health planning and research efforts in the region. Table 1
| 2022 Incidence | 2023 Incidence | 2024 Incidence | Total cases | Average Age | Laterality | Sex | Smokers (%) | Advanced/Metastatic at diagnosis | |||||||||||
| IDC | 1274 | 1733 | 1905 | 4912 | 51.1 | 52.32% (L) | 4878 F 35 M | 22.05% | 56.44% | ||||||||||
| ILC | 220 | 208 | 215 | 643 | 50.9 | 32.60% (L) | 641 F 2 M | 22.86% | 68.91% | ||||||||||
| Others | 19 | 23 | 33 | 75 | 49.4 | 43.1% (L) | 73 F 2 M | 17.33% | 51.72% |
Presentation numberPS5-06-17
Sentinel lymph node biopsy after neoadjuvant chemotherapy in patients with breast cancer using blue dye alone: Results from a multicenter study in Public Hospitals in Brazil
Rafael Henrique Szymanski Machado, Hospital Federal da Lagoa, Rio de Janeiro, Brazil
C. Lopes1, R. S. Machado2, S. Trota1, T. Orge3, L. Oliveira3, D. Torres2, F. Bacellar3, S. Paiva2, L. Piana2, J. Bines4; 1Instituto Oncologico Juiz de Fora, Juiz de Fora, BRAZIL, 2Hospital Federal da Lagoa, Rio de Janeiro, BRAZIL, 3Hospital da Mulher, Bahia, BRAZIL, 4Instituto Nacional do Câncer, Rio de Janeiro, BRAZIL.
Introduction: In recent decades, breast cancer surgery has evolved from radical procedures to more conservative approaches, with emphasis on sentinel lymph node biopsy (SLNB), which reduces morbidity without compromising oncologic control. The introduction of neoadjuvant chemotherapy (NAC) has expanded the use of SLNB, even in patients with initially clinically positive axilla (cN1/N2) who converted to clinically negative status after treatment. However, the safety of this technique in cN1/N2 scenarios remains debated, especially given the lack of resources such as dual tracer techniques (using both blue dye and technetium for sentinel node identification) and lymph node clip placement in public health systems.Methods: This multicenter retrospective study analyzed 369 breast cancer patients treated with NAC between 2017 and 2021 at three public hospitals in Brazil. Patients were grouped according to their pre NAC axillary status and the type of axillary surgery performed, either SLNB or axillary lymph node dissection (ALND). All patients had clinically negative axilla following NAC.Objective: The primary objective was to evaluate axillary recurrence in patients who were initially cN1/N2 and converted to cN0.Results: Among patients who underwent SLNB using blue dye alone as the sole mapping method, the axillary recurrence rate was 2.4% in the group initially with clinically negative axilla (cN0) and 2.6% in the initially cN1/N2 group, with no statistical difference. Overall survival (OS) was also similar, at 94.1% in the cN0 group and 89.7% in the cN1/N2 group, without statistical significance (p=0.255), as were the other survival outcomes analyzed. When comparing initially cN1/N2 patients who underwent SLNB or ALND, similar axillary recurrence rates were observed: 2.6% in the SLNB group and 2.3% in the ALND group. OS of 89.7% and 87.8%, respectively, also showing no statistically significant difference. Furthermore, when all 163 patients who underwent SLNB were grouped regardless of initial axillary status and compared to the 206 patients who underwent ALND, the axillary recurrence rates remained comparable: 2.5% in the SLNB group and 1.9% in the ALND group. Other outcomes, including local and locoregional recurrence, disease-free survival, distant disease-free survival and overall survival, also showed similar results between groups. Conclusion: SLNB with blue dye alone, even without complementary techniques, demonstrated acceptable oncologic safety in patients who converted to cN0 after NAC. These findings are especially relevant in resource-limited settings and support the use of less invasive strategies to reduce complications without compromising therapeutic effectiveness. Further studies are needed to validate these results.Keywords: Breast cancer; Sentinel lymph node biopsy; Neoadjuvant chemotherapy; Axillary lymph node dissection; Public Health System.
Presentation numberPS5-06-25
Investigating association of comorbidities and race with all-cause mortality outcomes of PI3K inhibitor use in metastatic breast cancer (mBC)
Tahj S Morales, Penn State College of Medicine, Hershey, PA
T. S. Morales1, A. Strong1, J. Petucci2, M. K. Vasekar3; 1Penn State College of Medicine, Hershey, PA, 2Institute for Computational and Data Sciences, University Park, PA, 3Penn State Milton S. Hershey Medical Center, Hershey, PA.
PI3K and AKT pathway inhibitors Alpelisib, Inavolisib, and Capivasertib (PI3K/AKTi) are FDA approved treatments with demonstrated significant clinical benefit, improving progression free survival for Hormone receptor positive (HR+) mBC. Hyperglycemia can commonly develop from these agents. While knowledge about disparities in outcomes based on race and comorbidities in certain mBC subtypes is emerging, real world data about PI3K/AKTi is limited. In this propensity score-matched cohort study we used TriNetX de‐identified EHR data from multiple health systems to compare mortality of patients with breast cancer diagnoses treated with PI3K/AKTi. We identified 2372 patients, female (F) and male (M), stratified in African American (AA; n= 262F, 8M), White (W; n=1933 F, 25M) and Asian (A, n=144F) cohorts. Qualification into the three race-based cohorts required presence of a C50 ICD-10-CM diagnosis code and treatment with PI3K/AKTi. Mortality outcomes were also compared in separate cohorts examining hazard ratios for conditions developed before or after treatment: diabetes (E08-E13), hypertension (I10-I15), obesity (E66), and hyperlipidemia (E78.4-5). Cohorts were matched for age, comorbidities, and treatment lines using 1:1 matching with a greedy nearest neighbor search. All-time Odds ratios (OR) and/or Hazard ratios (HR) with 95% confidence intervals are reported as an effect size and significance estimation. Compared to non-diabetic patients, those diagnosed with diabetes after treatment had higher odds of mortality post-matching (OR = 1.53, 95% CI 1.19-1.97), but those with pre-existing diabetes showed no significant difference in mortality (matched OR = 1.19, 95% CI 0.97-1.45). Both pre-existing hypertension and hypertension diagnosed post-treatment were associated with higher mortality compared to non-hypertensive patients: matched ORs = 1.23 (95% CI 1.02-1.47) and 2.02 (95% CI 1.36-3.00), matched HRs = 1.204 (95% CI 1.05-1.38) and 1.911 (95% CI 1.42-2.57), respectively. No statistically significant difference in mortality odds was seen comparing the AA cohort to W cohort (matched OR = 0.99, 95% CI 0.70-1.39) or A cohort to W cohort post matching (OR = 0.72, 95% CI 0.45-1.16). New-onset hyperlipidemia was not statistically significant after matching (OR = 1.15, 95% CI 0.91-1.45; HR = 1.15, 95% CI 0.97-1.35). No significant association was seen with obesity before or after treatment (all CI including 1). This study demonstrates that hypertension diagnosed before and after treatment with PI3K/AKTi is associated with higher mortality, as is diabetes diagnosis post-treatment. We did not identify disparities in mortality between our 3 racial cohorts. Future studies are needed to understand interactions between individual PI3K/AKTi agents and comorbidities to inform appropriate management strategies and ultimately improve patient outcomes.
| Analysis Date: September 22, 2025 | African American (AA) | White (W) | Asian (A) |
| Number of Patients | 270 | 1958 | 144 |
| Age at Index | 60.5 ± 12.6 | 63.7 ± 11.7 | 63.7 ± 11.7 |
| Gender (F) | 262 | 1933 | 144 |
| Diabetes mellitus (E08-E13) at Index | 71 | 332 | 30 |
| Hypertension (I10-I15) at Index | 163 | 852 | 59 |
| Median Survival (days) | 692 | 686 | 754 |
Presentation numberPS5-04-08
Abemaciclib plus endocrine therapy in hr+/her2− advanced breast cancer: insights from an italian retrospective observational study
Elisabetta Munzone, Division of Medical Senology, European Institute of Oncology IRCCS, Milan, Italy
E. Munzone1, V. Guarneri2, A. Ferro3, A. Beano4, G. V. Bianchi5, A. Fabi6, G. Bianchini7, F. Riccardi8, F. Giovanardi9, V. E. Chiuri10, E. Fiorio11, I. Portarena12, P. Tassone13, A. Avitabile14, S. Baffini14, M. A. Sarno14, A. Tamma14; 1Division of Medical Senology, European Institute of Oncology IRCCS, Milan, Italy, 2Medical Oncology 2, Veneto Institute of Oncology (IOV), IRCCS; Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy, 3Department of Medical Oncology, Santa Chiara Hospital; Rete clinica senologica e UO di Oncologia Medica, Azienda Provinciale per i Servizi Sanitari, Trento, Italy, 4SSD Oncologia Senologica, Dipartimento di Oncologia, AOU Città della Salute e della Scienza, Torino, Italy, 5SS Oncologia Medica Senologica, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy, 6Precision Medicine Unit in Senology, Department of Woman, Child and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy, 7Department of Medical Oncology, San Raffaele Hospital, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy, 8Oncology Unit, Antonio Cardarelli Hospital, Naples, Italy, 9Medical Oncology, Azienda Unità Sanitaria Locale-IRCCS, Reggio Emilia, Italy, 10Unit of Medical Oncology, Sacro Cuore di Gesù Hospital, Gallipoli, Lecce, Italy, 11Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital, Verona, Italy, 12Oncology Department, Policlinico Tor Vergata, Rome, Italy, 13Translational Medical Oncology Unit, AOU Renato Dulbecco; Magna Graecia University, Catanzaro, Italy, 14Eli Lilly Italia S.p.A., Sesto Fiorentino, Italy
Rationale: Abemaciclib, approved in Italy since January 2020, improves progression-free survival (PFS) and overall survival (OS) in women with hormone receptor-positive, human epidermal growth factor receptor 2-negative (HR+/HER2−) advanced breast cancer (ABC). This retrospective study described the demographic and clinical characteristics of patient with ABC treated with abemaciclib plus endocrine therapy (ET) and evaluated its effectiveness in routine clinical practice in Italy. Methods: This is an Italian, multicenter, retrospective study. Data on patients with HR+/HER2− ABC who started abemaciclib between Jun-21 and Jun-22 was collected from 13 sites. The study period spanned from initial diagnosis to 24 months post-abemaciclib initiation. The primary endpoint, 24-month PFS in real-world (rwPFS) including the rwPFS rate, was estimated using the Kaplan-Meier method. Co-primary endpoints include overall response rate (rwORR), clinical benefit rate (rwCBR), time to treatment discontinuation (rwTDT). Results: 199 patients, who started abemaciclib in combination with fulvestrant or an aromatase inhibitor (AI), were enrolled. At abemaciclib initiation, the mean age was 62 years, and most patients (97.5%) had an ECOG PS of 0-1. Endocrine resistant (ER) disease was observed in 44.2% of patients; the remaining 55.8% were endocrine-sensitive (ES), including 60 patients with de novo metastatic disease. The most frequent sites of metastasis were bone (61.8%) and visceral tissues (45.2%), with the liver being the most affected organ (51.1%). Brain metastases accounted for 4%. Abemaciclib was primarily administered as first line (88.4%) or second line (10%) therapy. The most common combination agent was fulvestrant (51.3%), mainly in ER or subsequent lines, followed by letrozole (45.7%), among AIs. The median rwPFS in the overall population was 22.2 months (95% CI: 19.2-25.3) and 18.3 (95% CI: 14.9-22.5) and 26.3 (95% CI: 22.9-29.8) months for ER and ES, respectively. rwPFS rate at 24 months was 45.7% (overall), 34.0% (ER), and 61.2% (ES), respectively. The rwORR was 46.1% (95% CI: 38.6-53.7), with rwCBR of 68.5% (95% CI: 61.2-75.3). Abemaciclib was initiated at 150 mg BID in 92.5% of cases. Dose modifications occurred in 60.8% of patients, and 53.2% (n=106) required dose reductions. Patients who reduced their dose had a longer rwPFS of 26.3 (95% CI: 21.6-31.0) months. Proper management supports staying on treatment and leads to improved outcomes. Approximately 58.3% (n=116) permanently discontinued abemaciclib during the observation period, while 41.7% (n=83) remained on treatment at final observation (24 months). Among those who discontinued abemaciclib, 67.2% stopped the combination agent simultaneously. Most discontinuations were due to disease progression (37.7%), with AEs accounting for 12.6%. Notably, following dose reduction, only 8% of patients discontinued treatment due to AEs. The median rwTDT was 21.2 months (95% CI: 17.2-25.2). Conclusion: These real-word results are consistent with those from the MONARCH 2 and MONARCH 3 trials, confirming the effectiveness of abemaciclib in combination with ET for patients with HR+/HER2− ABC, across ER and ES settings. Two years after treatment initiation, 45.7% of patients remained progression-free and 41.7% continued therapy, indicating that abemaciclib is an effective and manageable therapeutic option for patients with ABC. Patients who reduced their dose had a numerically longer rwPFS, reinforcing the importance of appropriate use of patient management strategies to improve abemaciclib treatment persistence and clinical outcomes.
Presentation numberPS5-12-05
Cdk4/6 inhibitors in outpatient gynecological oncology: identification of implementation barriers and opportunities for improving their use in the practice
Mohamed Elessawy, UKSH, Kiel, Germany
M. Elessawy, N. Maass, M. Mackelenbergh, H. Le; UKSH, Kiel, Germany
Background. The adjuvant therapy of breast cancer patients with cyclin-dependent kinase 4/6 inhibitors (CDKI) has been demonstrated to be remarkably efficacious, resulting in a notable enhancement in progression-free survival for patients afflicted with early-stage high-risk and locally advanced metastatic breast cancer. A notable benefit of CDKIs is their oral administration, which facilitates outpatient use. Nonetheless, the bulk of management of CDKI therapy in Germany occurs within the confines of hospitals, effectively impeding the access to treatment for a significant proportion of affected patients. The objective of this study is to ascertain the current state of knowledge and barriers to implementation, and subsequently, to develop support services that will enable an increase in the use of this class of drugs in gynecological practices. Methods. In a regional, multi-phase, anonymized, cross-sectional study, the level of knowledge, application experience, and implementation barriers of CDKI were recorded using a structured questionnaire among gynecological oncologists working in private practice in northern Germany. The data was evaluated to develop a structured practice guideline. In a subsequent survey, the impact of this practice guideline on willingness to treat was examined. In a third phase, the efficacy of structured training measures was examined using pre-post workshop evaluation with a structured questionnaire to quantify knowledge gain, self-confidence, and willingness to implement. Results. The initial survey revealed a discrepancy between the high level of interest in the use of CDKI (rather high/high: n =5 9/78; 75.6 %) and the level of knowledge regarding the subject (rather low/low: n = 63/78; 80.8 %). Despite the high level of interest, 50.1 % (no/rather no: n = 37/73) of practicing gynecologists reported a lack of confidence on using the drugs. The predominant rationale cited for the inadequate usage was a dearth of expertise in therapy management (n = 42/66; 63.6 %). To support these efforts, 69.9 % (n = 51/73) of participants indicated a preference for tailored information material. In response, a practice guide was developed for the use of CDKI, encompassing therapy management, dose adjustment in case of side effects, and practical application instructions. the findings of this study indicate that 73.0 % of gynecologists in private practice (n = 27/73) expressed a high level of confidence in independently treating their patients with CDKI, as indicated by their responses to the guide. The workshop evaluation revealed an initial low level of competence (mean overall score of 2.31 on a 5.0 scale), yet it demonstrated substantial impacts of structured training. A notable increase in interest was reported by 58.3 % (n = 7/12) of the participants, 41.7 % (n = 5/12) reported feeling better prepared for practical applications, and the overall workshop satisfaction was 3.67 on a 5.0 scale. The competence deficit was particularly pronounced in dosage management (2.09/5.0) and indication (2.15.5.0). Conclusion. The utilization of CDKI for the long-term adjuvant therapy of breast cancer presents a significant challenge in gynecological practices and is, as a result, employed only in limited capacities, despite the robust interest it has garnered. Primarily, the dearth of experience in its implementations constitutes an impediment that can be surmounted through the provision of information materials tailored to the outpatient section. The implementation of structured training measures, in conjunction with practice-oriented guidelines, has been demonstrated to be an effective intervention for enhancing competence and can measurably reduce implementation barriers, thereby contributing to the enhancement of outpatient care quality.
Presentation numberPS5-04-18
Real-world patterns of trastuzumab deruxtecan discontinuation in metastatic breast cancer: insights from a single-center experience
Luca Licata, IRCCS San Raffaele Hospital, Milan, Italy
L. Licata, G. Viale, F. Patanè, A. Chiavassa, C. Zanibelli, B. Galbardi, M. Mariani, M. Piras, I. Persano, G. Notini, M. M. Naldini, C. Bosi, A. Rognone, S. Zambelli, L. Sica, D. Aldrighetti, P. Zucchinelli, G. Bianchini; IRCCS San Raffaele Hospital, Milan, ITALY.
BackgroundTrastuzumab deruxtecan (T-DXd) is an approved standard-of-care treatment for patients (pts) with both HER2-positive (HER2+) and HER2-low, previously treated metastatic breast cancer (MBC). Clinical trials have shown that a substantial proportion of pts (approximately 30–50%) discontinue treatment for reasons other than disease progression, most commonly due to adverse events (AEs) or patient choice. Early discontinuation may negatively impact overall treatment efficacy in clinical practice, and discontinuation rates may be influenced by physician and institutional experience with T-DXd management. This study aimed to evaluate the reasons for T-DXd discontinuation in a real-world setting at a single institution with established experience in T-DXd management. MethodsWe conducted a retrospective observational study using the electronic health records of pts with MBC who received T-DXd at San Raffaele Hospital, Milan, between July 2018 and September 2025. Only pts who permanently discontinued treatment for any reason were included in this analysis. Tumors were categorized as HER2+ (IHC 3+ or 2+ ISH amplified) or HER2-low (IHC 1+ or 2+ ISH not amplified) based on at least one biopsy prior to T-DXd initiation, or HER2-0 if all biopsies showed IHC 0. Reasons for discontinuation were classified as clinical or radiological progressive disease (PD; including death due to progression), AEs (including death due to AE), patient decision, or other causes. Duration of treatment (DoT) was defined as the time from T-DXd initiation to permanent discontinuation. Rates and reasons for dose reductions in this population were also analyzed. ResultsA total of 89 pts permanently discontinued T-DXd up to September 2025. Among them, 71.9% (n=64) had HER2+, 25.8% (n=23) HER2-low, and 2.3% (n=2) HER2-0 tumors. All pts were female, with a mean age of 56.9 years [range 24–85]. T-DXd was administered as 1st line therapy in 5.6% (n=5), 2nd line in 16.9% (n=15), 3rd line in 31.5% (n=28), and ≥4th line in 46.0% (n=41) of pts. Visceral metastases were present in 74.2% (n=66), and 29.2% (n=26) had brain metastases.The median DoT among pts who discontinued T-DXd was 7.1 months [range 0.7–81.4] overall: 9.6 months [0.7–81.4] in HER2+, 5.6 months [0.7–15.1] in HER2-low, and 3.9 months [2.6–5.1] in HER2-0 tumors. Reasons for discontinuation were PD in 82.0% (n=73), AEs in 14.6% (n=13), patient decision in 1.1% (n=1), and non–treatment-related causes in 2.2% (n=2; one COVID pneumonia and one polytrauma).AEs leading to discontinuation included ILD in 11.2% (n=10), fatigue (n=1), liver function test (LFT) increase (n=1), and persistent neutropenia (n=1). Among the 10 ILD cases, 4 were Grade 1, 1 Grade 2, 2 Grade 3, and 3 Grade 5. The three pts with Grade 5 ILD were managed for this AE at facilities near their homes, since they lived far from our center. In one case, a concomitant opportunistic aspergillus infection was documented. Dose reductions were implemented in 21.3% (n=19) of pts before permanent discontinuation, mainly due to fatigue (9.0%, n=8), LFT increase (6.7%, n=6), and nausea (4.5%, n=4). ConclusionsIn this real-world study, most pts permanently discontinued T-DXd due to disease progression, while AE-related discontinuations were primarily driven by ILD. Discontinuations due to other AEs were uncommon, and those due to patient decision were also rare and occurred less frequently than in clinical trials. These findings suggest that treatment experience, along with improved prevention, monitoring, and management of AEs, may enhance treatment adherence and reduce unnecessary discontinuations. Given the retrospective design of this study, it is not possible to establish the most effective management strategies.
Presentation numberPS5-12-12
Internet use, access to care, and breast cancer screening among publicly insured women
Hanan Jalal Mohammad, University of Texas San Antonio School of Public Health, San Antonio, TX
H. J. Mohammad, M. T. Halpern; University of Texas San Antonio School of Public Health, San Antonio, TX
Introduction: Biennial screening of breast cancer is crucial for early cancer detection. Although both Medicare and Medicaid provide coverage for breast cancer screening, screening rates among Medicaid recipients have been consistently lower. Internet use to seek out health information has been linked to increased health literacy and preventive care but findings are inconsistent. Internet use may represent a pathway to improve breast cancer screening rates, although little is known about its role among publicly insured women. Methods: This cross-sectional study used data from the 2023 National Health Interview Survey (NHIS), a nationally representative household survey of the non-institutionalized U.S population. Women within the recommended breast cancer screening age, 40-74 years old, were included in the study population. Women in this study population had either Medicaid, Medicare, or Medicare + Supplemental insurance coverage. Women with a history of breast cancer or without valid screening information were excluded from the study. The primary outcome was up to date screenings, defined as receiving either a mammogram or a breast MRI within the past two years. Primary exposures included internet access at home and using the internet to seek health information. Base model covariates were age, education level, and income level; healthcare access variables (having a usual place of care, time since last doctor visit, and time since last wellness visit) were included in a second model. Analyses were conducted in Stata 18. Survey-weighted logistic regression models were estimated using NHIS weights, strata, and primary sampling units to account for the survey design and produce results generalizable to the U.S. population. Results: In the base model, internet use to seek health information was positively but not significantly associated with being up-to-date on breast cancer screening (p = 0.14) while home internet access was not significantly associated (p = 0.77). Compared to Medicare coverage, Medicaid recipients had significantly lower odds of being up-to-date with breast cancer screenings (p = 0.005); Medicare + Supplemental coverage was not significantly different (p = 0.82). Higher education (bachelor’s and above) was associated with increased odds of screening (p = 0.02). Age and income were not significantly associated with being up-to-date. When healthcare access variables were added to the model, internet use to seek health information and home internet access remained nonsignificant (p = 0.22 and 0.27, respectively) and Medicaid coverage was no longer associated with decreased screening odds (p = 0.48). However, access to care variables were strong predictors of breast cancer screening. Having a usual place for care (p = 0.003), a doctor visit within the past year (p = 0.002), and a wellness visit within the past year (p < 0.001) were all significantly associated with being up-to-date on breast cancer screenings. Conclusion: Home internet access and the use of the internet to seek health information were not significantly associated with being up to date on breast cancer screenings among publicly insured women. Instead, differences in screening were strongly explained by healthcare access factors. While Medicaid recipients initially had lower odds of screening compared to Medicare recipients, this association was no longer significant after accounting for access to care. Having a usual source of care, a recent doctor visit, and a recent wellness visit were the strongest predictors of being up-to-date. Given these results, interventions to improve breast cancer screening among publicly insured women may be more effective when focused on improving sources of usual care and access to annual care visits for publicly insured women.
Presentation numberPS5-04-19
70-gene signature high risk classification provides stronger prognostic value than histologic grade in hr+ her2- early breast cancer
Erin F Cobain, University of Michigan Medical School, Ann Arbor, MI
E. F. Cobain1, L. Pusztai2, C. L. Graham3, P. D. Beitsch4, C. R. C. Osborne5, R. L. Rahman6, N. M. Johnson7, A. Brufsky8, R. L. Mahtani9, V. K. Gadi10, K. Hoskins11, H. M. Linden12, R. A. Mukhtar13, E. A. Brown14, L. P. Gold14, J. Alberty15, S. Benjamin16, S. Lee17, J. V. Pellicane18, H. Ramaswamy19, R. Weber19, N. Stivers19, A. Menicucci19, W. Audeh19, J. O’Shaughnessy20; 1University of Michigan Medical School, Ann Arbor, MI, 2Yale School of Medicine, New Haven, CT, 3Piedmont Cartersville Breast Surgical Oncology, Cartersville, GA, 4Dallas Surgical Group, Dallas, TX, 5Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX, 6Texas Tech University, Lubbock, TX, 7Legacy Medical Group, Portland, OR, 8University of Pittsburgh, Pittsburgh, PA, 9Miami Cancer Institute, Baptist Health, Plantation, FL, 10University of Illinois Chicago, Chicago, IL, 11University of Illinois College of Medicine Chicago, Chicago, IL, 12Fred Hutch Cancer Center, Seattle, WA, 13University of California San Francisco, San Francisco, CA, 14Comprehensive Breast Care, Troy, MI, 15SUNY Downstate Health Sciences University, Brooklyn, NY, 16Upstate University Hospital, Syracuse, NY, 17Highland Medical, Hematology and Oncology, Nyack, NY, 18Bon Secours, Richmond, VA, 19Agendia, Irvine, CA, 20Texas Oncology, Dallas, TX
Background:Pivotal trials of immunotherapy and CDK4/6 inhibitors in early-stage hormone-receptor positive (HR+) HER2 negative (HER2–) breast cancer (BC) relied on histologic grade 3 to define high-risk disease, despite its variability and limited prognostic accuracy. The 70-gene MammaPrint® (MP) signature provides a genomic recurrence risk assessment and guides treatment. Patients (pts) classified as MP High Risk are typically recommended chemotherapy (CT). We evaluated distant relapse-free survival (DRFS) in CT-treated HR+HER2– pts classified as MP High Risk, further stratified into High Risk 1 (H1) and High Risk 2 (H2). We also compared MP risk groups with clinicopathologic factors, especially histologic grade, to assess prognostic value.Methods:This is part of the prospective, observational FLEX Study (NCT03053193), including pts with stage I–III disease who underwent MP testing and consented to transcriptome and clinical data collection. Pts with HR+HER2– genomic High Risk BC who received CT and had follow-up data were included (n=1407). Kaplan–Meier (KM) survival analyses were performed, with median follow-up of 3.2 yrs. DRFS was the primary endpoint per STEEP 2.0. Cox proportional hazards models assessed how grade (G), tumor (T) size, lymph node (LN), and menopausal status compared to MP risk stratification.Results:Of 1407 pts, 74% had H1 and 26% H2 BC. Pts with H2 BC were younger (mean 55 vs 58 yrs, p=0.004), more often G3 (62% vs 24%), and had larger tumors, while LN and menopausal status were similar. KM analyses showed pts with H2 BC had worse DRFS at 5 yrs (86.4, 95% CI 82.4–90.6) vs H1 (93.1, 95% CI 91.1–95.2), a diff of 6.7% (p<0.001). Although MP H2 was associated with poorer prognosis, we did not stratify outcomes by CT regimen; prior data suggest anthracycline-based CT improves outcomes in pts with MP H2 BC. When stratifying G3 pts by High Risk subgroups, H2 remained associated with worse outcomes (8.4% diff at 5 yrs, p=0.0029), showing that MP refines risk beyond histologic grade. Cox analyses confirmed higher recurrence risk for H2 vs H1 (uni: HR 2.37, p<0.001; multi: HR 2.14, p=0.005); T size and LN were also significant, while grade lost significance after adjustment.Conclusions:In this real-world cohort of HR+HER2– BC pts treated with CT, MP H2 was associated with significantly worse DRFS vs H1, even after adjusting for clinicopathologic factors such as grade. Notably, grade lost independent prognostic value when corrected for MP, underscoring the limits of histologic features alone. These findings support MP H2 as a superior prognostic biomarker to grade, offering greater accuracy for tailoring therapy in HR+HER2– BC. In addition, MP H2 tumors were previously shown to have an immune active state, supporting its use as a biomarker to select pts for immunotherapy trials, such as SWOG 2206.
| – | All Pts (n=1407) | Pts with Grade 3 disease (n=526) | ||||
| MP Risk | 5-yr DRFS | 95% CI | p value | 5-yr DRFS | 95%% CI | p value |
| H1 | 93.1% | 91.1-95.2 | p<0.001 | 93% | 89.3-96.8 | p=0.0029 |
| H2 | 86.4% | 80.8-98.7 | 84.6% | 79.8-89.7 |
Presentation numberPS5-04-20
Clinicopathologic Features and Outcomes of Breast Cancer in Young Women Treated at a National Oncology Institute in the Dominican Republic
Millyant B Rojas, Instituto de Oncologia Dr. Heriberto Pieter, Santo Domingo Distrito Nacional, Dominican Republic
M. B. Rojas, Evelyn Ruiz, Cindy Jiménez, Angela Grano de Oro, Vilma Núñez, Rosa Vassallo; Instituto de Oncologia Dr. Heriberto Pieter, Santo Domingo Distrito Nacional, DOMINICAN REPUBLIC.
Abstract Background: Breast cancer in women ≤40 years represents a minority of cases but carries a disproportionate burden due to its biological aggressiveness, psychosocial impact, and economic consequences. Young women often present with advanced disease and triple-negative or HER2+ subtypes, associated with higher recurrence rates. Evidence from Latin America and the Caribbean is scarce, and reports from the Caribbean are virtually absent. Characterizing this population is essential to guide regional cancer control strategies and improve outcomes. Methods: We conducted a retrospective cohort study of women aged 18-40 years diagnosed with breast cancer between 2015 and 2020 at the Instituto de Oncología Dr. Heriberto Pieter, the national reference center in the Dominican Republic. Demographic, clinicopathologic, and therapeutic data were collected. Survival was calculated in years and stratified by stage and subtype. Kaplan-Meier methodology was used to estimate overall and disease-free survival. Between 2015 and 2020, 3,074 breast cancer patients were diagnosed, of whom 22% (n=675) were young women. After applying criteria, 283 patients were included. Results: A total of 283 patients were analyzed (median age 35 years). Most presented with advanced disease: stage IIIA 20.6%, IIIB 27.3%, IIIC 2.8%, IIB 28.7%; 12.8% had de novo stage IV. Subtypes were HR+/HER2- 61.7%, triple-negative 16.7%, and HER2+ 21.7%. Within HR+/HER2-, most tumors were Luminal B, consistent with the higher prevalence of Luminal B vs Luminal A in young women. Thus, over one-third corresponded to aggressive subtypes (triple-negative or HER2+). Treatment included mastectomy (85%), neoadjuvant chemotherapy (80%), endocrine therapy (68%), and radiotherapy (73%). Only 50% of HER2+ patients received targeted therapy, underscoring limited access. After a mean follow-up of 5.7 years, recurrence occurred in 35% of stage I-III patients, predominantly distant. Lung was the most frequent site (43%), followed by bone (32%), liver (25%), and CNS (13%). Mean overall survival declined by stage: 7.1 years in stage II, 5 years in stage III, and only 2.5 years in stage IV. Conclusions: In this large single-institution cohort, most young women were diagnosed at advanced stages, with aggressive subtypes in more than one-third of cases. Recurrence was frequent and mainly distant, with lung and bone as leading sites. Survival was markedly reduced in advanced stages, particularly stage IV. The gap between HER2+ prevalence and limited targeted therapy reflects restricted access to modern agents, likely contributing to poor outcomes. These findings represent one of the first comprehensive reports on breast cancer in young women from the Caribbean and highlight the urgent need for early detection, equitable access, and policies tailored to this vulnerable group. Keywords breast cancer, young women, Latin America, survival, clinicopathologic features
Presentation numberPS5-12-14
Between Hope and Hardship: Patient Perspectives on Treatment Cessation in Metastatic Breast Cancer in Syria
Ayla Kouli, Damascus University, Damascus, Syrian Arab Republic
A. Kouli1, A. Al-Bitar1, L. Mardini1, s. Almansour1, O. Johar1, M. Saifo1, F. Kalam2, M. Hafez3; 1Damascus University, Damascus, Syrian Arab Republic, 2Ohio State University Comprehensive Cancer Center, Columbus, OH, 3St. Luke’s University Health Network, Bethlehem, PA
Background: Treatment decision-making in metastatic breast cancer (MBC) is complex. In low-resource and conflict-affected settings, studies rarely address the psychological burden and personal preferences of patients that shape MBC treatment decisions. In Syria, women face unique barriers, including financial hardship, limited access to modern therapies, and psychosocial strain. No prior studies have examined how Syrian women with MBC make treatment cessation decisions amid conflict-related healthcare disruptions. We aimed to characterize patient perspectives on pausing vs. continuing therapy in MBC within a conflict-affected, low-resource setting.Methods: We conducted a cross-sectional mixed-methods survey between August 2025 and September 2025 on Syrian MBC patients. Quantitative measures assessed sociodemographic and clinical factors, as well as Likert scale ratings of attitudes and illness impact. Qualitative responses were thematically analyzed and quantified to identify dominant concerns and support needs.Results: This study included 60 Syrian women with a confirmed diagnosis of MBC. Participants had a mean age of 48.2 years, most were married (75%), and were from low-income households (68.3%). Over 70% had lived with breast cancer >5 years. Hormonal therapy was the most common treatment (38.3%), while chemotherapy (5.0%), radiotherapy (8.3%), and immunotherapy/targeted agents (8.3%) were less accessible. Illness burden was high (Mean daily life impact score 6.9/10). Systemic barriers were striking: 40% reported incurring transport costs and financial burdens, and 17% noted a deterioration in medical services. 66.7% feared their cancer would progress if treatment were paused, and 75% cited treatment toxicity or cost as reasons to consider a break. 70% expressed a need for better psychological support and communication from physicians.Conclusion: Syrian women with MBC navigate the dual burden of treatment toxicity and collapse of the healthcare system, compounded by overlooked psychosocial needs. Most patients were unwilling to discontinue therapy despite hardships, a decision that might be influenced by the perception that access to treatment in Syria is a luxury and a privilege. Addressing psychological support, patient preferences, and equitable treatment access is essential to optimizing care in conflict-affected settings
Presentation numberPS5-04-21
Impact of Germline BCRA variants in the neoadjuvant treatment
Natalia Valdiviezo, Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
N. Valdiviezo1, I. Otoya1, P. Mora2, J. Herzog3, L. Reynaga3, S. Neciosup1, C. Calle1, S. Casavilca4, K. Roque1, Z. Morante1, H. Fuentes1, C. Castañeda1, T. Vidaurre1, V. Acuña5, M. Falla6, J. Galarreta6, S. Gruber3; 1Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 2Unidad Funcional de genética y biología molecular, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 3City of Hope National Medical Center, Duarte, CA, 4Equipo Funcional de Patología Quirúrgica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 5Universidad Nacional Mayor de San Marcos, Lima, Peru, 6Cirugía de Mamas y Tejidos Blandos, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
Background: Carriers of pathogenic BRCA1 and BRCA2 variants often develop breast cancers with distinct pathological features, frequently associated with more aggressive phenotypes such as triple-negative tumors. Previous studies have reported greater benefit from neoadjuvant chemotherapy and higher rates of pathological complete response in this population. Objective: To evaluate the response to neoadjuvant therapy and survival outcomes in breast cancer patients with germline BRCA1/2 variants compared with those with wild-type phenotypes. Methods: Women ≤45 years with breast cancer were included in the hereditary cancer protocol at INEN between 2013 and 2020. Genetic testing was performed at the City of Hope (COH). We selected patients who had received neoadjuvant treatment. Results: A total of 105 patients were analyzed, including 45 with germline BRCA1/2 variants and 60 wild-type. Most patients received standard chemotherapy with anthracyclines and weekly taxanes (85%), while only a minority (12%) had access to carboplatin due to policy and approval restrictions. The median age was 40 years and comparable between groups, as all participants belonged to the hereditary breast cancer screening program. No significant differences were observed in pathological complete response (pCR) between BRCA variant carriers and wild-type patients, although a correlation was noted between pCR and the triple-negative subtype (p=0.053). In terms of survival, BRCA1 carriers showed poorer outcomes, with a hazard ratio of 2.5 and a median overall survival of 6.9 years, while BRCA2 carriers had a median survival of 11.6 years, comparable to wild-type patients whose median survival has not yet been reached. It is likely that the suboptimal pCR rates in BRCA mutation carriers were influenced by the limited use of carboplatin, a drug shown in other studies to significantly improve response rates. Conclusions: In this cohort of 105 breast cancer patients, BRCA1 carriers had worse overall survival (median 6.9 years, HR 2.5) compared with BRCA2 and wild-type patients, despite similar pCR rates. Pathological complete response was mainly associated with the triple-negative subtype. Limited access to carboplatin likely contributed to suboptimal responses in BRCA mutation carriers, underscoring the need for broader availability of platinum-based therapy.
Presentation numberPS5-12-15
Outdoor ambient air pollution and breast cancer survival among Chinese patients: a multi-center analysis of 18553cases
Yuzhu Zhang, Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University,, Taizhou, China
Y. Zhang1, Y. Yuan2, W. Huang3, W. Wang2, Y. Kong2, Z. Hu1, Y. Pan1, Z. Zheng1, W. Liang1, C. Zheng1, L. Roche4, Z. Zheng1, X. Chen5, M. Peng6, Y. Guo3, C. Zhang2; 1Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University,, Taizhou, CHINA, 2School of artificial intelligence, Taizhou University, Taizhou, CHINA, 3Climate, Air Quality Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, AUSTRALIA, 4University Hospital for Gynecology, Pius Hospital, University Medicine, Oldenburg, GERMANY, 5Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, CHINA, 6Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA.
Background: Ambient fine particulate matter (PM2.5) is a Group 1 carcinogen associated with lung cancer, but evidence on its role in breast cancer prognosis remains limited.Although epidemiological studies suggest associations between PM2.5 exposure and breast cancer incidence, particularly for aggressive subtypes, few studies have evaluated its impact on survival outcomes, including progression-free survival (PFS) and overall survival (OS).Methods:In this prospective study we analyzed data from 18,553 breast cancer patients (2000-2019) sourced from two large-scale clinical databases, which included more than 40 medical centers in China. Long-term PM2.5 exposure was assessed using high-resolution spatial-temporal modeling (1 km resolution, R²=0.92). Cox proportional hazards models were used to evaluate associations between PM2.5 exposure and survival, adjusting for demographic characteristics, Clinical factors, and Socioeconomic status. Stratified analyses were conducted based on Molecular subtype, Age and Tumor characteristics.Results: Higher PM2.5 exposure was significantly associated with worse PFS and OS in dose-dependent manners. Each 10 μg/m³ increase in PM2.5 was associated with a 28% higher PFS hazard (HR=1.28, 95% CI: 1.20-1.37) and 47% higher OS hazard (HR=1.47, 95% CI: 1.33-1.63) in fully adjusted models. Kaplan-Meier curves showed widening survival gaps over time, with absolute differences in PFS rates between the highest and lowest exposure quartiles increasing from 8.4% at 5 years to 10.3 at 13 years. Subgroup analyses indicated strong associations among HER2-positive (PFS HR=2.13, OS HR=4.03) and triple-negative breast cancer (TNBC; PFS HR=2.53, OS HR=4.03) subtypes, as well as among older patients (>40 years) and those with high Ki-67 expression.Conclusion: This study is the first to demonstrate that long-term PM2.5 exposure is independently associated with poor prognosis in breast cancer, particularly for HER2-positive and TNBC subtypes. The findings highlight PM2.5 as a modifiable risk factor and emphasize the need to integrate environmental exposure into clinical risk assessment and public health strategies to mitigate breast cancer disparities.
Presentation numberPS5-12-26
Yoga in Breast Cancer Survivorship: Knowledge, Attitudes, <Perceived Benefits.
Prasenjit AH Chatterjee, MANIPAL HOSPITAL, KOLKATA, India
P. A. R. B. Chatterjee; MANIPAL HOSPITAL, KOLKATA, India
Aim: This study aimed to explore the knowledge, attitudes, and perceived effects of yoga among breast cancer survivors undergoing follow-up care. Methods: A single-arm, survey-based, retrospective study was conducted involving 35 breast cancer survivors at the Outpatient Department (OPD) of Manipal Hospital, Dhakuria. Eligible participants included women aged 18-70 years who had completed all treatment modalities, including surgery, radiotherapy, and systemic therapy, and were practicing yoga as part of their rehabilitation. A 20-point survey assessing knowledge, attitudes, and perceived effects of yoga was administered. Data collection spanned from January to August 2024, and statistical tools were applied for analysis. Results: Of the 35 participants, 32 (91.4%) were aware of yoga before starting therapy, and 85.7% recognized it as an accepted form of alternative therapy. Most participants (57.1%) practiced yoga daily, with 62.9% engaging for 15-30 minutes per session. Thirty participants completed six months of yoga during the study period and were included in the symptom assessment. Positive outcomes were reported by 28 (94.3%) participants, including improvements in physical and mental health, with 54.3% noticing benefits after three months of practice. Mild side effects were noted by 11.4% of participants, including body aches and chest wall muscle cramps. A significant reduction in symptoms such as breast/chest wall pain, lymphedema, fatigue, decreased appetite, mood, and sleep disturbances was observed. Conclusions: This study highlights yoga as a safe, accessible, and effective adjunct therapy for breast cancer survivors, with reported improvements in both physical and psychological well- being. High adherence and positive perceptions suggest its integration into survivorship care is both feasible and valuable. While limitations exist, these findings support further research into yoga-based rehabilitation as part of holistic cancer care strategies.
Presentation numberPS5-12-27
The impact of immediate contralateral breast symmetrization on body satisfaction among breast cancer survivors.
Andrea Cavalheiro Cubero, Department of Mastology, ABC School of Medicine, Santo André, Brazil
A. C. Cubero1, D. I. Cubero2, C. C. Mazzei3, F. C. Sandoval3, J. J. Romano3, I. T. Sant’Anna3, L. A. Sousa4; 1Department of Mastology, ABC School of Medicine, Santo André, Brazil, 2Department of Oncology and Hematology, ABC School of Medicine, Santo Andre, Brazil, Santo André, Brazil, 3ABC School of Medicine, Santo André, Brazil, 4Laboratory of Epidemiology and Data Analysis, ABC School of Medicine, Santo André, Brazil
Introduction: Contralateral breast symmetrization by reduction mammaplasy has been incorporated into oncoplastic surgery as a way of improving aesthetic outcomes and body image perceptions among women undergoing breast-conserving treatment for breast cancer. However, the actual impact of this approach to contribute effectively to women’s aesthetic satisfaction and self-image remains unclear. Objective: To evaluate whether immediate contralateral breast symmetrization is associated with greater satisfaction with self-image in women treated with breast-conserving surgery for breast cancer. Method: This cross-sectional study employed an approach involving retrospective clinical- epidemiological data collection and face-to-face application of the Body Dysmorphic Disorder Examination (BDDE) questionnaire, which has been validated in Portuguese. All consecutive women who underwent breast-conserving surgery either with or without contralateral symmetrization, performed by the same surgical team between 2008 and 2017 in a public hospital in Brazil, were included. The participants were divided into two groups: the control group (i.e., the CG, which included participants who underwent only breast-conserving surgery) and the study group (i.e., the SG, which included participants who underwent breast-conserving surgery with immediate contralateral symmetrization). The BDDE scores and aesthetic perceptions of participants in these two groups were compared after 12 months from the end of oncological treatment. Results: A total of 57 patients were evaluated, including 22 in the CG and 35 in the SG. Most participants in both groups (14 in the CG and 20 in the SG) reported satisfaction with the appearance of their breasts in the postoperative period, and no statistically significant differences were observed in this context. Only one patient obtained a score that suggested the presence of body dysmorphic disorder. Conclusion: Immediate contralateral symmetrization was not associated with increased body satisfaction among participants in this research. These findings suggest that this approach should be applied in a judicious and individualized manner; in addition, further studies are needed to better define its impact on patients’ quality of life more effectively.
Presentation numberPS5-12-28
Human Development Index and Breast Cancer Prognosis in Chinese Women: A Multiregional Survival Analysis
Wanting Wang, School of artificial intelligence, Taizhou University, Taizhou, China
W. Wang1, Y. Yuan1, A. Yuan2, Z. Hu2, C. Zheng2, Z. Zheng2, Y. Pan2, W. Liang2, B. Chen2, C. Zhang1, Y. Zhang2; 1School of artificial intelligence, Taizhou University, Taizhou, CHINA, 2Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, CHINA.
Background:Breast cancer is the most commonly diagnosed cancer among women in China and globally. Significant regional disparities in survival outcomes have been observed, potentially influenced by socioeconomic development. The Human Development Index (HDI) is a composite measure of health, education, and income that may reflect healthcare access and quality, yet its association with breast cancer prognosis remains unclear in China.Objective:To evaluate the association between regional HDI and survival outcomes in Chinese women with breast cancer, and to quantify the impact of incremental HDI improvement on prognosis.Methods:A multicenter retrospective cohort study included 25,514 patients from 40 cancer centers across 184 prefecture-level cities in China. Regional HDI values were extracted from the China Human Development Report Special Edition. The primary outcomes were overall survival (OS) and progression-free survival (PFS). Survival curves were estimated by using the Kaplan-Meier . Multivariable Cox proportional hazards models were used to calculate hazard ratios (HRs) and 95% confidence intervals (CI), adjusting for tumor stage, molecular subtype, Ki67 status, age, and pathological type. HDI was analyzed per 0.05-unit increase, corresponding to one standard deviation.Results:Patients in high-HDI regions had significantly better PFS and OS than they in low-HDI. 4-year PFS was 92.4% versus 88.4%, 8-year PFS 86.2% versus 72.6%, and 12-year PFS 83.2% versus 57.9% (log-rank P < 0.001). And 4-year OS was 97.0% versus 93.4%, 8-year OS 94.6% versus 87.0%, and 12-year OS 93.2% versus 84.5% (P < 0.001). After adjustments, each 0.05-unit increase in HDI was associated with a 14% reduction in mortality risk (HR = 0.861, 95% CI: 0.807-0.919; P < 0.001), and the similar reduction was observed for recurrence risk (HR = 0.884, 95% CI: 0.847-0.923; P < 0.001). The association remained consistent across molecular subtypes. We provide the first quantitative evidence of the independent association between HDI and both OS and PFS in a national breast cancer cohort.Conclusions:Higher regional HDI is independently associated with improved survival in Chinese women with breast cancer. Each 0.05-unit increase in HDI confers a clinically meaningful survival benefit. These findings underscore the role of socioeconomic development in cancer outcomes and support targeted interventions to reduce regional disparities in care access and quality.
Presentation numberPS5-12-29
Fertility Outcomes In Young Breast Cancer Patients And Their Prognosis And Offspring Health
Anqi Yuan, Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, China, Taizhou, China
A. Yuan1, Z. Hu1, C. Zheng1, Z. Zheng1, W. Liang1, Y. Yuan2, W. Wang2, C. Zhang2, Y. Pan1, Y. Zhang1; 1Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, China, Taizhou, CHINA, 2School of artificial intelligence, Taizhou University, Taizhou, CHINA.
Background: Most young breast cancer patients (≤40 years old) are in the golden period of fertility, but chemotherapy is prone to premature ovarian failure, and radiotherapy and long-term endocrine therapy further compress the fertility window. In addition, in response to the current controversy over fertility behavior on patient prognosis and the research gap in the long-term health of offspring with anti-tumor therapy, we designed this retrospective cohort study to clarify the fertility status of young breast cancer patients, verify the independent impact of fertility behavior on the prognosis of patients with different molecular types, and evaluate the association between treatment regimens and offspring health. Methods: This was a multicenter retrospective study on fertility outcomes and their prognosis and offspring health in young breast cancer patients. Based on the breast cancer database, we registered the baseline data of young breast cancer patients (≤40 years old), including age at initial diagnosis, molecular typing, tumor stage, treatment plan, fertility protection measures, etc., and followed up by telephone/email and other means to collect their fertility status, recurrence and offspring growth and development data. The primary endpoints were the disease-free survival (DFS), overall survival (OS) after childbirth and childrens’ growth and development. The secondary endpoints were the patient’s fertility live birth rate, pregnancy outcomes and complications, use of fertility preservation techniques, breastfeeding status, etc. We used the Kaplan-Meier method to draw the survival curve of the patients after childbirth, and analyzed the growth trajectory of the offspring born after diagnosis by the Lambda-Mu-Sigma method, and plotted the growth and development curve.We used Log-rank to test the significance of differences in pregnancy rates between variables. We used Univariate Cox regression analysis to screen factors related to prognosis, and then used multivariate Cox regression analysis to adjust for potential confounding factors to clarify independent prognostic indicators of disease. Finally, descriptive analyses were performed for other secondary outcomes.
Presentation numberPS5-12-30
Suicidal Ideation and the Role of Social Support Among Breast Cancer Patients in Syria
Ahmad Al-Bitar, Damascus University, Damascus, Syrian Arab Republic
A. Al-Bitar1, M. Doyya1, N. Battah1, S. Achkar1, F. Nahhat2, M. Saifo1; 1Damascus University, Damascus, Syrian Arab Republic, 2Mayo Clinic, Phoenix, AZ
Background: The diagnosis and treatment of breast cancer impose a significant psychological burden, which can include suicidal ideation. However, data from conflict-affected regions like Syria are scarce. This study aimed to determine the prevalence of suicidal thoughts and identify associated risk factors among Syrian patients with breast cancer. Methodology: An institution-based cross-sectional study was conducted at Al-Biruni University Hospital in Damascus, Syria. Between April and August 2025, a total of 319 breast cancer patients undergoing chemotherapy were recruited. Data were collected through structured face-to-face interviews and reviews of medical records. Standardized instruments included the Patient Health Questionnaire-2 (PHQ-2), the Multidimensional Scale of Perceived Social Support (MSPSS), and the Columbia-Suicide Severity Rating Scale (C-SSRS). Statistical analyses were used to identify factors associated with suicidal ideation. Results: The overall prevalence of suicidal ideation was 8.2%. Suicidal thoughts were significantly associated with depression (p=0.0014), anxiety (p<0.001), and low perceived family support (p=0.0293). Key clinical predictors included advanced disease stage (p=0.0275), metastatic disease (p=0.0251), and severe pain (p=0.0011). A higher prevalence was also noted in patients who had not undergone surgery (p=0.0057). Significant sociodemographic factors included being divorced (p=0.0117) and residency location (p=0.0039). Among those experiencing suicidal ideation, a small but significant portion progressed toward action, with approximately 5.2% to 5.6% reporting they had worked out details of a plan or had some intention of acting on their thoughts. Regarding deterrents, religious beliefs were the most common reason cited for not attempting suicide (58%), followed by personal reasons (46%). Conclusion: Suicidal ideation is a significant concern among Syrian breast cancer patients, strongly associated with psychological distress, advanced disease, severe pain, and specific sociodemographic vulnerabilities. These findings highlight the critical need to integrate mental health screening and targeted psychosocial support into routine oncological care, especially for high-risk individuals in this vulnerable population.
Presentation numberPS5-04-22
Interventions to Improve Hope and Psychosocial Outcomes in Patients with Breast Cancer: A Systematic Review
Rachel L Hoover, Albany Medical College, Albany, NY
R. L. Hoover1, A. S. Kaminsky1, A. Trees1, B. Suarez1, P. Shah2, C. Carlton1, K. Tepper2, B. Roberts3, C. L. Grimes1, M. Cohen1; 1Albany Medical College, Albany, NY, 2New York Medical College, Valhalla, NY, 3University of Chicago, Chicago, IL.
Title: Interventions to Improve Hope and Psychosocial Outcomes in Patients with Breast Cancer: A Systematic Review Authors: Rachel Hoover, Abigail S Kaminsky, Alanna Trees, Brittany Suarez, Pal Shah, Caitlin Carlton, Katharine Tepper, Brittany Roberts, Cara L. Grimes, Michael Cohen Abstract:Introduction: Breast cancer is the most frequently diagnosed malignancy in women worldwide and continues to be a significant cause of cancer-related morbidity. Beyond the physical burden of disease and treatment, patients can experience psychosocial distress that affects their overall well-being. Hope, defined as the capacity to maintain goal-directed thinking and motivation in the face of adversity, has the potential to be a strong protective factor in oncological care. Evidence suggests that higher levels of hope are associated with improved psychological adjustment, greater quality of life, and enhanced coping during cancer care. Among patients with breast malignancies, interventions that aim to increase levels of hope may reduce distress and improve psychosocial outcomes such as anxiety, depression, and perceived treatment efficacy. Objective: To systematically review interventions designed to improve hope in adults with breast cancer. Methods: Major databases including Scopus, PubMed, Embase, and CENTRAL were searched through July 2025. Inclusion criteria were adults with breast cancer undergoing an intervention designed to improve hope. Outcomes included hope measured by a validated hope scale, anxiety, depression, and treatment perceptions. English language randomized controlled trials, prospective or retrospective comparative studies, single-arm pre-post intervention studies were included. Abstracts and full texts were doubly screened and included manuscripts extracted and summarized. Risk of bias assessment was performed using ROB2 and ROBINS-I. Results: 42,111 abstracts screened, 61 full texts screened, and 39 studies included representing 2412 patients with breast cancer across a variety of geographical locations. Hope interventions included nursing-based crisis intervention and psychological interventions. The meta-analysis shows a significant overall effect of hope interventions on Snyder Hope Scale scores (Hedges’ g = -1.63, 95% CI -2.24 to -1.01, p < 0.001). Both intervention and control groups had changes, but the intervention group improved significantly more (group difference test, p = 0.05). Across studies, outcomes consistently demonstrated improvements in hope levels for patients undergoing hope-based interventions.
Presentation numberPS5-04-23
Patient priorities for breast cancer survivorship: a preliminary descriptive analysis of the health outcome goals that matter most to older adults breast cancer survivors
Basak Basbayraktar, University of Texas Health Science Center, Houston, TX
D. E. Giza1, A. Gonzalez1, I. Doostan1, B. Basbayraktar1, A. Mera2, G. Yalavarthy3, V. Kaklamani4, J. A. Barrera2, M. Karuturi5, S. Ghosh1, H. Holmes1, A. Naik1; 1University of Texas Health Science Center, Houston, TX, 2Institute of Aging, Houston, TX, 3Institute, Houston, TX, 4University of Texas San Antonio, Houston, TX, 5MD Anderson Cancer Center, Houston, TX.
Introduction: Older adult breast cancer survivors have an average of five chronic conditions, and frequently report worse physical functioning, greater fatigue, and emotional distress, which hinders their ability to perform activities of daily living and achieve whole-person, often function-oriented goals during survivorship care. The Patient Priorities Care (PPC) framework was initially developed on a cohort of older adult cancer survivors and offers a structured approach to: 1) elicit patients’ goals, and 2) align survivorship care with what matters most. Despite evidence that reduces treatment burden in patients with multiple chronic conditions, its application in breast cancer survivorship remains underexplored. Objective: To describe older adults’ goals for breast cancer survivorship care based on preliminary findings from a quality improvement initiative aimed at identifying older adult cancer survivors’ care priorities and aligning survivorship care using the Patient Priorities Care (PPC) framework. Methods: We conducted a randomized quality improvement project focused on older adults (≥65 years) with early stage breast cancer, post active cancer therapy (3 months after curative treatment and up to 10 years in their survivorship care) with evidence of burdensome care (having ≥ 3 comorbidities or taking ≥10 medications or seeing more than ≥specialists per year or having ≥ 2 emergency department/hospital visits per year). Using the PPC framework, with the help of a trained facilitator, the participants identified their care preferences and health outcomes goals, as well as barriers (symptoms, medical problems) that were in the way of achieving their goals. Interviews were conducted via telephone or the Teams online platform. A summary of the health priorities identification encounter was provided to the oncology team to use for further care alignment. Demographic data was gathered at baseline and evaluated through frequentist statistical methods. Goal attainment scaling was done at baseline and 3 months following the National Committee for Quality Assurance (NCQA) guidelines. Results: Since January 2025, a total of 32 patients have been enrolled, with a mean age of 73 years. The majority of participants (40%) were diagnosed with stage I breast cancer, followed by ductal carcinoma in situ (18%), stage II (31%), and stage III (9.3%). Regarding treatment received, 97% of patients underwent surgery, 81% received radiation therapy, and 78% received systemic treatment, including hormonal therapy (37.5%), neoadjuvant therapy (6%), and 6% adjuvant therapy (6%). Sixty percent of the patients reported that functional status and maintaining independence were their priority for survivorship care, leading participants to establish goals centered around these themes. Factors influencing goal-setting included physical function, social support, and the presence of chronic comorbidities. Among the 10 patients who reached the 3-month follow-up time point, 9 patients had successfully achieved their stated goal within that timeframe. Conclusions: Preliminary findings suggest that older adult cancer survivors value survivorship care that supports functional goals. The PPC framework may be a useful tool to guide individualized breast cancer survivorship planning and promote goal-concordant care. Future work should explore how these priorities evolve over time and influence decision-making. This project is supported through a K12 NIH Institutional Grant (5K12TR004908-02) and by NIA through a GEMSSTAR (1R03AG089057-01).
Presentation numberPS5-04-25
A phase II trial of platelet-rich plasma for breast cancer patients with genitourinary syndrome of menopause
Melissa Curran, University of Miami Miller School of Medicine, Miami, FL
M. Curran1, T. Wolde2, J. Firdman Moore3, G. Sierra4, N. Paez4, L. Pla4, A. Van Mossel5, K. Rojas2; 1University of Miami Miller School of Medicine, Miami, FL, 2Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 3Department of Obstetrics, Gynecologic & Reproductive Sciences, University of Miami, Miami, FL, 4Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 5Behavioral and Community-Based Research Shared Resource, Sylvester Comprehensive Cancer Center, Miami, FL.
Introduction: Genitourinary syndrome of menopause (GSM) is a common yet underrecognized effect of endocrine therapy leading to sexual dysfunction in breast cancer survivors. Vaginal hormones are the most effective treatment but are infrequently prescribed due to concerns regarding systemic absorption. Platelet-rich plasma (PRP) is derived from a patient’s blood and centrifuged to remove erythrocytes, and the concentrated growth factors promote tissue repair, angiogenesis, and collagen synthesis. It is unknown whether PRP, created and applied in a standardized approach, could improve vulvovaginal symptoms in patients on estrogen suppressive therapies. This phase II trial sought to determine whether 2 vulvovaginal PRP treatments could constitute a novel GSM therapeutic for breast cancer patients with GSM. Methods: A prospective phase II pilot trial included female breast cancer patients with GSM treated with two sessions of PRP one month apart. 4cc of PRP was created from 30cc of whole blood collected by venipuncture using the Magellan Autologous Platelet Separator System (Isto Biologics, Hopkinton, MA). PRP was injected into the posterior vagina, vestibule, and posterior fourchette. The primary outcome was a change in patient-reported GSM symptoms using the validated Vulvovaginal Symptom Questionnaire (VSQ), compared at baseline (T0) and one month after the treatment #2 (T1). Secondary outcomes included vaginal health index (VHI) collected on exam, as well as patient-reported sexual function (Female Sexual Function Index: FSFI) and sexual distress (Female Sexual Distress Scale-Revised: FSDS-R). Lower VSQ and FSDS correspond to less GSM symptoms and lower sexual distress, respectively, whereas higher VHI and FSFI correspond to less atrophy and greater sexual function. Exploratory analyses included outcome assessment at 6 months (T2). Paired t-tests compared mean score differences between timepoints. Results: 18 breast cancer patients were enrolled (2023-2025), 17 received 2 treatments. Median age was 51.5 years (38-66), 89% received chemotherapy, and all patients were on endocrine therapy during the trial. Mean VSQ, FSDS-R, and VHI significantly improved from T0 to T1 (Table 1), and remained significantly improved at T2 (n=11). Of 7 sexually inactive patients at enrollment, 5 resumed sexual activity during the study. Post-treatment pain scores were low, and one patient developed post-treatment itching requiring an antihistamine and fluconazole. Conclusion: Two treatments of vulvovaginal PRP improved breast cancer patient-reported vulvar symptoms and lowered sexual distress when applied in a standard application, and the improvements appear to be durable for 6 months after completing treatment. Funding: SCCC Cancer Survivorship Research Pilot Award (CC-CSRP-2023)
| Baseline | T1 (primary endpoint) | T2 | p-value | |
| Mean VSQ (SD) | 10.53 (2.72) | 3.87 (3.85) | 4.70 (5.03) | 0.003* |
| Mean FSFI (SD) | 9.84 (5.72) | 15.53 (9.66) | 14.38 (11.02) | 0.502 |
| Mean FSDS-R (SD) | 36.59 (14.66) | 26.13 (13.74) | 24.82 (11.64) | <0.001* |
| Mean VHI (SD) | 13.06 (3.75) | 17.63 (3.05) | 18.55 (4.08) | 0.034* |
| VSQ: Vulvovaginal Symptom Questionnaire; FSFI: Female Sexual Function Index, FSDS-R: Female Sexual Distress Scale (revised); VHI: Vaginal Health Index; * denotes significant p-values from baseline to primary endpoint (T1). |
Presentation numberPS5-04-26
Tailored Support for Young Women with Breast Cancer: Survey-Informed Program Development and Peer-Based Intervention
Shari Goldfarb, MSKCC, New York City, NY
S. Goldfarb1, C. Chang1, G. Mary2, T. Nicolas1, M. Ahmed1, A. Jamner1; 1Breast Medicine Service, MSKCC, New York City, NY, 2Breast Medicine Service, NYU, New York City, NY.
Background Low self-esteem and worse body image are common psychosocial challenges among young women with breast cancer. These issues are often exacerbated by treatment-related changes in physical appearance, such as alopecia, surgical scars, and weight fluctuations, which can impact identity, social interactions, sexual health, and overall QoL. Younger patients face additional challenges in maintaining peer and partner relationships, since treatment during young adulthood can feel isolating and disrupt a sense of normalcy. Emerging evidence supports integrating aesthetic and wellness-based therapies to improve body image and self-esteem during and after treatment among breast cancer survivors. Method Memorial Sloan Kettering Cancer Center’s (MSKCC) Young Women with Breast Cancer Program (YWBCP) launched in 2023 to address the unique needs of women diagnosed at age 45 or younger. To inform program development, YWBCP conducted online surveys at intake (n=1195) and six months into treatment (n=402). Surveys assessed psychological well-being, sexual health, body image, and preferences for psychosocial services. Paired responses were analyzed to evaluate changes over time and identify evolving patient needs. Survey findings also guided the design of a targeted wellness intervention. Results Paired survey data (n=328) revealed that 46% (n=147) of patients reported feeling less positive about their bodies six months after diagnosis, while 50% (n=161) expressed reduced appreciation for their body’s uniqueness. In response to the statement “I act as though I like my body,” 36% (n=115) reported a decline from baseline to six months. Patients’ responses shows that interest in sexual activity decreased (34%; n=107) more than increased (24%; n=75) over time. 55% (n=169) reported a decline in sexual satisfaction. At intake, patients most frequently prioritized individual therapy (23%, n=326), support groups (21%, n=301), and family support (19%, n=265). By six months, these preferences remained relatively consistent, with individual therapy (28%, n=86) and support groups (19%, n=59) still ranking highest, followed by increased interest in educational programs (14%, n=45). In response to survey findings, MSKCC hosted an in-person wellness and beauty event on June 27, 2025. The event featured therapeutic beauty services, educational workshops, group wellness activities, and a facilitated lunch with social work, fertility, sexual health and Integrative Medicine clinicians. Qualitative feedback was overwhelmingly positive. One participant shared, “The event was a deeply inspiring and empowering experience. It created a safe, supportive space for us women to connect with one another. I think it was a fantastic way to focus on self-care, confidence, and community.” Another noted, “Being surrounded by other young women going through the same thing reminded me I’m not alone.” A third reflected, “It made something that can feel extremely isolating a lot less lonely. I left the day with a bunch of new connections that can help guide and support me through my treatment.” She added, “The workshops helped me feel a lot more comfortable with the challenges I’m facing and more normal for what I’m going through.” Many attendees reported a strengthened sense of community, self-confidence, and body image. Quantitative evaluation is ongoing and will be available to present in December at the San Antonio Breast Cancer Symposium. Conclusion Survey data and patient feedback highlight the need for age-specific psychosocial programming that addresses evolving concerns around identity, body image, and emotional support. Integrating beauty and wellness services into psychosocial care may offer a meaningful way to enhance self-esteem, reduce isolation, and increase engagement with hospital-based supportive services.
Presentation numberPS5-04-27
Quality of life in patients with advanced breast cancer (aBC) treated with ribociclib (RIB) in the real-world RIBANNA trial
Peter A Fasching, University Hospital Erlangen, Erlangen, Germany
P. A. Fasching1, C. Brucker2, T. Decker3, A. Engel4, T. Göhler5, C. Jackisch6, J. Janssen7, A. Köhler8, K. Lüdtke-Heckenkamp9, D. Lüftner10, F. Marmé11, M. van Mackelenbergh12, B. Rautenberg13, R. Weide14, P. Wimberger15, E. Kisseleff16, C. Pfister16, C. Schneider-Walter16, A. Wöckel17, M. Schmidt18; 1University Hospital Erlangen, Erlangen, Germany, 2Paracelsus Medical University, Nuremberg, Germany, 3Oncology Ravensburg, Ravensburg, Germany, 4Winicker Norimed GmbH, Nuremberg, Germany, 5Oncology Center Dresden/Freiberg, Dresden, Germany, 6Evangelische Kliniken Essen-Mitte gGmbH (KEM), Essen, Germany, 7Medizinische Studiengesellschaft Nord-West GmbH, Westerstede, Germany, 8Practice for Hematology and Oncology, Langen, Germany, 9Niels-Stensen-Kliniken, Georgsmarienhütte, Germany, 10Medical University of Brandenburg Theodor Fontane, Rüdersdorf bei Berlin, Germany, 11Med. Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany, 12Universitätsklinikum Schleswig-Holstein, Kiel, Germany, 13Universitätsklinikum Freiburg, Freiberg, Germany, 14Institut für Versorgungsforschung in der Onkologie, Koblenz, Germany, 15Nationales Centrum für Tumorerkrankungen (NCT) Dresden, Dresden, Germany, 16Novartis Pharma GmbH, Nuremberg, Germany, 17University Hospital Würzburg, Würzburg, Germany, 18Klinik und Poliklinik für Geburtshilfe und Frauengesundheit, Mainz, Germany
Background: In addition to disease control, maintaining a high health-related quality of life (QoL) is a major treatment goal for patients (pts) with advanced breast cancer (aBC). In the pivotal MONALEESA (ML) trials, the CDK4/6 inhibitor ribociclib (RIB) in combination with an aromatase inhibitor (AI) or fulvestrant (F) demonstrated significant improvement of survival in pre- and postmenopausal pts with hormone receptor-positive (HR+), human epidermal growth factor receptor 2-negative (HER2−) aBC while maintaining QoL. Following approval of RIB in 2017, the non-interventional study (NIS) RIBANNA was initiated to collect real-world data on the safety and effectiveness of RIB plus endocrine therapy for first-line (1L) treatment of aBC. We have used the 7th interim analysis data to characterize the QoL of aBC pts treated with RIB in 1L. Methods: RIBANNA is a prospective, multicenter NIS investigating RIB for aBC in Germany. Here, we present patient-reported outcomes (PROs) based on the EORTC QLQ-C30 questionnaire as mean difference from baseline and mean first time to deterioration (TTD) by at least 10%. TTD is defined as the time from baseline to the first worsening by 10% of the baseline value. Furthermore, changes over time will also be reported for the EORTC subscales. All PROs constituted exploratory endpoints, and all analyses were descriptive and intended for hypothesis generation only. Results: At the data cutoff for the 7th interim analysis (Sep 30, 2024), 2,567 pts were enrolled in the study and followed up to 83.6 months. Enrolled pts were allocated to three treatment cohorts: RIB + AI/F (1,852 pts), endocrine monotherapy (183 pts) or chemotherapy (139 pts). Due to the small number of pts in the last two mentioned, the QoL analysis focuses solely on the RIB cohort. The study discontinuation rate due to any reason during 1L was 32.8% in the RIB cohort, while 20.8% of pts were still progression-free. The questionnaire completion rate was 83.5% at baseline and 42.6% at 72 months. In the overall RIB cohort, EORTC global health status (GHS) did not change significantly throughout the study, although an initial trend toward improvement (mean change from baseline at 7-12 months: + 3.9 points) was observed, followed by a gradual decrease over time (−2.5 points at 67-72 months). However, the magnitude of change did not reach the minimally important difference (MID) for the EORTC QLQ-C30 of 5-10 points change from baseline. In the overall RIB cohort, the median TTD of GHS by 10% was 12.2 months. Among EORTC subscales, role functioning (RF) in the total pt population showed a continuous decrease over time in the overall RIB cohort. However, only from 43-48 months onward, a deterioration of 7.8 points from baseline was observed, which, according to the EORTC guideline, is considered only a small change for RF. The median TTD for the RF subscale was 12.0 months. In addition to the analyses mentioned for the overall cohort, QoL results for pts subgroups will be presented. Conclusions: RIBANNA provides, for the first time, real-world QoL data on RIB in a large cohort of pts with HR+/HER2- aBC, confirming findings from the pivotal clinical ML trials. GHS changes over time were comparable to the randomized ML trials, where GHS was maintained throughout the study. Only after 12 months, 50% of pts experienced for the first time a deterioration in RF and GHS values of at least 10%. Stable mean changes from baseline in EORTC GHS and persistent RF over time suggest no relevant deterioration in QoL, which is a promising outcome in the real-world setting.
Presentation numberPS5-03-05
First-line (1L) datopotamab deruxtecan (Dato-DXd) vs chemotherapy in patients (pts) with locally recurrent inoperable or metastatic triple negative breast cancer (TNBC) for whom immunotherapy was not an option: Additional safety analyses from the TROPION-Breast02 study
Tiffany A. Traina, Memorial Sloan Kettering Cancer Center, New York, NY
T. A. Traina; Department of Medicine, Evelyn H. Lauder Breast Center, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: In the primary analysis of the phase 3 TROPION-Breast02 study (NCT05374512), 1L Dato-DXd showed statistically significant and clinically meaningful improvements in overall survival (HR: 0.79 [95% CI 0.64, 0.98]; p=0.0291) and progression-free survival (HR: 0.57 [95% CI 0.47, 0.69]; p<0.0001) vs investigator’s choice of chemotherapy (ICC) in pts with locally recurrent inoperable or metastatic TNBC for whom immunotherapy was not an option. Despite more than double the median duration of treatment (tx), rates of grade (G)≥3 and serious treatment-related adverse events (TRAEs) were similar, and discontinuations were lower, with Dato-DXd vs ICC. Here we report additional safety analyses. Methods: The TROPION-Breast02 study design has been previously reported; safety was a secondary endpoint. Exposure-adjusted incidence rates (EAIRs) of AEs were derived from a post-hoc analysis, and expressed as subject rate (per 100 pt years), calculated as number of pts with AEs divided by the total duration of treatment across all pts, x100. Tx-related AEs of special interest (AESIs) for Dato-DXd included oral mucositis/stomatitis, ocular surface events (OSEs), and interstitial lung disease (ILD)/pneumonitis. For prevention/management of AESIs in the Dato-DXd arm, steroid-based mouthwash 4x daily was highly recommended but not mandated, and pts were advised to use artificial tears and avoid contact lenses. Ophthalmological assessments were required at baseline, as clinically indicated and at end of tx; additional assessments were required every 3 cycles while on tx in the Dato-DXd arm only. Results: At data cut-off (DCO: 25 Aug 2025), 319 pts had received Dato-DXd and 309 had received ICC; median total tx duration was 8.5 months vs 4.1 months in the Dato-DXd vs ICC arm. Absolute incidence rates of TRAEs with Dato-DXd vs ICC were: all-grade, 92.8% vs 83.2%; G≥3, 32.9% vs 28.8%; serious, 9.1% vs 8.4%; leading to tx discontinuation, 4.4% vs 7.4%. EAIRs of TRAEs per 100 pt years were lower with Dato-DXd vs ICC: all-grade, 104.0 vs 175.9; G≥3, 36.9 vs 60.9; serious, 10.2 vs 17.8; leading to tx discontinuation, 4.9 vs 15.7. Tx-related oral mucositis/stomatitis was reported in 192 pts (60%) in the Dato-DXd arm; most events were G1 (78 pts [24%]) or G2 (87 pts [27%]), and no events led to tx discontinuation. Median time to onset (TTO) was 26 days (range 1-603). Among 114 pts with G≥2 oral mucositis/stomatitis, events had resolved to G≤1 in 103 pts (90%) at DCO; median time to resolution (TTR) was 40.5 days (range 4-510). OSEs were reported in 149 pts (47%) in the Dato-DXd arm, were mostly G1 (76 pts [24%]) or G2 (50 pts [16%]), and led to tx discontinuation in 3 pts (0.9%). Median TTO was 77.5 days (range 2-912). Among 73 pts with G≥2 OSEs, events had resolved to G≤1 in 49 pts (67%) at DCO; median TTR was 64 days (range 8-528). Adjudicated drug-related ILD/pneumonitis was reported in 9 pts (3%) in the Dato-DXd arm: G1 in 1 pt, G2 in 7 pts, and G5 in 1 pt (characterized by investigator as G3 pneumonitis, with death assessed as related to breast cancer). Adjudicated drug-related ILD/pneumonitis led to tx discontinuation in 3 pts (0.9%). Median TTO was 259 days (range 161-588); 4/9 events had resolved at DCO, and median TTR was 112.5 days (range 13-159). Conclusion: In TROPION-Breast02, pts were on tx for longer in the Dato-DXd vs ICC arm; exposure-adjusted rates of TRAEs were lower with Dato-DXd vs ICC. Tx-related AESIs with Dato-DXd were mainly G1/2, rarely led to treatment discontinuation, and were mostly resolved to G≤1 at DCO. These data further support Dato-DXd as the new 1L standard of care for pts with locally recurrent inoperable or metastatic TNBC for whom immunotherapy is not an option.
Presentation numberPS5-04-09
Pathological complete response rates, treatment patterns, and survival outcomes in high risk, neoadjuvant-treated HER2-positive early breast cancer: a Canadian chart review
Jan-Willem Henning, University of Calgary, Calgary, AB, Canada
W. Y. Cheung1, J.-W. Henning1, S. Shokar2, M. T. Warkentin1, Z. Senhaji Mouhri2, M. Badin2, A. Nam2; 1University of Calgary, Calgary, AB, Canada, 2AstraZeneca Canada, Mississauga, ON, Canada
Background: For patients with high risk HER2-positive (HER2+) early breast cancer (eBC), neoadjuvant therapy (NAT) aims to reduce recurrence and the need for treatment intensification. Patients typically undergo months of systemic therapy, which can be accompanied by toxicities, followed by surgery. Post-operative recovery may be complicated by surgical morbidity, and the need for further intensive adjuvant therapies can add to the burden. A pathological complete response (pCR) is regarded as a meaningful marker of therapeutic efficacy and is pivotal in the patient experience. However, real world evidence from Canada on treatment patterns, pCR rates, and key outcomes, including surgical type, complications, and subsequent adjuvant therapy, is limited for this population. Methods: We conducted a retrospective chart review of a random subset of adults (≥18 years) with non-metastatic, high risk (T0-4N1-3M0 or T3-4N0M0) HER2+ eBC treated with NAT (chemotherapy with or without anti-HER2 agents) at Alberta cancer centres between 2013 and 2022, identified from the Oncology Outcomes database, with follow up through August 2024. Primary objectives were to describe pCR rates and neoadjuvant and post-neoadjuvant treatment patterns. Secondary objectives included real world (rw) recurrence-free survival (rwRFS), event free survival (rwEFS), invasive disease free survival (rwiDFS), and overall survival (rwOS). Exploratory analyses examined associations between pCR and clinical factors with these outcomes. Descriptive statistics summarized baseline and treatment characteristics; time to event outcomes were analyzed with Kaplan Meier methods; associations with clinical factors were evaluated using Cox regression. Results: Among 606 patients (mean age 50.6 years; mean follow up 5.4 years), 65.2% (n=395) were hormone receptor (HR) positive, 34.8% (n=211) hormone receptor negative; 35.3% (n=214) received anthracycline containing NAT, 64.7% (n=392) non-anthracycline containing NAT; 95.0% (n=576) had >4 NAT cycles; 79.2% (n=480) received radiation therapy, 90.2% (n=433) within 12 weeks of surgery. Of 571 patients with known pCR status, 46.9% (n=268) achieved pCR. Breast conserving surgery was more common among those with pCR (53.3% [n=105]) vs without (46.7% [n=92]); mastectomy was more frequent among those without pCR (56.4% [n=211]) than with (43.6% [n=163]). Nodal dissection was less frequent in breast conserving surgery (27.4% [n=132]) vs mastectomy (72.6% [n=350]). Among 583 patients receiving adjuvant therapy, trastuzumab was most common (79.9% [n=484]), followed by trastuzumab emtansine (T-DM1, 11.9% [n=72]), pertuzumab + trastuzumab (2.8% [n=17]), and other therapies (1.7% [n=10]); T-DM1 was used almost exclusively in patients without pCR. At 5 years, patients with pCR had higher rwRFS (90.1% vs 80.8%), rwEFS (87.8% vs 78.2%), rwiDFS (87.3% vs 77.4%), and rwOS (91.3% vs 87.3%) compared to those without. In a Cox proportional hazards model (adjusted for age, HR status, immunohistochemistry, Charlson comorbidity index, institution type, NAT cycles), pCR was associated with reduced risk for all events (adjusted HR [95% confidence interval] for pCR vs non-pCR: rwRFS: 0.41 [0.26-0.66]; rwEFS: 0.46 [0.31-0.70]; rwiDFS: 0.44 [0.29-0.68]; rwOS: 0.52 [0.30-0.88]; all p<0.001). Conclusions: pCR following NAT is a key milestone for patients with high-risk HER2+ eBC, being associated with less intensive surgical management, a reduced need for further intensive adjuvant therapy, and improved survival outcomes. Overall rates of adjuvant T-DM1 use may reflect recency of public funding. Lower pCR rates, compared to clinical studies of HER2-based NATs, may reflect limited public funding for NAT pertuzumab in Canada during the study period.