Poster Session 4
Session Details
Presentation numberPS4-01-01
Multi-omics analysis of breast cancer with subtype classification
Tomomi Kon, Graduate School of Medicine, Tohoku University, Sendai, Japan
T. Kon1, N. Shoji-Harada1, H. Tokuno2, Y. Hamanaka1, A. Ebata1, M. Iida1, M. Sato1, M. Yanagaki1, A. Yamazaki1, A. Sakamoto1, M. Makita1, O. Hinano1, T. Abe2, M. Miyashita1; 1Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, JAPAN, 2Department of Clinical Biology and Hormonal Regulation, Graduate School of Medicine, Tohoku University, Sendai, JAPAN.
Background:In breast cancer, changes in the fecal metabolites and gut microbiota have been shown to influence drug sensitivity and resistance; however, differences in microbial composition between subtypes and their impact on cancer risk remain poorly understood. Here, we compared the fecal metabolites and gut microbiota between healthy controls(HCs) and breast cancer(BC) patients with subtype-specific analyses. Methods:43 HCs and 29 BC patients feces were collected. Samples from BC patients were collected before treatment initiation, when pre-treatment samples were unavailable, samples were collected during treatment. Metabolomic analysis was performed with LC-TOFMS/MS. 16S metagenomic analysis was performed and the composition was evaluated using ALDEx2. Statistical analysis was conducted using the Wilcoxon-Mann-Whitney test, and p values were adjusted for multiple comparisons using the Benjamini-Hochberg method.Results:The median age was 71 years (range: 36-95) for HCs and 57 years (range: 32-87) for BC patients. The subtypes included 18 cases of hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative, 6 cases of HER2-positive, and 5 cases of triple-negative breast cancer (TNBC). Concerning metabolomic analysis, the PCA plot indicated that the metabolomic profile was different between the HCs and BC patients. AMP, asparagine, diethanolamine, N-Acetylglucosamine 1-phosphate, NAD+, nicotinamide, riboflavin, terephthalate and UMP were increased in the BC patients. ROC analysis for these metabolites yielded an AUC over 0.79 in discriminating BC patients from HCs. In gut microbial analysis, β diversity assessed showed no significant difference between HCs and BC patients, while α diversity measured by the Chao1 was reduced in BC patients (q< 0.001). Similarly, the HR+/HER2- group showed no significant difference in β diversity, but α diversity assessed by Chao1 was reduced in the HR+/HER2- group (p< 0.05). Because the number of HER2-positive and TNBC was small, we couldn’t come to the conclusion about the difference in these two groups. ALDEx2 revealed that Clostridiales and Bacteroidales at the order level were changed when comparing HCs and BC patients (effect size > 0.4, p < 0.01, adjusted p = 0.27, 0.19). Furthermore, comparison between the HR+/HER2- and TNBC subtypes showed several changes in Clostridiales (effect size > 0.8, p < 0.05, adjusted p = 0.86). Even within the same species, the direction of change varied among subtypes.Conclusion:This study provides novel insights into the metabolome in breast cancer, highlighting as potential biomarkers. Microbial diversity was reduced in BC patients and the gut microbiota was different between breast cancer subtype.
Presentation numberPS4-01-02
A Comparative Analysis of TAR and NLR in Patients with triple negative breast cancer treated with as KEYNOTE-522 trial : Distinct Predictors of Pathologic Response and Survival
Airi Han, YUWMC, Wonju, Korea, Republic of
A. Han1, H. Choi1, J. Ahn2, H. Ahn2, J. Shin2, J. Song2, Y. Park2; 1Surgery, YUWMC, Wonju, KOREA, REPUBLIC OF, 2Internal medicine, SMC, Seoul, KOREA, REPUBLIC OF.
Background) Pathologic complete response (pCR) has been widely used as a surrogate marker for relevant outcomes in breast cancer patients undergoing neoadjuvant treatment. However, the prognostic value of pCR varies by subtype, with triple-negative breast cancer (TNBC) showing one of the strongest associations.Notably, some patients achieving pCR still experience recurrence, suggesting that pCR and survival outcomes such as event-free survival (EFS) may represent distinct biological endpoints from different processes.This study investigated whether the tumor-aorta ratio based on Hounsfield units (TAR) and the neutrophil-to-lymphocyte ratio (NLR) function as distinct predictive biomarkers for pCR and EFS in TNBC patients treated with the KEYNOTE-522 regimen. Materials and Methods) Patients with TNBC who received neoadjuvant systemic therapy according to the KEYNOTE-522 regimen were included. Clinicopathologic data, including TAR and NLR values, were collected and analyzed. Group comparisons were performed using independent t-tests or Mann-Whitney U tests, as appropriate. Statistical analyses were conducted using SPSS version 29, with a p-value <0.05 considered statistically significant. Results) Initial TAR was significantly associated with pCR (p = 0.022), supporting its role as a predictive biomarker for immediate tumor response. In contrast, EFS was more strongly associated with post-treatment NLR, a systemic immune marker (p < 0.001).TAR decreased consistently after treatment, regardless of pCR status or the occurrence of subsequent events, suggesting that TAR may reflect structural tumor regression.In contrast, NLR exhibited divergent post-treatment patterns. Among patients without events, NLR increased from 1.99 ± 0.85 to 2.50 ± 2.88. However, in patients who experienced events, NLR decreased from 2.26 ± 1.05 to 1.21 ± 0.45. Further analysis revealed that neutrophil counts changed in opposite directions depending on event occurrence, while lymphocyte counts decreased across both groups. Conclusion)This study suggests that pCR and EFS may require distinct predictive biomarkers, with TAR and NLR reflecting different biological processes: localized tumor response versus systemic immune activity. Importantly, in this uniform cohort of TNBC patients treated with an identical neoadjuvant regimen, an increase in NLR—driven primarily by increased neutrophil counts—was associated with better prognosis. These findings raise the possibility that neutrophil-mediated immune responses may contribute to improved clinical outcomes, underscoring the potential relevance of systemic immune dynamics in the context of outcome.
Presentation numberPS4-01-04
Circulating MIRNA Signature Predicts Breast Cancer Metastasis: A Pilot Study for Non-Invasive Risk Stratification
Veladi Sasimouli, Sri Shankara Cancer Hospital and Research Centre, bangalore, India
A. Korlimarla1, V. Sasimouli2, L. Krishna2, S. Shree3, H. P.S4, B. Sivaraman5, S. Appachu6, S. Srihari7, G. Guhan8, P. Advani9, S. B.S2; 1Molecular Oncology, Sri Shankara Cancer Hospital and Research Centre, bengaluru, INDIA, 2Surgical Oncology, Sri Shankara Cancer Hospital and Research Centre, bangalore, INDIA, 3Surgical Oncology, Sri Shankara Cancer Hospital and Research Centre, Bangalore, INDIA, 4Molecular Oncology, Sri Shankara Cancer Hospital and Research Centre, bangalore, INDIA, 5Molecular Oncology, Sri Shankara Cancer Hospital and Research Centre, BAngalore, INDIA, 6Medical Oncology, Sri Shankara Cancer Hospital and Research Centre, bangalore, INDIA, 7Pathology, Sri Shankara Cancer Hospital and Research Centre, bangalore, INDIA, 8Molecular Oncology, Sri Shankara Cancer Hospital and Research Centre, Bangalore, INDIA, 9Hematology and Oncology, Mayo Clinic, florida, FL.
Background: Breast cancer remains the leading cause of cancer-related mortality in women globally, with metastatic disease accounting for >90% of deaths. Current staging and histopathological features inadequately predict individual recurrence risk. Approximately 20-30% of early-stage patients develop distant metastases despite optimal therapy, while others are overtreated. Circulating microRNAs (MIRNAs) represent a paradigm shift toward liquid biopsy, offering real-time tumor assessment through minimally invasive blood sampling. These stable molecules reflect dynamic tumor behavior and metastatic potential, making them ideal for personalized surveillance strategies.Methods:. This prospective proof-of-concept study included 46 breast cancer patients from Sri Shankara Cancer Hospital, India: 37 with metastatic disease (plasma collected at progression, 2024) and 9 disease-free controls (≥3-year follow-up). MIRNA isolation used MIRVANA method with samples processed within 2 hours, stored at -80°C. Expression levels of MIR-21, MIR-30d, and MIR-425 were quantified by TaqMan qRT-PCR, normalized to MIR-16 control. Logistic regression analysis of the transcript levels yielded a probability of predictive accuracy ranging from 0-1. Cut-off of high expression was chosen based on the best sensitivity and specificity by ROC curve analysis. Clinicopathological parameters (CPP) were correlated with MIR expression(Mann-Whitney U test). A combined model of MIR and CPP was built using logistic regression and ROC curve analysis.Results: Patient demographics(in Table) Difference in median age, subtypes and nodal status are interesting to note. MIR-21 showed elevated expression in metastatic cases, particularly hormone receptor-positive tumors. MIR-30d demonstrated significant correlation with grade 3 and node-positive status (p=0.038 for N0 vs N1, p=0.006 for N0 vs N3), with high expression associated with poor progression-free survival (p=0.0057, HR=3.24). MIR-425 exhibited significant differential expression between metastatic and non-metastatic groups with strong correlation to advanced nodal stages (p=0.0018). Univariate analysis with all clinicopathological parametres with DFS revealed LN status as significant predictor (AUC=0.68). Similarly combined MIR score of MIR-425 and MIR-30d achieved 72% predictive accuracy of DFS (AUC=0.716). Interestingly, multivariate analysis of MIR score and LN with DFS, enhanced the AUC to 0.81.Conclusions: This proof-of-concept study suggests circulating MIR-30d and MIR-425 may serve as potential biomarkers for breast cancer recurrence risk stratification. The integrated signature shows promise for enhancing surveillance strategies. Assay also standardized methods for using plasma to measure circulating MIRNA levels. Validation in larger multicenter cohorts is underway for clinical assay development.
| characteristics | metastatic(n=37) | non metastatic (n=9) |
| Demographics | ||
| Median age, years (range) | 59(24-78) | 69(53-75) |
| menopasual status | ||
| premenopasual | 16(43%) | 2(22%) |
| post menopasual | 21(57%) | 7(78%) |
| Nodal status | ||
| Node positive | 34(92%) | 5(56%) |
| Node negative | 3(8%) | 4(44%) |
| Molecular Subtypes | ||
| Hormone receptor positive | 17(46%) | 4(44%) |
| Her2 positive | 13(35%) | 3(33%) |
| Triple negative | 7(19%) | 2(22%) |
| Tumour grade | ||
| Grade 1 | 2(5%) | 0 |
| Grade 2 | 24(65%) | 5(56%) |
| Grade 3 | 11(30%) | 4(44%) |
| Ki67, median % | 30 | 20 |
| Metastatic Disease Pattern | ||
| De novo Metastasis | 18(49%) | |
| Recurrent metastatic | 19(51%) | |
| CA15-3 elevated | 21(57%) |
Presentation numberPS4-01-05
Mitotic Circulating Tumor Cells Correlates with Poor Clinical Outcomes in Metastatic Breast Cancer Patients over a 3 Year Period
Alexis B Duffy, Creatv Microtech Inc., Monmouth Junction, NJ
A. B. Duffy1, M. Cristofanilli2, C. Reduzzi2, W. V. Williams3, G. Del Priore3, S. Chumsri4, C. Tang5, T. Iwase6, N. T. Ueno6, D. L. Adams1; 1Research, Creatv Microtech Inc., Monmouth Junction, NJ, 2Medicine, Weill Cornell Medicine, New York, NY, 3Management, BriaCell Therapeutics Corp., Philadelphia, PA, 4Oncology, Mayo Clinic Cancer Center, Jacksonville, FL, 5Management, Creatv Microtech Inc., Potomac, MD, 6Medicine, University of Hawai’i Cancer Center, Honolulu, HI.
Background: Currently, the gold standard in cancer diagnosis, prognosis, and treatment is assessing cancer cell mitosis in tumor tissue biopsies, which identifies patients with more aggressive cancer subtypes that may require more tailored treatment regimens. Circulating tumor cells (CTCs), cancer cells that detach from primary tumors, enter the bloodstream, and seed metastatic sites, are a well-known non-invasive blood biomarker which can also be used to aid in stratifying metastatic breast cancer (mBC) patients with highly aggressive subtypes. Initial pilot studies have identified a particular subtype of CTCs which are undergoing mitosis that appears to correlate with very poor survival outcomes compared to non-mitotic CTCs alone. In a multi-institutional prospective study, we isolated CTCs from the blood of n=167 mBC patients to categorize mitotic CTCs and evaluate their association with progression-free survival (PFS) and overall survival (OS) over a 3 year period. Methods: Peripheral blood samples (7.5ml) were collected from n=167 mBC patients progressing on systemic therapies and prior to new systemic therapy induction (i.e. chemotherapy n=102, hormone therapy n=13, immunotherapy n=63, targeted therapy n=62). Samples were processed using CellSieve microfilters to isolate CTCs via size exclusion and fluorescently stained for CD45, Cytokeratin, and DAPI. CTCs were imaged, enumerated, and subtyped based on the presence of ≥1 mitotic event, using previously established visual indicators. PFS and OS were assessed over 3 years by censored univariate and multivariate analyses. Results: CTCs were detected in ~47% (n=79/167) of patients, while mitotic CTCs were identified in a subset of this group, ~48% (n=38/79). The presence of any CTC was associated with poor outcomes (PFS HR=0.49, 95%CI=0.3-0.7, p=0.0002 and OS: HR=0.42, 95%CI=0.3-0.7, p=0.0002). However, the presence of mitotic CTCs correlated with significantly poorer outcomes compared to non-mitotic CTCs (PFS: HR=0.3, 95%CI=0.2-0.5, p<0.0001 and OS: HR=0.3, 95%CI=0.2-0.6, p=0.0002). Further, patients with no CTCs had a median PFS (mPFS) of 6 months and median OS (mOS) of 22 months, patients with non-mitotic CTCs had a mPFS of 4 months and mOS of 14.5 months; patients with mitotic CTCs had a mPFS of 2.4 months and mOS of 4.4 months. Notably, 95% of patients with a mitotic CTC progressed within 6 months, regardless of systemic therapy type. Interestingly, there were 5 patients who survived longer than 1 year with a mitotic CTC all of which were receiving specific targeted therapies. Conclusion: In this study, the detection of CTCs indicated poor outcomes in patients. Additional cytological analysis of CTCs further identified CTCs undergoing mitosis which correlated to highly aggressive disease with the poorest survival rates. These results indicate that while the presence of CTCs is a prognostic indicator for survival, subtyping CTCs and identifying mitosis may correlate to therapy response. This study highlights the need for larger, more comprehensive studies to expand upon and validate these findings.
Presentation numberPS4-01-06
Genomic alterations in young adults compared to average-onset patients with breast cancer
Nerea Lopetegui-Lia, The Ohio State University, Columbus, OH
N. Lopetegui-Lia1, A. Davenport1, J. Gill2, A. M. Roy1, S. Myers3, D. Agnese3, E. Burke3, T. Y. Andraos4, D. M. Schimming1, S. Hulett1, H. LeFebvre1, M. Gatti-Mays1, D. Stover1; 1Medical Oncology, The Ohio State University, Columbus, OH, 2Internal Medicine, Tennova North Knoxville Medical Center, Powell, TN, 3Surgical Oncology, The Ohio State University, Columbus, OH, 4Radiation Oncology, The Ohio State University, Columbus, OH.
Introduction: In the United States, breast cancer (BC) is the most common cancer and leading cause of cancer-related death among young women. Yet approximately only 17% of young adult (YA) patients (age < 40 years) carry a known cancer susceptibility mutation, and the full spectrum of somatic genomic alterations unique to this population remains poorly defined. YA also face worse outcomes than those with average-onset (≥40 years) BC, even after adjusting for tumor characteristics. How genomic features contribute to these disparities and influence outcomes is not well understood. This study aims to characterize clinical and genomic alterations in YA with BC at our institution.Methods: Patients consented to the Total Cancer Care protocol (as part of the Oncology Research Information Exchange Network) at the Ohio State University, whose tumors underwent RNA sequencing and tumor/germline whole exome DNA sequencing were included for analysis. YA were considered < 40 years, and average-onset were considered ≥40 years. We focused on clinical characteristics, as well as mutations, copy number alterations, and mRNA expression changes in genes relevant to BC. Descriptive statistics were used to characterize the frequency and co-occurrence of alterations. The Chi-square test was used to assess associations between categorical variables, and the Wilcoxon rank-sum test was applied to compare continuous variables between groups. Kaplan-Meier survival analyses and log-rank tests were conducted to explore associations between genomic alterations and overall survival (OS). Results: A total of 825 patients were included in the analysis: 98.2% (n=813) were women, 1.4% (n=12) were men, and 0.4% (n=3) had unreported sex. There were 10% (n=83) of patients aged <40 years, and 90% (n=742) were aged ≥40 years. The median age at diagnosis in the <40 cohort was 36.0 years, compared to 56.3 years in the ≥40 cohort. In both age cohorts, most patients were White: 89.3% (n=75) in the <40 cohort and 90.4% (n=672) in the ≥40 cohort. Black patients comprised 10.7% (n=9) and 7% (n=53) in <40 and ≥ 40 cohorts, respectively (p=0.974). Higher pathologic lymph node (LN) involvement was more common in younger patients: 20.3% (n=17) of <40 cohort had pN2 or pN3 disease versus (vs) 15.3% (n=114) in the ≥40 cohort (p=0.066). The median tumor mutational burden (TMB) was significantly lower in the <40 cohort: 5.5 vs 9.7 in the ≥40 cohort (p = 0.001). Genes significantly enriched in the <40 cohort (compared to ≥40 years) were: NBPF12 22.5% (n=16) vs 10.8% (n=73), p =0.007; CDK17 15.3% (n=11) vs 4.5% (n=30); ZNF280B 11.1% (n=8) vs 3.85% (n=26). Genes enriched in the ≥40 cohort were: CDH1 20.1% (n=136) vs 4.2% (n=3); p = 0.003; PIK3CA 39.1% (n=264) vs 18.1% (n=13), p = 0.0003. Looking at specific PIK3CA alterations, H1047 mutations were significantly more frequent in the ≥40 cohort. Lastly, the probability of OS was significantly worse in patients aged <40 compared to those ≥40 years (p= 0.02). Conclusion: While this analysis corroborates existing evidence that, compared to patients ≥40 years diagnosed with BC, YA patients exhibited higher rates of advanced LN involvement, lower TMB, and significantly worse OS, novel findings included differences in genomic alterations by age. Specifically, differences in the frequency of targetable alterations such as PIK3CA highlight biologically and clinically meaningful differences in BC by age, underscoring the need for age-specific therapeutic strategies and further investigation.
Presentation numberPS4-01-07
Prospective Validation of HER2DX in Early HER2+ Breast Cancer: Operational Feasibility of Centralized Genomic Screening in the DEFINITIVE Trial. Real-Time Insights from the First 80 Screened Patients
Marta Gonzalez Rodriguez, Hospital Clínic Barcelona, Barcelona, Spain
M. Gonzalez Rodriguez1, K. Amilliano2, S. Pernas3, P. Sánchez4, I. Blancas5, E. Gal-Yam6, M. Lopez-Flores7, E. Lopez-Miranda8, T. Curiel9, C. Rodriguez10, A. Pous11, E. Galve-Calvo12, E. Sanfeliu13, M. Pujol14, C. Polo15, C. Guardia16, L. Paré17, M. Marín-Aguilera17, A. Herrera18, A. Sebastian19, A. Prat1, O. Martínez-Sáez1, T. Pascual1; 1Medical Oncology, Hospital Clínic Barcelona, Barcelona, SPAIN, 2Medical Oncology, University Hospital Sant Joan de Reus, Tarragona, SPAIN, 3Medical Oncology, Catalan Institute of Oncology (ICO)-L’Hospitalet de Llobregat, Barcelona, SPAIN, 4Medical Oncology, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, SPAIN, 5Medical Oncology, San Cecilio University Hospital, Granada, SPAIN, 6Medical Oncology, Jusidman Cancer Center, Sheba Medical Center, Ramat Gan, ISRAEL, 7Medical Oncology, University Hospital of León, León, SPAIN, 8Medical Oncology, Hospital Universitario Ramon y Cajal, Madrid, SPAIN, 9Medical Oncology, University Hospital of Santiago de Compostela (SERGAS), Santiago de Compostela, SPAIN, 10Medical Oncology, Hospital Universitario de Salamanca-IBSAL, Salamanca, SPAIN, 11Medical Oncology, Catalan Institute of Oncology (ICO)-Badalona, Germans Trias I Pujol University Hospital, Barcelona, SPAIN, 12Medical Oncology, Basurto University Hospital, Bilbao, SPAIN, 13Pathology Department, Hospital Clínic Barcelona, Barcelona, SPAIN, 14Medical Oncology, Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN, 15Operational department, SOLTI Cancer Research Group, Barcelona, SPAIN, 16Scientific department, SOLTI Cancer Research Group, Barcelona, SPAIN, 17Scientific department, Reveal Genomics, Barcelona, SPAIN, 18Marketing department, Reveal Genomics, Barcelona, SPAIN, 19Operational department, Reveal Genomics, Barcelona, SPAIN.
Background: HER2-positive breast cancer (BC) accounts for approximately 20% of all breast tumors. Despite major advances in HER2-targeted therapies, many patients with early-stage HER2-positive BC remain overtreated, leading to unnecessary toxicity and increased healthcare costs. HER2DX is a genomic diagnostic tool that stratifies patients by risk of recurrence and likelihood of achieving pathological complete response (pCR) after neoadjuvant therapy. The DEFINITIVE trial (www.thedefinitivetrial.eu) evaluates the clinical utility of HER2DX to guide treatment de-escalation or escalation in early-stage HER2-positive BC. We report preliminary feasibility and performance metrics from the first 80 screened patients. Methods: DEFINITIVE (NCT06446882) is an international, multicenter, randomized, open-label, two-arm clinical trial designed to evaluate the impact of HER2DX-guided treatment versus standard of care on health-related quality of life, safety, and efficacy outcomes. Eligible patients include premenopausal or postmenopausal women and men with stage II-IIIA HER2-positive BC suitable for neoadjuvant therapy.This interim analysis includes all patients screened from study initiation on October 30, 2024, through June 19, 2025. All patients underwent centralized screening with HER2DX using formalin-fixed paraffin-embedded (FFPE) tumor samples shipped to a reference laboratory. Operational endpoints included the number of samples received, HER2DX testing success rate, and turnaround time (TAT) from sample receipt to result delivery. All data were collected prospectively as part of the trial workflow. Results:As of the data cutoff, 80 patients had been screened across 11 sites in 2 countries. A total of 80 samples were received by the central laboratory, and HER2DX testing was successfully completed in 78 cases, yielding a success rate of 97.5%. Test failures were due to insufficient tumor content or low RNA purity. The median TAT was 8 working days (interquartile range [IQR] 7-10; range 3-16), confirming the feasibility of rapid centralized testing.The median time from tumor biopsy to sample reception at the central lab was 29.5 days (IQR 22-42), reflecting real-world variability in local processing and logistics.Among the 78 patients with available HER2DX results, 42.3% were classified as having low pCR likelihood, 30.8% as intermediate, and 26.9% as high. Based on HER2DX risk score, 47.4% were considered low-risk and 52.6% high-risk. ERBB2 mRNA expression levels also reflected biological heterogeneity, with 11.6% classified as low, 25.6% as intermediate, and 62.8% as high expression. Conclusions: Centralized HER2DX-based screening in the DEFINITIVE trial has proven operationally feasible on an international scale. High testing success rates, acceptable TAT, and biologically informative distributions support the integration of genomic classifiers into real-time treatment decision-making in prospective clinical trials. Funding:This project has received funding from the European Union’s Horizon Europe research and innovation programme under grant agreement No. 101136953.
Presentation numberPS4-01-08
Ex vivo cytokine profiling of live triple negative breast cancer patient specimens from core needle biopsies illustrates proof of concept to assess tumor response to immune checkpoint inhibition
Pooja P Advani, Mayo Clinic, Jacksonville, FL
P. P. Advani1, J. Harvey1, J. Polley1, M. Weidner1, A. Arnold1, D. Haley1, R. Bollam1, R. Rao1, S. Chumsri1, K. Hugghins1, M. Acampora1, C. Vivelo2, H. Hernan3, L. Hrycyniak4, J. Zweng5, H. Gierman6, S. Bhattacharya1, D. Mukhopadhyay1; 1Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 2Medical Affairs, Elephas Biosciences, Madison, WI, 3Clinical team, Mayo Clinic, Madison, WI, 4R and D, Mayo Clinic, Jacksonville, WI, 5Hematology and Oncology, Elephas Biosciences, Madison, WI, 6Chief Scientific Officer, Elephas Biosciences, Madison, WI.
Background: Immune checkpoint inhibition (ICI) in combination with chemotherapy is associated with improved outcomes in a subset of patients with triple negative breast cancer (TNBC). However, biomarkers for predicting patient response, such as PD-L1 expression, have limited predictive efficiency. Voabil et al. has shown that ex vivo cytokine profiling of live tumor tissue resections treated with αPD-1 can predict patient response to ICI (Voabil et al., Nat Med. 2021). However, this method requires large amounts of tissue to address intra-specimen tumor heterogeneity, which is not feasible with standard of care (SOC) diagnostic core needle biopsies (CNBs) for breast cancer, where tissue is scarce. We show for the first time that ex vivo cytokine profiling is possible in live tumor fragments (LTFs) generated from CNBs in breast tissue.Methods: We present a cytokine profiling platform to assess response to ICI treatment in LTFs generated from CNBs. CNBs were collected from patients who were identified as potential candidates for SOC ICI (by treating physician) and enrolled in one of two prospective observational clinical trials (NCT05520099 and NCT06349642). CNBs were cut by an automated, proprietary cutting instrument to generate LTFs ofapproximately 300 µm in thickness. To address tissue heterogeneity and mitigate tissue scarcity in CNBs, we used a sequential treatment strategy in which control and ICI treatment were applied to LTFs in the same well, in a phased approach. This strategy provides a solution for the variability observed in cross-well comparisons. LTFs were encapsulated in hydrogel and treated with IgG control followed by αPD-1 (BioXCell). Cytokine production rates were measured longitudinally using multiplex assays assessing>45 cytokines, including 15 cytokines assayed by Voabil et al., and the fold changes of cytokine production rates in response to ICI over IgG control were calculated. Results: Using non-breast tumor tissue, we validated that our platform can measure changes in cytokine production in LTFs following sequential treatment, and that these measurements are comparable to those observed in a cross-well setting. In a small cohort of TNBC specimens, we show changes in cytokine production rates in response to ex vivo sequential control and ICI treatment. Ten cytokines, including IFN-γ and CXCL10, shown to be predictive of patient response to ICI in Voabil et al. were found upregulated in response to αPD-1 in TNBC specimens. Future determination of correlation with clinical response data (pathological complete response for early stage and RECIST 1.1 for metastatic patients) gathered from NCT05520099 and NCT06349642 will be used to validate platform responders and non-responders. To date, 11 patients with confirmed TNBC diagnoses have enrolled in these clinical studies. Conclusion: We report, for the first time to our knowledge, the ability to detect immune checkpoint inhibitor responses in live TNBC tissue obtained via CNBs. This proof-of-concept lays the groundwork for integrating functional tissue responses with clinical correlation data, with important implications for guiding current ICI therapies and advancing this platform for use in next-generation immuno-oncology treatments for TNBC.
Presentation numberPS4-01-09
<b>How does proteomics-based prognostic testing compare to genomic tests and online clinical risk prediction tools in early breast cancer patients?</b>
Manjiri Bakre, OncoStem Diagnostics Pvt Ltd, Bangalore, India
M. Bakre1, T. Durgekar1, S. BA1, P. Shrivastava1, G. Basaran2, G. Ozge2, T. Korkmaz3; 1R and D, OncoStem Diagnostics Pvt Ltd, Bangalore, INDIA, 2Medical Oncology, Acibadem University Scool of Medicine, Istanbul, TURKEY, 3Medical Oncology, Acibadem Maslak Hospital, Istanbul, TURKEY.
Objective: ~70% of HR+/HER2- early breast cancer have low risk of breast cancer recurrence. Hence, prognostication in these patients to assess chemotherapy benefit is a huge value add. Online tools such as Nottingham prognostic index (NPI), PREDICT etc are often used as they are quick and free. However use of genomic prognostic tests like Oncotype DX (ODX), Mammaprint (MP) etc is increasing. CanAssist Breast (CAB) is a proteomics-based prognostic test that uses an AI-based algorithm to segregate EBC patients as ‘low or high’ risk for recurrence. CAB is validated in global studies and clinically used on ~10,000 patients to date in the Indian subcontinent, UAE, Turkey, Iran, and Saudi Arabia. Comparative analysis of prognostic tests is important to evaluate the relative performance of different prognostic tests in a population and assess how a new test performs in relation to established, validated ones. We have compared CAB with NPI, PREDICT, ODX, and MP, and here we showcase the results of those comparative studies.Methods: A patient cohort of 1474 from Europe, India and US was used to compare CAB with NPI and PREDICT. NPI risk groups were categorized into three prognostic groups: good (GPG-NPI index ≤ 3.4), moderate (MPG 3.41-5.4), and poor (PPG > 5.4). Patients with chemotherapy benefit of 15.5) categories. Agreement between CAB and NPI/PREDICT risk groups were assessed by kappa coefficient. Retrospective comparison of risk stratification by CAB with ODX and MP was done with 109 (US and India) and 43 (EU) patients, and prospectively with a total of 116 Turkish patients- 58 patients in each group. Accuracy/ negative predictive value was calculated using MedCalc. Concordance of CAB with ODX or MP was calculated using the overall percentage agreement.Results: Risk proportions generated by all tests were: CAB low:high 74:26; NPI good:moderate:poor prognostic group- 38:55:7; PREDICT low:high 63:37. 65% of NPI-MPG patients were called low risk by CAB. From PREDICT high risk patients, CAB segregated 51% as low risk, thus preventing over-treatment in these patients. Overall, there was a fair agreement between CAB and NPI [κ=0.31(0.278-0.346)] / PREDICT [κ=0.398 (0.35-0.446)], with a concordance of 97% / 88% between CAB and NPI/PREDICT low risk categories. In cohorts with mostly T1N0 patients, NPI and PREDICT segregated more as low risk compared to CAB, suggesting that T1N0 patients with aggressive biology are missed by online tools but not by CAB. Comparison of CAB with ODX retrospectively (n=109) and prospectively (n=58) showed similar low:high risk proportions as 83:17 and 79:21 for CAB; 90:10 and 83:17 for ODX. An overall concordance of 75% and 65%, and low risk concordance of 82% and 77% was observed between both the tests in retrospective and prospective studies. Retrospective (n=43) and prospective (n=58) comparison of CAB with MP showed similar low:high risk proportions of 65:35 and 66:34 for CAB; 56:46 and 52:48 for MP. Overall concordance between the two tests was 75% in the retrospective study and 62% in the prospective study, while low risk concordance was 83% and 77%, respectively.Conclusion: CAB provided unbiased risk stratification across cohorts of various geographies with minimal impact by clinical parameters, whether compared with online tools and tumor based prognostic tests. CAB is useful for all EBC patients and specifically in NPI-MPG and PREDICT high risk patients for making accurate decisions on chemotherapy use. CAB shows good concordance with ODX and MP in low risk categories. This, coupled with high accuracy comparable with ODX, shows that CAB is an excellent, cost-effective, and quick alternative.
Presentation numberPS4-01-10
Characterizing CCR7 Gene Amplification and Protein Expression in Inflammatory and non-inflammatory Breast Cancer
Surbhi Shivhare, The University of Texas MD Anderson Cancer Center, Houston, TX
S. Shivhare1, C. Sanchez1, R. Larson1, L. Dobrolecki2, M. Lewis2, J. Chen3, M. Sebastian4, A. Evdokimova5, D. Goncharova5, A. Alexander6, A. Nasrazadani6, R. Layman6, B. Lim6, V. Valero6, A. Lucci3, The MDACC Inflammatory Breast Cancer Team, W. Woodward1; 1Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 3Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Department of Veterinary Medicine & Surgery and Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 5BostonGene RnD, BostonGene Corporation, Waltham, MA, 6Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Purpose: Inflammatory breast cancer (IBC) is a rare yet exceptionally aggressive subtype that accounts for a disproportionate number of breast cancer deaths. A significant barrier to effective treatment is its ability to invade the lymphatic system and evade immune suppression. Emerging research highlights the potential role of C-C chemokine receptor type 7 (CCR7) in driving tumor progression and modulating immune responses within IBC. Baseline changes in CCR7 gene expression have not been reported in IBC. We investigated CCR7 gene amplification, as well as protein expression and spatial distribution in patient tumors, patient-derived xenografts (PDXs), and IBC cell lines. Methods: PDX characterization of non-IBC models for CCR7 was performed using data from the BCM PDX portal (PDX Insights), and sections from selected PDX were stained for multiplex immunofluorescence (mIF) and imaged with a Nikon Ni-E microscope. Cell lines were subjected to immunoblotting for CCR7, and conditioned media were analyzed by ELISA for CCR7 ligands CCL19 and CCL21. Clinical analysis was performed retrospectively, based on an IRB-approved protocol using genomic data from 18 hormone receptor-positive (HR+) HER2-negative (HER2-) IBC and 24 HR-negative (HR-) HER2- IBC cases which were compared to 150 and 104 subtype-matched non-IBC cases, respectively. Results: CCR7 gene expression did not differ between IBC and non-IBC cases independent of subtype. Four IBC patients in each subtype had gain or amplification of CCR7, while six patients in each subtype had deletion. mRNA expression did not correlate with copy number alterations (CNAs). In triple-negative and HER2+ IBC cell lines, CCR7 protein expression was high. CCR7 ligands were not detected in conditioned media of IBC cell lines. Among non-IBC PDX models characterized from PDX Insights, 9/53 demonstrated gain or amplification of CCR7, while 25/53 demonstrated loss. CCR7 gene expression was not concordant with CNAs; we observed 11% CCR7 gene expression in HR+ HER2- IBC patients and 16% in HR- HER2- IBC patients. mIF analysis of PDX sections from BCM-3104, BCM-3807, and BCM-15029, representing CCR7 amplified/high expression (+/+), non-amplified/high expression (-/+), and non-amplified/low expression (-/-), respectively, revealed significant membranous expression only in the +/+ PDX context with concurrent high expression of podoplanin and ligand CCL21. CCR7 protein and gene expression were roughly correlated, with -/+ expression level being intermediate between -/- and +/+. H&E staining showed tumor necrosis and an increase in inflammatory infiltrate relative to CCR7 gene expression. Conclusion: Patient data suggest that CCR7 CNAs are common in both inflammatory and non-inflammatory breast cancer phenotypes within HR+ HER2- and triple-negative breast cancer subtypes and are not concordant with gene expression overall. In PDX tissues, membranous CCR7 was predominantly found in the amplified PDX context. Additional work is needed to distinguish the impact of membranous versus cytoplasmic CCR7 expression. CCL21 showed greater expression than CCL19, highlighting potential ligand-specific functionality of the CCR7 axis. These characterizations provide important baseline data for further consideration of CCR7 as a target for aggressive breast cancer.
Presentation numberPS4-01-11
Exploration of racial differences in variants of uncertain significance (VUS), ESR1 and PIK3CA mutation frequency, matched therapy use, and outcomes in metastatic breast cancer (mBC)
Andrew A Davis, Washington University in St. Louis, St. Louis, MO
A. A. Davis1, L. S. Welch2, J. Saha3, L. Bucheit2, M. D. Lipsyc-Sharf4, A. Bardia4, C. X. Ma1, M. Cristofanilli5, E. L. Podany1; 1Department of Medicine, Washington University in St. Louis, St. Louis, MO, 2Medical Affairs, Guardant Health, Palo Alto, CA, 3Outcomes and Evidence, Guardant Health, Palo Alto, CA, 4Department of Medicine, Division of Hematology/Oncology, UCLA Health, Santa Monica, CA, 5Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY.
Background: Black patients (pts) are more likely to be diagnosed with mBC and have higher mortality rates than White pts, in part due to social determinants of health, higher rates of de novo mBC, and possible genomic differences by race and ethnicity. Most variant classification efforts have focused on White populations, creating uncertainty about prevalence and significance of VUS in Black pts. It is also unclear if mutation frequency differences exist in genes with known targets, such as PIK3CA and ESR1. We examined a real-world (rw) database for racial differences in VUS, ESR1 and PIK3CA detected by circulating tumor DNA (ctDNA), matched therapy use, and outcomes by race for ESR1 and PIK3CA. Methods: Pts with mBC and ctDNA (Guardant360) results post-mBC diagnosis were identified from Guardant INFORM (n=46,286), which includes de-identified records with clinical genomic data, claims, and race when available. For Black and White pts (n=34,149), overall VUS rates were analyzed by race, as defined by OncoKB annotation. ESR1 and PIK3CA codons of interest [ESR1 ligand binding domain (LBDmut); PIK3CA helical or kinase domains (HKmut)], targeted therapy matching (elacestrant and alpelisib), and outcomes were evaluated by race. Outcomes were assessed using rw time to treatment discontinuation (rwTTD), time to next treatment (rwTTNT), and overall survival (rwOS) in months, stratified by race. Chi-squared tests compared proportions; log-rank tests compared time-to-event outcomes. Results: There were 12.5% Black (5,791/46,286) and 61.2% White (28,358/46,286) pts included. Mean VUS frequency was similar between groups (2.40 Black vs 2.47 White, p>0.05). While White pts with ESR1 mutations more frequently had ESR1 LBDmut vs Black pts, there was no difference in matched therapy use with elacestrant by race for ESR1 LBDmut (Table 1). Black pts with ESR1 LBDmut and 2L elacestrant had significantly shorter rwTTD (3.5 vs 4.2, p=0.024) and shorter numeric rwTTNT (5.9 vs 8.9, p=0.32). White pts also had significantly more PIK3CA mutations, yet among PIK3CA HKmut, there was no difference in matched therapy use with alpelisib by race (Table 1). Black pts with PIK3CA HKmut treated with 2L alpelisib compared with white pts had numerically longer rwTTD (4.4 vs 3.7, p=0.48), rwTTNT (8.7 vs 6.6, p=0.33), and rwOS (33.9 vs 25.9, p=0.38); none reached statistical significance. Conclusion:In this large, rw analysis of a diverse patient population, differences in VUS rate, ESR1/PIK3CA matched therapy use, and outcomes were not observed between Black/White pts, suggesting the need for additional exploration of race-based differences on mBC outcomes. This study underscores the need for continued inclusion of diverse populations in genomic discovery and prospective studies to clarify known outcome disparities.
| Black | White | p-value | |
| VUS rate-any gene | 3119/5971 (53.9%) | 15688/28358 (55.3%) | p<0.001 |
| VUS rate – mBC genes | 206/5585 (3.5%) | 873/28358 (3.1%) |
p=0.66 |
| Any ESR1mut | 1445/5791 (24.9%) | 7001/28538 (24.7%) | p=0.67 |
| ESR1 LBDmut | 1263/1445 (87.4%) | 6249/7001 (89.3%) | p=0.03 |
| LBDmut Therapy Matching | 184/1263 (14.6%) | 842/6249 (13.5%) | p=0.30 |
| Any PIK3CAmut | 1917/5791 (33.1%) | 10,114/25358 (35.7%) | p<0.001 |
| PIK3CA HKmut | 1383/1917 (72.1%) | 7557/10114 (74.7%) | p=0.19 |
| HKmut Therapy Matching | 176/1383 (12.7%) | 836/7557 (11.1%) | p=0.07 |
Presentation numberPS4-01-12
Androgen receptor and FOXA1 in breast cancer: Prognostic insights from a treatment-naïve historical cohort
Yuka Inoue, Cancer Institute Hospital of JFCR, Tokyo, Japan
Y. Inoue1, T. Osako2, M. Akiya2, T. Chiba2, Y. Haruyama1, U. Nakadaira1, J. Masuda3, Y. Kimura1, A. Iesato1, M. Nishimura3, Y. Ozaki3, T. Maeda1, N. Uehiro1, N. Yamashita1, T. Kobayashi3, T. Sakai1, T. Takano3, T. Ueno1; 1Breast Surgical Oncology, Cancer Institute Hospital of JFCR, Tokyo, JAPAN, 2Division of Pathology, Cancer Institute of JFCR, Tokyo, JAPAN, 3Breast Medical Oncology, Cancer Institute Hospital of JFCR, Tokyo, JAPAN.
Background: We are conducting an ongoing project using archival tissue samples from primary breast cancer cases treated with surgery alone at our institution, without any adjuvant systemic therapy. Tissue microarrays (TMAs) have been constructed, and various protein expressions are being re-evaluated using contemporary immunohistochemical techniques. This treatment-naïve cohort provides a unique opportunity to assess the intrinsic prognostic significance of biomarker expression, free from the confounding effects of systemic therapy.Androgen receptor (AR) is a steroid hormone receptor expressed in a subset of breast cancers and has attracted growing interest as a potential prognostic and therapeutic marker. Forkhead-box protein A1 (FOXA1) is a pioneer transcription factor known to act as a super-enhancer of the estrogen receptor (ER), playing a crucial role in mammary gland development. It is well established as a favorable prognostic marker in ER-positive, HER2-negative breast cancer. In recent years, however, FOXA1 expression has also been reported in other subtypes, including ER-negative tumors, although its clinical significance in these contexts remains uncertain. Whether FOXA1 functions primarily as a true prognostic marker or rather as a surrogate for endocrine responsiveness remains an open question. In this study, we report the results of immunohistochemical analyses for AR and FOXA1 in a treatment-naïve historical cohort, aiming to clarify their intrinsic prognostic relevance across breast cancer subtypes. Methods: We retrospectively analyzed primary breast cancer cases operated on at our institution between 1979 and 1982, from which TMAs were constructed by sampling three representative areas of the invasive tumor component. All pathological features, including immunohistochemical markers, were re-evaluated using current standardized protocols. Clinical outcomes were obtained from institutional records. Results: Among 935 eligible cases, 758 were available for AR and FOXA1 immunohistochemical evaluation. Subtype distribution based on ER and HER2 status was as follows: ER+/HER2− (n = 538, 71.3%), ER+/HER2+ (n = 47, 6.2%), ER−/HER2+ (n = 74, 9.8%), and ER−/HER2− (n = 96, 12.7%), comparable to current subtype frequencies.Using a 10% cutoff for positivity, AR expression was observed in 69.1% of ER+/HER2−, 57.5% of ER+/HER2+, 44.6% of ER−/HER2+, and 16.7% of ER−/HER2− tumors. In survival analysis, strong AR expression was significantly associated with improved distant recurrence-free survival (DRFS) (p = 0.0239).FOXA1 expression, using the same cutoff, was detected in 96.4% of ER+/HER2−, 100% of ER+/HER2+, 92.9% of ER−/HER2+, and 23.2% of ER−/HER2− tumors. Notably, FOXA1 was expressed in a subset of ER-negative tumors. FOXA1 expression was not significantly associated with DRFS, including in ER-positive tumors. In the ER−/HER2− subtype, although no significant difference in DRFS was observed based on Kaplan-Meier analysis, among patients who experienced recurrence, those with FOXA1-positive tumors had a significantly longer disease-free interval (DFI) (p = 0.0005). Conclusion: In this historical cohort of breast cancer patients untreated with adjuvant therapy, strong AR expression was significantly associated with improved DRFS. FOXA1 was expressed across all intrinsic subtypes and, contrary to previous reports, did not demonstrate prognostic significance in ER-positive tumors in the absence of systemic therapy. These findings suggest that FOXA1 may be more reflective of endocrine responsiveness rather than intrinsic tumor aggressiveness. The observation that FOXA1-positive tumors had a longer DFI suggests a potential role in late recurrence, particularly in ER-negative subtypes, and warrants further investigation.
Presentation numberPS4-01-13
Comparative efficacy of antibody drug conjugates (ADCs) in male versus female breast cancer across the spectrum of HER2 expression
Dario Trapani, University of Milan, milan, Italy
D. Trapani1, S. Deshmukh2, S. Wu2, J. Xiu2, N. Lin3, G. Curigliano1, P. Spanheimer4, S. Gandhi5, S. Chumsri6, S. Graff7, M. Lustberg8, G. Sledge Jr2, S. Tolaney3, J. Leone9; 1oncology and hematology, University of Milan, milan, ITALY, 2Caris Life Sciences, Caris Life Sciences, Phoenix, AZ, 3breast oncology, Dana farber cancer institute, Boston, MA, 4medical oncology, University of North Carolina at Chapel Hill, Chapel Hill, Chapel Hill, NC, 5medical oncology, Winship Cancer Institute of Emory University, milan, GA, 6medical oncology, Mayo Clinic, Jacksonville, FL, 7medical oncology, Lifespan Cancer Institute, Legorreta Cancer Center, Brown University, Providence, RI, 8medical oncology, Yale School of Medicine, New Haven, CT, 9breast oncology, Dana farber cancer institute, milan, MA.
Background: Antibody-drug conjugates (ADCs) are increasingly used in the treatment of breast cancer due to their superior efficacy compared to conventional chemotherapy. The management of male breast cancer is largely extrapolated from clinical data in females and evidence regarding the efficacy of ADCs in men remains limited. This study aimed to compare survival outcomes between male and female breast cancer patients treated with ADCs across the spectrum of human epidermal growth factor receptor 2 (HER2) expression. Methods: We included total of 2,954 (Male: 34, Female: 2,920 breast cancer samples) from patients who underwent treatment with ADCs. We excluded patients with triple-negative breast cancer status due to the rarity of this subtype in males and the worse prognosis of triple-negative breast cancer, so that the cohorts would be more homogeneous without overrepresentation of triple-negative breast cancer in females. We categorized patients by HER2 status as: HER2-null (if there was 0% stain on HER2 IHC), HER2-ultra low (if HER2 IHC was 0 but with stain 1-10%), HER2-low (if HER2 IHC was 1+ or 2+ with a negative chromogenic in situ hybridization [CISH] assay), and HER2-positive (if HER2 IHC was 3+ or 2+ with a positive CISH assay). Real-world median overall survival was derived from insurance claims and calculated from the start of first ADC with either trastuzumab deruxtecan (T-DXd) or trastuzumab emtansine (T-DM1) or sacituzumab govitecan (SG) to last contact using Kaplan-Meier. Statistical significance was assessed using chi-square and Mann-Whitney U. Results: The distribution of sex by HER2 expression group was: HER2-null: 8 males vs 612 females; HER2-ultra low 6 males vs 467 females; HER2-low 11 males vs 903 females; HER2-positive 9 males vs 938 females. Median survival from treatment initiation with either T-DXd or SG in the HER2-null cohort was 21.31 months in males vs 15.72 months in females (HR 0.82, 95% CI 0.33 – 2.01, p=0.67); in the HER2-ultra low cohort was 17.73 months in males vs 14.47 months in females (HR 0.84, 95% CI 0.26 – 2.63, p=0.76); in the HER2-low cohort was 11.08 months in males vs 18.62 months in females (HR 1.53, 95% CI 0.63 – 3.7; p=0.34). In the HER2-positive cohort, 5/9 males (55.5%) and 544/938 females (58%) were treated with T-DXd. Median survival from treatment initiation with either T-DXd or T-DM1 in the HER2-positive cohort was 36.55 months in males vs 46.52 months in females (HR 1.77, 95% CI 0.84 – 3.74, p=0.12). Conclusions: In this study, survival appears to be comparable between males and females with breast cancer across the spectrum of HER2 expression when both are treated with ADCs, although the survival in the HER2-positive cohort appears numerically lower in males, consistent with data on HER2-positive MaBC. An important limitation is the small sample size of the male breast cancer cohorts, and the lack of a categorization of HER2 in HR+ vs HR-, for the small MaBC sample size. However, to our knowledge this is the first analysis evaluating outcomes of men treated with ADCs across the spectrum of HER2 expression.
Presentation numberPS4-01-14
Evaluation of Metabolic Dysregulation, Endocrine Therapy Outcomes, and Tumor Biology in HER2-/HR+ Breast Cancer using a Multi-Omic, Real-World Analysis
Jasmin Hundal, Cleveland Clinic, Cleveland, OH
J. Hundal1, R. Plagens2, A. Elliott3, G. Sledge3, C. Travaline4, S. Graff5, M. Lustberg6, M. Vasekar7, J. Leone8; 1Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 2Taussig Cancer Institute, Caris Life Science, Irving, TX, 3Caris, Caris Life Sciences, Irving, TX, 4Hershey Cancer Institute, Penn State, Hershey, PA, 5Legorreta Cancer Center, Brown University, Providence, RI, 6Yale Cancer Center, Yale University, New Haven, CT, 7Hershey Cnacer Institue, Penn State University, Hershey, OH, 8Dana-Farber Cancer Institute, Dana-Farber Cancer Institute, Harvard Medical School,, Boston, MA.
Background: Metabolic dysregulation, including hyperglycemia (HG) and type 2 diabetes mellitus (T2DM), is increasingly prevalent among breast cancer (BC) patients (pts) and may influence treatment outcomes. In hormone receptor-positive (HR+) BC, metabolic comorbidities could affect tumor biology and response to endocrine therapy. This study integrates real-world clinical outcomes with multi-omic tumor profiling to evaluate the potential impact of HG/T2DM on endocrine therapy outcomes and tumor molecular signatures in pts with HER2-/HR+ BC. Methods: This retrospective analysis utilized BC samples profiled by Caris Life Sciences (Phoenix, AZ), including next-generation sequencing (592-gene panel or whole exome for DNA and whole transcriptome for RNA) and immunohistochemistry (IHC). Tumors were classified as HER2- (≤1+ intensity or ≤10% cells stained by IHC, or 2+/>10% and CISH-negative) and HR+ (ER or PR ≥1+/≥1%). Pts with no prior systemic therapy who initiated first-line aromatase inhibitor and/or fulvestrant (AI/Fulv) within 100 days post-biopsy were identified using linked claims data. ICD-10 codes were used to define HG and T2DM status, and medication exposure was assessed for insulin, metformin, and GLP-1 receptor agonists (Ins/Met/GLP-1RA). Clinical endpoints included overall survival (OS; time from first AI/Fulv to death or last contact) and time on treatment (ToT; from first to last AI/Fulv), as assessed via Cox proportional hazards models and log-rank tests. Among pts without reported exposure to Ins/Met/GLP1-RA, tumor molecular profiles were compared between pts with HG/T2DM and metabolically healthy controls (no HG/T2DM) using Mann-Whitney U and chi-square tests. Results: A total of 2,074 pts with HER2-/HR+ BC treated with first-line AI/Fulv were included in the analysis, comprising three cohorts: 1,119 pts without HG/T2DM and no exposure to Ins/Met/GLP-1RA; 398 pts with HG/T2DM and no exposure to these agents; and 557 pts with HG/T2DM who had received Ins/Met/GLP-1RAs. Median OS was 49, 48, and 52 months, respectively (p = 0.71), and median ToT was 17, 21, and 19 months, respectively (p = 0.37). Subgroup analyses stratified by CDK4/6 inhibitor exposure in first- or later-line therapy revealed consistent patterns, with no statistically significant differences in survival across metabolic strata. Molecular analyses identified notable distinctions in tumors from pts with HG/T2DM. Pathogenic mutations in PIK3CA (50.4% vs. 42.9%; p = 0.01), CASP8 (1.5% vs. 0.1%; p < 0.001), ACVR1B (2.1% vs. 0.2%; p = 0.02), and TSC2 (0.9% vs. 0.1%; p = 0.02) were more prevalent in the HG/T2DM group. Deletion events in CYLD (2.8% vs. 0.5%; p < 0.01), B2M (0.7% vs. 0.0%; p = 0.02), CDKN2C (1.1% vs. 0.1%; p = 0.02), RAD54L (1.1% vs. 0.1%; p = 0.02), and FANCA (2.8% vs. 0.9%; p = 0.03) were similarly enriched. Amplifications of HSP90AB1 and SS18 were detected only in HG/T2DM tumors (0.7% and 1.1%, respectively; p = 0.02 for both), while MDM2 amplification was more frequent in controls (3.1% vs. 0.7%; p = 0.03). Conclusion: Metabolic dysfunction was not associated with significant differences in clinical outcomes among pts receiving endocrine therapy. However, tumors from pts with HG/T2DM exhibited distinct molecular alterations, including higher frequencies of PIK3CA mutations, MDM2 and HSP90AB1 amplifications, and deletions in CYLD, B2M, and CDKN2C, suggesting enhanced proliferative signaling and reduced immune activation. These findings may contribute to endocrine resistance and underscore the need for biomarker-driven, metabolically informed treatment strategies. Prospective studies are warranted to validate these observations and inform future therapeutic approaches.
Presentation numberPS4-01-15
Acute and Early Circulating Tumor DNA Dynamics in Metastatic Triple-Negative Breast Cancer Targeted Therapy Phase Ib/II Clinical Trials
Briana To, The Ohio State University Comprehensive Cancer Center, Columbus, OH
B. To1, V. Prasath1, D. Veney1, C. Rolfo1, V. Adalsteinsson2, N. Lin3, S. M. Tolaney3, H. A. Parsons3, R. Wesolowski1, D. Quiroga1, D. Stover1; 1Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 2Cancer Diagnostics, Broad Institute of MIT and Harvard, Cambridge, MA, 3Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: Metastatic triple-negative breast cancer (mTNBC) is a poor prognosis subset ofbreast cancer characterized by shorter survival and higher circulating tumor DNA (ctDNA) ‘tumorfraction’ (TF) relative to other breast cancer subtypes. TF is the proportion of ctDNA originatingfrom tumors cells, and early changes in ctDNA TF may correlate with therapy response,potentially offering a rapid response biomarker for early phase studies. Furthermore, there is littleknown regarding TF dynamics in the hours immediately after initiation of systemic therapy –including whether there is a ‘surge’ in ctDNA.Methods: Banked plasma samples were identified from four completed phase Ib/II clinical trialsenrolling patients with mTNBC: heat shock protein 90 (HSP90) inhibitor onalespib with paclitaxel(NCT02474173), MEK inhibitor trametinib alone and in combination with AKT inhibitorGSK2141795/uprosertib (NCT01964924), multi-kinase inhibitor cabozantinib monotherapy(NCT02260531), and JAK1/2 inhibitor ruxolitinib monotherapy(NCT01562873). For theonalsepib+paclitaxel study, plasma was collected pre-infusion, end-of-infusion (EOI), then0.5/1/2/4/6/8/24 hours post-infusion for 1) onalespib alone (day -7); 2) paclitaxel alone (day 1);and 3) onalespib + paclitaxel (day 8). For the remaining studies, plasma was collected pre-treatment on cycle 1 day 1 (C1D1) and prior to cycle 2 day 1 (C2D1). Plasma samples underwentshallow whole genome sequencing (sWGS) and TF determination. The primary objectives wereto evaluate change in TF and association with best overall response and progression-freesurvival.Results: To evaluate acute ctDNA TF change in the onalespib+paclitaxel trial, ctDNA TF wassuccessful in 313/317 (98.7%) of available plasma samples from 14 patients. There wassignificant decline in TF from pre-infusion to 6 hours for paclitaxel alone (Wilcoxon signed rankp=0.03) but no significant change for onalespib alone or onalespib + paclitaxel from pre-infusion to 6- or 24-hours. There was significant decline in TF from pre-infusion day -7 to cycle 1, day 9(median TF 16% to 6.5%, Wilcoxon signed rank p=0.004). No significant ‘surge’ was identifiedand despite significant decline in TF over the first full cycle of therapy. To evaluate change in TFacross multiple Phase Ib/II clinical trials of targeted therapies in mTNBC, we identified 57 totalpatients across studies (onalsepib+paclitaxel trial n=8; trametinib trial n=22; cabozantinib trialn=19; ruxolitinib trial n=8) with available plasma sample prior to C1D1 (T1) and follow-up plasmasample 14-28 days after therapy initiation (T2). Change in ctDNA from T1:T2 was significantlyassociated with best overall response (ANOVA p=0.01). Clearance of ctDNA at T2 (TF=0) wasassociated with significantly longer PFS (median PFS 10.3 months clearance vs. 2.3mo noclearance; log-rank p=0.013). Similarly, TF that was stable or declined T1:T2 was associated withsignificantly improved PFS relative to TF increase (log-rank p=0.003).Conclusion: There was no significant ‘surge’ in ctDNA TF within minutes to 24-hours of infusionof a targeted therapy, cytotoxic chemotherapy, or both in combination. Stability or decline ofctDNA TF 14-28 days after initiation of targeted therapy trial was associated with objectiveresponse and prolonged PFS. This supports further research on ctDNA dynamics immediatelyafter systemic therapy initiation, particularly in the context of early phase trials for mTNBC.
Presentation numberPS4-01-16
The transcriptomic profile of circulating T lymphocytes is associated with response to neoadjuvant therapy in patients with early breast cancer
Benedetta Conte, Università del Piemonte Orientale, Novara, Italy
B. Conte1, S. D’Alfonso1, M. Mellai2, D. Cora2, F. Favero2, V. Martini2, N. Negrini1, I. Taglialatela1, T. Landolfo1, B. Ruffilli3, S. Nardin4, F. D’Avanzo4, V. Rossi4, C. Branni4, M. Graeser5, N. Harbeck5, A. Gennari1; 1Medicina Traslazionale, Università del Piemonte Orientale, Novara, ITALY, 2UPO-CAAD, Università del Piemonte Orientale, Novara, ITALY, 3Medical Oncology, Università del Piemonte Orientale, Novara, ITALY, 4Medical Oncology, Ospedale Maggiore della Carità, Novara, ITALY, 5Oncology, West German Study Group, Munich, GERMANY.
Transcriptomic profiling of circulating T lymphocytes predicts response to neoadjuvanttherapy in patients with early breast cancer.Benedetta Conte1, Sandra D’Alfonso2, Marta Mellai2, Davide Cora’1, Francesco Favero1, Veronica Martini3,Letizia Negrini1, Ida Taglialatela1, Tommaso Lupo Landolfo1, Beatrice Ruffilli1, Giulia Isingrini1, FrancescaD’Avanzo3, Valentina Rossi3, Simone Nardin3, Carmen Branni3, Monika Graeser4, Nadia Harbeck4,Alessandra Gennari1.1. Oncology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.2. Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.3. Division of Oncology, University Maggiore Hospital della Carità, Novara, Italy.4. West German Study Group, Moenchengladbach, GermanyBackground. In early breast cancer (eBC), anti-tumor immunity is a key determinant of response toneoadjuvant therapy (NAT). Immune profiling currently relies on static tissue biomarkers such astumor-infiltrating lymphocytes (TILs). However, the anti-tumor immune response is a dynamicprocess undergoing substantial changes during NAT. Compelling findings show that on-treatmentTILs are a stronger predictor of pCR than baseline TILs during NAT. Circulating immune cells mayoffer a non-invasive means to monitor systemic immune response during NAT. Here we present thepreliminary data for the development of a blood-based immune assay that analyzes transcriptomicdata from circulating immune cells.Methods. Baseline circulating T lymphocytes were collected from 95 eBC patients treated at ourinstitution between 2021–2023. The cohort included 39 HER2+ cases treated with NAT plus single(n=12) or dual HER2 blockade (n=27), 36 triple-negative cases treated with NAT alone (n=19) orNAT plus pembrolizumab (n=17), and 20 HR+/HER2- cases treated with anthracyclines-basedchemotherapy. Bulk RNA was extracted for gene expression analysis. 33 immune signatures werecalculated with known prognostic role in tumor tissue. Significant analysis of microarrays (SAM)was applied to identify differences in T cell polarization between the pCR and RD group.Results. 47 patients (49%) achieved pCR. The table below shows the genomic featuresdownregulated and upregulated in pCR. At SAM analysis, T cells of responders showed lowerexpression of genes linked to Tregs (FOXP3, CCR4), monocyte recruitment (CD163, IL1R2A), andsystemic inflammation (FGFR1, IL6R). A tissue-based gene signature associated to neutrophilactivation was upregulated in non-responders, along with chemokines linked to neutrophilrecruitment (CXCL8, CXCL2, CXCL1). Similarly, a tissue-based signature associated to IL6-JAK-STAT activation pathway was downregulated in T cells of responders.Conclusions. The transcriptomic profiling of T cells carries predictive information in eBC andreplicates the prognostic information captured by tissue-based signatures. This data supports thedevelopment of blood-based immune assays in eBC GENOMIC FEATURESlog2 FOLD CHANGEFALSE DISCOVERY RATECOX6CP1-0.542.09COL18A1-0.542.09CXCL8-0.502.09Neutrophil_Chemotaxis_NatureMed_20188-0.482.09S100A8-0.462.09S100A9-0.432.09FOXP3-0.422.09CCL4L2-0.402.09CXCL1-0.382.09CD163-0.382.09ITGA6-0.382.09CXCL2-0.372.09IGF1R-0.372.09BCL2-0.362.09CD14-0.362.09IL2RA-0.362.09CXCL3-0.332.09FGFR1-0.332.09COX7CP2-0.332.09PIM2-0.332.09CCDC154-0.322.09CCR2-0.312.09CD27-0.312.09IL6R-0.312.09IL6_JAK_STAT3_Pathway_GSEASig-0.292.09
Presentation numberPS4-01-17
Accelerating Access to Precision Medicine in Nigeria through Patient Engagement and Educational Materials
Runcie CW Chidebe, Project PINK BLUE – Health & Psychological Trust Centre, Abuja, Nigeria
R. C. Chidebe1, O. Akapo1, S. C. Ipiankama1, O. Uzodinma2, S. Bornengo3, H. Harnik4, I. I. Okoye5; 1Patients Care and Research, Project PINK BLUE – Health & Psychological Trust Centre, Abuja, NIGERIA, 2From Testing To Targeted Treatments, The Synergist, Dijon, FRANCE, 3From Testing To Targeted Treatments, The Synergist, Belguim, BELGIUM, 4From Testing To Targeted Treatments, The Synergist, Brussels, BELGIUM, 5UNN Centre for Clinical Trials, University of Nigeria Teaching Hospital, Enugu State, NIGERIA.
Background Globally, precision medicine (PM) is revolutionizing the treatment of different cancers, including metastatic breast cancer (mBC). However, patient education on PM is largely unknown in Nigeria. The study explored the perspectives of cancer patients on the PM materials and their access to targeted therapies. Methods Participants were (N=147) breast cancer (BC) patients recruited from two cancer support groups in Nigeria. Using a patient engagement approach, From the Testing to Targeted Treatments co-created patient educational materials (PEMs) on “biomarker testing for cancer treatment,” “targeted therapy for cancer,” and “questions to ask your healthcare provider.” The PEMs were adapted and reviewed at Project PINK BLUE. Using a mixed-methods design, 76 of the participants completed an online survey on the usefulness of PEMS, and 71 participated in three focus group discussions on access to targeted therapies. Results The majority of the participants, 71.1% reported an increase in awareness of PM, 80.3% found the materials helpful for making informed decisions, and 90% rated the materials highly for clarity. Themes were: “financial barriers to targeted therapies,” “poor awareness of targeted therapies,” and “lack of access to PEMs.” The qualitative findings showed that some of the cancer patients were taking targeted therapies but were unaware of their treatment. A participant said, “We feel the cost is too high, and even when we have the money, it’s still not affordable.” Conclusion Our finding underscores the urgent need to accelerate access to PM for patients in low-resource settings.
Presentation numberPS4-01-18
Prognostic Role of CXCR6 and RANKL for Bone Metastasis in Early Breast Cancer
Hanbaek Yi, Seoul National University Hospital, Seoul, Korea, Republic of
H. Yi1, K. Lee1, C. Park1, D. Lee1, S. Cho1, J. Koh2, H. Ryu2, S. Im1; 1Department of Internal Medicine, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF, 2Department of Pathology, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF.
Background: Bone is the most common site of metastasis in breast cancer, and patients developing skeletal-related events show poor prognosis. Bone metastasis is a complex process involving interactions between many different molecules, which include receptors and proteins related to chemokine signaling, cell adhesion and migration, or bone metabolism regulation. We aimed to demonstrate the correlation between bone metastasis and expression of these molecules in breast cancer. Methods: Clinicopathologic parameters were collected from 1,440 stage I-III breast cancer patients (1,436 female and 44 male, median age 50 years, range 25-90) who underwent curative surgery at Seoul National University Hospital from October 2000 to December 2013. Immunohistochemistry (IHC) for CXCR4, CXCR6, MGP, osteocalcin, periostin, PTH1R, PTHLH and RANKL were performed on tissue microarray (TMA) constructed from the resected breast cancer tissue. The expression levels of each marker were assessed as H-scores, and we applied cut-off values of 0, 100, 200, and 300 to dichotomize into positive or negative. Results: There were 966 patients (67.1%) with hormone receptor (HR)-positive and HER2-negative breast cancer, 225 patients (15.6%) with HER2-positive breast cancer, and 249 patients (17.3%) with triple negative breast cancer (TNBC). As of February 2024, at median follow-up duration of 138 months, 196 patients (13.6%) had experienced recurrence. This included 65 patients with local recurrence and 131 patients with distant metastasis (49 patients had bone-only metastasis and 82 patients had visceral metastasis as well). There were 186 patients (12.9%) who died from any cause. CXCR6 H-scores were higher in HR-positive breast cancer (mean H-score 33.90) compared to HER2-positive breast cancer (mean H-score 15.58, p=0.0006) or TNBC (mean H-score 16.78, p=0.0056). CXCR6 positivity was significantly associated with longer overall survival (OS) (5-year survival rate (5YSR) 95.8% vs. 95.0%, p=0.049 for cutoff 0). In HR-positive breast cancer, CXCR6 positivity was also correlated with longer bone-specific relapse-free survival (RFS) (5YSR 98.0% vs. 96.8%, p=0.024 for cutoff 0). In contrast, CXCR6 positivity was correlated with shorter bone-specific RFS in HER2-positive breast cancer and TNBC across various cutoff values. RANKL positivity was correlated with shorter OS (5YSR 95.6% vs. 93.9%, p=0.049 for cutoff 200) and RFS (5YSR 92.8% vs. 88.5%, p=0.026 for cutoff 200). In contrast, bone-specific RFS prolonged as RANKL positivity increased (5YSR 97.4% vs. 96.8%, p=0.044 for cutoff 100). The positive correlation to bone-specific RFS was maintained in RANKL-positive, HR-positive breast cancer (5YSR 98.2% vs. 96.3%, p=0.018 for cutoff 100). However, in TNBC, the RANKL score was negatively correlated with RFS across various cutoff values. Periostin was significantly less expressed in TNBC (mean H-score 23.79) compared to other subtypes (mean H-score 48.37, p<0.001 for HR-positive breast cancer and mean H-score 49.14, p<0.001 for HER2-positive breast cancer). Periostin positivity was related to longer OS (5YSR 97.7% vs. 95.6%, p=0.03 for cutoff 0) and bone-specific RFS (5YSR 99.5% vs. 96.7%, p=0.014 for cutoff 100) in HR-positive breast cancer, but not in other subtypes. Conclusion: Expression levels of CXCR6, RANKL, and periostin were correlated with prognosis and bone-specific survival in breast cancer. Interestingly, the clinical impact of each marker differed with respect to each breast cancer subtypes, possibly suggesting a different underlying mechanism of bone metastasis in HR-positive breast cancer compared to the others. Further investigations into the differences between subtypes and establishing proper cutoff values are warranted.
Presentation numberPS4-01-19
Isoform level RNA detection provides more detailed profiling of HER2 expression in breast cancer
Jacob Bradley, Wobble Genomics Ltd, Edinburgh, United Kingdom
Y. Cheng1, J. Bradley1, K. Morris1, I. Ivanova1, M. Barnett1, A. Séguret1, O. Eve1, A. Zyoud1, G. Benitez1, A. Robinson1, A. Turnbull2, J. Dixon2, R. Hockett1, H. Chuang1, R. Kuo1; 1Wobble Genomics, Wobble Genomics Ltd, Edinburgh, UNITED KINGDOM, 2Edinburgh Cancer Research Centre, MRC Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UNITED KINGDOM.
Background: Accurate quantification of tumor HER2 expression is essential for guiding breast cancer therapy selection, as HER2 status directly determines treatment decisions and eligibility for targeted therapies. Current HER2 testing methods, primarily based on IHC and ISH, often lack the precision to confidently distinguish certain HER2 statuses, particularly in borderline cases, which can lead to under- or overtreatment. Developing a more accurate and reliable HER2 quantification method will enable safer and more effective treatment choices, ensuring patients receive the most appropriate therapy while minimizing unnecessary exposure to side effects. Ultimately, this will lead to better clinical outcomes and improved quality of care for breast cancer patients. Methods: We have developed a novel platform that leverages long-read sequencing to capture full-length mRNA and generate isoform-level expression profiles. By integrating tumor and blood transcriptome data from breast cancer patients, we characterized the expression of HER2 and 728 other cancer-related genes (based on COSMIC Cancer Gene Census and established ADC targets) to identify patterns that can be used to distinguish patients with different HER2 statuses. Patients: Twelve tumor biopsies and matched blood samples were collected from nine breast cancer patients with HER2 IHC scores of 0 or 1+ (ongoing study; numbers reported as of abstract submission). In addition, blood transcriptome profiles were analyzed from a separate cohort of 95 control patients with benign breast condition or no cancer on imaging. Results: In the 12 tumor samples and 729 genes analyzed, 29,804 non-reference transcript isoforms which had not been previously reported in the reference human genome annotations were identified in multiple patients. Of these, 16,533 (55.5%) were not found in any of 95 control samples, representing novel isoforms potentially unique to breast cancer tumors. For 551 genes, we identified non-reference isoforms shared between tumor and matched blood samples, including in HER2. Comparative analysis of tumor samples with different HER2 statuses (IHC 1+ vs. IHC 0) revealed significant differences in HER2 expression aligned with the IHC scores. The IHC 1+ group exhibited not only higher expression levels (median 79.5 vs. 37.5 parts per million; p = 0.015), but also greater transcript isoform diversity (median 38 vs. 9 transcripts per sample; p = 0.015) at the HER2 locus. On average, 23.3% of HER2 isoforms identified in HER2 IHC 1+ patients contained novel combinations of known splice donor/acceptor sites, as opposed to 11.5% in HER2 IHC 0 patients. Notably, some of these alternative splicing patterns were also detected in matched blood samples, offering valuable insight that could facilitate non-invasive diagnosis of patient HER2 status from blood. Conclusions: Our novel approach utilizing full-length RNA sequencing enables more comprehensive characterization and precise quantification of tumor-associated expression of HER2 and other cancer-related genes, providing new insights into transcript diversity and expression patterns that are critical for improving diagnostic accuracy, refining treatment selection, and ultimately enhancing patient outcomes.
Presentation numberPS4-01-20
Clinico-pathological and genomic features of HER2-low early Breast Cancer (eBC). Results of retrospective analysis of seven adjuvant trials by the Hellenic Cooperative Oncology Group (HeCOG)
Sotirios Lakis, Hellenic Cooperative Oncology Group (HeCOG), Athens, Greece
S. Lakis1, E. Tsolaki1, N. Korfiatis1, A. Goussia1, H. P. Kourea1, A. Batistatou1, M. Bobos1, K. Papadopoulou2, A. Charchanti1, M. Bai1, O. Tzaida1, K. Petraki1, P. Arapantoni1, T. Koletsa1, D. Pectasides1, A. Koutras1, D. Kalapanida1, F. Dimitrakopoulos1, E. Aravantinou-Fatorou1, N. Spathas1, A. Psyrri1, H. Gogas1, F. Zagouri1, G. Fountzilas1; 1Data Office, Hellenic Cooperative Oncology Group (HeCOG), Athens, GREECE, 2Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Thessaloniki, GREECE.
Background: HER2-low breast cancers are a mixture of luminal and triple negative tumors. Precise molecular characterization can help refine treatment strategies with novel antibody-drug conjugates and improve outcomes in this patient subgroup. Methods: We used central ER/PgR (HR) and HER2/ERBB2 IHC/FISH assessment to define HER2-low (IHC 1+ or 2+/FISH non-amplified), HER2-zero (IHC 0) and Luminal A/ Luminal B/ TNBC subtypes across 7 prospective adjuvant studies in which eBC patients were treated with dose dense sequential chemotherapy. Retrospective analysis of clinico-pathological data and IHC markers from tissue microarray cores was performed on N=2,751 cases, whereas DNA next generation sequencing (NGS) data were available in a subset of N=1,120 tumors. Heterogeneity was investigated by comparing HER2-low concordant (HER2-low_c) cases, when all cores were scored consistently as HER2-low (>0) and HER2-low discordant (HER2-low_d) cases, when at least one core was scored “0”. Results: First, HER2-low tumors were more likely to be HR + (p<0.001). Second, within HR + disease only, the proportion of Luminal B was higher in HER2-low (51%) than HER2-zero (41.6%), whereas Luminal A showed the opposite pattern (p< 0.001); PIK3CA mutations on the other hand were more common in HER2-zero (33.6% vs. 24.7%, p=0.0036). Third, a statistically significant progressive increase in ERBB2 average copies was observed from HER2-zero to HER2-low_d and HER2-low_c irrespective of HR status (p<0.001). Finally, HER2 low was not associated with survival in either HR + or HR – disease. Conclusion: Our data suggests that HR + HER2-low and HER2-zero eBC differ with respect to the status of the estrogen receptor pathway. When accounting for inter-core variability, differences in HER2 IHC levels correlate with the underlying ERBB2 copy number; however, standard HER2 IHC scoring thresholds did not influence prognosis in this cohort.
Presentation numberPS4-01-21
Genomic characterization and genetic testing gaps in early-onset breast cancer in Colombia
Maria Alejandra Bravo, Fundación Centro de Tratamiento e Investigación sobre Cáncer(CTIC), Bogota, Colombia
M. A. Bravo, D. C. Sotelo-Rodríguez, J. Caicedo, S. Cervera, A. Ruiz-Patiño, W. A. Mantilla, S. X. Franco; Breast Cancer, Fundación Centro de Tratamiento e Investigación sobre Cáncer(CTIC), Bogota, COLOMBIA.
Abstract San Antonio Breast Cancer Symposium Title: Genomic Characterization and Genetic Testing Gaps in Early-Onset Breast Cancer in Colombia Background:Breast cancer (BC) in women under 50 years represents a biologically aggressive disease with distinct genomic features and higher hereditary burden. Latin American data on this subgroup remain limited. We aimed to describe the clinical and molecular characteristics of young BC patients in a comprehensive cancer center in Colombia, and to evaluate the yield of genetic testing. Methods:A retrospective cohort of 113 women <50 years diagnosed with BC at the Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC) between 2019 and 2024 was analyzed. Demographic, clinical, histopathologic, imaging, and molecular data were extracted. Tumor subtypes were defined based on ER, PR, and HER2 status. Germline sequencing was performed in 95% of cases (n=108). Chi-squared and t-tests were used to assess associations between mutation status, age, and clinical variables. Results:Among 108 women diagnosed with breast cancer at ≤50 years of age who underwent comprehensive germline genetic testing, 76 patients (70.4%) had complete immunohistochemical data, enabling subtype classification. The most common subtype was Luminal (n=36, 47.4%), followed by triple-negative (n=18, 23.7%) and HER2-enriched tumors (n=8, 10.5%). The luminal group showed a mutation frequency of 13.9% (5/36), while HER2-enriched tumors exhibited a rate of 12.5% (1/8). BRCA1 was the most frequent germline mutation in triple-negative tumors, identified in 18.2% (2/11) of cases. In contrast, Luminal and HER2-enriched subtypes showed a predominance of non-canonical mutations, including genes such as MUTYH, POLE, and APC, detected in 40.9% (9/22) and 14.3% (2/14) of cases, respectively. When considering all germline findings, 47 out of 108 patients (43.5%) harbored at least one germline alteration. The most frequently altered genes across the cohort were BRCA1 (7.4%), PALB2 (5.6%), BRCA2 (3.7%), and CHEK2 (1.9%). Additionally, 24.1% had variants in non-canonical genes, including both pathogenic and uncertain significance variants. Patients carrying germline mutations were diagnosed at a significantly younger age (mean 34.9 ± 6.1 years) compared to non-carriers (mean 41.8 ± 5.4 years; p < 0.001), emphasizing the contribution of hereditary predisposition to early-onset breast cancer and supporting the implementation of broad germline testing in all young patients, irrespective of tumor subtype. Conclusions:Germline pathogenic variants are frequent in early-onset breast cancer, particularly in triple-negative and Luminal subtypes. BRCA1 predominates in triple-negative cases, while PALB2 and CHEK2 are found in hormone receptor–positive tumors. Additionally, 24.1% of patients carried alterations in non-canonical genes, reflecting marked genomic heterogeneity. Mutation carriers were diagnosed at a significantly younger age, supporting early and comprehensive genetic testing in all young patients to guide personalized care and familial risk assessment.
Presentation numberPS4-01-22
The Utility of MT1-MMP in Predicting Treatment Efficacy with HER2-positive advanced or metastatic breast cancer: JBCRG-M06/EMERALD trial TR
Mio Yasukawa, Kanagawa Cancer Center, Kanagawa, Japan
M. Yasukawa1, S. Sato2, N. Saito3, S. Saji4, N. Masuda5, T. Yamanaka1, S. Fujiwara1, K. Goda1, N. Mori6, T. Takano7, K. Watanabe8, Y. Naito9, H. Tada10, M. Kitada11, T. Iwasa12, T. Morimoto13, A. Takahashi1, M. Isoda1, T. Yamashita1, D. Hoshino3; 1Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Kanagawa, JAPAN, 2Morphological Information Analysis Laboratory, Kanagawa Cancer Center Research Institute, Kanagawa, JAPAN, 3Cancer Biology Division, Kanagawa Cancer Center Research Institute, Kanagawa, JAPAN, 4Department of Medical Oncology, Fukushima Medical University, Fukushima, JAPAN, 5Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, JAPAN, 6Department of Breast Surgery, Seirei Hamamatsu General Hospital, Shizuoka, JAPAN, 7Department of Breast Medical Oncology, The Cancer Institute Hospital of JFCR, Tokyo, JAPAN, 8Department of Breast Oncology, National Hospital Organization Hokkaido Cancer Center, Hokkaido, JAPAN, 9Department of Breast and Medical Oncolog, National Cancer Center Hospital East, Chiba, JAPAN, 10Department of Surgery, Division of Breast and Endocrine Surgery, Tohoku University Hospital, Miyagi, JAPAN, 11Department of Breast Disease Center, Asahikawa Medical University Hospital, Hokkaido, JAPAN, 12Department of Medical Oncology, Kindai University Hospital, Osaka, JAPAN, 13Department of Breast Surgery, Yao Municipal Hospital, Osaka, JAPAN.
Background: Malignant tumor cells grow invasively and form distant metastases after moving through multiple tissue barriers. Invasion requires cell locomotion, accompanied by the degradation of the extracellular matrix (ECM) by matrix metalloproteinases (MMPs). MT1-MMP (Membrane Type 1 MMP) is an integral membrane protease that degrades a variety of protein components within the extracellular milieu. The substrates of MT1-MMP include a variety of components of the ECM membrane proteins, such as type 1 collagen. Type 1 Collagen is a physical varier not only affecting cancer cell invasion but also drug accessibility to cancer cells. It is reported that MT1-MMP regulates small compound drug resistances via activation of integrin signaling through type 1 collagen cleavage and higher expression of MT1-MMP was considered a marker of increased malignancy and poorer prognosis. However, it remains unclear whether larger drugs, such as antibodies, can be affected by MT1-MMP-related collagen remodeling. Here, we conducted a retrospective analysis of MT1-MMP on treatment effectiveness and survival using the JBCRG-M06/EMERALD clinical trial. The clinical trial is a Phase III study comparing two first-line chemotherapeutic treatments for HER2-positive local advanced or metastatic breast cancer: trastuzumab, pertuzumab, and taxane therapy versus trastuzumab, pertuzumab, and eribulin mesylate therapy. Method: In the EMERALD study, 231 specimens were collected from 231 patients who consented to biomarker research at the time of enrollment. Among these 182 specimens were collected from primary tumors, and 49 from metastatic sites. The tissue samples were immunostained with anti-MT1-MMP antibody and classified into low (score1), moderate (score2), and high (score3) expression based on staining proportion and intensity. We investigated the correlation between MT1-MMP expression and the therapeutic effects of trastuzumab and pertuzumab combination with eribulin mesylate or taxane, as well as Progression-Free Survival (PFS). Results: Of the 231 patients in this study, 123 were assigned to the eribulin group and 108 to the taxane group, with MT1-MMP scores 1/2/3 of 28/52/43 in the eribulin group and 26/43/39 in the taxane group, respectively. The median PFS in the eribulin group was 14.7 months and 13.4 months in the taxane group, and it was similar to the results for all patients in the EMERALD study. There was no difference between the eribulin and taxane groups. Whereas the median PFS for MT1-MMP scores 1/2/3 was 10.5/13.2/18.4 months, and it tended to be longer with higher MT1-MMP scores (p=0.0306). Although higher MT1-MMP scores tended to improve response rates overall, there was no significant difference between the two treatment groups. In this study of patients with HER2-positive local advanced or metastatic breast cancer patients, higher expression of MT1-MMP may be with better PFS and response rates. Conclusion: The MT1-MMP score is unable to distinguish the combined trastuzumab and pertuzumab with eribulin mesylate or trastuzumab and pertuzumab with taxane therapy. However, it may be a prognostic factor for HER2-positive local advanced or metastatic breast cancer patients treated with the trastuzumab and pertuzumab combination and eribulin mesylate or taxane.
Presentation numberPS4-01-23
Transcriptomic Profiling and Immune-metabolic Prediction Model Construction for Neoadjuvant Chemotherapy Response in Triple-Negative Breast Cancer Patients
Shuning Ding, Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, Shanghai, China
S. Ding1, J. Wu2, D. Liu1, Y. Fang1, W. Wang1, L. Zhu1; 1Department of Breast and Thyroid Surgery, Shanghai General Hospital,Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA, 2Comprehensive Breast Health Center, Ruijin Hospital,Shanghai Jiao Tong University School of Medicine, Shanghai, CHINA.
BackgroundTriple-negative breast cancer (TNBC) had great tumor heterogeneity and lacks effective therapeutic targets. The addition of immunotherapy to neoadjuvant chemotherapy (NAC) has dramatically improved pathologic complete response rates (pCR) in TNBC, but some patients may be overtreated as they could achieve pCR with NAC only. Thus, identifying the molecular features associated with efficacy of NAC and finding robust biomarkers to select patients who are sensitive to chemotherapy was crucial.Patients and MethodsRNA sequencing analysis was performed on paired tumor samples collected before and after neoadjuvant treatment with nab-paclitaxel and carboplatin from 44 TNBC participants enrolled in the prospective NeoPATH clinical trial (ClinicalTrial.gov identifier: NCT0390780). Transcriptomic features were compared in patients with different treatment responses. Changes in molecular biology after treatment were illustrated in matched pre- treatment and post-treatment samples as well. Univariate regression and lasso regression were utilized to construct the predictive signature for NAC. ResultsThe results showed that patients achieving pCR had upregulated immune-related signatures suggesting more active immune response and decreased metabolic signatures indicating less energy production. Increases in both anti-tumor immune cell infiltration and immune molecular expression were observed after neoadjuvant chemotherapy, inferring the activation of immune microenvironment. Multiple immune-related signatures were found to be positively associated with treatment response, while metabolic signatures were negatively associated with the response. Based on these findings, the immune-metabolic predictive signatures were further built and validated to predict the treatment response of neoadjuvant nab-paclitaxel and carboplatin for TNBC (AUC, 0.719 in the train set, 0.787 and 0.717 in the validation sets).ConclusionsOverall, our study delineates distinct molecular profiles between pCR and non-pCR TNBC patients following neoadjuvant nab-paclitaxel/carboplatin. The developed immune-metabolic signature effectively identifies treatment-sensitive tumors, while observed transcriptomic changes post-therapy may inform future targeted drug development.
Presentation numberPS4-01-24
Cytokine score based on expression levels of multiple cytokines is a prognostic indicator in metastatic breast cancer patients treated with chemotherapy
Junko Tsuchida, Hyogo Medical University, Nishinomiya, Hyogo, Japan
J. Tsuchida, M. Nagahashi, M. Komatsu, S. Urano, M. Kuroiwa, S. Yoshida, G. Sugimoto, Y. Togashi, A. Mitsuyoshi, H. Kanaoka, A. Hattori, T. Higuchi, A. Nishimukai, K. Murase, M. Shimoda, Y. Miyoshi; Division of Breast and Endocrine Surgery, Department of Surgery, Hyogo Medical University, Nishinomiya, Hyogo, JAPAN.
Background: The tumor immune microenvironment affects treatment response and outcomes in patients with metastatic breast cancer (MBC). Cytokines, such as tumor necrosis factor-α (TNF-α) and interleukin (IL)-6, play an important role in tumor progression and immune regulation, and they are associated with outcomes in patients with MBC. Although the significance of each cytokine as a prognostic factor has been reported, there is a paucity of reports on the relationship between the integrated indicator of multiple cytokines and outcomes. In this study, we established an integrated indicator of multiple cytokines and examined the relationship with patient outcomes. In addition, since inflammatory cytokines are thought to affect cancer progression and metastasis by acting on T cells, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs), we also examined the relationship between the integrated cytokine indicator and immune cell profile. Materials and Methods: This retrospective study included 43 patients with MBC treated with chemotherapy. Baseline values for cytokine levels, including TNF-α, soluble interleukin-2 receptor (sIL-2R), IL-6, IL-8, transforming growth factor (TGF)-β, and thymidine kinase 1 (TK1), were measured in peripheral blood on the day before the first treatment with chemotherapy. Each cytokine was classified into low and high groups using cutoff values defined by ROC curves for overall survival (OS), and the progression-free survival (PFS) and OS between low and high groups were compared by Kaplan-Meier plots. CD4+ and CD8+ lymphocytes, MDSCs, and Tregs levels were determined in the blood by flow cytometry analysis. Results: Of the patients, 46.5% (n = 20) were treated with eribulin, 9.3% (n = 4) with trastuzumab deruxtecan, 9.3% (n = 4) with paclitaxel and bevacizumab, 7.0% (n = 3) with trastuzumab emtansine, and 27.9% (n = 12) with other chemotherapeutic agents. Patients treated as first-line were 55.8% (n = 24), and those treated as second-line or later were 44.2% (n = 19). Patients with high levels of TNF-α, sIL-2R, IL-6, and TK1 showed a shorter OS compared to those with low levels (P = 0.01, 0.01, 0.01, and <0.01, respectively). IL-8 and TGF-β were not associated with OS (P = 0.07 and 0.06, respectively). Patients with high levels of sIL-2R, TGF-β, and TK1 showed a shorter PFS compared to those with low levels (P = 0.02, 0.01, and 0.04, respectively). TNF-α, IL-6, IL-8 and TK1 levels were not associated with PFS (P = 0.11, 0.10, 0.07 and 0.04, respectively). A “cytokine score” was developed based on the number of elevated cytokines among TNF-α, sIL-2R, IL-6, and TK1. Patients with 0-1 elevated cytokine were classified as low score, and those with 2-4 as high score. The high cytokine score group had shorter OS and PFS compared to the low score group (P < 0.01 and P = 0.03, respectively). Multivariable analyses revealed that cytokine score were an independent prognostic factor for both OS (P < 0.01) and PFS (P = 0.02). Furthermore, patients with high cytokine scores had lower proportions of CD4+ and CD8+ lymphocytes (P = 0.01 and 0.04, respectively), and higher levels of MDSCs (P < 0.01), with no difference in Tregs (P = 0.30). Conclusion: Our results show that the levels of TNF-α, sIL-2R, IL-6, and TK1 were associated with outcomes in patients with MBC treated with chemotherapy. The cytokine score, based on the number of elevated cytokines, is an important prognostic indicator for both OS and PFS. Furthermore, since a high cytokine score was associated with higher levels of MDSCs, and lower levels of CD4+ and CD8+ lymphocytes, the cytokine score might be associated with patient outcomes through immune reaction. Patients with high cytokine scores have worse outcomes, which may reflect a poor tumor microenvironment.
Presentation numberPS4-01-25
Noninvasive Early Detection of Breast Cancer via Copy Number Variation Profiling of Nipple Discharge DNA
Wen Wen, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
W. Wen1, P. Pu1, H. Xu1, T. Shang1, L. Cong1, G. Qiao2, Z. Jia1, Y. Liu1, Y. Huang1, R. Zhou1, Y. Li2, J. Liu1; 1Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 2Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, CHINA.
Background: Nipple discharge is a common clinical manifestation of breast disease, yet current diagnostic tools such as ultrasound, mammography, MRI, ductography, and cytology have limited accuracy in early cancer detection. Copy number variation (CNV) profiling of DNA from body fluids has shown promise in early diagnosis of cancers like bladder and cervical, but its application to nipple discharge remains unexplored. This study aimed to establish a noninvasive, CNV-based diagnostic approach that reflects primary breast tumor biology with high specificity and sensitivity. Methods: We analyzed CNV data from 1,086 tumor and 942 normal samples in the TCGA-BRCA cohort using GISTIC2.0, identifying recurrent CNV regions with high frequency and statistical significance (q50%, biological interpretability (oncogenes in amplifications, tumor suppressors in deletions), consistency with gene expression changes, and prognostic relevance. A final panel of 20 CNV features (12 amplifications, 8 deletions) was established and validated across multiple breast cancer cohorts and their matched normal tissues, when available. Diagnostic performance was further tested using nipple discharge DNA an in-house cohort of patients with benign and malignant breast disease. Additionally, single-cell RNA sequencing was performed on tumor tissues from five breast cancer patients in the cohort. Using inferCNV, the CNV features detected in nipple discharge were traced back to distinct epithelial subclusters using inferCNV, and the functional characteristics of the dominant CNV-contributing subpopulations were further analyzed. Results: In the TCGA-BRCA dataset, only 8 of 118 adjacent normal tissues were panel-positive, yielding a specificity of 93.22%. External validation demonstrated consistent diagnostic performance: GSE87048 (sensitivity 77.0% in tumor tissue, specificity 99.0% in matched peripheral blood mononuclear cells), GSE118527 (sensitivity 92.8%, specificity 100%), METABRIC (sensitivity 95.7%), and GSE31427 (Stage I-II; sensitivity 66.9%). In the MSK-CHORD PanCancer cohort, the panel showed the highest detection rate in breast cancer (78.3%) compared to colorectal, prostate, lung, and pancreatic cancers, supporting tissue specificity. In nipple discharge samples (n = 39 malignant, n = 18 benign), the panel achieved a sensitivity of 71.79% and a specificity of 94.44%, highlighting its potential for accurate, noninvasive early detection. In five patients from the in-house cohort, single-cell RNA sequencing of tumor tissues revealed that CNV features detected in nipple discharge originated from specific epithelial subclusters. These CNV-contributing subpopulations exhibited enrichment in pathways related to cell junction assembly and tissue morphogenesis. Conclusion: This study provides the first evidence that CNV profiling of nipple discharge DNA is a feasible and tumor-specific approach for early breast cancer detection. The selected CNV panel demonstrates robust diagnostic performance across independent datasets and early-stage tumors, supporting its clinical utility as a novel, minimally invasive screening tool.
Presentation numberPS4-01-26
Development and Validation of TROP2/HER2-low/Ki67/HR multiplex-immunofluorescence panel with membrane identification: enabling improved Antibody Drug Conjugate therapy selection
Yuting Liu, Akoya Biosciences, Marlborough, MA
Y. Liu, M. Parrella, S. Schwaegerle, S. Berry, C. Betts, M. Landers; Advanced Biopharma Solutions, Akoya Biosciences, Marlborough, MA.
Background Antibody-drug conjugates (ADCs) have emerged as a promising therapeutic class for solid tumors, especially in breast cancer (BC), with a focus on agents targeting HER2 and Trop2. Currently, two ADC drugs targeting HER2: Trastuzumab emtansine (T-DM1) and fam-trastuzumab deruxtecan-nxki (T-DXd), and two targeting Trop2: Sacituzumab govitecan (SG) and datopotomab deruxtecan-dlnk (Dato-DXd) have gained approval by the FDA in treating metastatic BC, with additional ADCs in development. Despite the advances, challenges remain in patient selection including identifying the HER2 “ultra-low” patient subgroup, characterizing HER2/Trop2 co-expression, and ensuring reliable evaluation of biomarkers on the cell membrane surface. Here, we present the fully validated Opal™ multiplex-immunofluorescence (mIF) BC-ADC panel, leveraging the IVD-grade Akoya PhenoImager HT imaging system. The BC-ADC panel provides simultaneous assessment of BC standard-of-care biomarkers with Ki67 and hormone receptor (HR), ADC-specific targets HER2 and TROP2, together with a proprietary membrane marker cocktail facilitating specific biomarker membrane evaluation. Methods A proprietary membrane cocktail, comprised of multiple membrane-specific biomarkers, was developed to specifically label and visualize cell membranes. The membrane cocktail performance supporting accurate cell definition and subcellular localization was assessed against ground truth boundaries created by board-certified pathologists. Biomarkers TROP2, HER2, Ki67, ER, PR and the membrane cocktail, plus DAPI were tested for staining order effects and spectral unmixing, then developed into an single slide Opal™ 6-color multiplex immunofluorescence panel. HER2-positive/HER2-negative breast cancer tissue was utilized for assay development and verification. Board-certified pathologists evaluated panel validation according to the pre-set acceptance criteria including: absence of spectral crosstalk or umbrella effect by antibody drop-test; assay robustness by inter/intra-day triplicate precision studies, and biomarker sensitivity and specificity confirmed by equivalency between “gold-standard” IHC and mIF. HER2-negative breast cancer samples utilized for testing contained samples spanning HercepTest score 0+ (null or ultra-low), score 1+ (low), and score 2+ (low). Assay HER2 signal sensitivity was evaluated by pathologists to differentiate HER2-negative scores qualitatively. Results The membrane cocktail consistently delineates cell membrane in both tumor and stromal compartments. The finalized mIF assay met the predefined validation criteria as assessed by board-certified pathologists. Antibody drop-testing revealed no umbrella effect or spectral crosstalk was observed between channels. The precision study demonstrated high reproducibility in inter-day comparison and intra-day comparison. Strong equivalency of each biomarker was observed between IHC and the mIF panel. Pathologist review confirmed the high sensitivity in differentiating HER2 expression levels across HER2-negative cases: null/ultra-low/low. Conclusion The validated 5-plex/6-color BC-ADC mIF panel demonstrated robust analytical performance: high sensitivity and specificity across all biomarkers, especially in HER2-negative samples, stratifying HER2 low and ultra-low subgroups. The mIF panel showed reliable isolation of markers and robust assay performance. Results also suggested that the custom membrane cocktail can enhance the subcellular evaluation of biomarkers. The study will be supplemented by an independent patient verification cohort to support forthcoming clinical-utility studies.
Presentation numberPS4-01-27
Lower Penetrance Germline Fanconi Anemia Variants in Breast Cancer: Opportunities for Expanded Genetic Testing and Therapeutic Targeting in the Updated INSPIRE Study Cohort
Leah Naghi, City of Hope, Duarte, CA
L. Naghi, K. G. Roth, S. Lindsey, A. Seuylemezian, J. Bonner, L. Kruper, S. B. Gruber, J. Mortimer, K. McDonnell; Medical Oncology, City of Hope, Duarte, CA.
Purpose: To characterize the prevalence of germline pathogenic or likely pathogenic (PLP) variants in Fanconi Anemia (FA) pathway genes among patients with invasive breast cancer (IBC) and evaluate opportunities for clinical actionability through germline and somatic analysis. Background: Disruption of the FA DNA repair pathway can impair homologous recombination (HR). While high-penetrance PLP variants in BRCA1, BRCA2, and PALB2 are established breast cancer susceptibility genes and contribute to HR deficiency (HRD), the clinical consequences of other FA germline PLP variants remain unclear. Using the updated cohort from the City of Hope INSPIRE Study, we assessed the prevalence and functional impact of germline PLP variants in 17 FA pathway genes. HRD, mutational signatures, and loss of heterozygosity were used to evaluate effects on HR integrity. These findings may have implications for HRD-related precision therapeutic intervention and clinical trial eligibility. Methods: This retrospective study included consented patients with histologically confirmed IBC enrolled in the INSPIRE Study as of January 14, 2025. Germline sequencing of 155 cancer predisposition genes was performed; patients with multiple FA variants were classified according to the FA gene of the highest penetrance. A cohort subset underwent paired tumor-normal whole exome sequencing (>400x tumor, >180x normal), including HRD scoring (scarHRD), LOH determination, and SBS3 assessment (SigProfiler). Clinical data were obtained from cancer registries and medical records. Clinical trial eligibility criteria were evaluated using ClinicalTrials.gov. Results: Of 8,242 patients with IBC, 7,982 underwent germline sequencing; 592 (7.4%) had a PLP variant in an FA gene, including 197 (33.3%) in non-high-penetrance (non-HP) genes. Among 1,833 patients with paired tumor-normal sequencing, 145 (7.9%) had an FA PLP variant, of which 53 (2.89%) were in non-HP genes. HRD scores were ≥42 in 45.3% of non-HP carriers (Table 1). 24 of 53 patients (45.3%) with a non-HP FA germline PLP variant had HRD-positive (≥42) tumors. HRD-positivity is more likely among patients with germline PLP variants in non-HP FA pathway genes compared to those without a variant in any FA gene (RR 1.51, 95% CI 1.12 to 2.05; p < 0.0077). Stage data were available for 1,548 patients; 128 (8.3%) were stage IV at diagnosis, including 7/128 (5.5%) with non-HP FA gene PLP variants. Conclusion: Germline PLPs in non-HP FA genes are present in a meaningful subset of patients with IBC and may be functionally relevant. These findings support the value of expanded germline testing and trial eligibility criteria that encompass the broader FA pathway and suggest that non-HP FA gene variants may serve as biomarkers for identifying patients likely to benefit from HRD-targeted therapies.
| Gene | Pts with germline PLPs n (% of total, N=1,833) | HRD score ≥ 42 |
| Highly Penetrant BC Predisposition FA Genes | ||
| BRCA2 (FANCD1) | 47 (2.56%) | 39 (82.9%) |
| BRCA1 (FANCS) | 30 (1.64%) | 26 (86.7%) |
| PALB2 (FANCN) | 15 (0.82%) | 13 (86.7%) |
| Other FA Genes | ||
| FANCA | 14 (0.76%) | 4 (28.6%) |
| FANCM | 13 (0.71%) | 6 (46.2%) |
| BRIP1 (FANCJ) | 7 (0.38%) | 5 (71.4%) |
| FANCC | 3 (0.16%) | 2 (66.7%) |
| FANCL | 3 (0.16%) | 2 (66.7%) |
| SLX4 (FANCP) | 3 (0.16%) | 0 (0.0%) |
| ERCC4 (FANCQ) | 2 (0.11%) | 1 (50.0%) |
| FANCD2 | 2 (0.11%) | 0 (0.0%) |
| FANCG | 2 (0.11%) | 2 (100.0%) |
| FANCI | 2 (0.11%) | 1 (50.0%) |
| FANCE | 1 (0.05%) | 0 (0.0%) |
| FANCF | 1 (0.05%) | 1 (100.0%) |
| FANCB | 0 (0.00%) | NA |
| RAD51C (FANCO) | 0 (0.00%) | NA |
Presentation numberPS4-01-28
Utility of MSK-ACCESS powered with SOPHiA DDM decentralized testing in patients with breast cancer.
Florian Klemm, SOPHiA GENETICS, Rolle, Switzerland
F. Klemm1, F. Mohammad2, A. C. Tuck1, X. Xing1, G. Morales2, A. Willig1, E. Smith1, M. Basgall1, A. Francois1, L. Canetti3, E. Jorge3, Z. Xu1, F. Penault Llorca4, M. Ferreira3; 1Data Science, SOPHiA GENETICS, Rolle, SWITZERLAND, 2Data Science, SOPHiA GENETICS, Boston, MA, 3Pathology Unit, OncoGèn Auvergne, Centre Jean Perrin, Clermont-Ferrand, FRANCE, 4Department of Pathology and Biopathology, Centre Jean Perrin, UMR INSERM 1240, University Clermont Auvergne, Clermont-Ferrand, FRANCE.
Background: Liquid biopsy (LBx) enables non-invasive molecular profiling and monitoring of cancer but has been impeded by operational, computational and financial barriers. While centralized LBx testing is widely used in clinical trials and specialized centers, locally or regionally performed decentralized testing has the potential to broaden access to molecular profiling and reduce turnaround times. We evaluated the analytical performance and real-world utility of the decentralized MSK-ACCESS® powered with SOPHiA DDM™ LBx assay in patients with breast cancer. Methods: Analytical characteristics were assessed by comparing results from the decentralized LBx assay against a reference orthogonal method in two cohorts: breast cancer (n=17) and prostate cancer patients (n=55). Metrics included the mean molecular coverage, number of variants per sample and mean variant allele frequency (VAF) for single nucleotide variants (SNVs) and insertions/deletions (indels). Similar analytical performance across both cohorts was used to support assay validity. Subsequently, mutational profiles were characterized in the breast cancer cohort, with a focus on clinically relevant alterations in ESR1 and PIK3CA. These findings were extended to a larger real-world dataset from the SOPHiA GENETICS user base. Results: The MSK-ACCESS® powered with SOPHiA DDM™ assay demonstrated comparable performance across both breast and prostate cancer cohorts, with similar mean duplex molecular coverage (nbreast=1791, nprostate=1638), median somatic variant counts (nbreast=3, nprostate=2.5), and median somatic VAFs (VAFbreast=0.6%, VAFprostate=0.6%). In breast cancer samples, recurrent ESR1 mutations (e.g. E380Q, Y537S and D538G) and hotspot PIK3CA mutations (e.g. H1047R) mediating resistance were observed down to 0.06% VAF. These variants were recapitulated in the larger real-world breast cancer cohort (n=219; ESR1mut VAFmin=0.11%, PIK3CAmut VAFmin=0.14%) confirming the solution’s reproducibility and robustness in routine decentralized use. Conclusions: The decentralized LBx assay showed consistent analytical performance across tumor types and reliably detected clinically relevant variants in breast cancer. These findings demonstrate the feasibility and clinical utility of the decentralized MSK-ACCESS® powered with SOPHiA DDM™ testing, supporting its broader implementation in precision oncology.
Presentation numberPS4-01-29
Tumor-derived ZAG expression is associated with immunologically cold tumors and poor response to neoadjuvant chemotherapy in TNBC
Toru Hanamura, Tokai University School of Medicine, Isehara, Japan
T. Hanamura1, K. Yokoyama1, S. Takahashi1, H. Kiyohara1, S. Nakagawa1, R. Ishida1, M. Terao1, T. Okamura1, N. Kumaki1, A. Katayama2, S. Kurozumi3, N. Niikura1; 1Department of Breast Oncology, Tokai University School of Medicine, Isehara, JAPAN, 2Department of Pathology, Shizuoka Cancer Center, Shizuoka, JAPAN, 3Department of Breast Surgery, International University of Health and Welfare, Narita, JAPAN.
Purpose: An immunosuppressive tumor microenvironment (TME) contributes to therapeutic resistance in breast cancer. Immune checkpoint inhibitors (ICIs) combined with chemotherapy have emerged as a promising strategy to overcome this barrier. Understanding immune regulation within the TME is essential for developing effective biomarkers and therapies. Our previous work suggested an immunomodulatory role for ZAG (AZGP1), with both clinical and in vitro findings consistently indicating its immunosuppressive activity, potentially contributing to therapeutic resistance. This study evaluates the relationship between ZAG expression and response to neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC). Methods: To determine the cellular source of AZGP1, we reanalyzed a public TNBC single-cell RNA-seq dataset using Seurat v5.2.1. UMAP clustering was used to annotate cell types. Feature plots and violin plots were employed to visualize cell type-specific AZGP1 expression. Gene set enrichment analysis (GSEA) was performed using bulk transcriptomic data from four NAC-treated TNBC cohorts to explore pathways associated with pathological complete response (pCR) and AZGP1 expression. ROC analysis was used to assess the predictive value of AZGP1 for pCR and to determine optimal cut-off values. For validation, we analyzed NAC-treated TNBC cases at our institution. Pre-treatment core needle biopsy (CNB) samples were evaluated for tumor-infiltrating lymphocytes (TILs) based on International TILs Working Group criteria. ZAG expression was assessed by immunohistochemistry. The ZAG score was defined as the percentage of tumor cells showing moderate-to-strong staining. ROC analysis was also performed to evaluate the ZAG score’s predictive value for pCR (ypT0/isN0, ypT0N0), overall ypN0, and ypN0 in cN1 cases. Results: Single-cell RNA-seq analysis confirmed that AZGP1 expression was restricted to tumor epithelial cells. GSEA revealed that pCR was associated with enrichment of immune-related gene sets such as ALLOGRAFT_REJECTION and INTERFERON_GAMMA_RESPONSE, while AZGP1 expression negatively correlated with these pathways. In four public TNBC cohorts, AZGP1 showed modest predictive value for pCR (AUC 0.557-0.599; not significant), but high-AZGP1 groups had significantly lower pCR rates in three cohorts (p < 0.05). In our institutional cohort of 51 cases (94.1% stage II-III, mean age 57.2), 86.3% received anthracycline-taxane regimens. Pathological CR (ypT0/isN0) was achieved in 31.4% of cases. ZAG scores were significantly higher in tumors with low TILs (p < 0.05). High ZAG expression was associated with poorer response to NAC, particularly in terms of nodal clearance in cN1 cases (AUC = 0.748, p < 0.05). The ZAG-high group showed significantly lower rates of pCR (ypT0N0), overall ypN0, and ypN0 in cN1 cases (all p < 0.05). Conclusion: ZAG (AZGP1), shown to be tumor-derived via single-cell RNA-seq, acts as a paracrine immune modulator in TNBC, supporting its validity as a tumor-intrinsic marker in bulk transcriptomic analyses. High AZGP1 expression correlated with suppressed immune gene programs and reduced chemotherapy sensitivity. Our data confirmed that elevated ZAG expression predicts poor NAC response and reduced nodal clearance, particularly in cN1 cases. ZAG represents a promising tumor-intrinsic biomarker and potential immunotherapeutic target in TNBC.
Presentation numberPS4-01-30
Identification and characterization of distinct chemoimmunomodulatory states in breast cancer via unsupervised transcriptomic analysis of pre- and post-treatment specimens
Mohammed O. Gbadamosi, University of Florida, Gainesville, FL
M. O. Gbadamosi, I. Lopes de Lima, K. H. Streeks, E. Molchan, M. S. Makarem, K. L. Coleman; Department of Pharmacotherapy and Translational Research, University of Florida, Gainesville, FL.
Background: PD-1 immune checkpoint inhibitors (ICIs) have reshaped cancer treatment paradigms. While PD-1 ICIs displays monotherapy efficacy in some tumor types, others, such as breast cancer (BC), require a synergistic combination of ICIs and chemotherapy for clinical benefit. Beyond cytotoxicity, chemotherapy synergize with ICIs via chemoimmunomodulation (CIM), which comprise the immunomodulatory signals elicited by chemotherapy. While many studies have examined molecular features influencing chemoresistance, few have focused on the molecular features that influence CIM, despite the overlap between CIM and the hallmarks of response to PD-1 ICIs. This paucity may be driven by inadequate means for phenotypically delineating CIM and presents a challenge to selection of the most synergistic agents for PD-1 ICIs for a given patient. Methods: We leveraged transcriptomic data from 20 pre-/post-treatment specimens obtained from BC patients (N = 2 reps/sample) treated with neoadjuvant chemotherapy, including anthracycline, cyclophosphamide, taxanes, and platinating agent, to identify CIM induction profiles. CIM induction was assessed using Δ log2(TPM+1) values for 377 genes obtained from gene sets and literature related to autophagy, lysosomes, cellular stress, and immune function. We developed a novel iterative k-means-based unsupervised clustering approach to identify discrete CIM induction states using CIM gene induction values. Overrepresentation analysis was used to identify transcriptomic programs induced in each CIM induction states. Differential gene expression (DEG) analysis and inferential statistics were used on matched pre-treatment specimens to evaluate baseline differences between states. The abundances of immune cell types were deconvoluted using CIBERSORT. Results: Our iterative unsupervised approached identified two discrete states: one with high (H-CIM; N = 10) and one with low CIM (L-CIM; N = 10) induction. Only one replicate from patients was separately clustered. Stability, robusticity, and the binary nature of the clusters were confirmed using standard cluster evaluation metrics. There were no significant differences in clinical characteristics or treatments between states. The H-CIM state had induction of antigen presentation, upregulation of T-cell populations, and phagocytosis transcriptomic programs while the L-CIM group was primarily characterized by the induction of MYC-governed reactive oxygen species (ROS) detoxification and proteostasis programs (FDR-adjusted P (adjP) < 0.05). Baseline DEG analysis identified upregulated immunosuppressive features such as S100A9 (Fold Change (FC) = 8.9; adjP = 0.04) and HP (FC = 21.2; adjP = 0.008) in the L-CIM group which was supported by a significantly higher baseline amount of M2 macrophages (P < 0.001). Proteostasis safeguards SEC61G (FC = 3.3; adjP = 0.04) and SEC31B (FC =2.6; adjP = 0.002) were also upregulated in the L-CIM state suggesting a predisposition for protection against cellular stress. Interestingly, there was no significant difference in baseline MYC expression between the L-CIM and H-CIM group (P = 0.34). Conclusion: Our unsupervised approach enabled classification of distinct CIM induction states using paired pre- and post-treatment transcriptomic analysis, allowing interrogation of CIM in a manner not captured via traditional chemoresistance studies alone. We uncover underappreciated molecular programs, like proteostasis and ROS detoxification, that may underlie blunted CIM induction in BC. Future work will focus on characterizing baseline molecular features that prompt drug-specific CIM induction states, with the goal of informing and guiding the personalization of chemoimmunotherapy in breast cancer.
Presentation numberPS4-02-01
Differential early response to neoadjuvant endocrine therapy in lobular versus non-lobular breast cancer on the Endocrine Optimization Pilot (EOP) of the I-SPY2 Trial
Rita Mukhtar, University of California, San Francisco, San Francisco, CA
R. Mukhtar1, P. Norwood2, A. Borowsky3, S. Alkhafaji4, K. Giridhar5, C. Vaklavas6, A. Elias7, M. Wei6, M. Goetz5, N. Onishi8, O. Olopade9, L. Van ‘t Veer10, L. Huppert11, W. F. Symmans12, L. Brown Swigart10, C. Wu1, C. Yau1, D. Yee13, M. Magbanua11, E. Price14, N. Hylton14, L. Esserman1, J. Chien11; 1Surgery, University of California, San Francisco, San Francisco, CA, 2Statistics, Quantum Leap Healthcare Collaborative, San Francisco, CA, 3Pathology, University of California, San Francisco, San Francisco, CA, 4Labaratory Medicine, University of California, San Francisco, San Francisco, CA, 5Oncology, Mayo Clinic, Rochester, MN, 6Medical Oncology, Huntsman Cancer Institute, Salt Lake City, UT, 7Medicine, University of Colorado Anschutz, Aurora, CO, 8Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 9Medicine, University of Chicago, Chicago, IL, 10Laboratory Medicine, University of California, San Francisco, San Francisco, CA, 11Medicine, University of California, San Francisco, San Francisco, CA, 12Anatomical Pathology, MD Anderson, Houston, TX, 13Medicine, University of Minnesota, Minneapolis, MN, 14Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA.
Background Invasive lobular carcinoma (ILC) differs from invasive ductal carcinoma (IDC) in its estrogen receptor (ER) signaling pathways, response to systemic therapy, and imaging features. We evaluated differences in early change in the proliferation marker Ki67 and features on magnetic resonance imaging (MRI) after novel endocrine based neoadjuvant therapy in lobular versus non-lobular cases on the I-SPY2 EOP Trial. Methods EOP is a prospective, randomized NET trial enriched for patients with ILC. Patients with stage II-III ER positive HER2 negative breast cancer with low molecular risk (Mammaprint low or Mammaprint Hi1 plus high Sensitivity to Endocrine therapy Index) were randomized to one of several endocrine therapy-based arms. Treatment arms include novel oral SERDS with or without CDK4/6 inhibitor, aromatase inhibitors, and SERMs with or without ovarian function suppression in pre-menopausal patients. Patients undergo repeat core needle biopsy with central Ki67 assessment after 3 weeks of treatment and serial breast MRI. Here, we compare endocrine therapy responsiveness as measured by change in Ki67 at 3 weeks and functional tumor volume (FTV) on pre-surgical MRI in lobular versus non-lobular cases. Results We evaluated 253 patients of whom 148 (58.5%) had non-lobular cancer (93.9% IDC), and 105 (41.5%) had lobular cancer (17.1% mixed ILC/IDC). Overall, mean age was 53.3 years, 63.8% of tumors were grade 2, and 86.5% were Mammaprint (MP) Low with the remaining being Hi1, with higher rates of MP Low in the lobular versus non-lobular cohorts (92.4% versus 83.4%, p=0.0358). At baseline, tumors in the lobular cohort had significantly lower mean Ki67 than those in the non-lobular cohort (11.2% [SD 12.8] versus 16.4% [SD 14.4%], p=0.0038). Consistent with this, the proportion with baseline tumor Ki67 <10% was significantly higher in the lobular cohort than the ductal cohort (60.4% versus 39.3%, p=0.001). At 3-week biopsy, mean Ki67 remained lower in the lobular versus non-lobular group (3.4% versus 5.4%, respectively, p=0.0415). The proportion of patients with Ki67 <10% at 3 weeks did not differ significantly between lobular and non-lobular cases (88.6% versus 79.7%, respectively, p=0.08). Among the 40 lobular cases and 88 non-lobular cases with baseline Ki67 ≥10%, week 3 Ki67 decreased to <10% in 90% of lobular cases and 72.7% of non-lobular cases (p=0.07). On MRI, mean FTV at baseline was similar among lobular and non-lobular cases (21 cc3 versus 16.3 cc3). On pre-surgical MRI, patients in the lobular cohort had significantly higher mean residual FTV than those in the non-lobular cohort (5.7 cc3 versus 3.8 cc3, p=0.034). Interestingly, when splitting the lobular cohort into mixed lobular and pure lobular, mean residual MRI FTV was 7.5 cc3 in mixed cases and 3.8 cc3 in pure lobular cases, reflecting that FTV decreased the least in mixed lobular cases (decrease of 51.1% in mixed cases, 69.7% in pure lobular cases, and 73.9% in non-lobular cases, p=0.0255). Conclusions These results highlight differences in lobular versus non-lobular breast cancer. Lobular cases had lower baseline Ki67, and similar rates of decrease in Ki67 after 3 weeks of NET than non-lobular cases. Despite this, mean FTV after completion of 6 months of NET was higher in lobular tumors. These findings may reflect differences in early versus late indicators of response to NET, or differential response patterns by histology. Alternatively, standard response measures such as Ki67 and FTV may need lobular-specific modifications. Ongoing work includes central assessment of Ki67 from surgical specimens, analysis of changes in gene expression signatures, and evaluation of surgical outcomes such as breast conservation rates by histologic subtype.
Presentation numberPS4-02-02
Ultrasensitive ctDNA-based MRD monitoring predicts relapse in postoperative HR+ inflammatory breast cancer
Ranjan Upadhyay, MD Anderson Cancer Center, Houston, TX
R. Upadhyay1, A. Alexander2, Q. Ye3, K. Chen3, C. Yam2, G. J. Hogan4, S. Zhang4, M. L. LaBella4, S. Ratzel4, The MDACC Inflammatory Breast Cancer Team, A. Lucci5, W. A. Woodward6, B. Lim2; 1Hematology/Oncology Fellowship Program, MD Anderson Cancer Center, Houston, TX, 2Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Department of Bioinformatics and Computational Biology, MD Anderson Cancer Center, Houston, TX, 4Research & Development, Myriad Genetics, Inc., Salt Lake City, UT, 5Department of Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, 6Department of Breast Radiation Oncology, MD Anderson Cancer Center, Houston, TX.
Background: Inflammatory breast cancer (IBC) is a rare, aggressive subtype characterized by rapid progression and a high risk of relapse despite curative-intent therapy. Outcomes are particularly poor for patients with residual disease following neoadjuvant treatment. Circulating tumor DNA (ctDNA)-based assays offer a noninvasive means of detecting minimal residual disease (MRD) and may enable real-time, risk-adapted decisions regarding adjuvant therapy. However, feasibility and performance data for personalized ctDNA monitoring in IBC remain limited. Methods: We prospectively enrolled adults with hormone receptor (HR)-positive (≥10% ER or PR) stage III IBC who did not achieve a pathologic complete response (pCR) to neoadjuvant chemotherapy to a single-arm phase II trial (NCT02971748). Eligible patients had no prior immunotherapy or active autoimmune disease and received pembrolizumab 200 mg IV every three weeks plus physician-chosen endocrine therapy for up to 24 months, starting during or after radiation. The primary endpoint was 2-year event-free survival (EFS); secondary endpoints included safety, toxicity, and overall survival (OS). As an exploratory analysis, the Myriad Precise ultrasensitive MRD assay was used to perform whole-genome sequencing of paired tumor/normal DNA and to subsequently design 1,000-variant capture panels. Plasma was collected postoperatively, serially during therapy, and at end-of-treatment (EOT) for ctDNA analysis. Quantitative tumor fraction (TF) and quality control (QC) metrics were tracked longitudinally. Results: Of 23 enrolled patients, 19 (83%) provided sufficient tumor or normal tissue for personalized panel design. A total of 153 out of 154 plasma samples (99%) passed all quality control (QC) metrics. ctDNA was detected in 42 of 154 samples (27%), with 23 (96%) ctDNA-positive calls below 0.01% tumor fraction (TF), underscoring the need for ultrasensitive detection. One patient who cleared ctDNA detection early remains recurrence-free (early molecular clearance). Two patients who were negative at baseline became MRD positive 10 and 15 months prior to clinical progression. For all patients with disease progression <24 months, we observed persistent and rising ctDNA levels ahead of radiographic relapse with lead times up to 18 months. In contrast, all patients who were MRD negative at the post-operative baseline and throughout treatment remained progression-free at 24 months. Correlative analyses of baseline MRD status and end-of-treatment (EOT) conversion patterns with EFS and overall survival (OS) are ongoing and will be presented. Conclusions: Personalized ctDNA MRD testing is feasible and highly sensitive in the postoperative HR-positive IBC setting, with >99% QC pass rate and detection capability below 0.001% TF. Preliminary data show that ctDNA kinetics correlate with clinical outcomes and ctDNA detection often precedes radiographic relapse by months. These findings support incorporating MRD monitoring into adjuvant trials for real-time therapeutic adaptation in high-risk breast cancer. Acknowledgments: The participants included in the current project were recruited through a Merck supported investigator-initiated trial (NCT02971748). The WGS and ctDNA analyses were performed by the Myriad and MDACC teams through a Myriad and MDACC strategic alliance. The Morgan Welch IBC Research and Clinic Program is funded by Texas State.
Presentation numberPS4-02-03
Tumor-educated platelet RNA captures treatment-induced transcriptomic changes and predicts response in luminal breast cancer.
IZASKUN URDANIBIA, IRB LLEIDA, LLEIDA, Spain
I. URDANIBIA1, A. VELASCO1, G. RODRIGUEZ2, N. TUSET2, A. GASOL2, S. MORALES2; 1Medical Oncology, IRB LLEIDA, LLEIDA, SPAIN, 2Medical Oncology, HospItal arnau de vilanova de lleida, LLEIDA, SPAIN.
Background: Tumor-educated platelets (TEPs) store cancer-related mRNAs and tolerate routine manipulation, providing a stable and minimally invasive window into disease biology. We wondered whether serial TEPs-RNA profiling reflects therapy-induced changes and predicts response in Luminal (HR+/HER2) breast cancer.Methods: In a prospective luminal cohort (n = 12), platelet-rich plasma was collected at diagnosis (T0), post-neoadjuvant therapy (T1) and post-surgery (TF). Fifty-five immune, proliferative and endocrine genes were quantified by RT-qPCR. Data were analysed with PLS-DA, limma differential expression, Spearman correlation with tumour shrinkage, and logistic regression for nodal status.Results: PLS-DA cleanly separated T0, T1 and TF, indicating stepwise transcriptomic re-programming. Twenty genes differed between T0 vs T1 and T0 vs TF (FDR < 0.05); five differed between T1 vs TF. Higher post-neoadjuvant IKBKG and VDR correlated with greater tumour reduction (r ≈ -0.6, p < 0.01). A two-gene model (CACNA2D1 + IKBKG) classified axillary involvement with 85% accuracy. A baseline five-gene signature (ADRB2, IL13, CACNA2D1, MET, ERBB2) was identified that distinguishes patients who achieved pathological complete response (pCR), showing an AUC of 0.87 and clear segregation in hierarchical clustering. Enrichment analysis revealed pathways related to cell cycle control, DNA repair, and p53 regulation, highlighting biological processes associated with treatment response.Conclusions: Sequential TEPs-RNA profiling reflects molecular effects of therapy and surgery and yields baseline biomarkers of complete response in luminal breast cancer. Platelet RNA warrants prospective validation as an accessible complement to tissue-based assays for personalized treatment.
Presentation numberPS4-02-04
Revealing the prognostic significance of dephosphorylation-related genes in breast cancer: comprehensive insightsbased on bulk RNAsequencingand single-cell RNA sequencingdata
Chen Wei, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
C. Wei, L. Mengbo, Z. Hui; Department of Breast Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, CHINA.
Background: Dephosphorylation regulates key signaling pathways in breast cancer, influencing tumor progression, metastasis, and treatment resistance. However, the role of dephosphorylation-related genes (DRGs) in breast cancer (BRCA) and their impact on patient outcomes has not been fully explored. This research sought to explore the prognostic relevance of NRGs in BRCA through the integration of bulk RNA sequencing (bulk RNA-seq) and single-cell RNA sequencing (scRNA-seq) data.Methods: The bulk RNA-seq and scRNA-seq data for BRCA, along with DRGs, were sourced from public databases. Utilizing differential expression analysis, Cox regression analysis, and a machine learning algorithm, this study identified prognostic genes. Leveraging prognostic genes, a risk model was crafted, further dichotomizing BRCA patients into two distinct risk groups. Moreover, enriched pathways and the immune microenvironment of the different risk groups were explored. Subsequent analyses involved examining the expression of prognostic genes at the single-cell level, identifying key cells, and conducting pseudo-time analysis.Results: NOS1, CTNNA2, ACTN2, and KRT5 were identified as prognostic genes in this study. The constructed risk model demonstrated robust predictive ability, with area under the curve (AUC) values surpassing 0.60 at 1-, 2-, and 3-year survival. Subsequently, the functional pathways were related to cellular functions and immune response (such as “apoptosis” and “chemokine signaling pathway”). Moreover, there were six types of immune infiltrating cells that showed significant differences between two risk groups. Additionally, scRNA-seq analysis identified eight cell types, with smooth muscle cells as the key cells, and the expression of KRT5 showed dynamic changes during the different stages of smooth muscle cells differentiation.Conclusion: NOS1, CTNNA2, ACTN2, and KRT5 were identified as prognostic genes for BRCA, highlighting the crucial prognostic role of NRGs in BRCA, which could be particularly important for identifying targeted therapeutic strategies.
Presentation numberPS4-02-06
Analysis of BRCA1 and 2 mutations as predictive biomarkers of response to trastuzumab deruxtecan in hormone receptor-positive, HER2-low metastatic breast cancer: a systematic review and meta-analysis
Lucas Pivetta Genovez, A. C. Camargo Cancer Center, São Paulo, Brazil
L. P. Genovez, H. J. Kim, S. M. Sanches, M. G. Cesca; Clinical Oncology, A. C. Camargo Cancer Center, São Paulo, BRAZIL.
Background: Trastuzumab deruxtecan (T-DXd), an antibody-drug conjugate targeting HER2, has shown significant clinical benefit in patients with HER2-low metastatic breast cancer. The DESTINY-Breast04 trial led to its approval in this setting, particularly among hormone receptor-positive (HR+) patients. Given its topoisomerase I inhibitor payload and DNA-damaging mechanism, tumors with homologous recombination deficiency, such as those harboring BRCA1/2 mutations, may be particularly sensitive to T-DXd. While prior studies have explored this hypothesis, the predictive role of BRCA mutations in the efficacy of T-DXd in HR+/HER2-low disease remains unclear and has not been systematically quantified. Objectives: To evaluate whether BRCA1/2 mutations are associated with improved efficacy of trastuzumab deruxtecan in patients with hormone receptor-positive, HER2-low metastatic breast cancer, by comparing objective response rate (ORR) and progression-free survival (PFS) between BRCA-mutated and BRCA wild-type populations. Methods: A systematic review and meta-analysis were conducted on June 23, 2025 according to PRISMA guidelines. Literature searches were performed in PubMed, Embase, Cochrane Library, and ASCO and ESMO websites databases. Eligible studies reported T-DXd outcomes stratified by BRCA mutation status in HR+/HER2-low metastatic breast cancer. Two reviewers independently performed study selection and data extraction. The primary endpoints were objective response rate (ORR) and progression-free survival (PFS). Analyses were performed using R software (v.4.2.2) with random-effects models. The protocol for this systematic review and meta-analysis was registered in the PROSPERO international prospective register of systematic reviews (ID: CRD420251089678). Results: A total of 3,194 records were retrieved, and after deduplication, 2,461 unique records were screened. Three studies were included: two randomized trials (DESTINY-Breast04 and DESTINY-Breast06) and one large real-world cohort (Tarantino et al., 2025). Patients with BRCA1/2 mutations demonstrated a significantly higher ORR compared to BRCA wild-type counterparts, with a pooled odds ratio of 4.48 (95% CI 1.66-12.09; I² = 0%). For PFS, the pooled hazard ratio favored BRCA-mutated patients (HR 0.56, 95% CI 0.15-2.04), although with substantial heterogeneity (I² = 90.3%). Conclusions: This is the first meta-analysis to evaluate the predictive impact of BRCA1/2 mutations on the efficacy of T-DXd specifically in HR+/HER2-low metastatic breast cancer. BRCA mutations appear to be associated with significantly higher response rates and a potential progression-free survival advantage. These results highlight the need to consider BRCA status as a predictive biomarker in future clinical trials of T-DXd and support its role in guiding therapeutic decisions in HER2-low disease.
Presentation numberPS4-02-07
A novel serum protein-based assay for monitoring response to therapy and recurrence of breast cancer.
Christine Chavany, Milagen, Inc., Emeryville, CA
J. Dea1, C. Chavany1, R. P. Valle1, R. Hernandez-Gonzalez2, M. Jendoubi1; 1Biomarker Assay Development, Milagen, Inc., Emeryville, CA, 2Departamento de Investigacion Experimental y Bioterio, Instituto Nacional de Ciencias Medicas y Nutricion, Mexico City, MEXICO.
Background: Breast cancer (BC) is the most frequent cancer in women from western countries. 30% of BC patients are likely to develop recurrence and risk remains indefinitely. Monitoring recurrence and evaluating response to therapy are important aspects of clinical decision making in the treatment of BC. According to clinical guidelines, BC follow-up is based on clinical examination and bilateral mammogram only. Serum tumor markers such as cancer antigen 15-3 (CA15-3) and other imaging techniques are used by many physicians, but without clear recommendations from clinical societies. We used an in-house developed modular proteomic array platform, the Matrix Protein Array to identify a protein biomarker (BF-09 antigen) that was differentially expressed in BC tissues versus normal counterparts, present in early and late stages and measurable in serum. The development of an ELISA assay to establish its diagnostic performance in discriminating BC patients from healthy individuals and benign cases was previously described 1-2. The present study is designed to assess the utility of our assay for monitoring BC recurrence and therapy response. Methods: The concentration of BF-09 antigen was measured by ELISA assay in 379 retrospective serial serum samples from 85 BC patients and changes in biomarker concentration were correlated to change in disease status as determined by standard of care. BC patient samples were obtained from a biobank at the IRCCS San Raffaele Pisana Research Center (Italy). Patients were followed for up to 10 years (median 23.1 months, range 2.9-203 months) and were examined periodically with imaging to document the remission or progression of the disease. 67% of patients were diagnosed with early-stage BC (I-III) while 33% had metastatic breast cancer (stage IV). Treatments received by patients were neoadjuvant therapy (NAT, 16.5%), adjuvant therapy (AT, 18.8%), metastatic breast cancer therapy (MBCT, 35.3%), or multiple therapies (AT± NAT and/or MBCT, 29.4%). A significant increase or decrease in BF-09 concentration was defined as a change equal or superior to 2.5 times the %CV of the assay as compared to a previous point and interpreted as progression or no progression, respectively. Sensitivity (SE), specificity (SP), negative (NPV) and positive predictive values (PPV) as well as overall concordance of the assay with disease status were calculated. Results: Total concordance of BF-09 serum level with disease status was not affected by BC subtypes (luminal A/B, HER2 positive or triple negative) or stage (Chi-square, p≥0.05). Overall performance of the assay in detecting progressive disease was 81.8% sensitivity at 92.6% specificity with 68.7% PPV and 96.2% NPV. The BF-09 assay could identify BC recurrence both in early BC patients following curative surgery/treatment and in metastatic BC patients during therapy follow up, in some cases years before patients started suffering from clinical symptoms. We were also able to show the utility of BF-09 antigen assay in monitoring therapy response in the neoadjuvant setting prior to surgery. Conclusion: Our findings show that the BF-09 biomarker assay quantitatively reflects therapy response both in the (neo)adjuvant therapy and metastatic therapy settings and detects cancer relapses in a timely manner. This appears to be the first time a protein biomarker has shown high accuracy and applicability for these clinical indications and this could offers new opportunities for BC patient management. Further testing is needed to confirm these results in a prospective trial population. 1 Chavany et al. New breast cancer marker BF-09 is overexpressed in tumor extracts and secreted in serum, Biochemistry and Biophysics Reports, Volume 43, 2025,102097. 2 Chavany et.al. A Serum Biomarker for the Early Detection of Breast cancer. Cancer Res (2024) 84 (9_Supplement): PO5-07-08.
Presentation numberPS4-02-08
Circulating tumor cell (CTC) transcriptional profiling identifies diverse CTC phenotypes in metastatic breast cancer.
Viridiana Carreno, University of Wisconsin, Madison, Madison, WI
V. Carreno, A. H. Chang, I. G. Fernandez, R. Qamar, M. T. West, K. B. Wisinski, M. N. Sharifi; Department of Medicine, University of Wisconsin, Madison, Madison, WI.
Background: Breast cancer is a heterogeneous disease with multiple transcriptionally defined subtypes including luminal A and B hormone receptor (HR) positive subtypes, the HER2 amplified subtype, and the basal subtype most commonly associated with clinically triple negative breast cancer (TNBC). These transcriptional subtypes have differing disease biology, clinical prognosis, and treatment vulnerabilities, and studies utilizing paired tissue biopsies have demonstrated that subtype switching/evolution can occur from primary tumor to metastatic recurrence, as well as in metastatic disease over time on treatment. However, understanding the role of subtype switching in treatment response and resistance has been limited by the challenges of obtaining serial tissue biopsies from patients. Liquid biopsies, including circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs), are a source of tumor-derived material shed into the blood of patients with cancer. Because liquid biopsies require only a peripheral blood draw, they are uniquely suited for longitudinal sampling to evaluate for subtype evolution in metastatic breast cancer. Here, we report the development of a targeted gene expression assay for genes associated with estrogen and HER2 signaling as well as luminal and basal identity in breast cancer CTCs, to address this need. Methods: 15mL peripheral blood was collected in EDTA vacutainer tubes from patients with metastatic breast cancer. CTCs were isolated with automated mTAE technology integrating live cell CTC capture using anti-EpCAM-conjugated paramagnetic particles with RNA extraction. CTC RNA underwent pre-amplification followed by qRT-PCR for genes related to epithelial cell identity, estrogen signaling, HER2 and other growth factors, basal differentiation, as well as controls for immune cell content and housekeeping genes. Expression values were calculated as 33-Ct values. Epithelial/CTC content score was calculated by summing the expression of the epithelial cell identity genes (EpCAM, KRT18, KRT19). Samples with epithelial content score > 10 were classified as having high CTC content. Results: CTCs were detected in 34/51 (66.6%) of HR+ samples, 3/3 (100%) of TNBC samples, and 2/3 (66.6%) of HER2+ samples. 27/51 (52.9%) of HR+ samples, 2/3 (66.6%) of TNBC samples, and 1/3 (33.3%) of HER2+ samples were classified as having high CTC content. In patients with progressing disease (24 samples from 21 patients), 20/24 (83.3%) had CTCs detected and 17/24 (70.8%) had high CTC content. In patients with stable disease (19 samples from 15 patients) 14/19 (73.7%) samples had CTCs detected and 8/19 (42.1%) samples had high CTC content. Hierarchical clustering of high CTC content samples by expression of the estrogen signaling, HER2/growth factor, and basal genes demonstrated clusters consistent with differing transcriptional subtypes. Among HR+ HER2 non-amplified samples (n=22), higher estrogen signaling gene expression was associated with shorter progression free survival (median 2.2 versus 6.9 months, log rank p<0.05) Conclusions: In a pilot cohort of 37 patients with metastatic breast cancer, we demonstrate the feasibility of detecting gene expression relevant to breast cancer transcriptional subtypes via liquid biopsy circulating tumor cell gene expression profiling and relatively high sensitivity even in patients with stable disease, with CTCs detected in 73% of samples including 42% with high CTC content. Interestingly, in HR+ breast cancer CTCs in this cohort, higher estrogen-regulated gene expression was associated with shorter progression free survival, suggesting that this assay could also be utilized to monitor response to anti-estrogen therapy in this patient population.
Presentation numberPS4-02-09
Symptom Clustering and Multi-Omics Signatures of Cancer-Related Fatigue in Breast Cancer: Findings from the China PERSEVERE Cohort
Zheng Qu, University of Pennsylvania, PHILADELPHIA, PA
Z. Qu1, S. Pei2, K. Li3, F. YI2; 1Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, PHILADELPHIA, PA, 2Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 3Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA.
Background: Cancer-related fatigue (CRF) is a debilitating and underrecognized symptom in breast cancer, often preceding treatment and co-occurring with psychological and physical complaints. However, its pathobiological basis remains poorly understood. We leveraged the China PERSEVERE study (NCT07010939), a national prospective cohort of early breast cancer patients, to investigate baseline symptom clusters and identify molecular correlates of CRF using multi-omics profiling. Methods: PERSEVERE is an ongoing multi-center cohort study designed to evaluate treatment-related toxicities and quality of life in women with stage I-III breast cancer in China. Baseline assessments, all conducted prior to surgery or neoadjuvant treatment, include detailed sociodemographic and clinical data, biospecimen collection (plasma, PBMCs, and tumor tissue), and validated patient-reported outcome (PRO) measures (EORTC QLQ-C30/BR23, FA12, IPAQ, HADS, PSS-10, PSQI, SSRS). This cross-sectional analysis included 168 newly diagnosed breast cancer patients enrolled to date, all of whom had complete baseline PROs and biospecimen collection. Clinically significant CRF was defined as a QLQ-C30 fatigue score ≥40 or any FA12 fatigue domain score ≥40. Symptom interrelationships were analyzed using Spearman correlation and hierarchical clustering. Multivariable logistic regression identified predictors of baseline CRF. A nested case-control subgroup of 14 participants (7 with high fatigue scores and 7 matched controls based on age and clinical stage) underwent untargeted baseline serum proteomic and metabolomic profiling. Results: At baseline, 39/168 participants (23.2%) met the criteria for clinically significant CRF. CRF scores showed moderate correlation with poor sleep quality (PSQI; r=0.37, p<0.001), anxiety (HADS-A; r=0.32, p<0.05), and perceived stress (PSS-10; r=0.25, p=0.01), forming a distinct symptom cluster. Patients with CRF also exhibited significantly higher systolic blood pressure, breast-specific symptom burden (BR23), and elevated IL-6 levels (all p<0.05). In adjusted models, BR23 symptom score and waist-to-hip ratio (WHR) remained independent predictors of CRF. In the multi-omics subgroup, 52 differentially expressed proteins were identified, enriched in cytokine signaling (e.g., IL-6 receptor activity) and mitochondrial pathways (e.g., oxidative phosphorylation). Metabolomic analysis revealed 76 altered metabolites, implicating disrupted kynurenine metabolism, glutathione balance, and neuroactive signaling. Integrated correlation analysis linked CRF severity with altered inflammatory markers (CRP, IL6R) and mitochondrial dysfunction (e.g., antimycin A1, tiopronin) across both omics layers. Conclusions: CRF is a core feature of a symptom cluster involving sleep and psychological distress, even before systemic therapy. This integrated clinical and molecular analysis from the PERSEVERE cohort highlights inflammation and mitochondrial dysfunction as plausible contributors to baseline fatigue. These findings inform early identification of high-risk individuals and suggest future intervention targets. Longitudinal follow-up is underway to validate predictive trajectories and biological correlates of CRF over time.
Presentation numberPS4-02-10
Agreement of cell-free DNA methylation-based molecular breast subtyping and tissue subtyping in hormone receptor positive metastatic breast cancer: a clinical cohort analysis
Letizia Pontolillo, Weill Cornell Medicine, New York, NY
L. Pontolillo1, L. Bucheit2, L. Tung3, E. Nicolò1, M. Serafini1, N. Bayou1, B. Pasto1, L. Gerratana4, E. Andreopolou1, E. Bria5, C. Reduzzi1, M. Cristofanilli1; 1Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, 2Medical Affairs, Guardant Health, Palo Alto, CA, 3Bioinformatics, Guardant Health, Palo Alto, CA, 4Department of Medicine, University of Udine, Udine, ITALY, 5Translational Medicine and Surgery, Catholic University of Sacread Heart, Rome, ITALY.
Background: Hormone receptor (HR) and HER2 status, typically assessed on tissue biopsies, are essential for breast cancer (BC) management. As multiple therapeutic options are available for receptors positive metastatic BC (mBC), ongoing subtype assessment may be recommended to monitor changes under the selective pressure and/or evolution of disease. Cell-free DNA (cfDNA) showed reliability for biomarker evaluation and high concordance with tissue-based methods and may be an opportunity for non-invasive, real -time subtype assessment. This study aimed to evaluate agreement between cfDNA methylation-based molecular breast subtyping (MBS) and tissue status in a clinical cohort who underwent cfDNA testing as part of routine clinical care. Methods: Blood samples drawn from mBC patients with HR+ disease on the most recent tissue biopsy and tested with Guardant360 at Weill Cornell Medicine from Nov 2024-June 2025 were queried. A novel cfDNA MBS signature was applied to test results through bioinformatics and/or residual sample testing. MBS confidence scores for HR+/HER2+ where tumor fraction (TF) >0.5% were applied. In this research setting, MBS predicted overall subtype was generated to compare it to the most recent tissue biopsy available. Clinical and genomic factors were annotated for correlation; Fisher’s exact and t-tests were used for statistical analysis.Results: 73 samples were screened of which 42 (57.5%) were evaluable for MBS (TF>0.5%). Of those not evaluable, 16/31 (51.6%) had maximum variant allele frequency <1% and two had no circulating tumor DNA detected. Notably, 24/31 (77%) non-evaluable samples had >1 genomic alteration annotated as clonal hematopoiesis. Two had undetermined results with TF>0.5% but confidence below MBS thresholds, thus 40 samples were included in agreement analysis. A total of 31/40 (77.5%) samples had initial MBS agreement with tissue of whom 27 (87.1%) were HR+/HER2+. The median confidence score from MBS for HR+ prediction was 80% (range: 30%->90%). Of 9/40 (22.5%) samples with initial MBS disagreement with tissue, one (11.1%) differed in HR status (MBS HR-/tissue HR+) and 8 (88.9%) differed in HER2 status (3 MBS HER2+/tissue HER2-; 5 MBS HER2-/tissue HER2+). For the HR case, concomitant genomic analysis did not identify any alterations typically associated with HR+ (ESR1, PIK3CA, PTEN, AKT1). When exploring clinical factors, among samples that had MBS/tissue agreement vs. disagreement, there was no significant difference in: average time from tissue to ctDNA testing (521 days vs. 494 days), number of metastatic sites (2.84 vs. 2.78), number of prior therapies (2.78 vs. 2.35) or prior exposure to CDK4/6 inhibitors (64.2% vs. 77.8%); all p>0.05. Although TF was higher in cases with agreement (mean: 23.4% vs. 20.5%; median: 8.06% vs. 5.26%) this difference was not significant (p>0.05). Upon clinical review of cases with disagreement,1 had MBS HR+/HER2- yet had HR+/HER2- on previous tissue biopsies, concordant with the MBS subtype. A second case had MBS HR+/HER2+ yet HR+/HER2- on the most recent tissue biopsy done in 2023; this case had previous HR+/HER2+ biopsies in 2017 (primary diagnosis) and 2020 (metastatic relapse) and received anti-HER2 therapies, suggesting MBS may capture heterogeneity even after therapeutic intervention. Thus, adjusted agreement of MBS with additional clinical information was 82.5% (33/40).Conclusions: In this mBC cohort, cfDNA MBS showed reliable agreement to tissue biopsy and replicated previously reported accuracy. Subtype switching, heterogeneity and/or lower levels of MBS confidence may contribute to disagreement with tissue. MBS may be a feasible non-invasive option for continuous subtype assessment for HR+ mBC.
Presentation numberPS4-02-11
Molecular and prognostic profiling of HR+/HER2+ Metastatic Breast Cancer (MBC): insights from circulating tumor DNA (ctDNA)-based genomic analysis
Lorenzo Foffano, University of Udine, Udine, Italy
L. Foffano1, A. Davis2, C. Reduzzi3, E. Podany2, A. Medford4, K. Clifton2, M. Velimirovic5, B. Pastò1, L. Pontolillo3, R. Occhiogrosso Abelman6, C. Gianni7, S. Tapiavala2, E. Molteni1, M. Lypsic-Sharf8, E. Nicolò3, E. Andreopoulou3, W. Gradishar9, F. Puglisi1, C. Ma2, A. Bardia8, L. Gerratana1, M. Cristofanilli3; 1Department of Medicine, University of Udine, Udine, ITALY, 2School of Medicine, Washington University, St Louis, MO, 3Department of Medical Oncology, Weill Cornell Medicine, New York, NY, 4Oncology, Massachusetts General Hospital, Boston, MA, 5Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 6Department of Oncology, Massachusetts General Hospital, Boston, MA, 7Department of Oncology, IRST Dino Amadori, Meldola, ITALY, 8Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 9Feinberg School of Medicine, Northwestern University, Chicago, IL, USA, IL.
Background: Hormone receptor–positive/HER2-positive metastatic breast cancer (HER2+/HR+ MBC) represents a biologically distinct subtype, characterized by extensive crosstalk between estrogen receptor (ER) and HER2 signaling pathways. The treatment landscape for HER2+ MBC is rapidly evolving, with recent data from the DESTINY-Breast09 trial supporting consideration of T-DXd in the first line and the PATINA trial demonstrating that adding the CDK4/6 inhibitor palbociclib to maintenance therapy significantly prolonged progression-free survival in patients with HR+/HER2+ disease. The aim of this analysis was to compare genomic profiles and outcomes across HR+/HER2–, HER2+/HR+, and HER2+/HR– subtypes through ctDNA assessment. Methods: This retrospective study analyzed a multi-institutional cohort of 1318 patients (pts) with MBC and ctDNA testing with the Guardant360 NGS panel within a large academic consortium (PMAC). HR and HER2 status were defined based on the most recent tissue biopsy. Associations between single nucleotide variants (SNVs), copy number variations (CNVs) and breast cancer subtypes were tested by multinomial logistic regression (MLR) in terms of Relative Risk Ratio (RRR). The impact of prognosis was evaluated through Cox regression for overall survival (OS), defined from time of baseline ctDNA collection. Results: Among 1318 pts with MBC, 1104 (83.8%) had HR+/HER2–, 127 (9.6%) HER2+/HR+, and 87 (6.6%) HER2+/HR- disease. Multivariable MLR, designed with HR+/HER2- as the reference subtype, investigated clinical variables and genomic alterations across subtypes. Clinically, the presence of nodal (RRR 1.82, p < 0.001) and soft tissue (RRR 2.60, p < 0.001) metastases were more common for HER2+/HR+ but not for HER2+/HR-, while central nervous system (CNS) metastases were associated with both HER2+/HR+ (RRR 3.41, p < 0.001) and HER2+/HR- (RRR 2.73, p = 0.006). Considering single genes alterations, at multivariable analysis, PIK3CA SNVs were less common (RRR 0.51, p = 0.010) and ERBB2 CNVs more common (RRR 41.17, p < 0.001) for HER2+/HR+, while ERBB2 CNVs were also associated with HER2+/HR- (RRR 85.64, p < 0.001). Considering pathway alterations, HER2+/HR+ had fewer PI3K SNVs (RRR 0.47, p = 0.003) and ER SNVs (RRR 0.52, p = 0.022), while in HER2+/HR- there were fewer ER SNVs (RRR 0.05, p = 0.004) and more MYC CNVs (RRR 5.10, p = 0.029). In OS analysis, both HER2+/HR+ (HR 0.55, p = 0.002) and HER2+/HR- (HR = 0.61, p = 0.027) had a significantly favorable prognostic impact compared to HR+/HER2-. Within the HR+/HER2- cohort, several pathway alterations were associated with worse prognosis, including ER SNVs (HR 1.33, p = 0.007), P53 SNVs (HR 1.46, p < 0.001), PI3K CNVs (HR 1.54, p = 0.028) and MYC CNVs (HR 1.84, p = 0.001). In HER2+/HR+, a significant prognostic impact emerged for PI3K SNVs (HR 2.66, p = 0.016), P53 SNVs (HR 2.46, p = 0.036) and RTK CNVs (HR 3.82, p = 0.004), while in HER2+/HR- it was observed only for PI3K CNVs (HR 79.65, p = 0.003). Conclusions: Our ctDNA-based analysis confirms that HER2+/HR+ MBC is a clinically and molecularly distinct subtype. Compared with HR+/HER2– disease, HER2+/HR+ MBC is characterized by higher ERBB2 amplification and a lower frequency of ER SNVs, suggesting reduced dependence on estrogen signaling despite hormone receptor positivity, and the presence of distinct mechanisms of endocrine resistance. Prognostically, both HER2+/HR+ and HER2+/HR– subtypes had significantly more favorable overall survival, suggesting an intrinsic advantage linked to the success of therapeutics aimed at targeting HER2. These findings therefore highlight the potential value of comprehensive baseline and longitudinal mutational profiling to guide initial and maintenance therapy selection and support personalized treatment approaches in HR+/HER2+ disease.
Presentation numberPS4-02-12
Combination of CDK4/6 and EZH2 Inhibitors Enhances Anti-Tumor Immunity in Triple-Negative Breast Cancer via the MAVS-TBK1-STAT1 Signaling Pathway and Synergizes with Anti-PD-1 Therapy
Sisi Li, Chongqing University Cancer Hospital, Chongqing, China
S. Li, Z. Ningning, S. Qing, W. Guanwen, Q. Fanli, W. Long, Q. Yang, Z. Xiaohua; Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, CHINA.
Background and Purpose: Although immune checkpoint inhibitors (ICIs) combined with chemotherapy have been established as the standard treatment regimen for triple-negative breast cancer (TNBC), the proportion of patients deriving significant clinical benefit remains limited. Identifying novel therapeutic targets and developing synergistic strategies to enhance the efficacy and broaden the applicability of ICIs represent critical areas of ongoing research. Methods: A series of comprehensive experiments, including 4T1 orthotopic breast cancer mouse models, tumor-T cell co-culture systems, transcriptome sequencing, flow cytometry, immunoblotting, and qPCR, were performed to systematically elucidate the anti-tumor immune mechanisms of the drug combination. Results: The combination of CDK4/6 and EZH2 inhibitors significantly inhibited 4T1 orthotopic tumor growth and promoted CD8+ T cell infiltration. Transcriptomic analysis identified differential gene enrichment in pathways related to cell adhesion and migration. qPCR demonstrated a notable upregulation of chemokine mRNAs (e.g., CXCL9, CXCL10, CXCL11) in the combination treatment group, while in vitro migration assays confirmed enhanced recruitment of CD8+ T cells. Mechanistically, the combination therapy activated the MAVS-TBK1 axis via dsRNA signaling (but not dsDNA) signaling, leading to STAT1 phosphorylation and increased IFN-β secretion, which subsequently upregulated chemokine expression (no such effects were observed with H151, a STING pathway inhibitor). Furthermore, the combination therapy synergistically enhanced the efficacy of anti-PD-1 monoclonal antibodies. Conclusion: This study demonstrates that the combination of CDK4/6 and EZH2 inhibitors induces chemokine expression and facilitates CD8+ T cell infiltration via the MAVS-TBK1-STAT1 axis, thereby enhancing anti-tumor immunity and providing a novel synergistic approach for TNBC immunotherapy.
Presentation numberPS4-02-13
Genes from epithelial-mesenchymal transition predict overall survival effectively in breast cancer:anovel risk model based on initial step of tumor metastasis
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen1, X. Qin1, X. Xu2, X. Lang3, G. Xie4, W. Liang1; 1Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA, 2Department of Thyroid and Breast Surgery, Northern Jiangsu People’s Hospital, Yangzhou, CHINA, 3Department of General Surgery, Sunshine Union Hospital, Weifang, CHINA, 4Department of Breast Surgery, The People’s Hospital of Chuxiong City, Chuxiong, CHINA.
Background: Accumulating evidence has demonstrated that epithelial-mesenchymal transition (EMT) plays a critical role in breast cancer (BRCA) initiation, invasion, metastasis, and prognosis. BRCA still has a high mortality rate owing to recurrence and metastasis. As a core component of the biological signal regulatory network, EMT urgently requires accurate biomarkers to evaluate the prognosis of BRCA. Methods: We obtained 1223 BRCA samples from The Cancer Genome Atlas and 1184 EMT-related genes from the dbEMT2.0 public database. Differentially expressed genes were filtered, and univariate Cox and LASSO-Cox regression analyses were performed to select prognosis-related gene signatures. A novel risk score model was developed and validated using external cohorts. Moreover, mutual confirmation between KEGG/GO and gene set enrichment analyses was performed to reveal potential molecular mechanisms. Finally, a nomogram including the risk score model and several clinical parameters was established to help individualize patients’ survival predictions.Results: A total of 381 differentially expressed EMT-associated genes (EAGs) were identified and 15 EAGs relevant to survival prognosis were used to calculate the risk score formula. Patients were divided into high- and low-risk groups based on the specific risk score model. Samples in the low-risk group had a higher overall survival. One UCSC cohort and three Gene Expression Omnibus cohorts verified the reliability of the model. A nomogram was established to predict survival, including the risk score, age, pathological N stage, and pathological M stage. Ultimately, the results of gene set enrichment analysis revealed that the potential molecular mechanism was mainly concentrated in the extracellular region.Conclusions: The 15-gene signature model based on EMT is significant for predicting overall survival and revealing possible molecular mechanisms as potential targets and tools for pharmacologic and regenerative medicine biomarkers.
Presentation numberPS4-02-14
Recognition of immunogenomic signature and prognostic value of the subtype of epithelial-mesenchymal transition in breast cancer
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen1, X. Qin1, X. Xu2, X. Lang3, G. Xie4, W. Liang1; 1Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA, 2Department of Thyroid and Breast Surgery, Northern Jiangsu People’s Hospital, Yangzhou, CHINA, 3Department of General Surgery, Sunshine Union Hospital, Weifang, CHINA, 4Department of Breast Surgery, The People’s Hospital of Chuxiong City, Chuxiong, CHINA.
Background: Accumulating evidence has revealed that epithelial-mesenchymal transition (EMT) plays a crucial role in tumor progression and the immune microenvironment, which further results in a high rate of recurrence and metastasis. The EMT immune signaling pathway provides a great perspective for designing personalized therapies. Methods: In this study, 1223 RNA-seq samples were obtained from the TCGA-BRCA dataset. A total of 381 EMT-related differentially expressed genes were analyzed and combined with clinical parameters, and the matrix was divided into training and testing cohorts at a ratio of 7:3. The training cohort was used to develop an EMT signature, including GKN2, FZD2, NDRG2, SCUBE2, ALX4, CCL19, SDC1, EZR, CPEB1, and HRG genes, and the accuracy of this signature was validated by testing and GSE158309 cohorts. Results: A risk score model and clinical parameters were used to establish a nomogram for predicting prognosis. The C-index (0.719), calibration curves, and model comparison with four previous studies demonstrated the reliability of the EMT signature, the biological phenotypes of which were tested for functional enrichment, immune infiltration, and tumor mutation. Additionally, patients’ responses to immunotherapy and chemotherapy were assessed. Our results showed that the low-risk group had higher immune infiltration, tumor mutational burden, microsatellite instability levels, immune checkpoint inhibitor expression, tumor immune dysfunction and exclusion scores, and immunophenoscores, which could predict patient sensitivity to immunotherapy. Moreover, low-risk patients exhibit better sensitivity to chemotherapy. Conclusion: This novel EMT signature offers excellent potential for predicting the prognosis, tumor immune heterogeneity, and therapeutic responses in breast cancer.
Presentation numberPS4-02-15
Sumo e3 ligase ranbp2 affects triple-negative breast cancer tumorigenicity by stabilizing birc5 expression
Yuxiang Lin, Fujian Medical University Union Hospital, Fuzhou, China
Y. Lin, J. Wu, Y. Li, F. Fu, C. Wang; Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA.
Background: Triple-negative breast cancer (TNBC) is more aggressive and has higher metastasis rate compared with other subtypes of breast cancer. RAN binding protein 2 (RANBP2) is a less-explored SUMO E3 ligase and has been shown to play key roles in various cellular cycle processes. However, its biological function and molecular mechanism in TNBC remain largely unclear. Methods: RANBP2 expression was evaluated in TNBC patients’ s tumor tissues with public database and immunohistochemical staining methods. Correlation between RANBP2 expression and patients’ prognosis was also analyzed. The effects of RNABP2 on TNBC cell proliferation ability were detected by CCK-8, colony formation, apoptosis and cell cycle analysis. Tumor formation assay was constructed to examine the effects of RANBP2 on TNBC growth in vivo. Proteomic profiling, Western blot, protein stability tests and rescue experiments were further applied to evaluate the detailed mechanism. Results: The expression of RANBP2 was indicated to be upregulated in TNBC tumor tissues and high expression of RANBP2 was correlated with poor prognosis of TNBC patients. Functional studies demonstrated that RANBP2 overexpression promoted TNBC cell proliferation both in vitro and in vivo, whereas RANBP2 knockdown inhibited proliferation, induced apoptosis, and caused G1-S phase cell cycle arrest. Baculoviral IAP Repeat Containing 5 (BIRC5) was identified as a downstream effector of RANBP2. Mechanistically, RANBP2 post-translationally regulated BIRC5 by facilitating its degradation via the proteasomal pathway. Functional rescue experiments further demonstrated that BIRC5 overexpression could partially reverse the anti-proliferative effects induced by RANBP2 knockdown in TNBC cells. Conclusion: RANBP2 serves as a critical regulator of TNBC development and progression. Targeting RANBP2/BIRC5 axis may provide new insights into therapeutic strategies and research directions for TNBC treatment.
Presentation numberPS4-02-16
Stress-induced adrenergic signaling promotes endocrine resistance in ER-positive breast cancer
Gilberto Gastelum Martinez, Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC
G. Gastelum Martinez, M. Rezaei, P. Miller, C. Pine, Y. Anbalagan, N. Bishopric, B. Hudson, M. Lippman; Department of Oncology, Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC.
Resistance to antiestrogen therapy is a major challenge to the treatment of estrogen receptor–positive (ER⁺) metastatic breast cancer (MBC). While endocrine therapies such as tamoxifen, aromatase inhibitors, and fulvestrant are initially effective, most patients ultimately develop resistance, leading to progressive metastatic disease. Psychosocial stress, widely reported in breast cancer patients, is a clinically relevant factor that worsens outcomes. Elevated chronic levels of norepinephrine (NE), a hallmark of stress physiology, correlate with poor prognosis and increased metastatic burden in breast cancer. We and others have shown the clear negative effect of chronic adrenergic stress in driving immunomodulatory changes that favor tumorigenesis. However, the direct, tumor-intrinsic effects of adrenergic stimulation remain underexplored and not well characterized.Here, we explore a previously unrecognized mechanism by which adrenergic stress drives ER⁺ breast cancer cells toward a therapy-resistant and metastatic phenotype. We first confirmed that human ER⁺ breast tumor, and culture murine and human ER+ breast cancer cells, express α2A-, β1-, and β2-adrenergic receptors at both transcript and protein levels using integrated transcriptomic analysis and validated through immunocytochemistry. Functionally, NE promotes proliferation in both estrogen-rich and estrogen-deprived conditions and cooperates with 17β-estradiol (E2) to significantly enhance clonogenicity, anchorage-independent sphere formation, CD44 expression, and migration—features consistent with cellular plasticity and partial cancer stem-like phenotypes. Crucially, we show that NE partially rescues ER⁺ breast cancer cells from growth inhibition by fulvestrant and tamoxifen, suggesting adrenergic signaling acts as a bypass mechanism that subverts endocrine blockade. We also observe that long-term exposure to NE plus E2 confers persistent cellular changes, as these cells maintain enhanced clonogenic potential even after withdrawal of stimuli. Pharmacologic interrogation using selective adrenergic antagonists abrogates these NE-mediated phenotypes in ER+ breast cancer cells and identifies α2A-adrenergic signaling as a key driver of endocrine resistance. Our findings establish that chronic adrenergic stimulation provides selective pressure on ER⁺ breast cancer cells and drives therapy-resistance, and transition of cells into stem-like states through tumor-intrinsic mechanisms independent of immune suppression. This work increases the potential role of stress in ER⁺ breast cancer and supports a new therapeutic paradigm—targeting adrenergic signaling not just to modulate the immune microenvironment, but to directly disrupt tumor plasticity and restore endocrine sensitivity. These insights provide strong rationale for incorporating adrenergic inhibitors into treatment strategies and open the door for potential clinical trials leveraging stress-targeted therapies to overcome therapeutic resistance for patients with endocrine-resistant MBC.
Presentation numberPS4-02-17
From Metabolites to Mechanisms: Linking Tryptophan Metabolism and AhR Activation in Breast Cancer
Yanming Wu, SUNY Downstate Health Sciences University, Brooklyn, NY
Y. Wu1, K. Wang2, Z. Li3; 1Department of Internal Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, 2Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, CHINA, 3Department of Breast Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, CHINA.
Background: Tryptophan (Trp), an essential amino acid, is metabolized through the kynurenine, indole, and serotonin pathways. Imbalances in Trp metabolism have been implicated in cancer progression. However, clinical trials targeting indoleamine-2,3-dioxygenase (IDO), a key enzyme in Trp catabolism, have yielded unsatisfactory results, prompting the exploration of alternative therapeutic targets within the Trp metabolic network. Emerging evidence discovers that several Trp-derived metabolites can bind to and activate the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor that is essential in immune response and tumorigenesis. Although AhR is widely expressed in various breast cancer subtypes, its precise role in breast cancer development remains unclear, likely due to disease complexity and the diversity of AhR ligands. Thus, investigating Trp metabolism in relation to AhR activation may provide novel insights into the role of Trp-activated AhR pathway in breast cancer progression. Methods: Clinical data and serum samples were collected from patients with breast cancer (n = 13) and age-matched individuals with breast fibroadenoma (n = 9). Eighteen Trp metabolites were quantified using Ultra-High Performance Liquid Chromatography (UHPLC). Metabolite levels were normalized using Z-score transformation and visualized via heatmap. Hierarchial clustering was performed to assess sample grouping. AhR mRNA expression levels were compared between primary breast cancer tissues (n = 1111) and benign tissues (n = 113) by retrieving RNA-sequencing data from The Cancer Genome Atlas (TCGA) Breast Invasive Carcinoma cohort. Results: Differential levels of Trp metabolites were identified between breast cancer and fibroadenoma groups, with Trp, cinnavalininate, 5-hydroxyindole-3-acetic acid and picolinic acid showing the most significant differences. Heatmap visualization revealed distinct Trp metabolite profiles across all samples. Hierarchical clustering showed partial segregation between the breast cancer and fibroadenoma groups. Analysis of TCGA data demonstrated significantly reduced AhR mRNA expression in breast cancer tissues compared to benign tissues. Conclusion: Trp metabolism is disrupted in breast cancer. The partial clustering of breast cancer and fibroadenoma samples reflects both the biological heterogeneity of breast cancer and the overlapping metabolic features between the two groups. The observed alterations in Trp metabolism, along with reduced AhR expression in breast cancer compared to benign tissues, suggest that impaired Trp-activated AhR signaling may play a critical role in breast cancer pathogenesis. These findings support further investigation into the Trp-activated AhR pathway as a potential therapeutic target in breast cancer management.
Presentation numberPS4-02-18
Detection of germline and somatic pathogenic variants in patients with de novo metastatic breast cancer (dnMBC) and assessment of their prognostic relevance: A Hellenic Cooperative Oncology Group (HeCOG) translational cohort study
Elena Fountzilas, Hellenic Cooperative Oncology Group (HeCOG), Athens, Greece
E. Fountzilas1, K. Papadopoulou1, N. Korfiatis1, A. Goussia1, A. Christopoulou1, E. Moirogiorgou1, N. Tsoukalas1, G. Petrakis1, C. Gouedard2, F. Zagouri1, D. Stefanou1, F. Kyriakou3, E. Vorrias3, S. Karageorgopoulou1, F. Dimitrakopoulos1, A. Vernadou1, S. Murray2, G. Fountzilas1; 1Data Office, Hellenic Cooperative Oncology Group (HeCOG), Athens, GREECE, 2Molecular Oncology, BioPath Innovations SA, Athens, GREECE, 3Medical Oncology, German Oncology Center, Limassol, CYPRUS.
Background: Robust clinico-genomic predictors of survival in dnMBC remain ill defined. We employed next generation sequencing (NGS) to explore the mutational spectrum of a Greek dnMBC cohort alongside key clinicopathological factors and evaluated their prognostic impact for overall survival (OS). Patients and Methods: Formalin-fixed, paraffin-embedded tumor tissue blocks were collected from the HeCOG Biobank for 106 patients with dnMBC alongside peripheral blood samples for 91 cases. NGS was performed on Ion Torrent platforms with AmpliSeq DNA panels targeting genes recurrently altered in BC. Variants classified as pathogenic or likely pathogenic (PVs) according to Clinvar and ACMG/AMP criteria were then processed for analysis. Cox proportional-hazards regression models were applied to assess prognostic relevance of both genomic and clinicopathological variables. Results: The total study population consisted of 106 women with a median age of 63 years (31-86); 67 patients (64%) were postmenopausal Overall, 62.5% presented with a PS of 0 and 74% with NST histology. Regarding tumor profiling, 82% were hormone receptor (HR) positive, 28.8% HER2+, 50% grade 3. Family history of cancer was positive in 17.3% of patients. Notably, 42% of patients underwent some type of breast surgery. First-line treatment included a combination of aromatase inhibitor (AI) and CDK4/6 inhibitor in 36 (34%) patients, chemotherapy (CT) alone in 27 (23.6%), combination of CT and other targeted agents in 30 (28.3%), hormonal therapy only in 7 (6.6%), while for 6 patients (5.6%) treatment data were missing. Moreover, 11 patients (10.3%) were diagnosed with locoregional (LR) disease only, 45 (42.5%) with distant and 42 (39%) with LR and distant metastases. Regarding IHC subtypes tested locally, 70 patients (66%) were classified as Luminal A/B, 31 (29.2%) HER2+ and 5 (4.7%) as TNBC. At least one PV was present in 69 of 106 tumors (65.1 %), with most carrying PVs in PIK3CA (40 cases; 37.7 %) and TP53 (29 cases; 27.4 %). Recurrent, albeit less frequent, PVs were also observed in ERBB2, GATA3, PTEN, CDH1 and AKT1 (each present in ≥ 2 tumors). Meanwhile, germline sequencing revealed single PVs in two patients, one in TP53 and the other in PTEN. In univariate analyses, none of the standard clinicopathological parameters (age, menopausal status, performance status, histology, T-stage, grade, ER/HER2 status) or individual PVs reached statistical significance for OS. No significant difference was found between Luminal and HER2+ subgroups. Conclusion: The mutational landscape of our Greek dnMBC series reveals a high prevalence of actionable PIK3CA, TP53 and ERBB2 alterations. These findings emphasize further the biological heterogeneity of dnMBC disease and the need for the identification of novel integrative biomarkers beyond standard clinico-genomic parameters to improve prognosis and guide therapeutic decisions. The study was registered on ClinicalTrials.gov under the identifier NCT05758948
Presentation numberPS4-02-19
Mutations in PIK3R1 Activate Multiple Pathways in Triple Negative Breast Cancer
Abde Abukhdeir, Rush University, Chicago, IL
A. Abukhdeir1, K. Cagin2, J. Borgia2, M. Cobleigh1; 1Department of Medicine, Rush University, Chicago, IL, 2Department of Anatomy and Cell Biology, Rush University, Chicago, IL.
Background: Approximately 6% of triple-negative breast cancers (TNBCs) carry mutations in the PIK3R1 gene (PIK3R1MUT). We previously showed that PIK3R1 knock-out causes increased phosphorylation of MEK and enhanced sensitivity to MEK inhibitors, trametinib and binimetinib. Alpelisib targets p110-α in the PI3K/AKT/mTOR pathway. We hypothesized that alpelisib might also inhibit growth of PIK3R1MUT TNBC. This study tested binimetinib, alpelisib, and their combination in PDX models with PIK3R1MUT. Methods: Four TNBC PDX models were studied (Table 1). All models had wild-type PIK3CA. Control mice had wild-type PIK3R1, while experimental mice had PIK3R1MUT. Animals were treated with vehicle control, binimetinib (10 mg/kg BID), alpelisib (35 mg/kg QD), or their combination for at least 28 days. Tumor volumes were recorded biweekly. The genetic backgrounds of each model are listed. All mice had co-mutations in PI3K or MAPK pathway genes but lacked co-mutations in both pathways. Tumors were analyzed using mass spectrometry to elucidate downstream protein and phosphoprotein expression. Results: Endpoint analyses showed no significant tumor growth reduction with single-agent binimetinib or alpelisib in Models A and B. Binimetinib alone resulted in a slightly significant effect in Model C (p<0.01). However, a highly significant effect of both single agents was observed in Model D (p<0.001). Combination therapy with binimetinib and alpelisib significantly reduced tumor growth in all models but was most effective in models C and D (p<0.001, p<0.001, p<0.0001, and p<0.0001 in Models A-D, respectively). The mass spectrometry results will be presented at the meeting. Conclusions: Mutations in PIK3R1 sensitize TNBCs to combined MEK and PI3K pathway inhibition. Co-mutations in PIK3R1 wt PDX models may also sensitize TNBC to MEK and PI3K inhibitors. These findings suggest that combining inhibitors of MEK and PIK3CA could inhibit growth of TNBCs. Disclaimer: AA is employed by AstraZeneca but contributed to this publication in his own capacity. The views expressed are his own and do not necessarily represent the views of AstraZeneca.
| Name | Age | Ethnicity | PIK3R1 DNA | PIK3R1 Protein | |||||
| PDX-A | 66 | Black or African American | wt | wt | |||||
| PDX-B | 66 | Caucasian | wt | wt | |||||
| PDX-C | 57 | Caucasian | wt | wt | |||||
| PDX-D | unknown | Caucasian | c.1705_1740del | p.Asp569_Tyr580del |
Presentation numberPS4-02-20
Clinical and genomic biomarkers of capivasertib response in patients with hormone receptor-positive, HER2-negative metastatic breast cancer (HR+/HER2- MBC)
Maxwell R Lloyd, Beth Israel Deaconess Medical Center, Boston, MA
M. R. Lloyd1, E. L. Podany2, Z. Bagheri3, E. Rizk1, A. Dedeoglu4, A. Putur4, C. Ping2, J. Hesse2, A. Golden5, S. Addison2, J. J. Tao6, N. Vidula4, R. O. Abelman4, K. K. Clifton2, I. Sanidas4, A. Varkaris4, D. Juric4, N. Vasan7, C. X. Ma2, G. M. Wulf1, L. W. Ellisen4, S. A. Wander4; 1Department of Medicine, Division of Hematology / Oncology, Beth Israel Deaconess Medical Center, Boston, MA, 2Department of Medicine, Division of Hematology / Oncology, Washington University in St. Louis, St. Louis, MO, 3Department of Graduate Education, Harvard Medical School, Boston, MA, 4Department of Medicine, Division of Hematology / Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, 5Department of Medicine, Division of General Medicine and Geriatrics, Washington University in St. Louis, St. Louis, MO, 6Department of Medicine, Division of Hematology / Oncology, Columbia University Irving Medical Center, New York, NY, 7Perlmutter Cancer Center, NYU Langone Health, New York, NY.
Introduction: HR+/HER2- MBC remains incurable, with therapeutic resistance limiting treatment durability. Capivasertib, the first FDA-approved AKT inhibitor for MBC with PIK3CA/AKT1/PTEN alterations, is now widely utilized. However, predictors of response remain poorly understood. This study examined baseline genomic alterations and prior therapy exposures for associations with progression-free survival (PFS) in patients treated with capivasertib. Methods: This retrospective cohort included patients from three U.S. centers treated between November 2023 (capivasertib approval) and June 2025 (data cutoff). Follow up continued until progression, toxicity-related discontinuation, death, or data cutoff. Eligible patients had HR+/HER2- MBC treated with capivasertib plus fulvestrant and baseline next-generation sequencing, as defined by testing within 6 months of therapy start. Two cohorts were analyzed: 1) patients with baseline circulating tumor DNA (ctDNA) sequencing only, and 2) patients with baseline ctDNA or tissue sequencing. Patients were identified via pharmacy claims; clinical and sequencing data were extracted and de-identified. The primary variable examined was the presence of baseline oncogenic genomic alterations; variants of uncertain significance and genes altered in <3 patients were excluded. Other exposures included prior lines of metastatic therapy and prior PI3K inhibitor (PI3Ki) use. The primary outcome was PFS, analyzed using the Kaplan-Meier method and Cox regression. Results: In cohort 1, we identified 45 patients treated with capivasertib and fulvestrant with baseline ctDNA-only sequencing. For this group, median age was 67, 96% were postmenopausal, and two-thirds had visceral and/or brain metastases. Median number of prior lines of therapy was 2; 96% had received a CDK4/6 inhibitor, 60% fulvestrant, 51% chemotherapy, and 22% prior PI3Ki. Median PFS in cohort 1 was 253 days (95% CI 147-392); 21 patients remained on therapy at data cutoff (approximately half had >6 months follow up). Baseline PIK3CA mutations (n=30) trended toward longer PFS (305 vs 194 days; HR 0.58, 95% CI 0.24-1.47); PTEN alterations (n=6) showed no PFS difference (HR 1.09, 95% CI 0.12-4.53), and AKT1 mutations (n=2) were too few to analyze. Pathogenic CCND1 alterations (n=4; 3 amplifications, 1 truncating mutation) were associated with shorter PFS (101 vs 305 days; HR 4.91, 95% CI 1.22-15.40), as were truncating ARID1A mutations (n=4; HR 6.35, 95% CI 1.19-23.45). ESR1 mutations (n=12), all co-occurring with a PI3K pathway alteration (PIK3CA, PTEN, or AKT1), trended toward worse PFS (147 vs 356 days; HR 1.86, 95% CI 0.66-4.82). PFS was numerically longer in patients with ≤1 prior line of therapy vs 2-3 prior lines (305 vs 202 days; HR 1.17, 95% CI 0.36-3.53) or ≥4 prior lines (305 vs 151 days; HR 1.44, 95% CI 0.51-4.06). Prior PI3Ki use trended toward shorter PFS (147 vs 305 days; HR 1.67, 95% CI 0.64-4.07). In cohort 2, 53 patients had baseline ctDNA or tissue sequencing; demographic and clinical features were consistent with cohort 1. FGFR1 alterations, too infrequent to analyze in the ctDNA-only group, trended toward shorter PFS (n=3; 88 vs 253 days; HR 2.33, 95% CI 0.58-6.92). Other clinical-genomic associations remained similar. Conclusions: In this study of patients with HR+/HER2- MBC treated with capivasertib and fulvestrant, alterations in CCND1 and ARID1A in ctDNA were associated with poorer PFS, while ESR1 and PI3K pathway co-mutations trended toward worse outcomes. Patients treated in the second-line or earlier, and those without prior PI3Ki exposure, may derive greater benefit. These findings are exploratory and warrant validation but suggest specific biomarkers that may correlate with risk for early progression on an AKT inhibitor.
Presentation numberPS4-02-21
Prognostic Significance of HER2 Heterogeneity in Early-stage and Locally advanced HER2-positive Breast Cancer
Takaaki Hatano, Osaka University, Suita-city, Japan
T. Hatano1, T. Tanei1, S. Seno2, Y. Sota1, N. Masunaga1, C. Mishima1, M. Tsukabe1, T. Yoshinami1, K. Shimazu1; 1Breast and Endocrine Surgery, Osaka University, Suita-city, JAPAN, 2Graduate School of Information Science and Technology, Osaka University, Suita-city, JAPAN.
Background: Breast cancer tumors often have intratumoral heterogeneity (ITH), which has been associated with therapeutic resistance. Tumors with high ITH show human epidermal growth factor receptor 2 (HER2) heterogeneity, impacting the effectiveness of HER2-targeted therapies. Our recent study identified HER2 ITH as an independent prognostic factor for poor outcomes in HER2-positive breast cancer. In this study, we investigated the association between HER2 ITH and resistance to Anti-HER2 Neoadjuvant Chemotherapy (NAC). Methods: The study included 97 patients of primary HER2-positive breast cancer treated with Anti-HER2 NAC. Breast tumor samples were obtained from vacuumassisted breast biopsy before NAC. HER2 gene amplification was assessed by fluorescence in situ hybridization (FISH), and histograms of HER2 gene copy numbers were generated. Using Gaussian mixture model, the histogram data were analyzed and classified into two groups, high HER2 heterogeneity (HH) and low HER2 heterogeneity (LH). The relationship between HER2 ITH and treatment response was evaluated with the pathological complete response (pCR) rate. Results: Of 97 patients,18 (18.6%) were classified into the HH group, and 79 (81.4%) into the LH group. The pCR rate in the HH group was significantly lower at 28% (5/18) compared to 65% (51/79) in the LH group (P = 0.01). Multivariate analysis of pathological parameters revealed that HER2 ITH was the significant predictor of pCR rate (P = 0.02), following estrogen receptor status. Conclusions: The assessment of HER2 ITH may be valuable in predicting therapeutic outcomes in HER2-positive breast cancer. Our novel approach of HER2 ITH method using FISH histograms could potentially serve as a useful tool in predicting resistance to anti-HER2 NAC.
Presentation numberPS4-02-22
Dynamics of the Intratumoral Microbiome Across Malignant Transformation and Treatment in Breast Cancer
liuliu quan, cancer hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
l. quan1, R. Huiteng2, y. jian1, y. peng1; 1Department of Medical Oncology, cancer hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 2Department of Data Science, School of Data Science, The Chinese University of Hong Kong, Shenzhen, shenzhen, CHINA.
IntroductionWhile breast cancer (BC) remains the most prevalent malignancy among women worldwide, the dynamics of the intratumoral microbiome during tumor initiation, progression, and treatment have been largely overlooked. Prior studies are predominantly cross-sectional and limited by indirect microbial inference from RNA-seq data.Method This study presents the first comprehensive longitudinal analysis of intratumoral microbiota across breast tissue samples using high-depth 16S rRNA sequencing (11W tags). Samples included: 165 benign nodules (82 non-transforming, 83 that later progressed to cancer with matched malignant tissues); 180 primary BC tissues and 165 benign controls; and 101 neoadjuvant therapy (NAT) specimens (15 pCR, 86 non-pCR, with paired pre/post-treatment samples). Microbial diversity, differential taxa, functional predictions, network structures, and correlations with clinical characteristics were systematically analyzed. In addition, two predictive models were developed to malignancy transformation and NAT response.ResultsCompared to NNT group, NDT group exhibited lower microbial diversity, with a relative enrichment of genera such as Bacteroides, Weissella, Paenibacillus, and Methyloversatilis, whereas the NNT group showed higher abundances of Halomonas, Dietzia, Nesterenkonia, and Aeromicrobium (P<0.05). Functional analysis revealed that amino acid and vitamin metabolism-related pathways were significantly more active in the NNT group, while lipid metabolism pathways were enriched in the NDT group (P<0.05). During the malignant transformation of nodules, the abundances of Aeromicrobium, Dietzia, Halomonas, and Nesterenkonia markedly increased, whereas Paenibacillus and Faecalibacterium significantly decreased (P<0.05). Second, in comparisons between BC tissues and benign lesions, genera such as Methyloversatilis, Weissella, Caulobacter, and Bosea were enriched in tumor tissues, while Delftia, Dietzia, Halomonas, Aeromicrobium, Nesterenkonia, and Acinetobacter were notably reduced (P<0.05). Furthermore, with advancing tumor stages, the relative abundances of Dietzia, Aeromicrobium, Delftia, Nesterenkonia, Halomonas, and Nitriliruptor gradually declined, suggesting that these taxa may be associated with the early-stage tumor microenvironment (P<0.05). Finally, following NAT, microbial diversity significantly increased, and the microbial community composition underwent a notable reconstruction. After treatment, alpha diversity rose, with genera such as Aeromicrobium, Halomonas, Dietzia, Nesterenkonia, and Nitriliruptor showing significant increases in abundance (P<0.05). Functional prediction analysis indicated enhanced activities in pathways related to amino acid metabolism, lipid metabolism, and terpene metabolism, while pathways involved in glycan metabolism were diminished (P<0.05). Importantly, we developed two predictive models with high clinical relevance: one stratifying malignancy risk in nodules (AUC=1.0), and another predicting NAT response (AUC=0.82). ConclusionOur findings highlight the clinical relevance of microbial signatures in early diagnosis, malignancy prediction, and response to therapy, offering novel insights for precision oncology and microbiome-based interventions in breast cancer.
Presentation numberPS4-02-23
Biomarkers of response and resistance to Sacituzumab Govitecan in HR+/HER2− advanced breast cancer: interim results from the phase II SOLTI ACROSS-TROP2 Trial
Eva Ciruelos, University Hospital 12 De Octubre / Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12) / SOLTI Cancer Research Group, Madrid, Spain
E. Ciruelos1, T. Pascual2, B. Adamo3, M. Alva Bianchi4, I. Blancas5, L. Carnerero6, R. Villanueva-Vázquez7, B. Fullana7, M. Melé8, J. Salvador Bofill9, J. M. Cejalvo10, A. Ferrando-Díez11, Y. Izarzugaza12, M. Borrell13, X. González Farré14, G. Villacampa15, M. Paes Dias16, F. M. Juan16, M. Oliveira13; 1Medical Oncology, University Hospital 12 De Octubre / Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12) / SOLTI Cancer Research Group, Madrid, SPAIN, 2Medical Oncology, Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona / Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS) / SOLTI Cancer Research Group, Barcelona, SPAIN, 3Medical Oncology, Institute of Cancer and Blood Diseases, Hospital Clinic of Barcelona / Translational Genomics and Targeted Therapies in Solid Tumors group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN, 4Medical Oncology, University Hospital 12 De Octubre, Madrid, SPAIN, 5Medical Oncology, Hospital Universitario Clínico San Cecilio / Medicine Department, Granada University / Instituto de Investigación Biosanitaria de Granada (ibs Granada), Granada, SPAIN, 6Medical Oncology, Hospital Universitario Clínico San Cecilio / Instituto de Investigación Biosanitaria de Granada (ibs Granada), Granada, SPAIN, 7Medical Oncology, Institut Català d’Oncologia (ICO Hospitalet), L’Hospitalet de Llobregat, SPAIN, 8Medical Oncology, Hospital Universitari Sant Joan de Reus, Reus, SPAIN, 9Medical Oncology, Hospital Universitario Virgen del Rocio, Instituto de BioMedicina de Sevilla (IBIS), Sevilla, SPAIN, 10Medical Oncology, Hospital Clínico Universitario de Valencia (INCLIVA) / SOLTI Cancer Research Group, Valencia, SPAIN, 11Medical Oncology, Institut Català d’Oncologia Badalona (ICO Badalona), Badalona, SPAIN, 12Medical Oncology, Hospital Universitario Fundación Jiménez Dí, Madrid, SPAIN, 13Medical Oncology, Vall d’Hebron University, Hospital and Vall d’Hebron Institute of Oncology (VHIO) / SOLTI Cancer Research Group, Barcelona, SPAIN, 14Medical Oncology, IOR – Instituto Oncologico Dr. Rosell, Hospital General de Catalunya / SOLTI Cancer Research Group, Barcelona, SPAIN, 15Clinical Research, SOLTI Cancer Research Group / Statistics Unit, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 16Clinical Research, SOLTI Cancer Research Group, Barcelona, SPAIN.
Background: Sacituzumab Govitecan (SG) is an antibody-drug conjugate (ADC) containing a TROP2-directed antibody bound to a topoisomerase I inhibitor payload. SG is approved for the treatment of patients (pts) with metastatic triple-negative breast cancer who have received at least two prior systemic therapies, including one for advanced disease, and pts with hormone receptor-positive (HR+)/HER2-negative metastatic breast cancer (mBC) after endocrine therapy and ≥2 systemic treatments in the advanced setting. SG efficacy does not seem to correlate to TROP2 protein expression, and no additional efficacy biomarkers have been identified. Methods: SOLTI ACROSS-TROP2 (NCT06236269) is a phase II, open-label, single-arm trial investigating biomarkers of efficacy and mechanisms of resistance to SG in 100 pts with HR+/HER2- mBC who progressed during or after treatment with CDK4/6 inhibitors and received up to one prior chemotherapy or ADC regimen for metastatic disease. Pts received SG at 10 mg/kg via IV infusion on Days 1 and 8 of each 21-day cycle. Mandatory tumor biopsies were obtained at baseline and after 2-3 weeks of treatment (C2D1). The primary objective is to evaluate the change in CelTIL score (a composite score of tumor cellularity and tumor infiltrating lymphocytes) between baseline and C2D1 tumor samples (Nuciforo et al, Ann Oncol 2019). Key secondary endpoints include progression-free survival (PFS), overall response rate (ORR), and safety. Here we present the planned interim analysis results from the first 50 enrolled pts. Results: Between April and December 2024, 50 pts were enrolled. Median follow-up was 8.6 months (m). Median age was 58 (range 30-79), all pts were female, most were postmenopausal (78%), had no visceral metastases (64%), and received SG as second line for mBC after chemotherapy or ADC (70%). At data cut-off (May-25), 18/50 (36%) remained on treatment and 43/50 (86%) were evaluable for the primary endpoint. The mean increase in CelTIL score from baseline to C2D1 was 4.33 points (95% CI 0.99-7.65), indicating a significant overall change, with a greater and significant increase in patients that achieved radiological response (6.33 vs 3.13 points). PFS events occurred in 28/50 pts (maturity 56%). Median PFS was 6m (4.2-NR) both overall and among pts treated in second line for mBC. ORR was 37% and 38%, respectively. Grade ≥3 treatment-related adverse events (TRAEs) occurred in 48.0% of pts. Most common TRAEs were fatigue (58%), anemia (58%), neutropenia (54%), nausea (46%) and diarrhea (38%). Treatment discontinuation rate due to TRAEs was 8%. Conclusions: In this interim analysis of SOLTI ACROSS-TROP2, one cycle of SG led to an increase in CelTIL score, particularly among patients who achieved a radiological response. Consistent efficacy was observed in terms of PFS and ORR. The final results from the full cohort of 100 patients, along with a comprehensive translational analysis of paired pre- and on-treatment biopsies, are expected to provide further insights into biomarkers of response and mechanisms of resistance to SG in HR+/HER2- mBC.
Presentation numberPS4-02-24
Identifying Prognostic Epithelial-Mesenchymal Transition and Tumor Stem Cell Markers in Breast Cancer Subtypes
Vanessa Dybal Bertoni, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA, Brazil), Salvador, Brazil
V. D. Bertoni1, B. Cavalcante2, G. Rocha1, C. Gurgel2; 1Human Pathology Pos Doc Program, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA, Brazil), Salvador, BRAZIL, 2LPBM, Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA, Brazil), Salvador, BRAZIL.
Identifying Prognostic Epithelial-Mesenchymal Transition and Tumor Stem Cell Markers in Breast Cancer SubtypesDybal, Vanessa1,2,3 & Cavalcante, Bruno1,3 & Rocha, Gisele1,3,4 & Gurgel, Clarissa1, 3,41.Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA, Brazil) ;2. Oncology Multidisciplinary Assistence (AMO, DASA, BA, Brazil)3.Department of Pathology and Forensic Medicine, School of Medicine, Federal University of Bahia, Brazil4.D ́Or Institute for Research and Education (IDOR), BA,Brazil Introduction: Breast cancer (BC) remains the most prevalent malignancy among women, excluding skin tumors. Despite significant therapeutic advancements, a high relapse rate, affecting approximately 20% of all cases, persists. To improve clinical outcomes and develop novel therapeutic strategies, a deeper understanding of mechanisms driving tumor resistance, such as epithelial-mesenchymal transition (EMT) and tumor stem cell (TSC) biology, is crucial.Objective: This study aimed to identify immunohistochemically targetable markers related to EMT and TSC in different breast cancer subtypes with prognostic significance.Methods: We analyzed a public The Cancer Genomic Atlas (TCGA) breast cancer patient database encompassing clinical, histopathological, transcriptomic, and prognostic data. Bioinformatics tools were employed to identify differentially expressed genes (DEGs) impacting survival, analyze associated biological processes, construct co-expression networks, and investigate interactions with EMT and TSC-related pathways. Patients were categorized by their clinical immunohistochemical subtypes: triple-negative (TNBC), HER2-positive, and hormone receptor-positive (HR+) tumors. Protein-protein interaction (PPI) network analysis was integrated to identify the most relevant proteins within each subtype that correlated with survival.Results: Analysis of 989 BC patients from the Firehose Legacy TCGA database revealed DEGs and co-expression gene communities correlated with breast cancer clinical subtypes. Within these communities, DEGs related to EMT and TSC were identified, and those with a significant impact on survival were selected. PPI analysis pinpointed proteins within each community exhibiting a high degree of interaction and clinical endpoint impact. Following integration with genomic findings, the following potential protein markers were identified for further investigation in paraffin-embedded patient material: CCNB1 and CDCA8 (TNBC); COMP, P4HA3, and ECM2 (HER2-positive tumors); PPARG, ADIPOQ, LIPE, LEP, FABP4, DGAT2; CAV1,KLF4, FOS, IGF1, CLDN5, FOB, ALDH1A1, EGR1,PGM5, AKAP12, CAV2 (HR+ tumors). Conclusion: We identified subtype-specific prognostic protein markers associated with EMT and TSC in breast cancer. These findings are currently undergoing validation through immunohistochemical analysis of paraffin-embedded material from Brazilian patients, Brazilian patients RNA analysis, and in commercial breast cancer tissue microarray.
Presentation numberPS4-02-25
Single Institution experience of longitudinal post-surgical circulating tumor DNA monitoring in patients with HER2+ breast cancer
Minhal Zaidi, Houston Methodist Neal Cancer Center, Houston, TX
Y. Gao1, M. Zaidi2, A. Naqvi2, F. Azim2, S. Satta3, C. Brewer3, C. Palsuledesai3, E. Kalashnikova3, K. Sun4, P. Niravath4, H. Mai4, J. McKenzie3, A. Rodriguez3, M. Liu3, A. Puri4; 1Department of Hematology/Oncology, Houston Methodist Neal Cancer Center, Houston, TX, 2Department of Internal Medicine, Houston Methodist Neal Cancer Center, Houston, TX, 3Oncology, Natera, Inc., Austin, TX, 4Breast Medical Oncology, Houston Methodist Neal Cancer Center, Houston, TX.
Background: Recent studies have demonstrated the utility of longitudinal circulating tumor DNA (ctDNA) testing for monitoring treatment response and predicting long-term outcomes in patients (pts) with breast cancer (BC). In this study, we evaluated ctDNA detection rates post-surgery in pts with HER2+ BC in the adjuvant treatment and surveillance settings. Methods: This retrospective study included 59 patients with stage I-IV HER2+ BC. Commercial CTDNA testing was performed postoperatively at the physician’s discretion. A clinically validated, tumor-informed ctDNA assay (SignateraTM, Natera, Inc.) was used to detect and quantify ctDNA in blood samples collected after primary resection and serially during surveillance. ctDNA status and dynamics were correlated with clinicopathological features and surgical outcomes. Results: The cohort included 59 patients with HER2+ BC; the majority of pts had clinical T1-3 stage disease at diagnosis (T1, 25%, 15/59; T2, 20%, 12/59; and T3, 24%, 14/59), the remaining pts were T4 (7%; 4/59) or had unknown stage (24%; 14/59). Lymph node involvement was reported in 46% of pts (27/59; stage N1, 22/59; N2, 3/59; N3, 2/59); 47% had grade 3 (28/59) and 46% had grade 2 (27/59) disease. Sixty-four percent of cases (38/59) were estrogen receptor (ER)-positive; 36% (21/59) were ER-negative. Neoadjuvant treatment was received by 71% (42/59) of pts, this information was unavailable for 4% (2/59); the rest of the pts (25%; 15/59) did not receive neoadjuvant treatment. Among pts receiving neoadjuvant therapy (n=42), residual cancer burden (RCB) class was available for 27 (RCB0: 22%, 6/27; RCB1: 19%, 5/27; RCB2: 40%, 11/27; RCB3: 19%, 5/27).Among pts with longitudinal ctDNA results and complete clinical information available (n=42), ctDNA-positivity any time after surgery was observed in 14% (6/42) with a median time to ctDNA-positivity of 34 months (range: 14-792) from surgery. Of these six ctDNA-positive patients, one experienced recurrence 13 months after the first positive ctDNA test, one experienced ctDNA clearance after adjuvant therapy and remained event-free for 4.6 mos, and four remained recurrence-free with a median follow-up since ctDNA-positivity of 6.7 months and ongoing monitoring. Among patients with a favorable response to neoadjuvant treatment (RCB 0/1; n=11), no ctDNA-positivity was observed after surgery. Among pts with residual disease at surgery (RCB 2/3; n=10), ctDNA-positivity was observed in 20% (2/10). Among pts with persistent ctDNA-negativity during surveillance (n=30, median follow-up of 15.4 months, range: 5.9-50.0 months), no clinical recurrences were reported. Conclusions: In addition to clinicopathological features and surgical outcomes, longitudinal ctDNA monitoring helps identify pts with HER2+ BC who may be at high risk of recurrence. Further research is needed to assess the true utility of a ctDNA-guided treatment approach in this patient population.
Presentation numberPS4-02-26
Gper signaling as a biomarker for endocrine therapy sensitivity in pik3ca mutant hr+ advanced breast cancer
Tenghua Yu, Jiangxi Cancer Hospital & Institute, Nanchang, China
X. Zeng, Z. Sun, Q. Hu, M. Tang, C. Liu, Z. Liu, T. Yu; Department of Breast Surgery, Jiangxi Cancer Hospital & Institute, Nanchang, CHINA.
The INAVO 120 clinical trial confirmed the efficacy of first-line palbociclib combined with inavolisib in hormone receptor-positive (HR+) advanced breast cancer patients harboring PIK3CA mutations who developed resistance to adjuvant endocrine therapy. However, the comparative effectiveness of alternative CDK4/6 inhibitors (e.g., abemaciclib, ribociclib) in the first line setting, or the potential of crossline therapy (using abemaciclib or ribociclib combined with inavolisib) following palbociclib resistance in PIK3CA-mutant patients remains unclear. Our prior research identified the G protein-coupled estrogen receptor (GPER) as a pivotal initiator of endocrine resistance in HR+ advanced breast cancer, further driving activation of the PI3K/AKT/mTOR (PAM) signaling pathway. We infer that GPER signaling modulates endocrine therapy sensitivity in PIK3CA-mutant HR+ advanced breast cancer and could serve as a novel biomarker to guide personalized clinical treatment decisions. To demonstrate this hypothesis, we identified MCF-7 (E545K-mutant) and T47D (H1047R-mutant) breast cancer cell lines harboring PIK3CA mutations, and developed tamoxifen-resistant (MCF-7/TAM-R, T47D/TAM-R) and palbociclib-resistant (MCF-7/Palb-R, T47D/Palb-R) models to recapitulate first- and second-line endocrine resistance in clinic. Interestingly, GPER expression was significantly upregulated in all above resistant cell lines and promoting ABCG2 efflux pump expression via the GPER/PI3K/AKT pathway, as validated by quantitative PCR and Western blot. In GPERhigh tamoxifen-resistant cells, combinations of inavolisib with abemaciclib or ribociclib exhibited superior antiproliferative effects compared to palbociclib-based regimens, which assessed by MTT assays, flow cytometry for apoptosis, and cell cycle analysis. Knockdown of GPER/ABCG2 axis using siRNA normalized the efficacy of palbociclib, abemaciclib, and ribociclib in combination with inavolisib, underscoring GPER’s critical role in diverse CDK4/6 inhibitors sensitivity. Notably, in palbociclib-resistant cells, combinations of inavolisib with abemaciclib or ribociclib significantly outperformed inavolisib monotherapy. Furthermore, GPER/ABCG2 knockdown not only partially reversed palbociclib resistance, but also enhanced sensitivity to abemaciclib- and ribociclib-based regimens, demonstrating reduced tumor growth and improved therapeutic response upon GPER signaling inhibition. Therefore, assessing GPER signaling activation in tumor biopsies may enable precise identification of PIK3CA-mutant HR+ advanced breast cancer patients likely to benefit from specific endocrine and targeted therapy combinations. Integrating PI3K, CDK4/6, and GPER-targeted therapies represents a promising strategy to overcome endocrine resistance in the future, offering a path toward personalized treatment paradigms with improved clinical outcomes.
Presentation numberPS4-02-27
Machine learning-enhanced ultra-deep sequencing for low-abundance circulating tumor DNA (ctDNA) in breast cancer
Yuwei Ni, Burning Rock Biotech, Shanghai, China
X. Li, Y. Ni, S. Zhang, C. Yang, G. Li, Y. Zhang, X. Chen, X. Yang, J. Su, B. Li; Innovation Center, Burning Rock Biotech, Shanghai, CHINA.
Breast Cancer is a major global health challenge and remains one of the most common cancers among women. Although traditional tissue biopsies provide valuable diagnostic insights, their invasive nature makes them unsuitable for long-term monitoring. Liquid biopsy, which analyzes circulating tumor DNA (ctDNA) in blood samples, offers a minimally invasive alternative with growing potential in breast cancer care. Recent studies have demonstrated that detecting specific mutations, such as ESR1, in ctDNA can guide treatment decisions, significantly reducing disease progression and mortality. Furthermore, ctDNA monitoring can accurately predict long-term outcomes, identifying patients at low risk of relapse. These findings highlight the importance of liquid biopsy in guiding personalized treatment strategies and improving patient outcomes, even in challenging low-tumor-burden scenarios. To establish an ultra-deep targeted sequencing for low-ctDNA detection, we collected blood samples from non-cancer controls and breast cancer patients. An advanced UMI-tagging protocol was applied to process 20 – 60ng of plasma cfDNA and 100ng of WBC genomic DNA, enabling targeted enrichment of 168 cancer-related genes through designed panels. Library sequencing was conducted on the NovaSeq6000 platform, producing a raw depth of 35,000X, and a mean unique molecular depth of 4000X. A multi-step data processing pipeline was implemented to eliminate technical noise and normalize biological heterogeneity. First, reads with identical UMIs and endpoints position were used to collapse and merge to generate high-accuracy consensus sequences, effectively remove random errors introduced during PCR amplification and sequencing. Building on this, a machine-learning-driven noise model was developed to systematically identify and filter recurrent artifacts, including DNA damage patterns and sequencer-specific error signatures. Additionally, paired WBC-sequencing was applied into the workflow to distinguish somatic mutations from non-cancer biological aberrations, such as clonal hematopoiesis-derived variants, ensuring specificity in detecting tumor-associated alterations. To evaluate analytical performance, ground-truth reference materials were utilized, with subsequent confirmation through a dilution series across different tumor allele fraction (TAF) levels. The limit of detection (LoD) was rigorously assessed across different variant classes at defined input masses. For panel-wide SNVs, the median LoD is 0.25% with a 20ng input, and this threshold could be further reduced to 0.15% with a 50ng input. For panel-wide indels, the median LoD improved from 0.25% at a 20ng input to 0.14% at 50ng input. Additionally, the limit of blank (LoB) was defined from 51 cancer-free donors with paired WBC controls, resulting in an exceptionally low error rate of 4.88*10-7 for both the SNVs and indels across the entire gene panel. This panel covers whole coding regions of key breast cancer-associated genes, such as PTEN, ESR1, PIK3CA, BRCA1, BRCA2 and PALB2. To further validate precision and accuracy, a combined evaluation was performed on 44 clinical samples derived from breast cancer patients with an expanded panel. This analysis showed a positive percent agreement (PPA) of 83.0% for panel-wide SNVs, 85.7% for panel-wide indels, along with a negative percent agreement (NPA) of 99.9%. In this study, we demonstrated that the accuracy and consistency of the advanced ultra-deep mutation sequencing technologies. Combined with machine-learning tools, this approach offers a practical, non-invasive diagnostic tool for mutation profiling, applicable even when tumor levels are low.
Presentation numberPS4-02-28
Outcomes in Metastatic Young Women Breast Cancer under the Personalized Onco-Genomics Program in British Columbia.
Zahi Mitri, BC Cancer Vancouver, Vancouver, BC, Canada
P. Chapani1, N. Pryma1, N. LeVasseur1, T. Nghiem1, M. Marra2, J. Laskin1, Z. Mitri1; 1Medical Oncology, BC Cancer Vancouver, Vancouver, BC, CANADA, 2Michael Smith Genome Sciences Centre, BC Cancer Vancouver, Vancouver, BC, CANADA.
Background. Young women breast cancer (YWBC), defined as diagnosed at age <40, represent 5% of all breast cancer cases, yet remains the leading cause of cancer related death in this age group. YWBC is more aggressive and associated with worse outcomes compared to breast cancer diagnosed in women age >40 (non-YWBC). Despite this, YWBC is underrepresented and understudied. In this study, we characterize clinical and pathologic features, as well as outcomes of YWBC enrolled in the Personalized Oncogenomics (POG) program at BC Cancer. Methods. Patients with metastatic breast cancer (MBC) enrolled in POG, a personalized oncology platform aimed at characterizing the molecular landscape of advanced cancers, were included and categorized into cohorts as YWBC or non-YWBC. Data collected included patient and tumor characteristics at initial diagnosis and metastatic recurrence, germline mutations, treatments received, and survival outcomes. Clinical tumor subtypes were classified as hormone receptor positive HER2 non-amplified, HER2-amplified, and triple negative breast cancer (TNBC). As all patients enrolled in the POG program undergo whole-genome and transcriptome analysis (WGTA), we evaluated for potential targetable alterations. Results. Patient Characteristics. Between 2012 and 2025, 249 MBC patients were enrolled, with 60 (24%) YWBC and 189 (76%) non-YWBC. Median age at breast cancer diagnosis was 35 (IQR 33 – 38) for YWBC and 52 (IQR 45 – 59) for non-YWBC. In this study population, 7.6% of patients had germline mutations in BRCA, PALB2, or TP53, with no difference between the cohorts. Clinico-Pathologic Features. Compared to non-YWBC, YWBC were more likely to have stage 2-3 disease (76.7% vs 58.7%, p = 0.07), node positive disease (68.3% vs 55%, p = 0.09) and more likely to receive chemotherapy (75% vs 63.5%, p=0.3). De-novo MBC was observed in 20% of YWBC, and 24.9% of non-YWBC. The distribution of tumor subtype at initial diagnosis was similar between the 2 cohorts (p =0.46). Notably, 13% of patients had a tumor subtype switch from initial diagnosis to MBC, partly driven by an increase in TNBC in YWBC, rising from 23.3% to 35%. Treatment and Survival Outcomes. Patients in both cohorts were enrolled to POG after a median of 2 prior lines of therapy for MBC. Compared to stage 1 at initial diagnosis, stages 2 and 3 were associated with shorter time to MBC development (p=0.06 and p=0.01, respectively). There was no significant difference in median overall survival from initial breast cancer diagnosis (7.97 vs 7.53 years, log-rank test p=0.86) or from MBC diagnosis (3.13 vs 2.85 years, log-rank test p=0.47) between YWBC and non-YWBC, respectively. Compared to other subtypes, TNBC was associated with a 79% (p <0.001) and 97% (p<0.001) increase in rate of death from metastatic and initial diagnosis respectively. Additionally, there was a non-significant trend towards increase in rate of death from metastatic diagnosis in patients who had tumor subtype switch and in those with 3 or more metastatic sites. Molecular Profiling. The most common mutations identified were in the PI3K/AKT/PTEN pathway in 50%, ESR1 mutations in 18%, and somatic BRCA mutations in 16% of metastatic samples. Notably, a composite score from WGTA identified 12% of tumors with potential sensitivity to immune checkpoint inhibitor therapy. Compared to non-YWBC, there was a non-significant trend for YWBC tumors to harbor more somatic BRCA mutations, and less alterations in the PI3K/AKT/PTEN pathway.Conclusion. In our cohort, YWBC were characterized by a higher stage at initial breast cancer diagnosis and enriched for TNBC tumors at time of metastatic recurrence. Efforts to characterize matched primary tumors aim to elucidate molecular factors driving metastatic progression in YWBC, and to identify potential therapeutic targets for this high-risk group are ongoing.
Presentation numberPS4-02-29
Development of an ultra-sensitive droplet digital PCR test for monitoring ESR1 variants in liquid biopsies
Leisa Jackson, Biodesix Inc., Louisville, CO
L. Jackson, G. A. Pestano; R&D, Biodesix Inc., Louisville, CO.
Background: Estrogen receptor 1 (ESR1) mutations are a critical mechanism of acquired resistance to aromatase inhibitor (AI) therapy in hormone receptor-positive (HR+) breast cancer, occurring in approximately 20-40% of patients during treatment. The detection of ESR1 mutations in circulating tumor DNA (ctDNA) through liquid biopsy represents a new approach to real-time monitoring of treatment resistance and therapeutic decision-making. Recent interim results from the landmark SERENA-6 Phase III trial demonstrated the potential clinical utility of ctDNA-guided treatment switching, where median progression-free survival was 16.0 months in the camizestrant group versus 9.2 months in the aromatase-inhibitor group when treatment was switched based on ESR1 mutation detection. This study represented the first global Phase 3 trial to demonstrate the potential clinical utility of using ctDNA for treatment guidance and breast cancer management.Methods: We have developed a droplet digital PCR (ddPCR) test for the detection of ESR1 variants in plasma-derived cfDNA. The ESR1 ddPLEX kitted assay, in combination with a supplementary 4-plex Expert Design assay, targets the 11 most prevalent ESR1 variants (97% coverage of ESR1 mutations in breast cancer). Analytical sensitivity studies were performed using synthetic DNA controls diluted into wild-type background DNA at varying input concentrations. Serial dilutions were tested to establish the limit of detection (LOD) across different DNA input amounts, with variant allele frequencies (VAF) ranging from 0.025% to 0.2%. Results: Our studies demonstrated robust analytic performance of the ESR1 ddPCR assay across multiple input concentrations. With 30 ng cfDNA input, we achieved analytical sensitivity ranging from 0.025-0.5% VAF, enabling detection of low-level ESR1 mutations present in early resistance development. When DNA input was reduced to 10 ng, reflecting real-world clinical sample limitations, the assay maintained excellent sensitivity of 0.025-0.1% VAF. The assay also demonstrated high specificity with no false positive calls in wild-type controls.Conclusions: We have successfully developed a highly sensitive and specific ddPCR test for the detection of multiple ESR1 variants in plasma. The assay performed as designed and meets the analytical requirements required for further clinical development. The achieved sensitivity of 0.025-0.1% with as little as 10 ng of cfDNA positions this ddPCR assay as a potentially valuable tool for liquid biopsy monitoring applications in breast cancer. Clinical validation studies may further support the initial utility of ctDNA guided treatment selection demonstrated in SERENA-6. The availability of this test could enable detection of emerging resistance mutations from AI therapy in HR+ breast cancer in advance of traditional monitoring methods, e.g. imaging.
Presentation numberPS4-02-30
Scoping Review of Biologic and Imaging Markers Associated with Aromatase Inhibitor Related Musculoskeletal Symptoms
Stephanie Rosenberg, University of New Mexico, Albuquerque, NM
S. Rosenberg, T. Escobar Gil, J. Nemunaitis; Internal Medicine, University of New Mexico, Albuquerque, NM.
Introduction: Aromatase inhibitors (AIs) are standard therapy for hormone receptor-positive breast cancer in postmenopausal women but are frequently associated with musculoskeletal symptoms (AIMSS) that impair quality of life and adherence. We summarize current evidence on biologic and imaging markers associated with AIMSS, to both clarify its underlying mechanisms and identify potential predictors, with the goal of guiding future strategies to improve AI adherence. Methods: We conducted a scoping review of original studies published between 2010 and 2025 that examined biologic and imaging markers for aromatase inhibitor related musculoskeletal symptoms in breast cancer patients, according to the PRISMA guidelines. Databases include PubMed, EMBASE, COCHRANE, JAMA, and Medline. Eligible studies reported on at least one of the following: breast cancer, aromatase inhibitors, arthralgia, biomarker, biological marker, inflammatory marker, imaging, radiology. Data were extracted in duplicate and synthesized narratively. Risk of bias was assessed using the ROBINS-I tool. Results: Thirteen studies met inclusion criteria. Participants were mainly postmenopausal women aged 50-67 with early-stage hormone receptor-positive breast cancer. Most studies evaluated markers after initiation of AI therapy, though a few included baseline assessments for comparison. Biomarkers included inflammatory markers (CRP, IL6, MCP-1), metabolic and lipidomic profiles (oxylipins, 8-HETE), hormonal markers (estradiol, sex binding protein), and genetic polymorphisms (CYP19A1, ESR1, HSD17B2). One study identified BMI as a clinical risk factor, and suggested IGF-1 as a potential biomarker, though it was not directly measured. Elevated inflammatory cytokines and lower estradiol levels were associated with AIMSS in several studies. Genetic findings were inconsistent, though newer studies identified novel variants like HSD17B2. Imaging assessments varied, with some studies performing baseline imaging and others focusing on early and serial imaging after AI initiation. Imaging methods included ultrasound, MRI, and shear wave elastography (SWE), with SWE showing promise as a quantifiable tool to detect early tendon stiffness. Discussion: Our review summarizes several markers that could be associated with in AIMSS, including inflammatory and hormonal pathways, genetic variants, and early imaging changes. However, these studies were limited by small sample sizes, heterogeneous methods, and have issues with reproducibility. While some markers show promise for predicting symptoms, the current evidence mainly advances understanding of AIMSS mechanisms. Future studies should integrate these markers to develop predictive tools, with the goal of improving AIMSS and AI adherence.
Presentation numberPS4-03-01
Tumorsphere Formation as a Liquid Biopsy Biomarker of Tumor Aggressiveness and Metastatic Burden in Breast
Monika Pizon, Laboratory Pachmann, Bayreuth, Germany
M. Pizon, D. Schott, K. Pachmann; Research, Laboratory Pachmann, Bayreuth, GERMANY.
Background:Breast cancer remains a leading cause of cancer-related morbidity and mortality worldwide. Emerging evidence suggests that a subpopulation of circulating tumor cells—circulating cancer stem cells (cCSCs)—plays a pivotal role in metastasis, therapy resistance, and disease relapse. However, functional assessment of these cells in a minimally invasive manner poses a significant clinical challenge. Tumorsphere assays offer a unique in vitro platform to enrich and evaluate the self-renewal capacity of cCSCs, potentially reflecting the biological aggressiveness of the tumor. Establishing a reliable method for cultivating tumorspheres from peripheral blood may provide a novel biomarker for monitoring tumor progression and facilitating patient stratification.Materials and Methods:A total of 98 breast cancer patients, representing stages I to IV, were enrolled in this study. Peripheral blood samples were collected from each patient to detect cCSCs using the stemtrac® method, followed by a standardized tumorsphere-forming assay.Results:Tumorsphere count showed a significant correlation with disease stage and metastatic status. Patients with stage IV disease exhibited a higher median tumorsphere count compared to those with stage I (30 vs. 10; p < 0.002). Metastatic patients had more tumorspheres than non-metastatic patients (30 vs. 15; p < 0.005), with the highest counts observed in cases with multiple metastases (median 61; p < 0.001). Additionally, higher tumorsphere counts were associated with features of tumor aggressiveness, including HER2/neu positivity (35 vs. 10; p < 0.05), increased Ki-67 index, and higher histological gradeConclusion:These findings underscore the clinical relevance of tumorsphere assays as a minimally invasive method for monitoring tumor biology. The number of tumorspheres derived from peripheral blood may serve as a surrogate biomarker for disease progression, metastatic potential, and tumor aggressiveness, offering promising implications for real-time patient stratification and personalized treatment planning in breast cancer.
Presentation numberPS4-03-03
Age-driven genomic differences in breast cancer: relevance for PI3K-pathway
Nerea Lopetegui-Lia, The Ohio State University, Columbus, OH
N. Lopetegui-Lia1, A. Davenport1, J. Gill2, A. M. Roy1, S. Myers3, D. Agnese3, E. Burke3, T. Y. Andraos4, D. M. Schimming1, S. Hulett1, H. LeFebvre1, M. Gatti-Mays1, D. Stover1; 1Medical Oncology, The Ohio State University, Columbus, OH, 2Internal Medicine, Tennova North Knoxville Medical Center, Powell, TN, 3Surgical Oncology, The Ohio State University, Columbus, OH, 4Radiation Oncology, The Ohio State University, Columbus, OH.
BackgroundThe rising incidence of Breast cancer (BC) in young women requires further genomic characterization, as only a fraction carry a germline mutation. Given the growing interest in therapies targeting the phosphoinositide 3-kinase/AKT (PI3K/AKT), mammalian target of rapamycin (mTOR), and phosphatase and tensin homolog (PTEN) pathways, central to BC pathogenesis, we evaluated whether PI3K/AKT/mTOR pathway alterations and gene expression differed by age at BC diagnosis.Methods: In this analysis of patients’ tumor/germline whole exome DNA and RNA sequencing data from the Ohio State University Total Cancer Care registry, 71 profiled patients were young adults (YA) aged < 40 years, and 675 were non-YA aged ≥40 years. Descriptive statistics characterized the frequency and co-occurrence of BC-specific alterations and mRNA expression profiles by age groups. ResultsThe prevalence of these BC-relevant mutations are reported in Table1. PIK3CA mutation occurred more frequently in non-YA patients (16.9% YA versus (vs) 39% non-YA; p<0.0002). The frequency of driver mutations were similar between cohorts (73% in YA vs 82% in non-YA). Most hotspot mutations were enriched in non-YA group, particularly H1047R (15.11% n=102 non-YA vs 4.23%, n=3 YA; p= 0.01). Others include: E545K (6.52%, n=44 vs 2.82%, n=2, p=0.302), E542K (4.59%, n=31 vs 1.41%, n=1; p=0.352); H1047L (1.93%, n=13 vs none, p=0.625) and N345K (1.78%, n=12 vs 1.41% n=1; p=1.00). There were no age-based differences in AKT1 mutation (5.6% YA vs 10.8% non-YA patients; p=0.220). Hotspot alterations E17K, E17G, and L52R were numerically enriched, albeit nonsignificant, in non-YA. There were no significant differences in the presence of mTOR by age (16.9% YA and 21.5% non-YA; p=0.450). Most alterations in both groups were variants of unknown significance (17/17 in YA, 157/159 in non-YA). YA and non-YA cohorts had similar frequency of PTEN mutations (12.7% YA vs 10.5% non-YA; p=0.550). The proportion of PTEN driver mutations also did not differ (82% YA and 93% non-YA). Conclusion PIK3CA mutations were more prevalent among non-YAs highlighting potential age-associated oncogenic drivers. Although AKT1, mTOR, and PTEN gene mutations did not differ between age groups, in general, oncogenic/hotspot mutations occurred more frequently in patients ≥40 years. As management of BC evolves to incorporate PI3K/AKT/mTOR inhibitors, age-specific somatic alterations may uncover therapeutic vulnerabilities and inform customized treatment. Table:
| Mutation | Age Group | % of Patients with Mutation | Fisher’s Exact Test p-value (<40 vs ≥ 40) | ||||
| PIK3CA | <40 | 16.9% (12/71) | 0.0002 | ||||
| ≥ 40 | 39% (263/675) | ||||||
| AKT1 | <40 | 5.6% (4/71) | 0.220 | ||||
| ≥40 | 10.8% (73/675) | ||||||
| mTOR | <40 | 16.9% (12/71) | 0.450 | ||||
| ≥40 | 21.5% (145/675) | ||||||
| PTEN | <40 | 12.7% (9/71) | 0.550 | ||||
| ≥40 | 10.5% (71/675) |
Presentation numberPS4-03-04
Hmgcs2 promotes malignant phenotype and apatinib resistance in her2-negative breast cancer via the rab23/β-catenin/tcf signaling axis
Jing Wu, Chongqing University Cancer Hospital, Chongqing, China
J. Wu1, M. Liu1, W. Li1, N. Zhang1, Y. Liu1, Y. Zhao2, X. Zeng1; 1Department of Breast Center, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, Chongqing University Cancer Hospital, Chongqing, CHINA, 2Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatmen, Chongqing University Cancer Hospital, Chongqing, CHINA.
Background Existing studies have primarily focused on the clinical efficacy of Apatinib, a selective VEGFR2 inhibitor, leaving a significant gap in understanding the biological mechanisms underlying its resistance. In this study, we identify 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), a rate-limiting enzyme in ketogenesis, as a novel biomarker associated with Apatinib resistance in HER2-negative breast cancer . Furthermore, we investigate its biological function and molecular mechanism.Methods Metabolomics analysis and immunohistochemical verification were performed on Apatinib-treated clinical samples to identify genes associated with Apatinib resistance. The biological role of these genes in HER2-negative breast cancer was validated through in vitro cell experiments and in vivo animal studies. The specific molecular mechanisms involved were elucidated using proteomics analysis , along with techniques such as co-immunoprecipitation and western blotting.Results HMGCS2 was found to be overexpressed in Apatinib-refractory tumors and was associated with resistance to Apatinib. Compared to normal mammary epithelial cells, HMGCS2 expression levels were significantly elevated in breast cancer cells. Knockdown of HMGCS2 via shRNA effectively suppressed tumor proliferation and invasion in HER2-negative breast cancer cell lines and reduced tumor burden in xenograft mouse models. Conversely, overexpression of HMGCS2 markedly enhanced malignant phenotypes in vitro. Mechanistic investigations revealed that RAB23-dependent β-catenin/TCF activation served as the primary driver of carcinogenesis.Conclusion Our study revealed a critical role of HMGCS2 in mediating Apatinib resistance in HER2-negative breast cancer. Mechanistically, HMGCS2 promoted malignant progression in HER2-negative breast cancer by activating the β-catenin/TCF transcriptional axis through RAB23-mediated signaling, highlighting HMGCS2 as a potential therapeutic target for precision oncology.
Presentation numberPS4-03-06
Aberrant KIF11 induction is a potential driver of aggressiveness in ER-a positive positive breast cancer
Seungpil Jung, Korea University Hospital, Seoul, Korea, Republic of
S. Jung1, J. Yisun2, S. Yang1, Y. Sun Young2, Y. Ji Young1, L. Eun-Shin1, O. Harim3, S. Jongmin3, K. Sangmin2, L. Jeong Eon2; 1Department of Surgery, Korea University Hospital, Seoul, KOREA, REPUBLIC OF, 2Department of Breast Cancer Center, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 3Department of Pathology, Korea University Hospital, Seoul, KOREA, REPUBLIC OF.
Kinesin family member 11 (KIF11), also known as Eg5, is a motor protein critical for mitotic spindle formation and proper cell division. While KIF11 has been implicated in promoting cell proliferation and invasion in various cancer types, its regulatory mechanisms in breast cancer remain poorly understood. In this study, we investigated the regulation of KIF11 expression and assessed the therapeutic potential of its specific inhibitor, ispinesib, in breast cancer models. Analysis of the GSE4922 dataset revealed that estrogen receptor-positive (ER+) breast cancer patients with high KIF11 expression exhibited significantly poorer survival compared to those with low expression. Moreover, KIF11 levels were elevated in lymph node-positive (LN+) patients and those with advanced-stage disease, suggesting its potential role in tumor progression. We further found that KIF11 expression was positively correlated with MAPK1 expression and negatively correlated with STAT3 expression. Pharmacological inhibition of MEK1/2 using binimetinib significantly reduced KIF11 expression, whereas overexpression of constitutively active MEK1 (CA-MEK1) increased its expression. KIF11 knockdown (KD) also induced G2/M phase arrest and inhibited cell growth in MCF7 breast cancer cells. Finally, we evaluated the pharmacological efficacy of ispinesib in regulating the cell cycle and cell growth. As expected, ispinesib induced G2/M phase arrest and completely inhibited cell growth in ER+ breast cancer cells. These findings support KIF11 as a prognostic marker and potential therapeutic target in ER+ breast cancer, regulated at least in part by the MAPK signaling pathway.
Presentation numberPS4-03-07
Association of clinical BRCA testing with multigene assay results
Seungji Lee, International St. mary’s hospital, Incheon, Korea, Republic of
S. Lee1, S. Lee1, I. Lee1, S. Park2, J. Kim3, N. Son4; 1Department of surgery, International St. mary’s hospital, Incheon, KOREA, REPUBLIC OF, 2Division of Breast surgery, Department of surgery, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 3Yonsei University College of Medicine, Yongin, International St. mary’s hospital, Yongin, KOREA, REPUBLIC OF, 4Department of Statistics, Keimyung University, Daegu, KOREA, REPUBLIC OF.
PurposeBRCA mutations significantly increase the lifetime risk of breast cancer. While they are strongly associated with triple-negative breast cancer, they are also found in hormone receptor-positive, HER2-negative subtypes, in which multigene assays are commonly used for risk stratification and treatment decision-making. However, the relationship between BRCA status and multigene assay outcomes remains unclear, particularly in Asian populations. This study investigates the association between clinical indications for BRCA testing and multigene assay results in Korean breast cancer patients. Methods This multicenter study included 2,542 patients with hormone receptor-positive, HER2-negative breast cancer who underwent multigene testing between 2013 and 2024. BRCA testing was performed in patients aged under 40 years, those with bilateral breast cancer, or those with a personal or family history of breast or ovarian cancer within third-degree relatives. Multi-gene assays included Oncotype DX, MammaPrint, EndoPredict, and OncoFREE, and each result was classified into high-risk or low-risk groups based on each test-specific criteria. Statistical analyses were performed using chi-square tests and logistic regression in SPSS. Results Patients were categorized as BRCA-tested group (n=798) or non-tested group (n = 1,596). In the BRCA-tested group, 19.5% were classified as high risk and 80.5% as low risk by multigene assay. pTstage, pNstage, histologic grade, progesterone receptor status, and Ki-67 level was independently associated with high risk and multigene assay result. BRCA mutation was identified in 44 patients (7 BRCA1 mutation, 37 BRCA2 mutation). BRCA mutation was independently associated with high-risk classification on multigene assay (OR 3.554, 95% CI 1.767-7.151, p < 0.001). In subgroup analysis of BRCA mutation group, among 30 patients with a family history, 20 (66.7%) were classified as high risk. Multivariate analysis also confirmed that familial history of breast or ovarian cancer was independently associated with high-risk in BRCA-mutation patients. (OR 9.752, 95% CI 1.449-65.651, p <0.05). An association between BRCA testing indications and high-risk multigene assay results was not statistically significant when only one indication was present. Conversely, the presence of two or more indications demonstrated a potential correlation with high-risk classification. (OR 5.282, p <0.061) Conclusion In Korean patients with hormone receptor-positive, HER2-negative breast cancer, BRCA mutation was significantly associated with high-risk classification on multigene assay. In BRCA-mutated patients, the presence of a family history further increased the likelihood of high-risk classification. Additionally multiple BRCA testing indications predicted high-risk classification in BRCA mutation patients, but this trend was not observed in those without a detected mutation.
Presentation numberPS4-03-08
The role of platelets in masking circulating tumor cells
Monika Pizon, Laboratory Pachmann, Bayreuth, Germany
D. Schott, M. Pizon, A. Riess, K. Pachmann; Research, Laboratory Pachmann, Bayreuth, GERMANY.
Background:The most critical and life-threatening feature of malignant tumors is their ability to metastasize to distant organs, ultimately leading to patient mortality. Most metastases become clinically detectable only months or even years after the surgical removal of the primary tumor, indicating that they originate from tumor cells disseminated into the circulation and persisting in the body as minimal residual disease (MRD).It is widely assumed that the majority of circulating tumor cells (CTCs) rapidly disappear from the bloodstream following surgery, with a reported half-life of only a few hours. However, some of these cells clearly survive, either by recirculating or by entering protective niches within the body. Using the standardized maintrac® method, we demonstrated that substantial numbers of viable tumor cells can still be detected in peripheral blood several days after surgical intervention in patients with both lung and breast cancer. These findings strongly suggest that a fraction of CTCs is indeed capable of escaping rapid elimination.This raises an important question: why do most conventional CTC detection methods fail to identify these cells, both before and after surgery? How do tumor cells released into the bloodstream manage to remain undetected?Methods:Using fluorochrome- labeled anti-EpCAM antibody cells were dyed with the maintrac® approach right after blood draw and after resting the samples for up to 24 hours at room temperature on the shelf. At all times the presence of platelets was monitored using red fluorescing anti CD36 or anti-CD41/CD61 antibody.Results: Immediately after blood draw, no EpCAM-positive (green-fluorescing) CTCs were detectable. However, clusters of red-fluorescing platelets were observed, which, under transmitted light, were shown to be adherent to the circulating tumor cells. After 24 hours of resting at room temperature, a substantial number of viable EpCAM-positive tumor cells became detectable, with platelets visibly detached. Time-lapse imaging over 24 hours revealed progressive detachment of platelets from tumor cells, accompanied by increased anti-EpCAM staining intensity. These findings indicate that platelets form a cloak around circulating tumor cells in circulation, physically obstructing antibody binding and thereby preventing detection. Only upon platelet detachment could the tumor cells be reliably identified.Conclusion:Circulating tumor cells may evade detection by being cloaked in platelets. This platelet-mediated masking may not only hinder diagnostic detection but could also play a functional role in immune evasion and metastasis formation. Platelets expressing immune checkpoint molecules like PD-L1 may further suppress anti-tumor immune responses. These findings underscore the importance of considering platelet interactions in liquid biopsy techniques and highlight a potential target to improve cancer diagnostics and therapy.
Presentation numberPS4-03-09
Positron emission tomography imaging detected enhanced anti-tumor immunity in triple-negative breast cancer mediated by glutamine antagonism
Hoon Choi, University of Pennsylvania, Philadelphia, PA
H. Choi1, B. Hassan1, R. O’Connor2, F. Michael1, D. Mankoff1, R. Zhou1; 1Radiology, University of Pennsylvania, Philadelphia, PA, 2Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA.
OBJECTIVES: Glutaminolysis is a metabolic pathway deployed by aggressive cancers including triple-negative breast cancer (TNBC) to support their growth and progression. Immune checkpoint inhibitors (ICI) are FDA approved treatment modalities for TNBC; however, treatment responses are modest and variable. To overcome the heterogeneity in response, there is a critical need to understand the unique characteristics underlying immune activity responding to ICI. Our prior study has shown that TNBC tumors are enriched with myeloid-derived suppressor cells (MDSCs), which were significantly reduced by a pan-glutamine metabolic inhibitor JHU083, a pool compound of DRP-104/ Sirpiglenastat being evaluated in clinical trials. JHU083/DRP-104 is the prodrug form of DON (6-Diazo-5-oxo-L-norleucine). Here we examined the impact of glutamine antagonism in cancer versus immune cells traversing the tumor microenvironment. PET imaging is an important tool to identify patients most suited to the therapeutic promise of ICI. Leveraging the recent development of a radio-labeled antibody against CD69, a well characterized early-activation cell surface marker, we tested whether this immune-PET marker can detect the enhanced activation of T cells in the TNBC tumor immune microenvironment. METHODS: Murine (4T1, EO771) and human (HCC1806) TNBC and ER+ (MCF7) cell lines obtained from ATCC, T cells and MDSCs isolated from the spleen, were treated with JHU083 (0.1µM) to assess ROS and lipid ROS levels using Dihydroethidium and C11BODIPY assays. The impact of MDSC on T cell activation was examined by co-culturing T cells with MDSCs for one day in culture media containing 0.05 µg/ml PMA and 0.1 µg/ml ionomycin and either 0.1 µM DON or without DON. For in vivo imaging studies, 4T1 cells were inoculated into the mammary fat pad of BALB/c mice. When tumors grew to approx. 100 mm3, mice were randomized and enrolled in control (no treatment), ICI (treated with PD-1 at 250 and CTLA4 at 100 μg/mouse twice/week i.p.) or ICI plus JHU083 (1.82 mg/kg, i.p. every other day from day 0) group. After one week treatment, each mouse received a 100 ~ 300 µCi of [89Zr]-DFO-H1.2F3 (anti-CD69 mAb) via tail vein injection and PET scan was performed at 48 and 72 hours post-injection. After PET imaging, FACS analysis was performed to profile immune cells in the tumor. RESULTS: 0.1 µM DON does not induce cytotoxicity in TNBC cells, MDSCs or T cells, however it markedly increases cellular reactive oxygen species and lipid peroxidation in cancer cells and MDSCs but not in the activated or naïve T cells. CD69 PET signals (%ID/g) in 4T1 tumors are significantly higher after ICI (13%, p<0.05) or JHU083 plus ICI treatment (15%, p<0.05) compared to controls (9%). FACS analysis of tumors after PET imaging reveals elevated CD69+ cell population in treated tumors compared to controls. Meanwhile the CD45+CD11b+ cell fraction in ICI treated tumors (76%) remains similar to controls (84%) but decreased substantially after JHU083 plus ICI treatment (19%). In co-cultures, the presence of MDSCs reduced the fraction of CD69+ and granzyme B+ CD8+ T cells, confirming MDSC’s immune suppressive activity. However, adding 0.1 µM DON in co-culture reversed the functional competence of T cells. DISCUSSIONS & CONCLUSION: By selectively disrupting metabolism in cancer cells and MDSCs while sparing T cells that provide anti-tumor control, glutamine antagonism has immediate translational relevance for adoptive immunotherapies. Although ICI therapy removes PD-1 and CTLA4 mediated inhibition on T cells, it fails to inhibit tumor growth likely due to predominant presence of MDSCs which suppress CD8+ T cells as demonstrated by co-culture data. Combining JHU083 with ICI reduces MDSC infiltration to the tumor, leading to enhanced anti-tumor immunity detected by [89Zr]CD69 PET.
Presentation numberPS4-03-10
Assessing the impact of real-time serial circulating tumor cell results on the management of patients with metastatic breast cancer
Karthik V Giridhar, Mayo Clinic, Rochester, MN
K. V. Giridhar1, D. Zahrieh2, M. Hanna3, A. Singh4, E. Al-Hattab5, P. Schumacher2, J. Carroll1, J. Hoppenworth1, T. Haddad1, A. Tevaarwerk1, B. Rudder4, G. M. Choong1, S. Basu5, R. A. Leon-Ferre1, T. O’Brien4, A. Lewis3, F. de Snoo6, L. Stork-Sloots6, F. K. Kuhr7, S. Lazaro8, R. Prendergast8, D. S. Spinner9, M. P. Goetz1; 1Medical Oncology, Mayo Clinic, Rochester, MN, 2Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 3Medical Oncology, Mayo Clinic Health System, Albert Lea, MN, 4Medical Oncology, Mayo Clinic Health System, Mankato, MN, 5Medical Oncology, Mayo Clinic Health System, Eau Claire, WI, 6Medical Affairs, Medex15, Amsterdam, NETHERLANDS, 7Medical Affairs, Menarini Silicon Biosystems, Huntingdon Valley, PA, 8Clinical Laboratory, Menarini Silicon Biosystems, Huntingdon Valley, PA, 9Market Access & Reimbursement, Menarini Silicon Biosystems, Huntingdon Valley, PA.
Introduction Serial assessments of circulating tumor cells (CTCs) in metastatic breast cancer (MBC) provide independent prognostic significance for progression free survival and overall survival. A low CTC burden (<5 CTCs / 7.5mL blood) at baseline is favorable prognostically, and reduction of CTCs during treatment indicates response to treatment (Janni, CCR 2025). Despite increasing evidence supporting its clinical validity, real-time serial CTC enumeration has not been utilized broadly in clinical practice in the U.S. The ACT-MBC study is a prospective observational study to evaluate the impact of serial CTC testing on treatment decisions and response assessment (NCT05662345). Provider (physician or advanced practitioner) perception of CTC testing is measured with questionnaires at baseline (PQ1), and for each pt during the first restaging visit (PQ2), and at the end of the study (PQ3). We previously reported PQ1 results (Giridhar, SABCS 2024). Here, we report on results of PQ2. Methods ACT-MBC prospectively enrolled patients (pts) at Mayo Clinic Rochester and Mayo Clinic Health Systems sites (Mankato, Albert Lea and Eau Claire). Pts with MBC were enrolled at time of progressive disease during any line of therapy (TNBC) or ≥2nd line metastatic therapy for ER+/HER2- disease. Pts were tested for CTCs using the FDA-cleared CellSearch® assay which included enumeration and biomarker testing on CTCs for ER, HER2, and PD-L1. The treating clinician determined the regimen and timing of restaging scans. CTC testing was performed once per cycle (every 3-4 weeks) with results returned to the treating provider within 7 days. We descriptively summarized PQ2 responses from providers who had at least one on-treatment CTC result available by the first restaging visit. We report the proportion (90% CI) of providers who agreed or strongly agreed that assessing CTCs enhanced their ability to determine response to therapy and that the CTC results helped guide their decision to continue or change therapy (co-primary endpoints) while accounting for within provider correlation. Results As of June 25, 2025, 15 providers enrolled a median of 2 pts each (range: 1 to 15 pts) and completed PQ2 surveys on 40 pts. 7 providers (47%) practiced in the community setting and 4 (27%) were nurse practitioners. Most pts were female (98%), White (98%), and had ER+/HER2- MBC (80%). Baseline (C1) CTC results were elevated (≥5 CTCs) in 24 (60%) pts (CTC range 0-609). At C2 (n=39), CTC values remained undetected in 7 pts, decreased in 21 pts, increased in 8 pts, and remained unchanged in 3 pts. At PQ2 (n=40), based on clinician interpretation of radiographic findings, results showed 2.5% of pts with complete response, 20% with partial response, 25% with stable disease, 17.5% with mixed response, and 35% with progressive disease. Clinicians reported that CTC results were concordant with imaging results in 34 (85%) responses, discordant with imaging in 7.5% of responses, and unclear in the remaining 7.5% of responses. Most clinicians (82.5%) agreed or strongly agreed that assessing CTCs enhanced their ability to determine response to therapy (90% CI: 68.3%, 96.7%). Further, 70% of providers agreed or strongly agreed that the CTC results helped guide their decision to continue or change therapy (90% CI: 50.8%, 89.2%). 32.5% of the time, CTCs results influenced the timing of the first restaging scans. In 11/25 pts without progressing disease, CTC results influenced the timing of future scans. In the 7/8 pts with a mixed response at first restaging scans with decreasing CTCs, 4 remained on the same systemic therapy. Conclusions Oncologists in academic and community practice responded that serial CTC assessments enhance ability to determine response of therapy, guide decisions to continue or change treatments, and influence timing of radiographic assessments.
Presentation numberPS4-03-11
Uncovering ctDNA-detected co-mutational patterns in HR+/HER2− metastatic breast cancer to inform treatment sequencing
Konstantinos Venetis, European Institute of Oncology IRCCS, Milan, Italy
K. Venetis1, A. Marra2, D. Presti3, D. Trapani2, V. Peruzzo1, R. Adorisio1, G. Castellano2, A. Ranghiero1, A. Borghi1, D. Vacirca1, E. Giordano2, S. Perazzo2, K. Qeraj2, P. Zagami2, M. Lombardi1, E. Munzone3, G. Curigliano2, E. Guerini Rocco1, N. Fusco1; 1Division of Pathology, European Institute of Oncology IRCCS, Milan, ITALY, 2Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, ITALY, 3Division of Medical Senology, European Institute of Oncology IRCCS, Milan, ITALY.
Introduction: Clinically actionable mutations in ESR1 and genes within the PI3K/AKT/PTEN pathway inform the use of selective estrogen receptor degraders (SERDs), PI3K inhibitors, and AKT inhibitors in patients (pts) with hormone receptor-positive, HER2-negative (HR+/HER2−) metastatic breast cancer (mBC). While the prevalence of such mutations detected through circulating tumor DNA (ctDNA) varies by treatment type and line, the broader landscape of co-occurring actionable mutations remains poorly characterized. Methods: Clinical and genomic data were retrospectively collected from patients with HR+/HER2− mBC who underwent ctDNA testing at the European Institute of Oncology (IEO) between November 2023 and June 2025, upon progression to endocrine therapy plus CDK4/6 inhibitors. Genomic profiling was performed using a 77-gene liquid biopsy (LB)-based next-generation sequencing (NGS) assay. Progression-free survival (PFS) and overall survival (OS) were compared between groups using Cox proportional hazards models. Results: Among the 256 patients who underwent ctDNA testing, 53% were tested after first-line (1L) therapy, 24% after second-line (2L), and 23% after third-line or beyond (≥3L). PIK3CA mutations were detected in 33% of pts (n=84), ESR1 in 32% of pts (n=82), PTEN in 5.9% of pts (n=15), and AKT1 in 3.9% of pts (n=10). PIK3CA mutations were more frequently identified in patients tested at ≥3L, while ESR1, PTEN, and AKT1 alterations were more frequently observed in patients tested at 1L. However, these distributional differences across treatment lines did not reach statistical significance. Other clinically relevant genes mutations were also observed in TP53 (n=47, 18.4%), ERBB2 (n=16, 6.3%), and RB1 (n=10, 3.9%). Co-mutations in both ESR1 and PIK3CA were identified in 30.4% (n=25) of ESR1-mutant patients, more frequently when tested after the 1L (31.2%) and ≥3L (42.1%). ESR1 co-mutation with AKT1 and PTEN was less frequent, occurring in 6.1% and 3.6% of ESR1-mutant patients, respectively. To explore the clinical relevance of these molecular profiles, PFS and OS were compared across three molecular subgroups: (i) ESR1-mutant/PI3K-pathway-wild-type, (ii) ESR1-wild-type/PI3K-pathway-mutant (PIK3CA, AKT1, or PTEN), and (iii) double-mutant (ESR1-mutant/PI3K-pathway-mutant). No associations between co-mutations and clinical outcomes were found. Conclusions: ctDNA-guided molecular profiling is a fundamental tool for guiding clinical decisions among patients with HR+/HER2− who experience progression on ET. In our cohort, co-mutations occurred in almost 1/3 of patients and seems not to influence prognosis. More data are needed to clarify the prognostic role of co-mutations and to determine the most appropriate molecular driver for therapeutic choices.
Presentation numberPS4-03-12
Quantifyher (trial in progress): quantitative immunofluorescence and/or messenger rna to measure her2 expression in patients with metastatic breast cancer treated with trastuzumab deruxtecan (tbcrc066)
Eleanor Plaut Taranto, University of Pennsylvania, Philadelphia, PA
E. P. Taranto1, W. Fawn1, C. Desirae1, E. Walinsky1, S. Deluca1, L. Bayne1, E. P. Wileyto2, A. Shirali3, C. J. Robbins3, Y. Abdou4, J. Anampa5, M. Balic6, J. V. Canzoniero7, N. C. Chen8, F. Howard9, L. Huppert10, A. Kahn11, C. Mainor12, J. Mouabbi13, V. Nair14, T. Degeer15, J. Weidler16, T. McNicholas15, C. Serway17, J. Reischl18, M. Bates16, R. Monroe17, D. L. Rimm3, A. DeMichele1; 1Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, PA, 2Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, 3Department of Pathology, Yale School of Medicine, New Haven, CT, 4Division of Oncology, University of North Carolina School of Medicine, Chapel Hill, NC, 5Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, New York, NY, 6Division of Oncology, Hillmans Cancer Center, University of Pittsburgh, Pittsburgh, PA, 7Division of Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, MD, 8Department of Medicine, Section of Hematology and Oncology, Baylor College of Medicine, Houston, TX, 9Biological Sciences Division, Section of Hematology/Oncology, University of Chicago, Chicago, IL, 10Department of Medicine, University of California San Francisco, San Francisco, CA, 11Department of Medicine, Division of Medical Oncology and Hematology, Yale School of Medicine, New Haven, CT, 12Department of Internal Medicine, Division of Hematology/Oncology, Georgetown University School of Medicine, Washington, DC, 13Department of Breast Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, 14Division of Hematology/Oncology, Fred Hutch/University of Washington School of Medicine, Seattle, WA, 15Open Innovation, Leica Biosystems, Richmond, IL, 16Medical/Scientific Affairs and Strategy, Oncology, Cepheid, Sunnyvale, CA, 17Oncology, Danaher Diagnostics, Washington, DC, 18External Innovation Diagnostics, Danaher Diagnostics, Washington, DC.
Background: Antibody drug conjugates (ADCs) can be effective therapeutic options for some patients (pts) with metastatic breast cancer (MBC). However, given serious potential toxicities, identifying pts most likely to benefit from ADCs is critical to optimizing quantity and quality of life. Trastuzumab-deruxtecan (T-DXd), a HER2-targeted ADC, is approved for pts with MBC expressing low/ultra-low levels of HER2 by immunohistochemistry (IHC 1+/2+, FISH-neg, or IHC 0 with faint membrane staining in 400 retrospective samples, ~50% of HER2 IHC 0 cases had HER2 QIF levels above the limit of quantification, with a significant relationship demonstrated between HER2 QIF and time-to-next-treatment in a retrospective cohort of pts treated with T-DXd. Retrospective data using Cepheid’s Xpert Breast Cancer STRAT4 mRNA assay have also demonstrated high ERBB2 concordance with HER2 IHC, with potentially greater range. Both assays hold promise to improve measurement of surface protein expression at lower limits of detection than traditional IHC and may better select pts likely to respond to T-DXd. The primary objective of QuantifyHER is to determine the relationship between HER2 QIF and/or ERBB2 mRNA levels and real-world objective response (rwOR) to T-DXd. Secondary objectives include evaluating the relationships between TROP2 and rwOR, HER2 QIF/mRNA and real-world progression-free survival, determining response thresholds and the relationship with ER status, and evaluating a joint QIF/mRNA approach. Methods: QuantifyHER (TBCRC066/NCT06551116) is a multi-site prospective cohort study. Key eligibility criteria include biopsy-proven HER2 IHC<2+ MBC, available tissue, measurable disease, and initiation of T-DXd within 30 days of registration. Pts receive T-DXd per standard of care and are followed for real-world endpoints. Up to 10 unstained FFPE/1 H&E section from an IHC <2+ archival specimen (most recent metastatic biopsy preferred) are sent to the Quantitative Diagnostics in Anatomic Pathology (QDAP) CLIA lab at Yale for analysis with both TROPLEX (returning HER2 and TROP2 QIF levels in amol/mm2) and the Cepheid Xpert STRAT4 Research Use Only (RUO) assay (returning ERBB2 mRNA delta Cycle threshold (dCt) levels). Patient data (demographics, clinical features, prior treatment, follow-up) are abstracted from the medical record, with clinical endpoints assessed by local investigators using real-world response categories and toxicities collected only as related to T-DXd discontinuation or modification. The planned sample size is 200 evaluable pts, providing 80% power (with a 2.5% 1-sided alpha), to detect a difference in proportions of 9-10 % per standard deviation QIF/mRNA increase, corresponding to an odds ratio of 1.6. A non-binding interim review at n=70 will assess preliminary biomarker distribution for any necessary sample size adjustment. QuantifyHER is open within the Translational Breast Cancer Research Consortium (TBCRC). As of 7/1/25, 10/12 planned sites are open and 23 pts have enrolled.
Presentation numberPS4-03-13
Septin9 isoforms predict taxane efficacy in triple negative breast cancer
Jycole Bush, UT Health Science Center San Antonio, San Antonio, TX
J. Bush1, J. Essif1, K. Lathrop2, R. Kannaiyan2, A. L. Risinger1; 1Cancer Pharmacology, UT Health Science Center San Antonio, San Antonio, TX, 2Oncology, UT Health Science Center San Antonio, San Antonio, TX.
Microtubule stabilizing taxanes are a mainstay of the treatment of breast cancer in both the adjuvant and neoadjuvant settings, including as backbone chemotherapy in combination with targeted agents. Despite their ubiquitous use and well-understood mechanism of action, there are currently no biomarkers to inform which tumors are most likely to respond to taxane treatment. A class of cytoskeletal associated small GTPases critical for multiple oncogenic processes, the septins, can directly interact with microtubules in a taxane-dependent manner with distinct septin9 isoforms having opposing effects. The longest isoform, sept9_i1, is associated with tumor progression, promoting oncogenic activity and taxane resistance through direct binding to microtubules. In contrast, the sept9_i2 isoform lacking this microtubule binding domain reduces migration with lower levels in tumors as compared to normal tissue. We hypothesized that the relative expression of these mechanistically distinct septin9 isoforms that differentially interact with microtubules could serve as a clinical biomarker of taxane response. We conducted a retrospective analysis of the relationship between septin9 isoform expression in tumor biopsies and the overall survival of taxane-treated breast cancer patients post-metastatic diagnosis. Septin9 isoform expression did not differ between primary and metastatic biopsies or between tumors of ductal versus lobular origin. However, tumors from triple-negative breast cancer (TNBC) patients expressed significantly lower levels of septin9_i1 and higher levels of septin9_i2 than ER and HER2 positive breast tumors. Elevated expression of septin9_i2 was correlated with a significant increase in overall survival specifically in TNBC patients with no correlation observed for septin9_i1. To further interrogate the role of septin9 isoform expression in the response of breast tumors to taxane-containing neoadjuvant chemotherapy we conducted a prospective clinical study in previously untreated patients where isoform expression was evaluated in pre and post-treatment biopsies and correlated with pathological response. Consistent with the retrospective study, we found a trend toward elevated levels of septin9_i2 expression in patients who had a pathologic complete response (PCR) to taxane-containing chemotherapy and a reduction in expression post-treatment in patients without a PCR. Comparative studies found a high correlation between septin9 isoform expression at the mRNA level using qRT-PCR and the protein level by immunohistochemistry using isoform-specific antibodies.To further interrogate the potential underlying mechanism of septin9 isoforms in the response of breast cancer cells to taxane treatment we generated isogenic breast cancer cell lines where specific isoforms were stably knocked down or overexpressed. Consistent with published results, we found that reduced septin9_i1 slightly improves the efficacy of taxane treatment in vitro however the beneficial role of septin9_i2 observed in the clinical studies was not evident in vitro. We further interrogated how alterations in septin9 isoform expression regulate the ability of taxanes to stabilize cellular microtubules and demonstrate that drugs that differentially alter microtubule structure and dynamics effectively shift the association of oncogenic septins between the microtubule and actin cytoskeleton. Together, these data support further interrogation of the role of septin9 isoforms in tumor models and clinical samples to determine whether septin9_i2 has a causal role in the response of tumors to taxane-containing chemotherapy and, if so, identify whether clinically approved chemotherapeutics that work through distinct mechanisms of action would have benefit in tumors with reduced expression of this isoform.
Presentation numberPS4-03-15
Repurposing eliglustat as an anti-cancer agent for triple-negative breast cancer
Johannes Francois F Fahrmann, UTMDACC, Houston, TX
J. Vykoukal1, Y. Chen1, M. Zuo1, R. Ballaro1, M. Hong1, H. Krishna1, D. Rodriquez-Perera1, H. Katayama1, E. Irajizad2, R. Wu1, R. León-Letelier1, J. Dennison1, A. Gutierrez3, A. Paulucci-Holthauzen4, Y. Cai1, F. Hsiao1, S. Park1, B. Arun3, S. Hanash1, J. F. Fahrmann1; 1Clinical Cancer Prevention, UTMDACC, Houston, TX, 2Biostatistics, UTMDACC, Houston, TX, 3Breast Medical Oncology, UTMDACC, Houston, TX, 4Genetics, UTMDACC, Houston, TX.
Triple-negative breast cancer (TNBC) represents a heterogeneous subtype of breast cancer characterized by high pathological grade, strong invasiveness, local recurrence, high metastasis rate, and poor prognosis. Chemotherapy and in some cases, immunotherapy, remain the main choice of systemic treatment. However, response is variable, lacks durability, and acquired resistance to chemotherapy is a frequent manifestation. There remains an unmet need to establish novel alternative therapies that can both exert targeted effects on cancer cells as well as stimulate an anti-cancer immune response. We have uncovered an onco-metabolic biochemical feature in TNBC wherein cancer cells increase chemical conversion of sphingomyelin to ceramide with subsequent glycation to glycosphingolipid derivatives. Our mechanistic studies have established that this biochemical program is essential for survival mitophagy. The activation state of this program in cancer cells opens a metabolic vulnerability that is targetable through biochemical inhibition of glucosylceramide synthase (UGCG), a rate-initiating enzyme that mediates glycation of ceramides to glycosphingolipid derivatives. Our preclinical studies using orthotopic syngeneic mouse models of Brca-deficient TNBC as well as human cell line and patient-derived xenograft models of TNBC have demonstrated that targeting of cancer cell UGCG via repurposing eliglustat, an FDA-approved selective UGCC inhibitor for long-term use in patients with Gaucher’s disease, at clinically achievable doses elicits potent anti-cancer effects by targeting cancer cell mitochondria, resulting in accumulation of mitotoxic ceramides, induction of compensatory mitophagy, and induction of cyclic GMP-AMP Synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway activities to promote an anti-cancer immune response. Importantly, assessment of eliglustat-associated toxicity revealed that extended-use of eliglustat was safe and tolerable in treated mice with histopathological examination of kidney and liver from treated mice revealing no remarkable differences compared to saline control. Clinical chemistry tests following eliglustat treatment indicated dose-dependent elevations in blood ALT and AST levels, consistent with clinical studies. Our study highlights utility of eliglustat as a potential anti-cancer agent for the treatment of TNBC.
Presentation numberPS4-03-16
Characterization of the cytokine profile of metastatic Breast Cancer, from mediators of chronic inflammation to potential biomarkers
Maria M Caffarel, Biogipuzkoa Health Research Institute, Donostia / San Sebastian, Spain
M. M. Caffarel, U. Alvarez, P. Azcoaga, A. Romo, S. Manzano, P. Petit, I. Garmendia, M. Garzón, Z. Telletxea, M. Otaño, A. Urruticoechea, I. Alvarez-Lopez; Oncology, Biogipuzkoa Health Research Institute, Donostia / San Sebastian, SPAIN.
Inflammation is a natural immune response that can protect against cancer in early stages but, when becomes chronic, creates an immunosuppressive tumour microenvironment (TME) that promotes tumour progression. Recognized as a hallmark of cancer, chronic inflammation offers potential for new therapeutic strategies, particularly immunotherapy. However, some solid tumours, such as breast cancer, show partial resistance to these treatments. Cytokines are key regulators of inflammation, influencing cell communication and immune responses within the TME (1,2). Importantly, current clinical classification of BC often neglects the immune landscape, limiting the ability to predict responses to immunotherapy.Our hypothesis is that the cytokine profile of metastatic breast cancer possesses prognostic and predictive value and that cytokines can be potential biomarkers in breast cancer. Understanding how cytokine loops are coordinated within the breast TME could also lead to novel cytokine-based therapeutic strategies. Cytokine levels in serum and fresh tissue samples from metastatic BC patients (n=150) and in serum samples from metastatic cancer patients from other solid tumour types (n=186) were measured by Olink cytokine panel, which quantitatively assess 40 cytokines. Cytokine levels were correlated with clinical data, including overall and relapse free survival. The statistical analysis was done using different mathematical modelling methods. Our unpublished results show that the serum and tumour cytokine profiles do not correlate significantly, supporting the idea that some cytokines act locally in the TME. In addition, comparison with other metastatic tumour samples, led to the identification of some cytokines that are differentially expressed in BC. Integration of these data will shed light on the characterization of cytokine interactions in metastatic BC, helping to define its immune landscape. 1.Soler MF, Abaurrea A, Azcoaga P, Araujo AM, Caffarel MM. New perspectives in cancer immunotherapy: targeting IL-6 cytokine family. J Immunother Cancer. 2023;11(11):e007530; 2.Manzano S, Caffarel MM. Cytokine-centered strategies to boost cancer immunotherapy. Mol Oncol. 2025;19(3):579-583
Presentation numberPS4-03-18
Homologous Recombination Deficiency and DNA-Damage Response in Breast Invasive Lobular Carcinoma
Christopher J Schwartz, Memorial Sloan Kettering Cancer Center, New York City, NY
C. J. Schwartz1, D. Muldoon1, H. Dopeso1, A. Gazzo1, P. Terraf1, M. Repetto1, B. Weigelt1, D. Mandelker1, L. Norton2, H. Y. Wen1, H. Zhang1, D. Ross1, B. Edi1, A. Mamtani3, S. Shen2, C. Bandlamudi1, F. Pareja1; 1Pathology, Memorial Sloan Kettering Cancer Center, New York City, NY, 2Medicine, Memorial Sloan Kettering Cancer Center, New York City, NY, 3Surgery, Memorial Sloan Kettering Cancer Center, New York City, NY.
Background: Identification of genetic alterations in homologous recombination deficiency (HRD) and DNA damage response (DDR) genes in breast cancer (BC) is key for prognostication and potential selection for targeted therapies, such as PARPi. While HRD/DDR somatic oncogenic alterations are generally associated with ductal BCs, their roles in invasive lobular carcinoma (ILC) remain unclear. Here, we sought to investigate the spectrum of HRD/DDR somatic genetic alterations in ILC and their histologic features of these BCs. Design: We interrogated 1,076 ILC previously subjected to paired tumor-normal targeted sequencing (MSK-IMPACT) for oncogenic somatic genetic alterations in 22 HRD/DDR genes. Loss of heterozygosity (LOH) was assessed using FACETS, and mutational signatures using SigMA. HRD status was evaluated using an algorithm optimized for MSK-IMPACT. Histologic assessment was performed as per the WHO. ER/HER2 status was determined per ASCO/CAP guidelines, and follow-up data retrieved from medical records. Results: We identified 53 ILC, including 14 primary ILCs and 39 recurrent/metastatic ILCs harboring somatic oncogenic alterations in HRD/DDR genes, with BRCA2 (38%) and ATM (23%) being the most frequently affected. Other recurrently altered (≥2 cases) HRD/DDR genes were CHEK2, PALB2, BRCA1, BARD1, RAD50, RAD51C, NBN, RECQL and ABRAXAS1. LOH analysis revealed that 53% of HRD/DDR genetic alterations in ILCs were bi-allelic, including 75% of BRCA2 and 70% of ATM cases.Most (71%) primary ILCs were of pleomorphic type or exhibited pleomorphic features (Pleo-ILC), histologic grade 2/3 (93%) and ER+/HER2- (93%). Of the metastatic ILCs, 59% were pleomorphic, and most were ER+/HER2- (74%), followed by HER2+ (18%). Median follow-up for the HRD/DDR-mutant primary ILC cohort was 72 months (range 21-150); 6 patients (43%) had no evidence of disease, 5 were deceased (36%), and 3 were alive with disease (21%). Median follow-up time for the HRD/DDR-mutant metastatic ILC cohort was 84 months (range 19-288); 62% of patients were deceased. ILCs with biallelic inactivation of BRCA2 were classified as HRD by our algorithm in 60% of cases, and half of them showed a dominant HRD mutational signature, whereas the majority of those with ATM biallelic alterations (83%) were classified as non-HRD and exhibited either dominant APOBEC or clock signature (60%). The most frequent non-HRD/DDR affected genes in the cohort were CDH1 (76%), PIK3CA (48%), and FOXA1 (21%).Conclusion: Our findings show that, although at a lower frequency than that reported for ductal BC, DNA repair deficiency including HRD/DDR defects can occur in a subset of ILC. Somatic alterations affecting HRD/DDR genes in ILC are often biallelic, with BRCA2 and ATM being the most altered genes. While these tumors are phenotypically diverse, they often display pleomorphic histology and appear to be associated with more aggressive clinical behavior. These findings highlight the potential clinical relevance of HRD/DDR profiling in ILC, which may help inform treatment strategies.
Presentation numberPS4-03-19
Machine learning based inference of real-time HER2 activity from gene expression profiling in breast cancer to inform HER2 targeted therapy selection
Saumya D. Sisoudiya, Foundation Medicine Inc., Cambridge, MA
S. D. Sisoudiya1, J. S. Ross2, R. B. Keller-Evans3, R. S. Huang3, A. B. Schrock3, G. Frampton1, E. M. Ebot1, E. S. Sokol1, S. Sivakumar1, A. M. Kahn4, M. Lustberg5; 1Computational Discovery, Foundation Medicine Inc., Cambridge, MA, 2Medical Affairs, Foundation Medicine Inc., Cambridge, MA, 3Clinical Development, Foundation Medicine Inc., Cambridge, MA, 4Center for Breast Cancer, Yale School of Medicine, New Haven, CT, 5Yale Cancer Center, Yale School of Medicine, New Haven, CT.
Background: Recent advances in antibody-drug conjugates have demonstrated clinical benefit in patients with HER2-positive breast cancer as well as HER2-low and ultra-low expression tumors. However, prior studies have shown that only a fraction of patients positive by IHC will respond to HER2-targeted therapy. Orthogonal methods to determine HER2 activity such as gene expression have the potential to improve patient selection. We utilized gene expression profiling to build a multi-gene machine learning model to predict HER2 activity, which may aid the identification of patients most likely to benefit from HER2 targeted therapies, particularly for those with HER2-low tumors. Methods: Our cohort comprised 477 patients with breast cancer who underwent RNA-based gene expression profiling of 1,517 genes using FoundationOne®RNA (a laboratory developed test by Foundation Medicine, Inc.). HER2 IHC status was abstracted from pathology reports (IHC 0: 182; 1+: 137; 2+: 128; 3+: 30). Differentially expressed genes (DEGs) between HER2 IHC 0 (label: no activity) and 2+/3+ (label: high activity) groups were identified based on an FDR p-value <0.01 and log2(fold change)>1. A random set of 25% of 0 and 2+ samples and 50% of 3+ samples were held out, prior to modeling. Using a 70:30 (training:test) split of the remaining samples, lasso regression was applied on the expression of DEGs (quantified as transcripts per million, TPM) to train the model. Held-out samples were used to assess the proportion of samples predicted to have high HER2 activity across all IHC categories. This was repeated for 50 iterations and summary metrics were calculated across all iterations. Results: A total of 44 DEGs were used to train the model; ERBB2, TTYH1 and GRB7 were some of the top predictors. Running the model on held-out samples across 50 iterations, IHC 3+ had the highest proportion of samples predicted as “high activity” (median proportion 87%, IQR [87%-93%]), with decreasing proportions in 2+ (58% [47%-63%]), 1+ (41% [35%-44%]) and 0 (22% [17%-28%]). Notably, HER2 3+ samples with no predicted activity had a significantly lower median ERBB2 expression (4,825 TPM [IQR: 4,502-5,167]) than high activity samples (34,146 [IQR: 21,568-44,604]) (P<0.0001) and were most frequently ERBB2 amplification negative (OR = 14.2, P < 0.0001). Additional performance assessment in an independent validation cohort will be presented. Conclusion: A multi-gene expression signature could identify signaling pathways that may better inform HER2 dependency. The machine learning-based model developed here showed a higher proportion of HER2 activity cases with increasing HER2 IHC staining, with potential utility to identify patients who may benefit from HER2 targeted therapies. Exploration in a clinical cohort is warranted.
Presentation numberPS4-03-20
Spatial and biomarker associations of CD83, a promising CAR-T target for breast cancer
Maia Homsi, Roswell Park Comprehensive Cancer Center, Buffalo, NY
M. Homsi1, M. DaVila2, R. Brentjens3, B. Betts4, S. Kabraji1; 1Department of Breast Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Department of Medicine – Translational Research, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 3Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 4Department of Medicine – Transplant & Cellular Therapy Center, Roswell Park Comprehensive Cancer Center, Buffalo, NY.
Background: We previously reported that CD83, a member of the immunoglobulin super family, is a putative target for CAR-T therapy in breast cancer based on preclinical activity against breast cancer cell lines and expression by IHC in a breast cancer tissue microarray. Here we use high-plex tissue imaging (cyclic immunofluorescence) to describe the single-cell and sample based expression of CD83 in tissue compartments and measure its association with canonical breast cancer biomarkers, estrogen receptor, HER2 receptor. Methods: Tissue microarrays underwent dewaxing and antigen retrieval using a BOND auto stainer in the Advanced Tissue Imaging Shared Resource (ATISR). Tissues then underwent bleaching followed by iterative staining of unconjugated or conjugated antibodies. We completed six cycles of staining for all tissues, comprising 18 markers per tissue. Images were acquired at 20x on Leica Thunder Tissue Imager (ATISR). Images were processed using the MCMICRO pipeline and downstream analysis performed in R and Scimap. Results: After filtering for cellularity and technical artifacts, we analyzed 81 TMA cores (41 patients) of which 48 cores were HER2+ (IHC 3+) and 33 were HER2-low (30 HER2 2+ FISH negative, 3 HER2 IHC 1+). 42 cores were ER+ while 28 were ER neg by IHC with 11 not classified. We analyzed a total of 5.0 x105 cells of which 34% were tumor cells (pan-cytokeratin+), 37% were immune cells (CD45+), with the remainder being stromal cells(alpha-smooth muscle actin+). We found that single cell CD83 expression was positively correlated with pan-CK and CD45, consistent with CD83 expression (fluorescence) in both tumor and immune cells. Median CD83 single cell expression was negatively correlated with tumor ER expression (r -0.34, p= 0.0002) but not correlated with median tumor HER2 expression (r=0.03, p=0.7) by CyCIF or IHC (1+, 2+, 3+). Mean CD83 tumor cell expression was higher in ER neg (<1% by IHC) vs ER pos tumors (0.18 +/- 0.1 vs 0.13 +/- 0.7, p <0.001) while CD83 immune cell expression was higher in ER+ tumors (0.24 vs 0.21, p<0.001). Conclusions: Together, these data suggest that tumors that are ER negative may express highest CD83 in the tumor compartment. Given our previous work showing activity of CD83 CAR-T against LM2 TNBC murine tumor model, these data suggest that ER negative tumors may be more likely to respond to CD83-targeting CAR-T cells. Further spatial analysis and correlation of CD83 with outcome will be presented at the conference.
Presentation numberPS4-03-21
Neoadjuvant chemotherapy impacts her2 status as defined by sataloff classification and consequent follow up in a lower middle-income country
RIM BOUCHELAGHEM, Anticancer Center Annaba, Annaba, Algeria
R. BOUCHELAGHEM, I. Boulouh, W. Bechairia, M. Ghassoul, S. Bouacha, S. Bouharoud, S. Mohammed Benali, R. Hachichi, A. Kitouni, N. Néche, H. Djedi; Medical Oncology, Anticancer Center Annaba, Annaba, ALGERIA.
Neoadjuvant chemotherapy impacts HER2 status as defined by Sataloff classification and consequent follow up in a lower middle-income country Rim Bouchelaghem 1, Ibtissem Boulouh1,2, Wafa Bechairia 1, Marwa Ghassoul 1, Salsabil Bouacha 1, Safia Bouharoud 1, Soumaya Benali 1, Rayene Hachichi 1, Ahmed Kitouni 1, Nadia Néche 1,2, Hanene Djedi 1,2 1.Medical oncology Departement, Anticancer Center, Annaba, Algeria2.Faculty of medicine, Badji Mokhtar University, Annaba, Algeria Keywords: Breast cancer, HER2, Sataloff classification, Neadjuvant chemotherapy. Introduction: Neoadjuvant chemotherapy (NACT) followed by surgery remains a common therapeutic strategy for the management of breast cancer (BC) in Algeria. NACT can induce molecular heterogeneity of BC, which presents a challenge in achieving a complete response and impacts patient follow-up, particularly when it affects tumor HER2 expression and modifies patient eligibility for targeted therapies. This evidence can be notably demanding in Algerian public hospitals, where it presents a consequential issue in the face of a shortage of health products, especially for targeted therapies such as HER2 therapy. We therefore assessed post-NCAT HER2 status change in response to different protocols proposed to patients with BC.Methods: This study aimed to assess the change in HER2 status before and after NCAT, as determined by immunohistochemistry, for 61 patients managed at our single institution. According to guidelines, all patients received NACT, with or without anti-HER2 (Trastuzumab with or without Pertuzumab). Pathological response (pR) was evaluated according to Sataloff classification. All patients underwent post-NCAT surgery. Descriptive statistics and the Chi-square test were used to determine statistical differences of pR. Results: The mean age of patients was 50,01 ± 13.39 years (range 24-80). Among the 61 BC, 41 tumors were invasive carcinoma of no specific type (67%), 20 carcinomas (33%) were 10 ductal (16%), five lobular (8%), two mixed (3%), and three mucinous (5%). Hormone receptors (HR) were positive in 43 patients (70%) and negative in 12 patients (20%). Six patients were HER2-enriched. Of all evaluable tumors, the HER2 score was 0 in 18 patients (30%), low in 33 patients (54%), and 3+ in 10 patients (16%). There were 4 complete pR (TA) out of 61 (6,55%). Indeed, there were 7 grade TA (11%), 27 (44%) TB, 17 (28%) TC, and 10 (16%) TD. Among post-NACT-evolved HER2 tumors, 33 (54%) maintained their HER2 level, whereas 28 (46%) exhibited a modified HER2 level. Among 18 HER2-0 tumors (30%), 10 (16%) remained HER2-0, 7 (11%) transitioned to HER2-Low expression, and one to HER2-3+ expression. Among 33 HER2-low tumors (54%), 14 tumors (22%) remained HER2-low, 15 tumors (24%) transitioned to HER2-0, and 3 (9%) transitioned to HER2-3+. Among the 10 HER2-3+ tumors, 8 (80%) remained HER2-3+, while 2 (20%) transitioned to HER2-0 and Low status. Interestingly, there was a statistically significant difference in the percentage of tumors with better pR that maintained HER2 status grade TA (10%), TB (30%), TC (11%), TD (3%) in comparison to tumors that exhibited a changed HER2 status with no grade TA, TB (16%), TC (16%) and TD (13%) (p < 0,0001). Conclusion: Our results support the well-established post-NACT heterogeneity. This reflects a real issue for achieving a pR for patients and managing BC in our country. Indeed, we found for the first time that concomitant analysis of Sataloff response and HER2 changes can represent an endpoint surrogate for treatment achievement after NACT in BC, and a favorable prognosis is associated with HER2 upregulation. The assessment of HER2 after NCAT in combination with Sataloff response is a prerequisite for managing the follow-up of patients. Further studies are needed to characterize this finding.
Presentation numberPS4-03-22
High throughput analysis in locally advanced triple negative breast cancer TNBC: pathological complete response pCR and mutational status.
Ida Paris, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
I. Paris1, G. Mantini2, T. D’Angelo1, A. Minucci3, G. Luciano2, C. Parrillo2, M. De Bonis4, D. Generali5, G. Raspaglio4, M. De Donato4, F. Pavese1, C. Nero1, A. Fabi6, A. Piermattei7, P. Fuso1, V. Ricozzi1, L. Carbognin6, S. Rotondaro1, T. Pasciuto8, M. Marrazzo9, A. Mulè7, G. Garganese1, A. Di Leone10, M. Sanchez10, A. Migliore11, D. Giannarelli12, G. Franceschini10, A. Fagotti13; 1Gynecologic Oncology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 2Bioinformatics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 3Genomics Core Facility, Gemelli Science and Technology Park (G-Step), Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 4Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 5Department of Medical, Surgery and Health Sciences, Cremona Hospital, University of Trieste, Cremona, ITALY, 6Department of Woman and Child Health and Public Health, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 7Gynecopathology and Breast Pathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 8Data Collection Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 9Data Collection Core Facility, Gemelli Science and Technology Park (G-STeP),, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 10Breast Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 11Department of Woman and Child Health and Public Health, Marche Polytechnic University and Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, ITALY, 12Facility of Epidemiology and Biostatistics, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY, 13Head of Gynecologic Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, ITALY.
BackgroundPathologic complete response after neoadjuvant treatment is a strong prognostic biomarker associated with improved survival in breast cancer patients, especially in triple negative breast cancer and HER2 positive subtypes. Despite high response rates to neoadjuvant chemotherapy, triple negative breast cancer has been linked to higher rates of recurrence and poorer outcome. The aim of this study is to evaluate the genomic landscape of tissue samples from locally advanced breast cancer patients treated in a neoadjuvant setting and to correlate the genomic analysis to response and clinical outcome. Methods We retrospectively evaluated 48 core needle biopsies before chemotherapy from a cohort of 136 patients with locally advanced triple negative breast cancer. Pathological chemotherapy response and not responders was assessed by surgery. DNA extracted from archival formalin-fixed paraffin-embedded samples was subjected to sequencing using the TruSight Oncology 500 panel (Illumina, San Diego, USA; 523 genes, size: 1.94 Mb), following the manufacturer’s protocol, which assesses microsatellite instability status, tumor mutation burden, recurrent somatic copy number variations, somatic sequence variants as single nucleotide variants and Insertion/Deletion. Libraries were sequenced on NovaSeq 6000 Illumina platform to reach a minimum of 500× read depth. 67 (51.3%) out of 136 patients received a standard anthracycline and taxane regimen and 69 (50.7%) patients received a platinum-containing chemotherapy regimen. Somatic mutations classified as Oncogenic or Likely Oncogenic according to OncoKB with a variant allele frequency ≥0.03 were included. Differences of mutational profile were assessed using Chi-squared test. Statistical analysis was performed with R Studio (version 4.2.2). ResultsIn total we analyzed samples from 48 patients. The median age was 46 years. 22 of 48 (46%) were HER2 low, 42 (87%) with G3, 27 (56%) with stage II and 16 (33%) with stage III. 28 out of 48 patients (58.3%) not obtained pCR.We showed statistically significant correlations between PIK3CA mutations and PI3K pathway mutations with the absence of pathological complete response with p-values of 0.013 and 0.024 respectively. Genomic analysis of the entire cohort revealed the most frequently altered genes to be TP53 (94%), PIK3CA (21%), BRCA1 (15%), along with amplifications of MYC (23%) and GATA3 (17%) and MCL1 (15%). The most aberrant pathways in not pCR population are PI3K and RTK-RAS signaling.Alterations in PIK3CA was observed in 39% of not responders and absent in responder patients (p-value 0.05, reflecting its key role in PI3K-AKT-mTOR signaling pathway. The lollipop plots show how missense (H1047R, E542K, R88Q, V344G) and inframe deletion (G106_E109del, E110del) mutations are presents in not responder patients after neoadjuvant chemotherapy.Interestingly, median TMB in not responder’s cohort is 5.5 while in chemo sensitive patients is 4.8 (p-value 0.93).ConclusionsIn this preliminary report, PIK3CA mutations and related pathways may impact the effectiveness of neoadjuvant chemotherapy in achieving pCR. A larger sample size is needed to further evaluate the significance of mutations and co-alterations in different subgroups. We would like to acknowledge the contribution of the Multispecialistic Biobank Research Core Facility of Fondazione Policlinico Universitario A. Gemelli Biobank, for providing the bioresources.
Presentation numberPS4-03-23
Detection of Treatment-Resistant ESR1 Mutations by Ultra-sensitive SuperRCA Technology
Duy Nguyen, Quest Diagnostics, San Juan Capistrano, CA
D. Nguyen1, L. Chen2, V. Radic3, C. Ma1, F. Racke3; 1Molecular Oncology, Quest Diagnostics, San Juan Capistrano, CA, 2R&D, Rarity Biosciences, Uppsala, SWEDEN, 3Hematology and Oncology, Quest Diagnostics, San Juan Capistrano, CA.
Introduction The ESR1 gene encodes the estrogen receptor alpha (ERα), a transcription factor that regulates gene expression in response to estrogen. Mutations in ESR1 can lead to constitutive activation of ERα, allowing estrogen receptor-positive (ER+) breast cancer cells to proliferate even in the absence of estrogen; this proliferation contributes to resistance against standard hormone therapies such as treatment with aromatase inhibitors (AI), which function by reducing estrogen levels. ESR1 mutations are typically acquired during treatment and are most frequently observed in metastatic breast cancer. Detecting ESR1 mutations, often through circulating tumor DNA (ctDNA) in liquid biopsies, is critical for tracking disease progression and optimizing therapeutic strategies. As reported at the recent ASCO 2025 meeting, treatment switch before clinical progression based on the detection of ESR1 mutations in ctDNA can potentially improve outcomes in HR+/HER2- advanced breast cancer. Thus, identifying ESR1 mutations enables clinicians to make informed treatment decisions and supports the development of targeted approaches to overcome endocrine resistance. Molecular assays such as digital PCR (dPCR), real-time PCR (RT-qPCR), and next-generation sequencing (NGS) are commonly employed for detection of ESR1 mutations, with each platform offering distinct advantages in sensitivity and mutation coverage. However, a common limitation of these technologies is their sensitivity, which is compounded by the limited quantity of ctDNA that can be isolated from human specimens. To address these limitations, we developed a novel assay specifically designed for the detection of ESR1 mutations; the assay leverages the high specificity and sensitivity of the SuperRCA® technology developed by Rarity to enable accurate identification of low-frequency ESR1 variants. Methods The SuperRCA® uses 2 consecutive rolling circle amplification (RCA) reactions, which enable highly specific genotyping and accurate quantification of low-frequency variants. The sensitivity of the assay for ESR1 was assessed by testing a quantitative, serial dilution of synthetic controls of commonly found resistant variants and numerical positive signals detected using flow cytometry. Assay specificity was assessed by the number of positive signals from cfDNA extracted from normal individuals. Results The assay was capable of detecting rare ESR1 mutations at a sensitivity of 0.01% VAF down to 0.001% VAF. The number of positive signals was essentially zero for normal individuals , indicating high specificity. Conclusions These results demonstrate the SuperRCA® can be used to detect clinically relevant ESR1 variants, which may offer a powerful tool for improving the management of endocrine-resistant breast cancer. References: [1] ESR1 mutations in HR+/HER2-metastatic breast cancer: Enhancing the accuracy of ctDNA testing. Konstantinos Venetis et al. Cancer Treat Rev. 2023. PMID: 37864956[2] ESR1 mutations and therapeutic resistance in metastatic breast cancer: progress and remaining challenges. Sarah K Herzog et al. Br J Cancer. 2022. PMID: 34621045 [3] ESR1 mutations-a mechanism for acquired endocrine resistance in breast cancer. Rinath Jeselsohn et al. Nat Rev Clin Oncol. 2015. PMID: 26122181 [4] Camizestrant + CDK4/6 inhibitor (CDK4/6i) for the treatment of emergent ESR1 mutations during first-line (1L) endocrine-based therapy (ET) and ahead of disease progression in patients (pts) with HR+/HER2 – advanced breast cancer (ABC): Phase 3, double-blind ctDNA-guided SERENA-6 trial. Nicholas C. Turner et al. ASCO 2025. Abstract LBA4
Presentation numberPS4-03-24
Association of HER2 0 vs HER2 low with median OS in patients with breast cancer and BRCA1, BRCA2, and PALB2 pathogenic mutations.
Darya Kizub, MD Anderson Cancer Center, Houston, TX
D. Kizub1, A. Gutierrez2, R. K. Murthy1, C. Albarracin3, B. K. Arun1; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology-Research, MD Anderson Cancer Center, Houston, TX, 3Pathology, Anatomical, MD Anderson Cancer Center, Houston, TX.
Background and objective: HER2 0 compared to HER2 low (1+ or 2+) is associated with worse survival in HER2-negative metastatic breast cancer (MBC). We explored the impact of HER2 status on survival in patients withe HER2-negative breast cancer (HER2-) and germline BRCA1, BRCA2, and PALB2 mutations. Methods: We evaluated outcomes among women with HER2- patients with breast cancer treated at MD Anderson Cancer Center from August 1996 to May 2024. Patient characteristics were analyzed using descriptive statistics and standard tests of association. Cox proportional hazard models evaluated factors associated with overall survival (OS). Results: PALB2. Among 93 patients with PALB2, median age at diagnosis was 49.5 years old (st.dev 10.5). 20 (21.5%) had Stage I, 21 (22.6% Stage II), 18 (19.4%) Stage III, 4 (4.3%) de novo Stage IV disease. ER was weakly positive in 4 (4.3%), negative in 28 (30.1%), positive in 61 (65.6%). PR was negative in 41 (45.2%), positive in 51 (54.8%). 48 (51.6%) were HER2 0, while 45 (48.3) were HER2 low (1+ or 2+). Of the 48 patients who were HER2 0, 5 died at last follow-up, with OS 89.6%. Of the 45 patients with HER2 low, 4 died with OS 91.1%. Median OS for HER2 0 patients was 130.06 months (95% CI 114.65, 145.465 months) compared to 261.96 months for HER2 low (95% CI 219.21, 297.89) (p < 0.05). BRCA1 and BRCA2. For 752 patients with BRCA1 and 2, median age was 44.5 years (st.dev 11.20). 224 (29.8%) had Stage I disease, 332 (44.1%) Stage II, 144 (19.1%) Stage III, and 42 (5.6%) de novo Stage IV. ER was negative in 374 (49.7%), positive in 377 (50.1%). PR was negative in 460 (61.2%) and positive in 289 (38.4%). HER2 was 0 in 443 (58.9%), 1+ in 208, 2+ in 101 (13.4%). BRCA1. Among 424 patients with BRCA1, 266 (62.7%) were HER2 0 and 158 HER2 low. Among 266 who were HER2 0, 49 died, with OS 81.6%. Among those who were HER2 low, 26 died, with OS 83.5%. Median OS was 289.34 (95% CI 229.66, 548.80) for patients with HER2 0 compared to 175.54 (95% CI 158.05,193.03) for HER2 low (p<0.05). BRCA2. Among 327 patients with BRCA2, 177 (54.1%) were HER2 0 and 150 HER2 low. Among 177 who were HER2 0, 26 died with OS 85.3%. Among those with HER2 low, 25 have died with OS 83.3%. For Her2 0, median OS was 384.46 months (95% CI 199.54, 441.99) compared to 172.927 for HER2 low (95% CI 107.98, 291.4) (p > 0.05). Conclusions: HER2 0 was associated with worse OS compared to HER2 low in patients with PALB2 mutations and with better OS compared to HER2 low in patients with BRCA1 and BRCA2, though the association for BRCA2 was not statistically significant. Future studies should address the impact of targeted therapy and tumor biology in these populations.
Presentation numberPS4-03-25
Targeting Pro-Metastatic Stress Kinase Networks in Triple-Negative Breast Cancer Using Patient-Derived Models
Margarite Matossian, University of Chicago, Chicago, IL
M. Matossian1, M. Bungert1, C. Nguyen1, G. Yerradoddi2, M. East3, D. Okumu3, R. Nanda1, G. Johnson4, M. Rosner5; 1Medicine, University of Chicago, Chicago, IL, 2Bioinformatics, University of Chicago, Chicago, IL, 3Pharmacology, University of North Carolina, Chapel Hill, NC, 4Pha, University of North Carolina, Chapel Hill, NC, 5Pathology, University of Chicago, Chicago, IL.
Background: Metastatic triple-negative breast cancer (mTNBC) is one of the most challenging forms of breast cancer to treat due to profound heterogeneity and drug resistance. There have been very limited advancements in approval of targeted treatment regimens that are not chemotherapy-based for mTNBC in the past decade. Kinases are tractable therapeutic targets, but while kinase inhibitors have improved outcomes in many metastatic solid tumors including other breast cancer subtypes, they have not improved patient outcomes in TNBC clinical trials. We posit this is because we do not leverage understanding of complex kinase signaling networks and how we utilize kinase inhibitors in TNBC is not optimized. Further, high doses of kinase inhibitors to achieve cytotoxicity have unacceptable side effects for patients limiting real-world use. Chronic stress states such as those initiated by hypoxia, poor nutrient delivery and higher metabolic demands promote pro-metastatic transcriptional programs in TNBC. Our preclinical research and secondary analyses from clinical trial data reveal stress kinase signaling networks, and not individual pathways, drives TNBC metastatic transformation. Our central hypothesis is the RKIP/BACH1 signaling axis is a regulator of the chronic stress response to suppress TNBC metastasis and can be leveraged to produce biomarkers for risk stratification and a novel combination regimens. Methods: Prior work showed activation of the physiologic metastasis suppressor RKIP effectively reduced TNBC metastasis in cell line models through inhibition of stress MAPK networks (p38, JNK, ERK1/2, MLK) resulting in decreased transcription of the pro-metastatic transcription factor BACH1. We utilized mass spectrometry-based techniques with a kinase inhibitor system (MIB-MS) to characterize the activity of kinase signaling networks in heavily treated mTNBC organoids and examined kinase networks prominent in BACH1-high TNBC from publicly available phosphoproteomics datasets. We utilize live/dead, proliferation assays and transwell invasion assays to test efficacy of our stress kinase inhibitor regimen. Results: We show that combinations of 3 kinase inhibitors used in early phase clinical trials (Ralimetinib (p38), JNK-in-8 (JNK) or FDA approved (Trametinib (MEK & ERK1/2)) inhibit stress kinase networks, pro-motility gene expression and TNBC cell invasion. Low dose inhibition of multiple kinase nodes of networks, rather than single kinase nodes, more effectively reduces oncogenic signaling output and prevents compensatory signaling activation and potentiates suppression of organoid tumor initiating capacity when used in combination with chemotherapy. Using patient-derived organoids that recapitulate heterogeneous mTNBC molecular characteristics we successfully generated stable BACH1-knockout organoids which will be used to further identify BACH1-dependent critical kinase networks. Conclusions: Inhibition of these stress kinase networks should enable effective reprogramming of metastatic or high-risk cancers to more benign states and make tumors more susceptible to approved anti-proliferative therapies such as chemotherapy or immunotherapy while limiting toxicities from kinase inhibitors using a low-dose, multi-drug combination approach.
Presentation numberPS4-03-26
Cell-free DNA BRCA copy number loss and/or deletions in patients with metastatic breast cancer: incidence and association with clinical and genomic features
Neelima Vidula, Massachusetts General Hospital, Harvard Medical School, Boston, MA
N. Vidula1, D. Giannarelli2, A. Putur1, L. Pontollilo3, E. Nicolo3, C. Reduzzi3, E. Podany4, G. Cunningham4, A. Davis4, E. Blouch1, M. Cristofanilli3; 1Hematology and Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 2Oncology, Instituto Nazionale Tumori Regina Elena, Rome, ITALY, 3Hematology and Medical Oncology, Cornell University, New York, NY, 4Hematology and Medical Oncology, Washington University School of Medicine, St. Louis, MO.
Background: BRCA mutations may impact outcomes in metastatic breast cancer (MBC). While germline and somatic BRCA mutations have been studied, the incidence and impact of cell-free DNA (cfDNA) BRCA copy number loss and/or deletions (CNL/D) are not known. We evaluated cfDNA BRCA CNL/D, and the association with clinical/genomic features in MBC. Methods: Patients with MBC with cfDNA (Guardant360 next-generation sequencing) results at 3 academic centers in the Precision Medicine Action for Cancer consortium from 12/2021 (when Guardant360 began reporting BRCA CNL/D) to 1/2024 were identified. A retrospective analysis was conducted to identify clinical/genomic features associated with cfDNA BRCA1 and/or 2 CNL/D. Median therapy durations and overall survival (OS) after detection of cfDNA BRCA CNL/D were estimated with the Kaplan-Meier method. Results: Among 1,198 patients with MBC who had cfDNA testing, 65 (5.4%) had cfDNA BRCA CNL/D. In the cohort with cfDNA BRCA CNL/D, median age at MBC diagnosis was 60 years. Subtypes included 47/65 (72%) hormone receptor (HR)+/HER2-, 15/65 (23%) triple negative, and 3/65 (4.6%) HER2+. Prior to cfDNA testing, patients received a median of 1 hormone therapy (HR+/HER2-) and 0 chemotherapy agents (all subtypes) for MBC. Table 1 shows the types of cfDNA BRCA CNL/D that were detected. Of patients with cfDNA BRCA CNL/D, 16/65 (25%) were BRCA1, 37/65 (57%) BRCA2, and 12/65 (18%) BRCA1&2. Thirteen of 65 (20%) patients had a co-existing pathogenic cfDNA BRCA mutation, 40/65 (62%) had a cfDNA TP53 mutation, and 43/65 (66%) had cfDNA high tumor mutation burden (TMB) >/= 10 mut/Mb. Ten of 32 (31%) patients with tumor tissue genotyping results had a concomitant tissue BRCA1/2 mutation. Five of 45 (11%) patients with germline genetic testing results had a concomitant germline BRCA1/2 mutation. Median duration on first therapy post detection of cfDNA BRCA CNL/D was 94 days (95% confidence interval (CI): 54-134 days). Post detection of cfDNA BRCA CNL/D, 5 patients received platinum chemotherapy with a median response duration of 168 days (range 32-582 days), and 4 patients received a PARP inhibitor with a median response duration of 105 days (range 26-231). Median OS after detection of BRCA CNL/D was 490 days (95% CI: 232-748 days). Conclusions: CfDNA BRCA CNL/D were detectable in a subset of patients with MBC, with most present in non-germline BRCA carriers. CfDNA BRCA CNL/D co-occurred frequently with TP53 mutations and elevated TMB, suggesting the possibility that cfDNA BRCA CNL/D could arise from increased mutagenesis in MBC. Further research is needed to define the impact of platinum and PARP inhibitor therapy in this cohort.
| Type of BRCA copy number loss and/or deletions | # of patients (total N=65 but some patients had greater than or equal to 1 type of BRCA copy number loss and/or deletions) |
| BRCA single copy number loss | 20 (31%) |
| BRCA single copy deletion | 17 (26%) |
| BRCA biallelic loss (single copy deletion & co-occurring loss of function variant) | 9 (14%) |
| BRCA biallelic loss (homozygous deletion) | 4 (6%) |
| BRCA exon deletion | 4 (6%) |
| BRCA copy number loss not otherwise specified | 15 (23%) |
Presentation numberPS4-03-27
M2 Macrophages Predict Response to Neoadjuvant Chemotherapy in Triple Negative Breast Cancer Patients
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen, Q. Shao, Z. Wang, B. Zhu; Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA.
Background: Macrophages, the predominant immune-related stromal cells within tumor microenvironment, could be polarized into M1 and alternative M2 macrophages. M1 macrophages are pro-inflammatory and eliminate cancer cells, whereas M2 macrophages are immunosuppressive and facilitate tumor growth. Mounting evidence has shown that M2 macrophages are responsible for a poor prognosis in triple negative breast cancer (TNBC) patients. Since neoadjuvant chemotherapy is becoming the foundation treatment for TNBC, the relationship between M2 macrophages and response to neoadjuvant chemotherapy remains to be elucidated. Methods: Infiltration of M2 macrophages were checked by analyzing the CD163 levels on surgical resection specimens from 42 Chinese TNBC patients treated with neoadjuvant chemotherapy. Evaluation of M2 macrophages together with other immune markers including CD8-positive cytotoxic T cells and PD-L1 expression in TNBC and its association with pCR after neoadjuvant chemotherapy were performed. Results: Pathological complete response (pCR) was significantly associated with younger age, higher tumor or stromal PD-L1 expression, higher levels of tumor or stromal CD8-positive cytotoxic T cells, but lower infiltration of CD163-positive M2 macrophages, with the level of CD163-positive M2 macrophages in stromal area as the strongest factor. Conclusions: Our study showed that high infiltration of CD163-positive M2 macrophages was negatively associated with the chemotherapeutic response to neoadjuvant chemotherapy in Chinese TNBC patients and could be used as a promising prognostic candidate for Chinese TNBC patients treated with neoadjuvant chemotherapy.
Presentation numberPS4-03-28
Hypoxia and lactate metabolism-relatedgene signature for prognosis prediction in triple negative breast cancerand hub gene COL5A3 promotes cancer progressionvia PI3K/AKT pathway
Wei-xian Chen, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
W. Chen, Q. Shao, Z. Wang, B. Zhu; Department of Breast Surgery, Changzhou No.2 People’s Hospital, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, CHINA.
Background: Triple negative breast cancer (TNBC) is a highly aggressive subtype distinguished by the lack of hormone receptors and specific molecular targets, leading to limited treatment options, high recurrence rates, and poor clinical prognosis. The Warburg effect in cancer cells generates a tumor microenvironment characterized by hypoxia, low glucose, and high lactate, which severely promote chemo-resistance, immune escape, and distant metastasis. Nevertheless, prognostic significance of hypoxia and lactate metabolism-related genes in TNBC remains largely unclear, and more accurate prediction models are urgently needed. Methods: Clinical information and gene expression profiles of TNBC patients were obtained from the online database. LASSO and multivariate Cox regression analysis were used to establish the hypoxia and lactate metabolism-related prediction model, and prognostic value was further validated by Kaplan-Meier plotter, receiver operating characteristic curves, and nomogram. Gene set enrichment analysis was performed to evaluate the pathways and molecular functions of hypoxia and lactate metabolism-related gene signature. Tumor mutation, microsatellite instability, RNA expression-based stemness scores, immune infiltration, and drug susceptibility analysis were applied to identify potential therapeutic targets. A series of in vitro experiments were performed to evaluate the biological function of the selected gene. Results: Five genes including COL5A3, LRRC8D, IGFL1, SEPTIN3, and PEG10 were identified to establish the hypoxia and lactate metabolism-related risk score. TNBC patients divided into high-risk or low-risk group by risk score exhibited substantial differences in survival outcome, tumor mutation, immune infiltration, and drug sensitivity. Gene set enrichment analysis revealed that difference between these two groups may be related to extracellular matrix, intercellular links, intercellular information transfer, immune-related pathways, angiogenesis and metabolism. The constructed model showed strong predictive performance, with receiver operating characteristic curves showing area under the curve values of 0.82, 0.86, and 0.90 at 1, 3 and 5 years, respectively. In vitro studies indicated that hub gene COL5A3 could promote tumor proliferation, migration, and drug resistance via PI3K/AKT pathway in TNBC cells. Conclusion: A novel five hypoxia and lactate metabolism-related gene signature could be used for prognosis prediction in TNBC patients, and hub gene COL5A3 promotes TNBC progression via PI3K/AKT pathway. Further research is needed to explore the biological roles of these genes in TNBC aiming to refine therapeutic approaches.
Presentation numberPS4-03-29
N-nose as a non-invasive biomarker for predicting neoadjuvant chemotherapy response in breast cancer patients
Akira Nakakami, Gifu university Hospital, Gifu, Japan
A. Nakakami1, Y. Tokumaru1, Y. Niwa1, R. Mori1, M. Okawa1, Y. Sato2, H. Hatakeyama3, T. Hirotsu4, E. di Luccio3, N. Matsuhashi2, M. Futamura1; 1Breast surgery, Gifu university Hospital, Gifu, JAPAN, 2Gastroenterological Surgery and Pediatric Surgery, Graduate School of Medicine, Gifu University, Gifu, JAPAN, 3R&D department, Hirotsu Bio Science Inc., Tokyo, JAPAN, 4Head office, Hirotsu Bio Science Inc., Tokyo, JAPAN.
(Background)Breast cancer (BC) remains a leading cause of cancer-related deaths despite advances in diagnosis and treatment. Accurate evaluation of response to neoadjuvant chemotherapy (NAC), especially in HER2-positive and triple-negative subtypes, is critical. In recent years, liquid biopsy techniques, which analyze tumor-derived materials in body fluids, have emerged as promising non-invasive tools for cancer detection and monitoring. Nematodes-NOSE (N-NOSE) is a novel cancer screening test that utilizes the chemotaxis index of the nematode Caenorhabditis elegans (C. elegans). The N-NOSE method is a non-invasive diagnostic approach that falls within the broader category of liquid biopsy, which analyzes tumor-derived materials in body fluids. C. elegans possesses a highly sophisticated olfactory system and has been reported to exhibit attraction toward the urine of cancer patients while avoiding that of healthy individuals. In the current study, we aimed to clarify the ability of the N-NOSE method to predict the response to NAC in BC patients. (Materials and Methods)A total of 36 BC patients scheduled to undergo NAC followed by surgery at our institution between August 2020 and May 2023 were prospectively enrolled in this study. All patients were histologically confirmed to have BC and were deemed eligible for NAC based on clinical staging and risk stratification. Urine samples were collected from each patient at three distinct time points: prior to the initiation of NAC, immediately before surgery, and four weeks after surgery. For chemotactic assays, 0.5 μL of 1 M sodium azide, an anesthetic used to minimize the effects of adaptation, was spotted onto four points on the agar plate (urine side and non-urine side), and 1 μL of urine sample diluted 100-fold with ultra-pure water was added to two points (urine side). Approximately 100 adult nematodes were placed in the center of the plate. After roaming for 30 minutes, the number of nematodes present in area A (urine side) and the number of nematodes present in area B (non-urine side) were counted. The chemotaxis index (CI) was calculated using the following equation: Index = (Number of nematodes in area A – Number of nematodes in area B)/Total number of nematodes. The change in CI (Index Reduction Score, IRS) was calculated for three intervals: before and after NAC (IRS1), before and after surgery (IRS2), and before NAC and after surgery (IRS3). The association between IRS and treatment efficacy was statistically evaluated using receiver operating characteristic (ROC) curve analysis. (Results)Among the 36 patients enrolled, the median age was 51 years (range: 35-77). Thirteen patients (36.1%) achieved pathological complete response (pCR), while sixteen patients (44.4%) achieved pathological partial response (pPR). When the treatment response included CR or PR, the area under the ROC curve (AUC) for IRS1, IRS2, and IRS3 were 0.53 (95% CI: 0.19-0.78), 0.76 (95% CI: 0.56-0.96), and 0.66 (95% CI: 0.37-0.96), respectively. Notably, when only pCR cases were analyzed, the AUCs were 0.58 (95% CI: 0.34-0.82) for IRS1, 0.64 (95% CI: 0.40-0.88) for IRS2, and 0.75 (95% CI: 0.54-0.95) for IRS3, indicating that IRS3 had the highest predictive value for pCR. In this study, some pCR cases exhibited minimal residual intraductal disease before surgery, which was subsequently removed during surgery, potentially resulting in a higher IRS3 compared to IRS1. (Conclusion)The IRS measured by the N-NOSE method may accurately reflect the efficacy of NAC in BC patients. Furthermore, N-NOSE may be able to detect minimal or subclinical residual tumors after NAC, which are usually grouped clinically but may differ in their biological characteristics. Further large-scale, multicenter prospective studies are warranted.
Presentation numberPS4-03-30
Desmoplakin, DSP, is a potential tumour suppressive protein in human breast cancer
Hong Yu Xiang, Cardiff University, CARDIFF, United Kingdom
H. Y. Xiang1, H. Y. Xiang2, X. Li1, C. Wang1, T. A. Martin1, E. Davies3, K. Mokbel1, Y. H. Liu2, W. G. Jiang1; 1School of Medicine, Cardiff University, CARDIFF, UNITED KINGDOM, 2Department of Thyroid and Breast Surgery, Peking University First Hospital, Beijing, CHINA, 3Wales Breast Centre, University Hospital Llandough, Cardiff, UNITED KINGDOM.
Introduction. Desmoplakin (DSP) is a membrane subcoat plaque protein, primary co-located within the desmosomes, which are membrane structures found in tissues such as skin and muscles. Known DSP interactive proteins include desomosome cadherin desmogleins (DSG)and desmocollins (DSC). Although an early indication of a possible loss of the DSP protein, particularly in high grade tumours in human breast cancer was reported (Davies et al 1999), the impact of DSP on the clinical course and prognosis in breast cancer has not been fully explored. Here, we investigated the expression of DSP together with the membrane integral demososome protein DSG1 (desmoglein-1), aiming to establish a possible prognostic significance in clinical breast cancer. Methods and Results. The expression of DSP gene transcript was quantitatively evaluated in a cohort of clinical breast cancer with a ten-year followup. The expression levels were fully analysed against the clinical and pathological factors and the long term clinical outcome of the patients. Where appropriate, the prognostic significance was conducted using Kaplan-Meier model and Bayes analyses. The most significant finding was that the expression levels of DSP is linked to the clinical outcome of the patients. Breast tumours which developed distant metastasis, local recurrence and in patients who died from breast cancer exhibited a significant reduction of DSP expression when compared with tumours from those who remained disease free (p=0.036, p=0.020 and p=0.022, respectively, versus disease free). The reduction of DSP was seen profoundly in ER positive tumours and HER2 positive tumours. Survival analysis indicated that DSP expression is a favourable factor for overall survival (>OS) with an AUC at 0.241 (p<0.0001) and hazard ratio (HR) at 0.063 (95% confidence internal 0.008-0.477, p=0.007 by Cox regression), a connection supported by an impressive Bayes factor (192.1). The connection with OS was observed across the molecular subtypes and when the molecular subtypes and hormone receptor status were considered, DSP remained a significant OS indicator in multivariant analysis. Very similarly, DSP expression is also a favourable indicator for disease free survival (DSF) (p=0.006), again seen in most subtypes. Significant correlation between expression pattern of DSP and DSG1 was not observed. Finally, evidence suggests that DSP expression level is a good indicator for patient response to both neoadjuvant and adjuvant chemotherapies in patients with breast cancer.Conclusion. Desmoplakin exhibits characteristics as a favourable prognostic indicator for patient overall and disease free survival in breast cancer patients. The connection, which is less influenced by the molecular subtypes, also has value in predicting the chemotherapeutic response of the patients. Reference:Davies EL et al. 1999. The immunohistochemical expression of desmoplakin and its role in vivo in the progression and metastasis of breast cancer. Eur J Cancer, 35, 902
Presentation numberPS4-05-01
Postpartum Associated Breast Cancer and Its Survival Impact among Young Women.
Benjamin Walbaum, Hospital Clinic Barcelona, Barcelona, Spain
B. Walbaum, A. Morell, J. Muñoz-Carrillo, L. Arenas, A. Del Val, M. Rey, P. Gimenez-Xavier, N. Chic, F. Brasó-Maristany, E. Segui, R. Gómez-Bravo, I. García-Fructuoso, T. Pascual, F. Schettini, M. Bergamino, M. González-Rodríguez, B. Adamo, M. Muñoz, E. Sanfeliu, A. Prat, O. Martínez-Sáez, M. Vidal; Oncology, Hospital Clinic Barcelona, Barcelona, SPAIN.
Background: Over half of women diagnosed with breast cancer (BC) before the age of 45 fall within the postpartum window, generally defined as a diagnosis made up to 10 years after last delivery. Postpartum has been associated with poorer outcomes and may be a key determinant of the worst prognosis described among young BC patients. Diagnostic delays and biological changes in tumor microenvironment could explain this increased risk. Moreover, delayed childbearing can intensify the postpartum risk and forestall the long-term protective effects of pregnancy. We herein present a retrospective analysis of postpartum-associated BC (PPBC) from a single institution.Methods: Retrospective analysis including a cohort of 223 women under 45 years diagnosed at the Hospital Clinic of Barcelona (2010-2023) with complete pregnancy history. Patients were stratified into PPBC (n=113) defined as BC diagnosed postpartum and up to 10 years after last delivery, and non-PPBC (n=110), either nulliparous or diagnosed after 10 years of last delivery. Analyses evaluated disease-free survival (DFS), distant disease-free survival (DDFS), and overall survival (OS) in early-stage patients. Subgroup analyses in PPBC explored the effect of time since delivery (30 years). PAM50/PROSIGNA was used to determine intrinsic subtypes and risk of recurrence (ROR). Clinicopathological features and survival outcomes were analysed using Chi-square tests, Kaplan-Meier, and Cox regression models.Results: PPBC cases were slightly older at diagnosis than non-PPBC (median 38.0 vs. 36.8 years, p=0.017). Stage distribution was similar, though stage IV disease at diagnosis was numerically higher in PPBC (9.7% [n=11] vs. 5.5% [n=6], p=0.262). Immunohistochemistry (IHC)-based subtypes were comparable (p=0.857), with 59.3% of PPBC and 57.3% of non-PPBC patients having estrogen receptor-positive /HER2-negative (ER+/HER2−) subtype. No other significant differences were observed regarding tumor size, nodal status, histologic type or grade, or Ki67 index. Among patients with available PROSIGNA (n=65; 35 PPBC and 30 non-PPBC), no significant differences for intrinsic subtypes (p=0.892) or ROR categories (p=0.344) were found.At a median follow-up of 85 months, 7-year DFS and DDFS were numerically lower in PPBC compared to non-PPBC (DFS: 75.1% vs. 83.3%; DDFS: 76.2% vs. 89.5%), though only DDFS approached statistical significance (hazard ratio [HR] 1.90, 95% CI 0.976-3.702; p=0.054). Noteworthy, OS was significantly reduced in PPBC patients (7-year OS: 89.6% vs. 95.6%; HR 3.01, 95% CI 1.00-9.79; p=0.042), with persisting differences in the full cohort including de novo stage IV cases (HR 3.4, 95% CI 1.23-9.38; p=0.01). Within PPBC, those diagnosed <5 years postpartum had numerically worse DFS (HR 1.72, 95% CI 0.84-3.53; p=0.136), while no survival differences were observed by age at first pregnancy (p=0.610).In multivariate Cox regression models excluding metastatic cases and adjusting for age at diagnosis, tumor size (cT 2-4 vs. 1), nodal status (cN 1-2 vs. 0), and IHC subtype, PPBC was independently associated with worse DFS (HR 2.63, 95% CI 1.18-5.89; p=0.019) and DDFS (HR 2.82, 95% CI 1.24-6.45; p=0.014). Tumor size was also an independent predictor of recurrence across all models. Regarding OS, PPBC remained an independent risk factor (HR: 6.96 95% CI 1.58-30.71; p=0.010), while increasing age was associated with better survival (HR: 0.81 95% CI 0.68-0.96; p=0.018).Conclusions: PPBC is an independent predictor of poor survival among young women. In the absence of differences in clinical stage or tumor subtype, these findings suggest that postpartum-specific biological mechanisms may underlie the worse outcomes observed. Further investigation is needed to refine risk stratification and improve care in this distinct population.
Presentation numberPS4-05-03
Pro-c3 as a circulating biomarker of tumor fibrosis in breast cancer: functional insights from studying cancer associated fibroblasts in vitro
Marco Pannone, Nordic Bioscience, Herlev, Denmark
M. Pannone, A. Hettich, R. Vestermark, M. Karsdal, N. Willumsen; Oncology, Nordic Bioscience, Herlev, DENMARK.
Background: Extracellular matrix (ECM) remodeling is a hallmark of tumor progression, driven in part by cancer-associated fibroblasts (CAFs). The N-terminal pro-peptide of type III collagen (PRO-C3), released during collagen formation, is a circulating biomarker reflecting active tumor fibrosis (Nissen et al., 2022). Elevated pre-treatment PRO-C3 levels have been associated with poor outcomes in metastatic HER2+ breast cancer treated with Trastuzumab (Lipton et al., 2018). Additionally, in a Phase II trial of tetrathiomolybdate in high-risk triple-negative breast cancer, patients who remained disease-free exhibited lower PRO-C3 levels (Liu et al., 2021), suggesting a link between reduced collagen formation and improved outcomes.Methods: To investigate the cellular origin and regulation of type III collagen production in breast cancer, we quantified PRO-C3 in conditioned medium (by competitive ELISA) using primary breast CAFs and primary benign breast fibroblast cultured in Ficoll-containing media with ascorbic acid, according to the in vitro Scar-in-a-jar fibroblast model system (Chen et al., 2019). Fibroblasts were stimulated with profibrotic (TGF-β1, PDGF-AB) or inflammatory (IL-1α, IL-6) cytokines, and treated with antifibrotic agents (ALK5i, Fresolimumab) to assess potential changes in PRO-C3.Results: PRO-C3 was elevated intrinsically in CAFs compared to benign fibroblast. Breast CAFs exhibited high basal PRO-C3 levels and with minimal response to TGF-β1, while benign fibroblasts showed low basal levels with marked induction upon stimulation with TGF-β1. TGF-β1 was the dominant driver of PRO-C3 compared to the other cytokines. Both antifibrotic treatments effectively suppressed PRO-C3 to baseline in both CAFs and TGF-β1-induced benign fibroblast.Conclusions: PRO-C3 reflects both intrinsic and inducible fibrotic activity in breast CAFs and may serve as a surrogate marker of fibroblast activation in breast tumors. PRO-C3 is a clinically relevant biomarker of tumor fibrosis in breast cancer, with elevated circulating levels associated with poor prognosis. Our data support a fibroblast-derived origin of PRO-C3 and highlight TGF-β1 as a key upstream regulator. The observed findings underscore the need for individualized anti-fibrotic strategies in breast cancer management. Integration of PRO-C3 in clinical workflows could enable non-invasive monitoring of fibroblast activity and fibrotic burden, informing prognosis and therapeutic response.References: Nissen et al., 2022, PMID: 35159087Lipton et al., 2018, PMID: 29923614Liu et al., 2021, PMID: 34426581Chen et al., 2019, PMID: 19785660
Presentation numberPS4-05-05
EGFR amplification and PI3K pathway mutations identify a subset of breast cancers that synergistically respond to EGFR and PI3K inhibition
Stan Lipkowitz, Center for Cancer Research, NCI, NIH, Bethesda, MD
D. J. Wiisniewski1, D. J. Voeller1, Y. A. Addissie1, S. K. Deshmukh2, S. K. Wu3, M. B. Lustberg4, D. Wangsa5, D. Wangsa5, K. Heselmeyer-Haddad5, G. Sledge6, S. Lipkowitz1; 1Women’s Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 2Clinical and Translational Research, Caris Life Sciences, Phoenix, AZ, 3Clinical and Translational Research, Caris Life Sciences, Irving, TX, 4Medical Oncology, Yale Univerrsity School of Medicine, New Haven, CT, 5Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, 6Chief Medical Officer, Caris Life Sciences, Irving, TX.
Background: Epidermal Growth Factor Receptor (EGFR) family signaling is commonly dysregulated in cancer by amplification or activating mutations. Although studies have investigated dual EGFR/PI3K inhibition in breast cancer, they have not determined biomarkers that predict success. Here, we present a subset of patients with EGFR amplification and PI3Kinase pathway mutations in breast cancer that can be synergistically targeted by dual EGFR/PI3K inhibition. Methods: Frequency of EGFR amplification and PI3K pathway mutations (PIK3CA, PIK3R1/2, PTEN, AKT1/2/3, TSC1/2/3) were studied through datasets from cBioPortal and Caris Life Sciences. Triple negative breast cancer (TNBC) cell lines used include BT20 (amplified EGFR, PIK3CA activating mutation), MDA-MB-468 (amplified EGFR, PTEN deletion), and MDA-MB-231 (no EGFR or PI3K alterations). EGFR amplification was determined by immunoblot and fluorescent in situ hybridization, and PI3K mutations by sequencing. Signaling was determined by immunoblot, drug synergy by cell viability, cell death by propidium iodide staining, cell cycle analysis by flow cytometry, and xenograft animal study. Caris Life Science CODEai was used to evaluate real-world overall survival (OS) obtained from insurance claims and calculated from date of tumor biopsy to last contact using Kaplan-Meier estimates. Statistical significances were determined by chi-square and Mann-Whitney U test, student’s t-test with 2 tailed comparisons assuming equal variance, one-way or two-way ANOVA were performed as indicated. P-values of ≤ 0.05 were considered significant. Results: EGFR amplification occurs in approximately 1-2.5% of breast cancer patients, more frequently in TNBC (2.45-6.7%) and ER-/HER2+ (1.3-6.5%) breast cancers, and in the molecular basal (2.33-8.1%) and HER2 enriched (1.87-5.4%) subtypes. EGFR amplified patients in cBioPortal and in the Caris datasets had worse median OS compared to those with non-amplified EGFR (mOS: 111.1 m vs 164.6 m, p=0.0269, and 21.6 m vs 32.9 m, HR 0.69, 95% CI 0.58 – 0.82, p<0.0001 for the cBioPortal and Caris data, respectively). cBioPortal and Caris databases showed that 54-71% of EGFR amplified tumors have activating mutations in the PI3K pathway. Our in vitro studies showed combination EGFR and PI3K inhibitors more dramatically reduced MAPK, mTOR and AKT signaling in the BT20 and MDA-MB-468 cells, whereas the inhibition of downstream signaling was less significant in MDA-MB-231 cells. Combination of EGFR and PI3K inhibitors reduced cell viability in these three cell lines, but inhibition was greater, statistically significant, and synergistic in MDA-MB-468 and BT20 compared to MDA-MB-231. Only MDA-MB-468 and BT20 cells had an increased fraction of apoptotic cells with combined pathway inhibition. EGFR or PI3K inhibition alone in a BT20 xenograft model reduced tumor volume, however the combination induced the only statistically significant reduction in tumor volume when compared to vehicle control. Conclusions: EGFR/PI3K inhibitor combination causes apoptosis and reduction in tumor growth in cells with EGFR amplification and PI3K alteration. EGFR amplification with coincident PI3K pathway mutations are drivers in a subset of breast cancers and identify a subgroup of breast cancers that are more likely to respond to dual targeted therapy.
Presentation numberPS4-05-06
Comparative Analysis of PIK3CA, AKT1, and PTEN Reporting Across Commercial NGS Tests in Breast Cancer (BC)
Hope Rugo, City of Hope, Duarte, CA
H. Rugo1, A. Schrock2, J. Lee2, R. Graf2, M. Gearing3, A. Heilmann4, A. Gasco2, N. Vasan5, J. Quintanilha2; 1Department of Medical Oncology & Therapeutics Research, City of Hope, Duarte, CA, 2Clinical Development, Foundation Medicine, Inc, Boston, MA, 3Medical Affairs, Foundation Medicine, Inc, Boston, MA, 4Translational Oncology & Clinical Reporting, Foundation Medicine, Inc, Boston, MA, 5Department of Medicine, New York University, New York, NY.
Introduction: Genomic alterations in PIK3CA, AKT1 and PTEN are biomarkers for the AKT inhibitor Capivasertib (Capi), an approved treatment for metastatic BC (mBC) patients who progressed on ≥1 endocrine therapy or recurred during/within 12 months of adjuvant therapy. Several NGS assays include these genes in their panels; however, it is often unclear which types of alterations are evaluated, particularly PTENloss, and how the results compare across platforms. This study aimed to retrospectively compare two commercially available tissue NGS tests for the detection of short variant mutations (mut) in PIK3CA, AKT1, and PTEN, as well as homozygous PTEN copy loss (PTENloss) and PTEN rearrangements (re) in patients with BC: FoundationOne/FoundationOne®CDx (F1CDx®) and Caris NGS tests. Methods: This study included patients with BC who underwent F1CDx tissue comprehensive genomic profiling and had Caris tissue NGS testing. Data was obtained from the U.S.-wide de-identified Flatiron Health and Foundation Medicine real-world clinicogenomic breast database (CGDB), from ~280 U.S. cancer clinics (~800 sites of care) between 01/2011 and 12/2024. A comparison between F1CDx on-label Capi alterations and Caris NGS, abstracted from electronic health records (EHR)1, was conducted for patients with both tests performed on tumor tissue specimens collected on the same day. Caris NGS specific mutations are not available, so we could not assess if they were on-label for Capi. Positive percentage agreement (PPA) was calculated using F1CDx as a reference. Caris IHC PTEN protein loss results abstracted from EHR were also assessed when available. Results: A total of 74, 85 and 80 patients had PIK3CA, AKT1, and PTEN NGS results available from both assays, respectively, and 68 patients also had a Caris IHC PTEN result available. For PIK3CAmut, we observed an agreement of 98.6 % (73/74), with 53 (71.6%) negative and 20 (27%) positive for both tests, and 1 patient F1CDx-/Caris+ (100% PPA). For AKT1mut, the agreement was 100% with all cases negative for both tests. For PTEN, a 95% (76/80) agreement was observed, with 75 (93.7%) being negative for both tests, 1 (1.25%) with both positive, and 4 (5%) F1CDx+/Caris- (20% PPA). No PTENloss or PTENre was reported by Caris NGS, while 3 patients (3.7%) were positive for PTENloss and 1 patient (1.25%) for PTENre by FMI (0% PPA). Of 8 patients with a PTEN alteration not reported by Caris NGS test and detected on F1CDx, 6 had an IHC PTEN protein loss reported by Caris. Additionally, 4 patients were identified to have PTEN protein loss by Caris’s IHC PTEN results, but no PIK3CA/AKT1/PTEN alterations were identified by either NGS test and two functional copies of PTEN were detected on F1CDx. Conclusions: 29% (8/28) BC patients with on-label Capi alterations detected with F1CDx were not reported by Caris NGS tissue testing. 89% (8/9) of PTEN alterations were not reported by Caris NGS tissue testing, including all PTENloss and PTENre. 75% (6/8) Caris IHC PTEN results reported PTEN protein loss (not a current Capi indication). Further investigation on variations in reporting of different NGS assays in both tissue and plasma and their ability to identify on-label alternations are needed to understand true differences between testing results. Footnotes1 All data analyzed in this study related to the Caris tissue test is based on reported data only.
Presentation numberPS4-05-07
Integrated Genomic Profiling Identifies Predictive Biomarkers for Neoadjuvant Therapy Response in Chinese Breast Cancers
Zhonghua Wang, Fudan University Shanghai Cancer Center, Shanghai, China
X. Ying, K. Zhang, X. Zhu, S. Jiang, X. Hu, C. Chen, Z. Wang; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
BACKGROUNDNeoadjuvant therapy (NAT) has emerged as a standard treatment strategy for locally advanced breast cancer (BC), yet effective predictive markers are still lacking. This study aimed to investigate genomic biomarkers of NAT efficacy in a Chinese BC cohort and characterize subtype-specific molecular signatures associated with differential treatment responses.METHODS A large-scale NAT cohort comprising 1,161 patients with primary BC was analyzed, including 1,145 patients who underwent comprehensive genomic profiling using the FUSCC-BC targeted panel. Mutational signatures predictive of pathological complete response (pCR) and metastatic recurrence were identified. Machine learning models were developed for NAT response prediction. RESULTSThe FUSCC-BC neoadjuvant cohort study integrated clinicopathological analysis and targeted sequencing of breast cancer subtypes (36.9% HER2+, 39.8% HR+/HER2-, 23.3% triple-negative) to identify genomic biomarkers associated with differential pCR rates. We revealed shared resistance mechanisms across subtypes, notably PIK3CA mutations and PI3K pathway activation predicting non-pCR in both HR+/HER2− and triple-negative breast cancer (TNBC), while ERBB2 and GRINA alterations emerged as a HER2-enriched subtype-specific predictor of therapeutic resistance. In patients who fail to achieve pCR, multivariate analysis revealed that multiple genomic alterations (e.g., TP53 and TOP2A) were independently associated with elevated risk of metastatic recurrence. To optimize clinical applicability, a machine learning framework integrating somatic mutation profiles and clinicopathological variables demonstrated robust predictive performance, achieving area under the curve (AUC) values of 0.82 in the training set and 0.81 in the test set for neoadjuvant therapy response prediction.CONCLUSIONOur study identified subtype-specific biomarkers predictive of the response of BC patients to neoadjuvant therapy and established a predictive framework to optimize NAT strategies, providing molecular guidance for personalized treatment regimens that may enhance clinical outcomes. 1
Presentation numberPS4-05-08
Perineural invasion increases the risk of lymph node metastasis in early-stage operable breast cancer:a large-scale propensity-matched cohort study and meta-analysis
Weifeng Cai, Fujian Medical University Union Hospital, Fuzhou, China
W. Cai, Y. Wang, Y. Qiu, S. Liu, Q. Cai, P. He, Y. Lin, M. Chen, L. Chen, F. Fu, C. Wang; Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA.
Background: Recently, perineural invasion (PNI) has emerged as a promising biomarker and a potential therapeutic target in cancer. PNI has been shown to provide prognostic value on locoregional recurrence and distant metastasis in the breast cancer. However, its role in the the progression of lymph node metastasis remains controversial. This study aims to explore the relationship between PNI and the extent of axillary lymph node metastasis (ALNM) in breast cancer. Methods: Patients with early-stage, operable invasive breast cancer who underwent surgery between June 2011 and June 2023 were enrolled. The patients were stratified into PNI-positive and PNI-negative groups based on postoperative pathology. To control for confounding factors and simulate a randomized controlled trial, we used propensity score matching (PSM), inverse probability of treatment weighting (IPTW), and overlap weighting (OW) methods between two groups. Multivariate logistic regression analysis was performed to evaluate the association between PNI and both ALNM and lymph node ratio (LNR). A meta-analysis was conducted to further validate the effect. Results: Among 4,156 early-stage operable breast cancer patients, 1,223 (29.4%) were PNI-positive. Multivariate logistic regression analysis revealed that PNI is independently associated with ALNM (P = 0.002) and a high-risk lymph node ratio (LNR > 0.2) (P = 0.012). These associations remained consistent after sensitivity analyses using PSM, IPTW, and OW methods (all P < 0.05). The meta-analysis included 16 eligible studies with 17,451 participants, further confirmed the significant association between PNI and ALNM (P < 0.001). Additionally, the meta-analysis demonstrated that PNI is linked to worse disease-free survival (DFS) (P < 0.001) and overall survival (OS) (P = 0.007) in patients with breast cancer. Conclusion: PNI is associated with a greater burden of ALNM in patients with early-stage operable invasive breast cancer, underscoring its potential value for risk stratification in the era of axillary surgery de-escalation.
Presentation numberPS4-05-10
Plasma neurofilament light chain levels are prognostic in patients with primary breast cancer and are associated with CTC.
Michal Mego, Faculty of Medicine, Comenius University, National Cancer Institute, Bratislava, Slovakia
M. Mego1, J. Hanes2, V. Parrak2, G. Minarik3, M. Karaba4, B. Mravec5, P. Turcani6, K. Kalavska1, L. Kucerova1, N. Zilka7; 12nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Bratislava, SLOVAKIA, 2Institute of Neuroimmunology, Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, SLOVAKIA, 3Medirex Group Academy, Medirex Group Academy, Bratislava, SLOVAKIA, 4Department of Oncosurgery, National Cancer Institute Slovakia, Bratislava, SLOVAKIA, 5Department of Physiology, Faculty of Medicine, Comenius University, Bratislava, SLOVAKIA, 61st Department of Neurology, Faculty of Medicine, Comenius University, Bratislava, SLOVAKIA, 7Institute of Neuroimunology, Institute of Neuroimunology, Slovak Academy Sciences, Bratislava, SLOVAKIA.
Background: Increased tumor innervation has been associated with more aggressive tumor behaviour and worse clinical outcomes across multiple cancer types. Emerging evidence suggests that heightened neural infiltration may lead to axonal damage. Neurofilaments (NfL), key structural components of neurons, have been proposed as biomarkers of neuronal damage, but their prognostic role in solid tumors remains unclear. This study aimed to assess the clinical significance of plasma neurofilament light chain levels in early-stage breast cancer.Methods: We prospectively analyzed plasma neurofilament light chain concentrations in 80 patients with non-metastatic breast cancer prior to systemic treatment that underwent surgery at National Cancer Institute, Slovakia from 2013 to 2015. NfL level was determined by ELISA. Associations with clinicopathological features and circulating tumor cells (CTCs) were evaluated using descriptive and inferential statistics. Survival outcomes were analyzed using Kaplan-Meier curves and log-rank tests, with a cut-off set higher than 95th percentile of the normative distribution.Results: NfL light chain levels were not significantly associated with tumor size, nodal status, histological grade, hormone receptor, HER2, or Ki-67 status. However, higher NfL levels were significantly associated with negative Bcl-2 expression (mean 13.1 vs. 9.2 ng/mL, p = 0.012). Moreover, patients with high NfL plasma level had detectable CTCs with epithelial-mesenchymal transition phenotype compared to patients with low NfL level (66.7% vs. 25.4%, p = 0.01). Patients with low plasma neurofilament levels had significantly improved disease-free survival (HR = 0.23, 95% CI 0.05–1.08, p = 0.0006) and overall survival (HR = 0.19, 95% CI 0.03–1.16, p = 0.0005) compared to those with high levels. Multivariate analysis confirmed prognostic value of NfL light chain levels independently of tumor stage, nodal status, oestrogen receptor and/or HER2 status.Conclusions: Plasma NfL light chain level concentration is a promising prognostic biomarker in non-metastatic breast cancer, independently of conventional pathological features. Its association with poor survival and CTC EMT phenotype suggests a potential link to tumor aggressiveness and dissemination potential
Presentation numberPS4-05-11
Breast Cancer Drives Detectable Reprogramming in White Blood Cells and Platelets
OLESYA A KHARENKO, Syantra Inc, Calgary, AB, Canada
O. A. KHARENKO1, K. Norek1, J. Kennard1, K. Fuh1, R. D. Shepherd1, K. D. Rinker2; 1Syantra Inc, Syantra Inc, Calgary, AB, CANADA, 2Syantra Inc, Syantra Inc, University of Calgary, Calgary, AB, CANADA.
Background: Tumour-induced immune reprogramming is a key mechanism by which cancers can evade immune surveillance and promote disease progression. In recent in vitro studies, we demonstrated that triple-negative breast cancer (TNBC) cells can modify gene expression in THP1 monocytes, through phenotypic and transcriptomic changes consistent with immune dysfunction. These effects included activation of oncogenic signalling pathways, upregulation of IL6, and enhancement of proliferative signals. Here we report on molecular changes in white blood cells and platelets in women with breast cancer. Methods: Blood samples were collected from participants in the IDBC clinical study (clinicaltrials.gov/study/NCT04495244) prior to biopsy. Samples were fractionated into white blood cells (WBCs) and platelets. RNA was extracted, transcriptomic analysis with RNA sequencing performed, and expression profiles compared between cancer and non-cancer samples. These samples were also compared to those from a model system of THP1 monocytes co-cultured with MDA-MB-231 TNBC cells that was used to identify key regulatory genes. Differential gene expression and pathway analyses were conducted using Ingenuity Pathway Analysis (IPA). Results: For clinical samples, 11,355 differentially expressed genes (DEGs) were identified between cancer and non-cancer: 1,741 were unique to the cancer WBC fraction and 2,070 were unique to the cancer platelet fraction. Key shared upstream regulators between clinical WBCs and platelets and in vitro model THP1 included TRAF2, ZNF281, BMP4, SMAD3, and ITGA11 (WBCs), as well as IFNG, STING1, CDK6, RIPK2, CD44, TGM2, and STAT1 (platelets). In WBCs, the upstream regulators point to a transcriptional landscape shaped by TGF-β signaling, cellular stress response, and integrin-mediated remodeling, consistent with immune tolerance and altered cell trafficking. In contrast, platelet-derived signatures were enriched for IFNG, STING1 and related factors, suggesting a shift toward innate immune sensing, inflammatory signaling, cell cycle activity and adhesion dynamics. Conclusions: WBCs and platelets exhibited differential expression of key immune-related transcripts in cancer versus non-cancer clinical samples, and were in alignment with tumor-induced changes in a in vitro co-culture model. These results demonstrate the presence of tumour-induced blood reprogramming that is detectable in blood, with WBCs reflecting chronic signaling modulation and platelets demonstrating acute immune activation and vascular interaction. This supports the biological basis for using tumor-educated immune changes as a foundation for enabling early detection and therapeutic strategies.
Presentation numberPS4-05-12
Region-specific collagen morphometric parameters in HER2+ breast cancer versus TNBC: insights from SHG/TPE and AI-based analysis
Kutbuddin Akbary, HistoIndex, Singapore, Singapore
K. Akbary1, L. Jiaojiao1, R. Yayun1, D. Tai1, C. Hsiao2, K. Huang2; 1Research and Development, HistoIndex, Singapore, SINGAPORE, 2Dept of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, TAIWAN.
BACKGROUND: Triple-Negative Breast Cancer (TNBC) and HER2-positive (HER2+) breast cancer represent biologically distinct subtypes with differing molecular drivers, clinical behaviours, and responses to therapy. Despite these differences, both subtypes may be associated with significant tumor-stroma interactions and fibrotic remodelling, which may contribute to disease progression and therapeutic response differences. However, comparative spatial characterization of collagen architecture across different tissue compartments in these subtypes remains limited. Second harmonic generation/two-photon excitation (SHG/TPE) microscopy, coupled with AI-based image analysis, enables quantitative assessment of collagen morphometry beyond conventional histopathology. This study aims to compare region-specific collagen features between TNBC and HER2+ breast cancer biopsies and identify distinguishing fibrosis patterns across tumor and non-tumor regions. METHODS: Unstained breast cancer biopsies from TNBC (n=21) and HER2+ (n=158) patients from Dept of Surgery, National Taiwan University Hospital were imaged using SHG/TPE microscopy. Collagen morphometric features were identified using a proprietary AI-based analysis platform (Genesis®200, HistoIndex Pte Ltd, Singapore). Four anatomically distinct tissue regions were assessed: tumor, lobule and duct, stroma-fat, and stroma-fibrosis. Each region comprised of 65 collagen morphometric parameters, and was compared between TNBC and HER2+ groups within each region using normalized Median Relative Differences. Statistical comparisons were performed using the Wilcoxon rank-sum test to evaluate feature-level differences between the two subtypes. RESULTS: Region-specific collagen morphometric profiles demonstrated distinct differences between TNBC and HER2+ breast cancer biopsies. Within the tumor region, collagen parameters were largely comparable between the two groups, with no major differences observed. In the stroma-fibrosis compartment, TNBC samples showed elevations in parameters related to collagen fibres but these trends were not statistically significant. Statistically significant reductions (p < 0.05) of specific collagen parameters were observed in lobule and duct regions of HER2+ biopsies versus TNBC biopsies as follows; 25% decrease in number of thick strings per unit tissue area, a 17% reduction in string perimeter, and decrease of 20% and 18% in width of all strings and width of aggregated strings per unit tissue area, respectively. CONCLUSIONS: Quantitative SHG/TPE imaging combined with AI-based collagen morphometric profiling reveals distinct fibrosis patterns between TNBC and HER2+ breast cancer subtypes. The lobule and duct region in HER2+ biopsies demonstrates a unique collagen architecture when compared with rest of TNBC biopsies. These spatially localized differences in extracellular matrix structure may reflect subtype-specific stromal remodelling and could offer utility in subtype classification, prognosis, or therapeutic response prediction. Further validation of these imaging-derived biomarkers in larger cohorts is warranted.
Presentation numberPS4-05-13
Germline microRNA-based variants and lymph node burden in breast cancer
Joanne Weidhaas, UCLA, Los Angeles, CA
J. Weidhaas1, K. McGreevy2, J. Le3, M. Alcaraz1, E. Rietdorf1, M. Ensenyat-Mendez4, J. Baker5, M. Dinome4, D. Marzese4, D. Telesca6; 1Radiation Oncology, UCLA, Los Angeles, CA, 2Statistics, MiraDx, Los Angeles, CA, 3Surgery, UCSD, San Diego, CA, 4Surgery, Duke, Raleigh, NC, 5Surgical Oncology, UCLA, Los Angeles, CA, 6Statistics, UCLA, Los Angeles, CA.
Background: Germline variants in microRNAs (mirSNPs) have been found to shape tumor biology and to predict both outcome and response to therapy. Here, we tested the hypothesis that mirSNPs predict high versus low lymph node burden in breast cancer patients. Methods: DNA was isolated from microdissected tumor specimens from 73 breast cancer patients with invasive ductal carcinoma (IDC), who underwent axillary lymph node dissection. The mean age at diagnosis was 59.1 years (SD 12.9), and the cohort was racially diverse: 43.8% White, 23.3% Asian, 12.3% Black, 12.3% Hispanic, and 8.2% Middle Eastern. The average BMI was 27.3 (SD 7.3). The majority of patients had HR+/HER2- breast cancer (72.6%), with fewer HER2+ (15.1%) or triple-negative (12.3%) disease. Tumors were primarily grade 2 (41.1%) or grade 3 (46.6%), with a mean Ki67 proliferation index of 29.1% (SD 20.2). T2 was the most common clinical T stage (57.5%), and the mean tumor size at diagnosis was 28.3 mm (SD 19.7). Most patients were clinically node (cN) positive (91.8%), and 57.5% had four or more positive lymph nodes. Low nodal burden was defined as N1 (<3 axillary lymph nodes) and high nodal burden N2/N3 (>4 axillary lymph nodes).The germline mirSNP dataset was filtered prior to modeling, with 22 SNPs demonstrating an association with nodal burden at a Fisher exact p-value cutoff of 0.2. mirSNPs were further reduced by iteratively removing the bottom five predictors by AUC impact and re-fitting models, resulting in a final set of 17 mirSNPs. A set of clinical variables was selected by iterative boosted tree modeling, with features retained if they had non-zero median relative importance across five leave-one-out cross-validation (LOOCV) rounds. This resulted in retention of age at diagnosis, tumor size, BMI, race, Ki67, clinical T stage, cancer subtype (TNBC, HER2+, HR+/HER2-), tumor grade, IDC with lobular features, and use of adjuvant therapies (endocrine, chemotherapy, radiotherapy). These selected clinical features were then used alone and in conjunction with mirSNP genetic data in subsequent predictive modeling. Results: Elastic net regression using the 17 mirSNPs yielded strong predictive accuracy for high nodal burden, with AUCs ranging from 0.76 to 0.79 and specificity values between 0.81 and 0.87. Importantly, the predictive accuracy did not correlate with the number of nodes sampled, and misclassifications by the model were predominantly concentrated around the clinical threshold of four positive nodes- rather than being biased by the depth of nodal sampling. When mirSNPs were combined with clinical variables in the predictive models, the resulting performance was lower than for genetics alone, with AUCs ranging from 0.67 to 0.71. Models using only clinical variables performed the worst, achieving an AUC of 0.55. The most significant mirSNP in the model was in P2RX7, a gene whose expression has been previously associated with the risk of bone metastases in breast cancer, and the second most significant mirSNP in the model was in CSMD1, a gene that is linked to the spread of breast cancer to lymph nodes. Conclusions: Our findings support the potential of mirSNPs as an additional tool to help identify patients at increased risk of high nodal burden in breast cancer. Our mirSNP model outperformed clinical models in this data set. Studies investigating the benefit of incorporating mirSNPs with other biomarkers of lymph node burden, the association of this signature with outcome, as well as further validation of these findings are ongoing.
Presentation numberPS4-05-14
Integrated NGS-Based Tissue and Plasma Profiling to Advance Precision Oncology in Breast Cancer
Dimitrios Mavroudis, University General Hospital of Heraklion, Heraklion, Greece
D. Mavroudis1, D. Jinga2, E. Razis3, S. Giassas4, N. Touroutoglou5, C. Bilir6, M. Ozdogan7, I. Ökten8, S. Karageorgopoulou9, E. Galani10, G. El Hachem11, D. Tzanninis12, T. Floros13, G. Lazaridis14, P. Vlachostergios15, I. Hacibekiroglu16, E. Maragkouli17, I. Gioulbasanis18, A. Tsantikidi19, S. Maxouri19, C. Florou-Chatzigiannidou19, V. Metaxa-Mariatou19, E. Papadopoulou19, D. Grigoriadis20, A. Papathanasiou20, G. Nasioulas19; 1Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, GREECE, 2Oncology Department,, Neolife Medical Center, Bucharest, ROMANIA, 33rd Oncology Department, Hygeia Hospital, Marousi, Athens, GREECE, 4General Maternity and Gynecology Clinic, Iaso General Clinic, Marousi, Athens, GREECE, 5Oncology Department, European Interbalkan Medical Center, Thessaloniki, GREECE, 6Clinic of Medical Oncology, VM Medical Park Pendik Hospital, Istanbul, TURKEY, 7Clinic of Medical Oncology, Memorial Antalya Hospital, Antalya, TURKEY, 8Department of Medical Oncology, Göztepe Prof. Dr. Süleyman Yalçın City Hospital, Istanbul, TURKEY, 93rd Oncology Clinic, Iaso General Clinic, Marousi, Athens, GREECE, 102nd Oncology Clinic, Perseus Healthcare Group SA, Metropolitan Hospital, Neo Faliro, Athens, GREECE, 11Department of Hematology and Medical Oncology, Saint George Hospital University Medical Center, Beirut, LEBANON, 12Oncology Department, Athens Medical Center, Marousi, Athens, GREECE, 13Department of Oncology, Athens Naval and Veterans Hospital, Athens, GREECE, 14Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, GREECE, 15Oncology Clinic, IASO Thessalias S.A. – General Hospital, Larissa, GREECE, 16Faculty of Medicine, Department of Medical Oncology, Sakarya University, Sakarya, TURKEY, 17Medical Oncology Dpt and Clinic, General Hospital of Trikala, Trikala, GREECE, 18Department of Oncology, Thessalia General Clinic, Larissa, GREECE, 19Molecular Oncology, Genekor Medical SA, Gerakas, Athens, GREECE, 20Department of Bioinformatics, Genekor Medical SA, Gerakas, Athens, GREECE.
Introduction: The integration of broad genomic testing has become increasingly vital for personalized treatment planning in breast cancer patients. In this study, we evaluated the analytical performance and clinical relevance of a certified for in vitro diagnostics (CE-IVD), 1021-gene comprehensive genomic profiling panel in detecting actionable tumor biomarkers across a diverse cohort of breast cancer cases. The study aimed to validate the robustness of the assay in both tissue and liquid biopsy formats, and to explore its clinical utility in real-world patients with advanced breast cancer. Methods: Tumor and blood samples from 353 patients were analyzed, including 197 FFPE tumor and 156 plasma cfDNA. Concurrent molecular profiling of tissue and plasma was conducted in 23 cases to evaluate the efficacy of liquid biopsy although tissue samples may have originated from prior procedures. DNA extracted from leukocytes was used as a control to prevent the detection of false-positive results due to clonal hematopoiesis mutations. Targeted-capture sequencing was performed using the Oncology Multi-Gene Variant Assay (GenePlus) which is a qualitative in vitro diagnostic test for detection of variants in 1021 tumor-related genes, and gene fusions in 38 genes. Sequencing was carried out on the MGI DNBSEQ-G400 platform. NGS data were analyzed via a dedicated bioinformatics pipeline on the Gene Box platform (GenePlus), enabling detection of all major genomic alterations, including gene fusions, as well as assessment of tumor mutational burden (TMB) and microsatellite instability (MSI). Results: Regarding the analysis of 197 FFPE samples, at least one on-label actionable biomarker was identified in 54.8% of cases. Among these samples, PIK3CA mutations were the most frequently detected in 27.4% of biomarker-positivethe cases. ESR1 mutations were present in 16.2% Mutations in PTEN and AKT1 genes were also observed in 7.6% and 3% of cases, respectively. Additionally, 20% of patients harbored a suspected germline variant in BRCA1/2, PTEN and CDH1 genes. In the analysis of 156 plasma samples, 46% were positive for an on label-biomarker. Among these, PIK3CA alterations were identified in 17.9% and ESR1 mutations were present in 17.3% of the cases. At lower frequency, variants were also observed in BRCA1/2 (3.8%), PTEN (3.2%), AKT1(1.3%) genes. ERBB2 (1.3%) amplification and an NTRK fusion (0.6%) were also detected. Approximately 20% of plasma samples harbored verified germline variants, most frequently in BRCA1/2 and CHEK2 genes. In a subset of 23 mBC patients, concurrent analysis of tissue and liquid biopsy demonstrated a 70% overall concordance rate on on-label biomarker analysis. In two cases, FFPE analysis identified additional on-label mutations not detected in plasma, whereas in one case plasma analysis revealed a second ESR1 mutation not detected in the FFPE sample. In two patients, plasma analysis was negative, while tissue profiling identified alterations in ESR1 and PIK3CA genes. Importantly, in two cases, liquid biopsy surpassed tissue analysis by detecting an ESR1 variant and an NTRK fusion. Conclusions: This study demonstrates the clinical utility of comprehensive multigene NGS profiling in both tissue and plasma samples from patients with breast cancer by revealing a high rate of actionable variants. Parallel analysis of matched tissue and plasma samples revealed a high concordance rate, indicating complementary role of both approaches in genomic profiling. In conclusion, integrated NGS analysis of tissue and liquid biopsies provides a powerful platform for precision oncology in breast cancer, supporting therapeutic decisions, resistance monitoring, and hereditary cancer risk assessment. The combined use of both sample types maximizes diagnostic yield and clinical impact, especially in the context of advanced disease.
Presentation numberPS4-05-15
Immunohistochemical characterization of the tumor cells present in four body fluids from patients with metastatic breast cancer in the context of the UPTIDER program
Camille Carette, KU Leuven, Leuven, Belgium
C. Carette1, G. Zels1, A. Pabba1, M. Maetens1, K. Borremans1, K. Van Baelen1, J. Van Cauwenberge1, T. Geukens2, M. De Schepper3, H. Nguyen1, M. Nysen4, H. Hoogsteyn1, T. Van Assche1, A. Mahdami1, S. Leduc1, P. Vermeulen5, P. Neven6, H. Wildiers2, W. Van Den Bogaert7, G. Floris3, F. Richard1, S. Hatse4, C. Desmedt1; 1Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, BELGIUM, 2Department of General Medical Oncology, University Hospitals Leuven, UZ Leuven, Leuven, BELGIUM, 3Department of Pathology, University Hospitals Leuven, UZ Leuven, Leuven, BELGIUM, 4Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, BELGIUM, 5Translational Cancer Research Unit, ZAS Augustinus, Antwerp, BELGIUM, 6Department of Gynaecology and Obstetrics, University Hospitals Leuven, UZ Leuven, Leuven, BELGIUM, 7Department of Forensic Medicine, University Hospitals Leuven, UZ Leuven, Leuven, BELGIUM.
Background. In patients with metastatic breast cancer (BC), tumor cells can be observed in blood as well as in other fluids such as pleural fluid (PF), ascites (ASC), pericardial fluid (PERI) and cerebrospinal fluid (CSF). Here, we aimed at evaluating the association between presence of tumor cells in non-blood liquid biopsies (LB) and presence of metastases in surrounding tissues (aim 1), and assessing the heterogeneity of BC biomarkers (estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2) and KI67) in these non-blood LB (aim 2) from patients with metastatic BC included in our post-mortem tissue donation program, UPTIDER (NCT04531696, PMID: 38658604). Methods. 38 patients were included in aim 1. When possible, non-blood LB samples were collected during autopsy and instantly centrifuged. Cell pellets were formalin-fixed, paraffin-embedded, and evaluated histologically for tumor cellularity. Associations between the presence of tumor cells in LB and solid metastases in surrounding tissues were explored with a Fisher’s exact test. All LB with ≥5% tumor cellularity and primary tumor samples were considered for aim 2 and immunohistochemically stained for ER, PR, HER2 and KI67. Results. Aim 1. 155 samples (52 PF, 30 ASC, 34 PERI and 39 CSF) were analyzed with a median of 4 LB per patient (range 2-7). Malignant PF was observed in 29 out of 31 patients with collected PF. All except one patient (28/29) had pleural, lung and/or mediastinal metastases. Malignant ASC was observed in 18 from 27 patients with collected ASC. 17/18 had peritoneal, liver and/or gastro-intestinal metastases. Malignant CSF was detected in 6 out of 36 patients with collected CSF. Four (4/6) had brain and/or meningeal metastases. Malignant PERI was observed in 19 out of 34 patients with collected PERI, of whom 5/19 had pericardial or heart metastases. No statistical evidence was found for association of presence of tumor cells in PF, ASC or CSF and solid metastases in surrounding tissues (Fisher’s exact p-value (p) = 0.13, 1.00 and 0.37, respectively). However, a trend was seen for PERI and pericardial/heart involvement (p = 0.053). Aim 2 was evaluated on 69 samples from 27 patients. Among them, 20 presented with primary hormone receptor (HR)+/HER2- BC (18 ER+/PR+, 1 ER+/PR- and 1 ER-/PR+), and 7 patients had primary HR-/HER2- BC. Regarding intra-patient heterogeneity across different LB, the median variation in expression was 5% for ER, 0.5% for PR, 0% for HER2, and 13% for KI67 with maximum differences of 90%, 50%, 60% and 70% respectively. Of note, 10/19 (53%) patients diagnosed with primary ER+ BC, had at least 1 LB that lost ER-expression. Similarly, 14/19 (74%) patients with primary PR+ BC, lost PR-expression in at least 1 LB. All patients had HER2 non amplified primaries, but 11/27 (41%) had at least 1 LB with HER2-low and/or HER2-ultralow expression, while the other 16 patients (59%) showed no detectable HER2 expression in any of their LB. KI67 value from the primary was available for 17 patients. Twelve out of 17 patients presented with low KI67 (≤15%) at primary diagnosis of whom 5 had at least one LB with high KI67 (>15%). The remaining 5 patients presenting with high KI67 at primary diagnosis all had at least one LB with low KI67. Conclusions. This study reports on the detection of tumor cells and expression of clinically relevant biomarkers in LB from patients with metastatic BC. Our findings suggest that a multi-fluid biopsy approach, when possible, may improve the characterization of the metastatic disease heterogeneity and aid in selecting the best next-line treatment options.
Presentation numberPS4-05-16
Multi-omics analysis identifies EFR3B as a driver of chemoresistance in breast cancer through epithelial-endothelial cell communication
Yilong Lin, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
Y. Lin1, S. Lin2, Y. Zhang3, J. She1, R. Zhao1, A. Qiu1, L. Zhang1, Q. Yang1; 1Department of Breast Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, CHINA, 2School of Medicine, Xiamen University, Xiamen, CHINA, 3Medical college, Guangxi University, Nanning, CHINA.
Background: Breast cancer is one of the most common malignancies in women. Although previous genome-wide association studies (GWAS) have identified multiple susceptibility loci, they account for only a small portion of the heritable risk. Transcriptome-wide association studies (TWAS), which integrate GWAS and expression quantitative trait loci (eQTL) data, offer a more effective strategy for uncovering functional genes involved in complex traits. This study aims to systematically identify breast cancer susceptibility genes through multiple TWAS frameworks and to elucidate their biological roles using multi-omics integration. Methods: Firstly, we performed cross-tissue TWAS for breast cancer using both UTMOST and JTI frameworks. Next, single-tissue associations were evaluated via FUSION and validated using MAGMA. We further prioritized key genes through Mendelian Randomization (MR) and colocalization analyses, followed by experimental validation. Single-cell and spatial transcriptomic data were employed to delineate gene expression patterns, intercellular communication, and spatial heterogeneity. Additionally, drug resistance profiles were constructed using TCGA transcriptomic data and verified across multiple pharmacogenomics databases. Results: Cross-tissue TWAS analysis identified 29 candidate susceptibility genes for breast cancer. The FUSION method detected 1,768 genes with FDR < 0.05 in at least one tissue, while MAGMA analysis revealed 354 breast cancer-associated genes. By integrating results from four analytical approaches, we prioritized 13 high-confidence susceptibility genes, including EFR3B, CASP8, and XBP1. MR and colocalization analyses further confirmed EFR3B and CASP8 as causal genes with shared genetic architecture in breast cancer. EFR3B, a susceptibility gene for both ER-positive and ER-negative breast cancer, was experimentally validated to be overexpressed in breast cancer cell lines and tumor tissues using Western blot, real-time PCR, and immunohistochemistry. Single-cell transcriptomic analysis revealed that EFR3B was predominantly expressed in specific epithelial and endothelial subpopulations within breast tumors, exhibiting notable expression heterogeneity. Analysis of VEGFA ligand-receptor signaling indicated enhanced intercellular communication between EFR3B-positive epithelial and endothelial cells. Spatial transcriptomics demonstrated heterogeneous expression of EFR3B in tumor tissues and showed that EFR3B-positive epithelial cells are spatially co-localized with vascular endothelial cells, suggesting a potential role for EFR3B in modulating the tumor microenvironment. Finally, drug sensitivity analysis revealed that high EFR3B expression is significantly associated with resistance to paclitaxel and anthracycline-based chemotherapies. Conclusions: This study identified 13 breast cancer susceptibility genes through integrative TWAS approaches, providing new insights into the genetic architecture of the disease. EFR3B emerged as a key risk gene involved in tumor microenvironment modulation and chemotherapy resistance, representing a potential therapeutic target for personalized treatment strategies in breast cancer.
Presentation numberPS4-05-18
The predictive value of tumor-to-gland density ratio in pathological complete response to Neoadjuvant chemotherapy in breast cancer
Xiaoyun Mao, The First Affiliated Hospital of China Medical University, Shenyang, China
W. Shijing, W. Hongrui, X. Mao; Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, CHINA.
Objective: Identifying effective predictors of response to neoadjuvant chemotherapy (NAC) is essential for personalized breast cancer treatment. Our previous observations suggested that a higher tumor-to-gland density ratio (TGDR) on mammography may predict poorer therapeutic response. This study evaluated the predictive value of TGDR in NAC and developed models based on various pathological complete response (pCR) criteria. Methods: This retrospective study included 303 breast cancer patients who received NAC at the First Affiliated Hospital of China Medical University from January 2020 to July 2024. pCR was defined by four criteria: pCR1 (ypT0/is), pCR2 (ypT0/is ypN0), pCR3 (ypT0), and pCR4 (ypT0 ypN0). Patients were randomly divided into training and test sets (7:3). Logistic regression analyses identified independent predictors for each pCR criterion, and multiple logistic regression (LR) models were constructed. Model performance was assessed using AUC, F1 score, DeLong test, DCA, NRI, and IDI. Four machine learning algorithms (SVM, K-nearest neighbor, XGBoost, and LightGBM) were applied to compare predictive performance. Results: For pCR1, 140 patients (46.2%) achieved response. TGDR, ADC, ER, HER2, histological grade, and molecular subtype were independent predictors. Combined-variable models (AUCs: 0.840-0.852) outperformed single-variable models and demonstrated good clinical utility. Model 3 was selected for its simplicity and MRI independence; the LR algorithm performed best. For pCR2 (n=127), similar predictors were identified. Combined models (AUCs: 0.847-0.859) showed strong performance; SVM performed best. For pCR3 (n=118), model 2 (TGDR + pathological features) outperformed TGDR alone (AUC: 0.848 vs. 0.705); XGBoost performed best. For pCR4 (n=109), model 2 (TGDR, ER, HER2) outperformed model 1 (TGDR alone) in both training and test sets; the LR model performed best. Conclusion: TGDR is a novel imaging biomarker with predictive performance comparable to ADC and is an independent predictor of NAC response. Across all pCR criteria, TGDR-based models combined with clinicopathological features consistently outperformed TGDR alone. TGDR offers a practical alternative for NAC response prediction, particularly in settings with limited MRI access.
Presentation numberPS4-05-19
Enumeration of circulating tumor cell (CTC) clusters as a function of age or neutrophil -to-lymphocyte-ratio (NLR) in women with metastatic breast cancer
Marcus Vetter, Cantonal Hospital Baselland, Liestal, Switzerland
M. Vetter1, T. Kanthaiah1, E. Chiru1, C. Francesc2, I. Krol2, A. Wicki3, A. Ring4, B. Nguyen-Straeuli5, V. Heinzelmann-Schwarz5, W. Weber6, C. Kurzeder6, N. Aceto2, F. Schwab5; 1Cancer Center and Center for Oncology and Hematology, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 2Institute of Molecular Health Sciences, ETH Zürich, Zürich, SWITZERLAND, 3Comprehensive Cancer Center, University Hospital Zürich, Zürich, SWITZERLAND, 4Medical Oncology, University Hospital Zürich, Zürich, SWITZERLAND, 5Gynecology and Obstetrics, University Hospital Basel, Basel, SWITZERLAND, 6Breast Clinic, University Hospital Basel, Basel, SWITZERLAND.
Background: CTC clusters are established mediators of metastasis in breast cancer and are associated with unfavorable clinical outcomes. However, their prevalence and phenotypic characteristics in older patient populations remain inadequately studied. Systemic inflammation, as measured by the NLR, may influence CTC dynamics. Methods: A prospective translational analysis was conducted in women with metastatic breast cancer (mBC), stratified into two cohorts: Group A (age ≥70 years) and group B (age <70 years). Peripheral blood samples were processed using the FDA-cleared Parsortix® system for CTC enrichment, followed by immunofluorescence staining (EpCAM/HER2/EGFR/CD45) to identify single CTCs and CTC clusters (homotypic and heterotypic). Baseline clinical characteristics, including histological subtype, stage, tumor grade, Ki-67 index, and sites of metastasis, were documented. NLR ≥4 was defined as elevated. Primary endpoints included CTC cluster frequency and size; secondary endpoints assessed correlations with age group and NLR status. Results: A total of 155 patients were evaluable, including 58 patients in group A (mean age: 78 years). A trend for elevated NLR being more frequent in Group A compared to Group B was visible (53.2% vs 36.8%, P = 0.075). No statistically significant differences were observed between groups regarding histological subtype, stage at diagnosis, or tumor grade. The Ki-67 proliferation index was significantly elevated in group B (mean 37%) compared to Group A (mean 21%, P < 0.001). The pattern of metastatic spread was comparable, although a trend toward increased brain metastases was observed in Group B. A trend showing a higher proportion of patients exhibiting ≥1 CTC in Group B was detected (43.5% vs. 29.3%, P = 0.082). CTC clusters were identified in 6.9% of Group A and 12.5% of Group B (P = 0.275). Elevated NLR (≥4) was not significantly associated with CTC cluster presence (11.1% vs. 8.2%, P = 0.594). Conclusions: While older patients with mBC (Group A) exhibited a trend for higher systemic inflammation as indicated by NLR, no statistically significant differences in CTC cluster frequency or size were observed compared to younger patients (Group B). Moreover, elevated NLR was not independently predictive of increased cluster prevalence. These findings underscore the need for further investigation into age-associated CTC phenotypes and their potential clinical implications in larger patient populations.
Presentation numberPS4-05-20
Genomic Diversity of Breast Carcinomas Exhibiting Biomarker Triads Derived from Quantified Levels of ER, PR and HER2 Proteins or from Expression of Their Cognate Genes
Michael W Daniels, University of Louisville, Louisville, KY
M. W. Daniels1, J. L. Wittliff2; 1Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, 2Biochemistry & Molecular Genetics and Institute for Molecular Diversity & Drug Design, University of Louisville, Louisville, KY.
Background: Clinical assessment of breast cancer progression and therapeutic response typically relies on qualitative immunohistochemistry results for ER, PR, and HER2 proteins, which may be improved by adherence to ASCO/CAP Guidelines. To identify more robust, clinically relevant molecular markers, we conducted in silico analyses of a comprehensive, de-identified database that integrates quantified protein biomarker levels, qPCR, and microarray gene expression data, as well as long-term clinical outcomes. Because TNBC represents an aggressive disease that is difficult to treat, our multifarious investigation focuses on differentiating biomarkers for this subtype that improve clinical outcomes. Methods: Biomarker Triads were created from ER and PR proteins quantified by radio-ligand binding (n = 13,966) and enzyme immunoassays (n = 4,408), expressed as fmol/mg cytosol protein. HER2 protein was quantified in 504 and 901 specimens using either ELISA or EIA, respectively, and expressed as HNU/mg membrane protein. Clinical data were available for 1,194 ER/PR cases, and complete ER/PR/HER2 protein Triads were available for 189 of these. However, survival outcomes (PFS and OS) were only available for a subset of these patients. Expression levels of these genes and 82 other cancer-associated genes were validated by RTqPCR of 580 specimens after RNA extraction and purification. Microarray analysis (~22,000 genes) was conducted on RNA from each of 247 LCM-isolated carcinoma samples. Biomarker Triads were defined as ER+/PR+/HER2+ (TPBC) or ER-/PR-/HER2- (TNBC), using either protein levels or cognate gene expression (ESR1, PGR, ERBB2). Differential gene expression was assessed using nonparametric tests and Spearman correlations in R (version 4.4.2). For the subset of patients with survival data, exploratory Kaplan-Meier and Cox proportional hazards models were used to assess associations between gene expression and progression-free survival (PFS) or overall survival (OS). Results: Neither ER/PR assay platform influenced ER+/PR+ or ER-/PR- status, and neither ER nor PR levels were associated with HER2 expression. TPBC biopsies showed elevated expression of BL2 (CADM1), CAXII, ERBB2, and ERBB4 compared to TNBC, supporting their use in subtype definition. TPBC also exhibited increased IL10 and VEGFATV2, while TGFB1 was downregulated in both TPBC and ER+ cases. BRCA1/2 expression was reduced in ER-/PR- tumors, while NAT1/2 and COMT were elevated in ER+, ER+/PR+, and TPBC samples. FOXA1 and ART3 emerged as potential targets from gene-based Triad definitions using microarray data. In a subset of cases with available clinical outcomes, exploratory survival analysis suggested that TPBC triads were associated with more favorable PFS and OS compared to TNBC, and elevated expression of certain genes (e.g., ESR1, FOXA1) trended with improved survival outcomes. Conclusions: This integrative dataset, which links rigorously collected clinical biopsies, precise biomarker protein quantification, and gene expression profiles from LCM-procured carcinoma cells, enabled the discovery of genes differentially expressed across biomarker-defined subtypes. Biomarker Triads based on protein and gene expression levels in primary breast tissue biopsies revealed a landscape of genomic diversity in carcinomas, which improved molecular subtype classification. Select genes, including the transcription factor FOXA1 and ART3, reported to be involved in cell migration, represent probable targets for drug development and/or companion diagnostics in ER+ and triple-negative breast carcinomas.
Presentation numberPS4-05-21
The Molecular and Immune Landscape of HER2 Positive Breast Cancer
Yuan Gao, Houston Methodist Hospital, Houston, TX
Y. Gao1, M. Weitz2, S. Fragkogianni2, K. Layng3, H. Mai1, J. Chang1; 1Neal Cancer Center, Houston Methodist Hospital, Houston, TX, 2Data Science, Tempus AI, Chicago, IL, 3Medical Affairs, Tempus AI, Chicago, IL.
Background Recent molecular profiling advances have transformed breast cancer treatment. These breakthroughs have largely benefited patients with hormone receptor-positive and triple-negative breast cancers, while HER2-positive (HER2+) breast cancer remains less understood molecularly. We evaluated the tumor immune microenvironment of HER2+ breast cancer, assessing immune cell infiltration, TMB, PD-L1 status, and HLA allele prevalence, to uncover biomarkers for treatment guidance. MethodsWe used Tempus Lens (Tempus AI, Inc., Chicago, IL) to identify 455 female patients with HER2+ breast cancer and xT and xR testing. Patients were classified as having localized (stage 1-3, N = 124) or de novo metastatic disease (stage 4, N = 331). Wilcoxon rank sum, Fisher’s exact, and Pearson’s Chi-squared tests were used to compare groups. Immune cell proportions were estimated using quanTIseq. TMB, PD-L1, HLA, MSI, and MMR were also analyzed. Real-world progression-free survival (rwPFS) was time from therapy (neoadjuvant or first-line) start to progression. Real-world overall survival (rwOS) was time from first-line therapy start to death or loss to follow up. Median rwOS and rwPFS were estimated with Kaplan-Meier (KM) and hazard ratios (HR) using Cox proportional hazard models (CoxPh). Results In patients with localized disease, the median proportion of tumor-infiltrating B cells was significantly higher than in those with metastatic disease (p < 0.001). A similar pattern was observed for natural killer (NK) cells, CD8⁺ T cells, and regulatory T cells (Tregs), with enrichment in localized stage patients (p<0.001, p<0.001, and p 10 mutations/Mb) was observed in 6.7% of patients with localized disease and 10% of patients with de novo metastatic disease. 29% of patients with localized disease were PD-L1 IHC positive, as were 11% of patients with de novo metastatic disease. Interestingly, in metastatic patients who received standard chemotherapy and/or anti-HER2 therapy as first line treatment, positive PD-L1 status and high TMB were significantly associated with worse median rwOS (p<0.021 and p<0.001, respectively). Across all HER2+ breast cancer patients with patient-reported race information available, the HLA-A *02:01 allele exhibited the highest overall frequency (43%). Stratified by race, this allele was most prevalent in White patients (48%), with significantly lower frequencies observed in Asian (25%) and Black (24%) patients (p=0.018), consistent with previous studies. In contrast, the HLA-A *33:03 allele demonstrated a markedly higher frequency in Asian patients (33%), while its prevalence was substantially lower in Black (17%) and White (1.4%) patients (p<0.001). Similarly, the HLA-A *23:01 allele was more frequent in Black patients (36%) compared to White (5.2%) and Asian (0%) patients (p<0.001). Conclusions Our findings suggest a potential therapeutic benefit of incorporating immunotherapy into the treatment paradigm for HER2+ breast cancer, in both localized and metastatic disease settings. Moreover, the ethnically enriched HLA polymorphisms within our patient cohort may help elucidate observed disparities in clinical outcomes and offer valuable insights for the development of population-tailored immunotherapeutic strategies.
Presentation numberPS4-05-22
Eprs1 induces ac<sup>4</sup>c modification of mRNAs encoding pluripotency factors in breast cancer cells
Jeetendra K Nag, Cleveland Clinic Research, Cleveland Clinic, Cleveland, OH
J. K. Nag, G. Subedi, I. Zin, P. L. Fox; Cardiovascular and Metabolic Sciences, Cleveland Clinic Research, Cleveland Clinic, Cleveland, OH.
Breast cancer remains a formidable clinical challenge, underscored by the persistence of cancer stem cells (CSCs) that drive tumorigenesis and therapeutic resistance. CSCs are characterized by elevated expression of pluripotency-associated transcription factors, including SOX2, OCT4, and NANOG (SON factors), which orchestrate self-renewal and tumor maintenance programs. The downstream transcriptional targets of the SON factors have been investigated in detail; however, the upstream regulation of the factors is less well understood. We previously reported that activation of AKT, or chemical or genetic inhibition of PTEN, induces nuclear translocation of EPRS1 (glutamyl-prolyl tRNA synthetase) a cytoplasmic protein essential for accurate interpretation of the genetic code during translation (1). The contribution of EPRS1 to breast cancer progression is supported by reports that EPRS1 mRNA and protein are elevated in human breast tumors, and associated with reduced overall survival of breast cancer patients (2, 3). In view of reports that AKT activation is a principal determinant of expression of SOX2 (4), the master switch of pluripotency factors, we considered the potential role of AKT-induced nuclear localization of EPRS1 in SON factor expression. siRNA-mediated knockdown of EPRS1 in PTEN– MDA-MB-468 and HCC38 cells markedly inhibited expression of the mRNAs encoding the three SON factors, as shown by RT-qPCR; protein expression was similarly diminished. Knockdown of other aminoacyl-tRNA synthetases, e.g., LARS1 (leucyl-tRNA synthetase), was ineffective, thereby showing specificity. Ablation of EPRS1 in breast cancer cells reduced stemness, migration, invasion, and clonogenic potential. Moreover, limited dilution assays in mice showed EPRS1 knockdown significantly reduced tumor mass and tumor-initiating cell number. A proteomic analysis of EPRS1-interacting proteins in nuclear lysates of MDA-MB-468 cells gave an insight into the mechanism underlying regulation of SON expression by EPRS1 (1). Mass spectrometric analysis revealed interaction of nuclear EPRS1 with RNA cytidine acetyltransferase (NAT10), the only enzyme known to acetylate RNAs, including mRNAs, at the 4-cytidine position, i.e., ac4C modification (5). The interaction was validated by co-immunoprecipitation. Knockdown of NAT10 reduced ac4C modification of SON mRNAs as determined by ac4C-RIP (RNA immunoprecipitation) as well as SON mRNA expression, indicating an important role for ac4C modification in SON expression. Importantly, EPRS1 knockdown likewise inhibited ac4C modification of SON mRNAs, revealing for the first time a role for EPRS1 in mRNA modification. These results implicate EPRS1 and ac4C modification of SON factors as key drivers of cancer stem cell activity and suggest new targets for breast cancer treatment. References: 1. I. Zin et al., AKT-dependent nuclear localization of EPRS1 activates PARP1 in breast cancer cells. Proc. Natl. Acad. Sci. U. S. A. 121, e2303642121 (2024). 2. I. Katsyv et al., EPRS is a critical regulator of cell proliferation and estrogen signaling in ER+ breast cancer. Oncotarget 7, 69592-69605 (2016). 3. L. Qi et al., Significant prognostic values of differentially expressed-aberrantly methylated hub genes in breast cancer. J. Cancer 10, 6618-6634 (2019). 4. Z. Wang et al., AKT drives SOX2 overexpression and cancer cell stemness in esophageal cancer by protecting SOX2 from UBR5-mediated degradation. Oncogene 38, 5250-5264 (2019). 5. D. Arango et al., Acetylation of cytidine in mRNA promotes translation efficiency. Cell 175, 1872-1886 e1824 (2018).
Presentation numberPS4-05-23
Investigating Interferon Pathway Biomarkers as Predictors of Disease Free Survival in Triple Negative Breast Cancer
Kathleen Yuan, Washington University in St. Louis, Saint Louis, MO
K. Yuan1, S. Humble2, A. Mabry1, R. Kladney1, L. Maggi1, C. X. Ma1, J. D. Weber1, G. A. Colditz3; 1Division of Oncology, Washington University in St. Louis, Saint Louis, MO, 2Division of Public Health Sciences Department of Surgery, Washington University in St. Louis, Saint Louis, MO, 3Division of Public Health Sciences in the Department of Surgery, Washington University in St. Louis, Saint Louis, MO.
Background: Triple-negative breast cancer (TNBC) accounts for approximately 15-20% of all breast cancers, characterized by aggressive clinicopathology, limited targeted therapies, and high rates of early recurrence. Immune activation, particularly type I interferon (IFN) signaling, has emerged as a key modulator of tumor behavior, response to therapy, and patient outcomes in TNBC. However, the impact of IFN pathway regulators on long-term prognosis remains unclear.As modulators of immune activation, ADAR1 and ISG15 are IFN pathway genes that may influence the tumor-immune microenvironment and clinical outcomes in TNBC. This study investigates whether ADAR1 and ISG15 expression are associated with disease-free survival in TNBC, aiming to clarify their prognostic significance and potential interplay as immune-modulatory biomarkers in this high-risk breast cancer subtype. We hypothesized that the expression of ADAR1 and ISG15 would be linked to DFS events.Methods: This cohort was prepared by the St. Louis Breast Tissue Registry, derived from tissue microarrays (TMAs) constructed from surgical tumor blocks of 326 patients with TNBC diagnosed between 1973 and 2022 who underwent curative surgery as the immediate therapy at the Barnes-Jewish Hospital and Siteman Cancer Center. All clinical data and tissue specimens were collected by the Washington University Institutional Review Board (IRB #201102394). Information was deidentified prior to distribution of TMA to investigators. For this study, disease-free survival (DFS) is defined as the time from diagnosis to recurrence, second primary malignancy, death or last disease-free follow-up. ISG15 and ADAR1 expression were examined by immunohistochemistry and scored by Allred and HALO. Clinical and pathological data were systematically annotated and analyzed using custom R scripts. Descriptive statistics were employed to summarize clinical and molecular variables. Cox proportional hazards models were generated to assess the association of biomarker expression with DFS. Kaplan-Meier survival analysis was used to evaluate clinical outcome and additional models were constructed to explore the impact of clinical stage and race on DFS.Results: The median age was 56.5 years, with 225 White and 101 Black patients. 221 patients were disease-free at follow-up. Median follow-up time was 10 years. In this cohort, stage was prognostic for DFS in multivariate analysis. Cytoplasmic ADAR1 and ISG15 expression were significantly higher in Black patients compared to White patients (6.49 vs. 5.95, 0.92 vs. 0.84, p = 0.044, 0.041). However, race was not independently predictive of disease-free survival in this model (p = 0.80).Higher ISG15 expression was associated with improved DFS through univariate analysis (p = 0.03) but not in multivariate analysis that included stage. Interestingly, patients who were disease-free at follow-up had significantly higher ADAR1 expression through univariate analysis (mean difference = 0.571, 95% CI: [-1.09, -0.05], p = 0.030). Kaplan-Meier analysis confirmed that high expression was correlated with DFS (p = 0.012). However, in this cohort, patients with higher ADAR1 expression also have a higher rate of adjuvant chemotherapy (p=0.01) and those treated with chemotherapy are more likely to be disease-free than patients who did not undergo chemotherapy (p=0.012).Conclusion: To our knowledge, this study is the first to examine the association between IFN pathway regulators, ISG15 and ADAR1, and TNBC prognosis. The finding of increased ISG15 in association with improved DFS is consistent with the likely activated IFN pathway state for these tumors. Additional analysis is ongoing to examine the association of these markers with immune cell infiltration and the expression of immune checkpoints.
Presentation numberPS4-05-24
Development of a Novel Platelet RNA Based Biomarkers for Breast Cancer Detection
Jong-Ho Cheun, SMG-SNU Boramae Medical Center, Seoul, Korea, Republic of
J. Cheun1, C. Lee2, D. Shin2, S. Park3, T. Ahn4, W. Han2, H. Kim2; 1Surgery, SMG-SNU Boramae Medical Center, Seoul, KOREA, REPUBLIC OF, 2Surgery, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF, 3Integrated science, Handong Global University, Pohang, KOREA, REPUBLIC OF, 4Life Science, Handong Global University, Pohang, KOREA, REPUBLIC OF.
Emerging evidence suggests that tumor-educated platelets (TEPs) undergo dynamic RNA splicing changes in response to cancer-derived signals, offering a minimally invasive avenue for cancer detection. In this study, we leveraged platelet RNA sequencing (RNA-seq) data to identify breast cancer-associated exon-exon splice junctions whose expression is significantly altered in cancer patients compared to healthy controls. Using a stepwise feature selection method guided by the Akaike Information Criterion (AIC), we identified 11 discriminatory splice junctions from platelet RNA profiles. These junctions were used to train a logistic regression classifier, which achieved strong predictive performance with an AUC of 0.873 on public test data and 0.949 on newly generated clinical samples, highlighting the model’s robustness and clinical applicability. Dimensionality reduction (t-SNE) and heatmap visualizations confirmed distinct separation between breast cancer and control samples based on these junction features. Gene Ontology enrichment analysis of the corresponding genes revealed significant enrichment in pathways such as endothelial cell development, establishment of endothelial barrier, and regulation of protein localization to the cell surface (FDR < 0.05), suggesting that platelet splicing alterations may reflect tumor-induced vascular and immune interactions. Our findings demonstrate that platelet-derived exon-exon junction expression profiles serve as promising biomarkers for breast cancer detection.
| Data set | Sensitivity | Specificity | Balanced Accuracy | AUC |
| Public data | 0.742 | 0.905 | 0.824 | 0.873 |
| Clinical data | 0.857 | 0.897 | 0.877 | 0.949 |
Presentation numberPS4-05-26
Classivus: a clinical platform for reclassifying uncertain variants in hereditary breast and ovarian cancer in a Brazilian cohort
Elisângela Paula Silveira-Lacerda, Federal University of Goiás, Goiânia, Brazil
B. E. Nascimento1, L. P. Lacerda2, P. F. Silva1, R. M. Goveia1, I. M. Calassa1, K. M. Veiga1, R. M. Rahal3, W. D. Oliveira1, D. C. Maia4, E. P. Silveira-Lacerda5; 1Genetics, Federal University of Goiás, Goiânia, BRAZIL, 2Medicine Faculty, UNICEPLAC – Universidade do Planalto Central, Gama, BRAZIL, 3CORA, Clinical Hospital, Federal University of Goiás, Goiânia, BRAZIL, 4oncology, Federal University of Ceará, Fortaleza, BRAZIL, 5Center for Human Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, BRAZIL.
Variants of Uncertain Significance (VUS) represent a major challenge in clinical genetics, particularly in the context of hereditary breast and ovarian cancer (HBOC). The underrepresentation of Latin American populations in global genomic databases contributes to a higher prevalence of inconclusive results, hindering genetic counseling and risk-reduction interventions. Efforts to improve variant interpretation through population-specific evidence and clinical integration are urgently needed. To reclassify VUS identified in patients suspected of HBOC by applying the structured ACMG/AMP guidelines, integrating computational prediction, population data, and clinical context; and to develop and implement a dedicated institutional platform (CLASSIVUS) to record, standardize, and track the classification of genetic variants in a Brazilian clinical genetics center. We analyzed 105 patients with a confirmed diagnosis of breast cancer who met the NCCN criteria for HBOC. Genetic testing was performed using next-generation multigene panel sequencing covering key susceptibility genes. VUS were selected based on ACMG criteria and evaluated using in silico predictors (PolyPhen-2, SIFT, MutationTaster), population frequency databases (gnomAD, ABraOM), and clinical annotation platforms (ClinVar, Franklin by Genoox, Varsome). Each variant was scored according to the ACMG criteria, and a final classification was determined. A web-based institutional database, CLASSIVUS, was developed to register variants, document the applied criteria, associate the responsible professionals, and enable filtered queries and reanalyses. Thirteen VUS were identified in 10 patients (9.5%) across seven genes (ATM, BRCA1, BRCA2, BRIP1, BARD1, CHEK2, TP53). Five variants (38.5%) were reclassified as likely benign based on ACMG scoring, while eight (61.5%) remained classified as VUS. The CLASSIVUS platform enabled centralized storage of variant records, real-time retrieval of previous classifications, and standardized documentation of scoring justification. This system reduced analytical redundancy and ensured traceability and reproducibility among professionals, offering a scalable solution for routine clinical workflows. The integration of ACMG guidelines with computational tools, clinical annotations, and local population frequency data enabled reliable reclassification of VUS in a Brazilian HBOC cohort. The CLASSIVUS platform proved to be a valuable tool for standardizing variant interpretation and supporting safe, reproducible, and transparent clinical decisions. This approach strengthens precision medicine practices in underrepresented populations and may serve as a model for other institutions seeking to enhance their genetic curation capabilities.
Presentation numberPS4-05-27
Longitudinal multi-omic profiling of tissues and blood to track the molecular evolution of metastatic breast cancer
Henry Kaplan, Swedish Cancer Institute and Paul G Allen Research Center, Seattle, WA
H. Kaplan1, A. Dowdell2, J. Wagner3, J. Welle3, S. Reynolds3, C. Carney1, D. Page4, A. Conlin4, K. McCormick5, F. Yan5, E. Johnston6, D. File5, T. Wahl7, J. Lopez6, Z. Topp4, K. Paulson1, B. Xie7, S. Montgomery6, A. Bartlett8, C. Bifulco9, B. Piening9; 1Medical Oncology, Swedish Cancer Institute and Paul G Allen Research Center, Seattle, WA, 2Molecular Genomics Lab, Earle A Chiles Research Institute and Providence Genomics, Portland, OR, 3Molecular Genomics Lab, Providence Genomics, Portland, OR, 4Medical Oncology, Earle A Chiles Research Institute and Providence Cancer Center, Portland, OR, 5Medical Oncology, Swedish Cancer Institute, Seattle, WA, 6Medical Oncology, Swedish Cancer Institute, Edmonds, WA, 7Medical Oncology, Swedish Cancer Institute, Issaquah, WA, 8Medical Oncology, Earle A Chiles Research Institute and Providence Genomics, Portland, OR, 9Medical Oncology, Earle A Chiles Research Institute and Providence Genomics, Portland, OR.
Background Despite treatment advances, long-term survival for patients with metastatic breast cancer (mBC) remains low. While many tumors exhibit an initial response to conventional or targeted therapies, this is typically followed by the acquisition of resistance. While a subset of these acquired resistance events are driven by known mechanisms (e.g. ESR1 mutation on hormone therapy), others are driven by mechanisms that remain unsolved. Moreover, the trajectory and timeline by which tumors acquire resistance remains challenging to predict. To address this, we developed a study following the molecular evolution of mBC in tissues and circulating tumor DNA (ctDNA) combining both DNA sequencing and DNA methylation analysis. Methods We developed a protocol enrolling patients with a new diagnosis of de novo or first recurrence HER2-negative mBC. For enrolled patients, primary tumor and one or more metastatic sites are profiled using comprehensive genomic profiling (CGP) (ProvSeq Solid Tumor) and DNA methylation (DNAm) (Twist Human Methylome Panel). Patient blood samples are collected using Streck tubes at enrollment and at 12-week intervals for 3 years. Blood ctDNA is extracted and profiled using CGP (ProvSeq Liquid) and DNA methylation (Twist Human Methylome Panel). Mutations and methylation signatures are assessed in primary and metastatic tissue, and over time in ctDNA. Results While recruitment and longitudinal sample collection is ongoing, we report preliminary analyses from the first 40 participants enrolled in the study. Using CGP testing across solid tumor and liquid biopsy samples, we found 735 unique reportable variants, with 165 variants annotated as clinically-significant (OncoKB database), with 16 patients harboring a pathogenic alteration in PIK3CA. Solid tumor variant detection via CGP was highly accurate down to 5% variant allele fraction and down to 0.5% variant allele fraction in liquid CGP. For patients with both primary and metastatic tissue samples (n=20) mean concordance of variants was 86%. For variants deemed clinically significant in the tissue, 36% were also found in the closest ctDNA and 40% were found in ≥ 1 ctDNA sample over the monitoring period. Thus far, two patients (both receiving hormone therapy) have demonstrated ESR1 mutations. One was detected in the first ctDNA and progressed within 3 months. The other was detected 3 months after the diagnosis of metastatic disease, but did not progress clinically until 5 months later when the VAF had increased from 3% to 13%. Both solid tissue and liquid methylation analysis yielded high coverage across genome-wide CpG sites assessed in the panel, and current efforts are focused on investigating methylation changes in non-mutational acquired therapeutic resistance. Conclusions The combination of comprehensive genomic profiling and genome-wide methylation profiling is highly feasible in both tissue and liquid biopsies and may represent a synergistic strategy for maximizing diagnostic yield. Given the incomplete portrait of genomic evolution contributing to therapeutic response in mBC, our ongoing efforts seek to provide more comprehensive maps of mutational and epigenomic changes that may yield better treatment strategies in this population.
Presentation numberPS4-05-28
Impact of Neoadjuvant Endocrine Therapy on MammaPrint Index in Hormone Receptor-Positive, HER2-Negative MammaPrint Low Risk Early-Stage Breast Cancer
Saya Jacob, University of Pennsylvania, Philadelphia, PA
S. Jacob1, C. Wu2, A. Glas3, C. Yau4, L. Brown-Swigart5, G. Hirst6, T. Haddad7, K. Giridhar7, A. Elias8, R. Mukhtar9, L. Huppert10, K. Albain11, D. Yee12, L. van’t Veer5, L. Esserman9, J. Chien13; 1Medicine, University of Pennsylvania, Philadelphia, PA, 2Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 3Biomarkers and Innovation, Quantum Leap Healthcare Collaborative, San Francisco, CA, 4Surgery, University of California San Francisco, San Francisco, CA, 5Laboratory Medicine, University of California San Francisco, San Francisco, CA, 6Surgery, Div of Surgical Oncology, University of California San Francisco, San Francisco, CA, 7Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 8Department of Medicine, Medical Oncology, University of Colorado, Aurora, CO, 9Surgery, University of California San Francisco, San Francisco, CA, 10Medicine, University of California San Francisco, San Francisco, CA, 11Medicine, Loyola University Chicago Stritch School of Medicine, Maywood, IL, 12Medicine and Pharmacology, University of Minnesota, Minneapolis, MN, 13Medicine, University of California San Francisco, San Francisco, CA.
Intro: The 70-gene MammaPrint (MP) assay is prognostic for distant recurrence and predictive of benefit from adjuvant chemotherapy (CT) benefit established on surgical tissue of systemically untreated patients (pts). The MP index range is from -1 to +1 with higher index indicating lower risk of early recurrence. Risk is categorized as MP High Risk, MP Low Risk, and MP UltraLow Risk, and results can guide treatment decisions. Pts with MP index >0 are classified as Low Risk and derive little benefit from CT. There are clinical scenarios in which the only available tissue for MP or other genomic testing has been exposed to neoadjuvant endocrine therapy (NET). Few studies have investigated the impact of ET exposure on MP index. Methods: Pts were identified through the I-SPY Low Risk Registry (LRR), an observational sub-study which enrolled pts with hormone receptor positive (HR+)/HER2-negative early breast cancer ineligible for the I-SPY2 trial due to MP Low Risk or UltraLow Risk status, between 2010-2020. Pts were treated at the discretion of their provider. We conducted a retrospective analysis of MP index from paired tumor tissue obtained from systemically untreated diagnostic core biopsies and surgical specimens after NET. MP categories and index before and after NET were compared. Change in MP index was assessed for an association with changes in clinicopathologic factors obtained through the LRR using the paired-t test. Results: 28 pts with paired tumor samples were included in the analysis. Median age was 48 (range 31-75) and 18 (64%) were premenopausal. At baseline, 13 (46%) were clinically node-positive and 22 (76%) had ductal histology. Tumor grade was 1 in 6 pts (21%), 2 in 20 pts (71%), and 3 in 2 pts (7%). ER expression was >95% in 25 pts (range 60-100%) and PR expression was >90% in 14 pts (range 0-100%). NET regimens included tamoxifen +/- ovarian function suppression (OFS) in 3 pts (10.7%), aromatase inhibitor (AI) +/- OFS in 15 pts (53.6%), tamoxifen followed by or in combination with AI +/- OFS in 8 pts (28.6%), fulvestrant in 1 pt (3.6%) and OFS alone in 1 pt (3.6%). 11 (61%) of premenopausal pts received OFS. 96% (27/28) of pts were classified as Low Risk both pre- and post-NET. Only one pt had MP change from Low Risk to High Risk. Three pts had change from Low Risk to UltraLow Risk or vice versa. As a continuous variable, a small average increase in MP index was observed after NET; but, there was a range of responses. 19 pts (68%) had an increase in MP index after NET, while 9 pts (32%) had a decrease. In those with a decrease in MP index, the average ER expression decreased by -13.6%, whereas in those with an increase in MP index, ER expression was unchanged (-0.18%) (p=0.049). There was no significant difference in changes in PR expression between tumors with increased vs decreased MP indices. At a median follow-up of 35 months, 4 distant recurrences were observed. Of the 9 pts with a decrease in MP index, 3 pts (33%), including the one who changed from Low Risk to High Risk, experienced a distant recurrence. In contrast, in those with an increase in MP index, 1 of 19 (5%) experienced a distant recurrence. Conclusion: In pts with early-stage HR+/HER2- MP Low Risk or UltraLow Risk breast cancer, NET had minimal impact on the tumor MP classification. These data suggest that the MP assay could be used for tissue exposed to NET if untreated tissue is not available. Further investigation in larger cohorts is needed to validate these findings. The observation that there were more distant recurrences in pts with tumors that had an increase in MP after NET is hypothesis generating, and potentially clinically relevant if validated in a larger study. The potential of MP as a dynamic risk marker to stratify recurrence risk following NET is being evaluated in the ongoing I-SPY2 Endocrine Optimization Pilot.
Presentation numberPS4-05-29
Homologous Recombination Deficiency in Breast Cancer: Exploratory Analysis of a Brazilian Cohort with Germline Genetic Testing.
Andreza Karine de Barros Almeida Souto, Oncoclínicas&Co, Brasília, Brazil
A. K. Souto1, F. C. Koyama2, S. S. Koide2, B. B. Souza1, J. D. Massaro2, L. J. Oliveira2, L. Yamamoto2, T. A. Santana3, M. L. Bulcão4, C. B. Fernandes2, C. A. Resende1, M. S. Mano5, L. C. Landeiro6, G. O. Bretas1, C. B. Miranda7, R. Dienstmann2, R. L. Sandoval8; 1Oncology, Oncoclínicas&Co, Brasília, BRAZIL, 2Oncology, Oncoclínicas&Co, São Paulo, BRAZIL, 3Oncology, Oncoclínicas&Co, Aracaju, BRAZIL, 4Oncology, Oncoclínicas&Co, Rio de Janeiro, BRAZIL, 5Oncology, Hospital Albert Einstein, São Paulo, BRAZIL, 6Oncology, Oncoclínicas&Co, Salvador, BRAZIL, 7Oncology, Oncoclínicas&Co, Vitória, BRAZIL, 8Oncology, Hospital Sírio Libanês, Brasília, BRAZIL.
Introduction: Breast cancer (BC) is the most prevalent malignancy among women, and 5-10% are associated with germline pathogenic/likely pathogenic variants (PVs) in high-penetrance BC genes, mostly BRCA1/2. These genes encode proteins involved in the homologous recombination repair (HRR) pathway, whose dysfunction leads to homologous recombination deficiency (HRD). Germline PVs, somatic alterations, and/or epigenetic modifications in HRR pathway genes can induce the HRD tumor phenotype. While HRD is well-characterized in ovarian cancer and is a predictive biomarker for Parp inhibitors in this setting, data in BC remains scarce, especially in the Brazilian population. Objective: To evaluate HRD status in breast cancer samples from a Brazilian cohort. Methods: This retrospective single-center study included women with BC treated between 2020-2023 whose formalin-fixed paraffin-embedded tumor samples were available for analysis at Oncoclínicas Precision Medicine laboratory (OCPM). The laboratory database was used to identify eligible samples. All tumor samples were submitted to pathology review. HRD analysis assessed genomic scars (loss of heterozygosity, telomeric allelic imbalance, and large-scale state transitions) by sequencing 13,809 single nucleotide polymorphisms, plus mutational profiling of 15 genes involved in the HRR pathway. HRD status was defined using a validated assay by OCPM. Genomic instability score cutoff of 65 or higher. HRD profiles were described in relation to clinicopathological and molecular features. Results: Among 134 identified samples, 55 patients were consecutively selected for tumor analysis according to the study budget. Eight samples were excluded due to inadequate material for molecular analysis. Among 47 cases eligible for HRD analysis, 9 samples were excluded due to post-analytical failures. HRD profile was performed in 38 samples. The mean age of BC diagnosis was 51.4 years (range 49,8-52,2 years); 52.6% were premenopausal. Regarding molecular subtype distribution: 47.4% were Luminal B-like, 31.6% triple-negative, 15.8% Luminal A-like, and 2.6% HER2-positive. Half of the samples were from primary tumor sites and the remaining from metastatic sites. Most tumors were stage III (31.6%) at diagnosis followed by stage II (26.3%), IV (18.4%), and I (15.8%). Regarding germline status, 26.3% of patients harbored germline PVs, among these, 40% in BRCA1/2, 40% in other HRR genes, and 20% in non-HRR genes. The overall HRD prevalence was 31.6% (12/38). TNBC had a higher rate of HRD (66.7%; 8/12), followed by Luminal B-like (25%) and Luminal A-like (8.3%). None of the HER2-positive tumors had HRD phenotype. Patients with HRD tumors (n=12) were predominantly premenopausal (75%), had a younger age at BC diagnosis (mean age 49.8 years), presented more locally advanced disease (41.7% stage III), and in 75% of these cases, tumor samples were from the primary site. Thirty-three percent of patients with HRD tumors harbored germline PVs in HRR genes (2BRCA1,1BLM;1RAD51C); 60% of those were truncating variants (nonsense and frameshifts). Conclusions: This is the first Brazilian study to evaluate HRD in breast cancer, revealing a 31.6% prevalence consistent with international data. HRD was more common in aggressive tumors subtypes, younger patients, and those with germline PVs in HRR genes. The identification of alterations beyond classical BRCA1/2 mutations expands the molecular spectrum of HRD.
Presentation numberPS4-05-30
Maged1 inhibition enhances sensitivity to parp inhibitors in brca-mutated breast cancer cells
Lee In Hee, Kyungpook National Chilgok University Hospital, Daegu, Korea, Republic of
L. In Hee1, L. Soo Jung1, K. Byeongju2, L. Jeeyeon2, P. Ho Yong2, M. Joon Suk2, L. Yangsoo3, Y. Jung Dug4, K. Eun Ae5, J. Seolhwa5, K. Jieun5, C. Yee Soo1; 1Oncology/Hematology, Kyungpook National Chilgok University Hospital, Daegu, KOREA, REPUBLIC OF, 2Breast Surgery, Kyungpook National Chilgok University Hospital, Daegu, KOREA, REPUBLIC OF, 3Surgery, Kyungpook National Chilgok University Hospital, Daegu, KOREA, REPUBLIC OF, 4Plastic and Reconstructive Surgery, Kyungpook National Chilgok University Hospital, Daegu, KOREA, REPUBLIC OF, 5Breast Cancer Precision Medicine Institute, Kyungpook National Chilgok University Hospital, Daegu, KOREA, REPUBLIC OF.
Background: MAGED1 (Melanoma Antigen Gene Family, Member D1) is a member of the MAGE gene family, which is notable for its involvement in various cellular processes, including apoptosis, cell cycle regulation, and differentiation. Furthermore, previous studies showed that MAGED1 is required for double-strand breaks (DSB) repair via homologous recombination repair (HRR) and downregulation of MAGED1 sensitizes cancer cells to DNA-damaging agents. In this study, we aimed to investigate the role of MAGED1 in HRR in TNBC and BRCA1/2 mutations breast cancer. Methods: In this study, we validated MAGED1 function in HRD using siRNA in TNBC and BRCA mutant breast cancer cell lines. Cell viability was assessed by CytoFLEX after treatment with drugs (cisplatin, olaparib and niraparib) at several concentrations (0–300 µM) in Hs578T, HCC-1937 (BRCA1 5382insC), and BT-474 (BRCA2 c.0391C>A) breast cancer cell lines. Cells were transfected with control siRNA (si-Cont) or two independent siRNAs (si- MAGED1 #1, si-MAGED1 #2) targeting MAGED1. Result: Silencing of MAGED1 by siRNA in Hs578T (TNBC), HCC-1937 (BRCA1 5382insC), and BT-474 (BRCA2 c.0391C>A) cells led to enhanced sensitivity to the PARP inhibitors olaparib and niraparib, as assessed by flow cytometry using CytoFLEX. The synergistic effect between MAGED1 suppression and PARP inhibitors was more pronounced in BRCA-mutant cell lines (HCC-1937 and BT-474) compared to the TNBC Hs578T cells. In contrast, MAGED1 knockdown did not enhance sensitivity to cisplatin. Combined treatment with cisplatin and MAGED1 knockdown resulted in an unexpected increase in cell viability, warranting further investigation. To further elucidate the role of MAGED1 in HRD, additional functional studies are being conducted involving RNF8, BARD1, RAD51, and ATM. Conclusions: Our study demonstrates that silencing of MAGED1 enhances sensitivity to PARP inhibitors in BRCA1/2-mutant breast cancer cell lines. These results suggest that MAGED1 is involved in HRD and could be a therapeutic target to enhance PARP inhibitor response in breast cancer.
Presentation numberPS4-04-01
A 15-Gene Predictive Signature for Pathologic Complete Response in Triple-Negative Breast Cancer: Validation Across Institutional and TCGA Cohorts
Chi-Wen Luo, Kaohsiung Medical University Hospital, Kaohsiung city, Taiwan
M. Hou1, M. Pan2, S. MOI2, F. Chen1, C. Luo1; 1Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung city, TAIWAN, 2Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung city, TAIWAN.
Background: Pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) is a strong prognostic marker in triple-negative breast cancer (TNBC). However, predictive biomarkers for pCR remain limited. This study aimed to identify gene expression profiles associated with pCR in TNBC to aid treatment stratification. Methods: Tumor samples were obtained from 16 TNBC patients treated with NAC at Kaohsiung Medical University Hospital (KMUH), including 5 pCR and 11 non-pCR cases. RNA sequencing (RNA-seq) was performed, and differentially expressed genes (DEGs) were identified using DESeq2 (|log₂FC| ≥ 2, adjusted p < 0.05). Expression profiles were compared with a validation cohort of 27 chemotherapy-responsive TNBC cases from The Cancer Genome Atlas (TCGA). Overlapping DEGs were identified via Venn diagram analysis. Drug-gene interaction databases were queried to assess therapeutic relevance. Results: A total of 175 DEGs were found in the KMUH cohort, with 146 upregulated and 29 downregulated in non-pCR tumors. Fifteen DEGs showed consistent expression patterns between KMUH non-pCR tumors and both KMUH pCR and TCGA responder cohorts. These genes were enriched in pCR samples and may serve as predictive biomarkers. Several candidates were identified as potentially druggable, suggesting relevance for therapeutic targeting. Conclusions: We identified a 15-gene signature associated with pCR in TNBC, validated across independent cohorts. These biomarkers may inform treatment decisions, improve patient stratification, and offer new directions for targeted therapy development in chemoresistant TNBC.
Presentation numberPS4-04-02
Improving HER2 Status Assessment in Breast Cancer with MammaTyper®: Focus on Equivocal Cases
ANA VELASCO, IRB LLEIDA, LLEIDA, Spain
A. VELASCO1, I. URDANIBIA1, G. RODRIGUEZ1, N. TUSET2, G. ARIADNA3, S. MORALES3; 1Medical Oncology, IRB LLEIDA, LLEIDA, SPAIN, 2Medical Oncology, HOSPITAL ARNAU DE VILANOVA DE LLEIDA, LLEIDA, SPAIN, 3Medical Oncology, HospItal arnau de vilanova de lleida, LLEIDA, SPAIN.
Background: Accurate HER2 status assessment is crucial for guiding treatment decisions in breast cancer. MammaTyper® is a CE-marked in-vitro diagnostic device utilizing real-time quantitative PCR (qRT-PCR) to quantify mRNA transcripts of ERBB2, ESR1, PGR, and MKI67. It provides an objective and standardized classification of breast cancer into intrinsic subtypes (Luminal A, Luminal B, HER2-enriched, and Triple-Negative), offering prognostic and predictive information that complements traditional immunohistochemistry (IHC). MammaTyper®’s high reproducibility and sensitivity are particularly valuable when subjective interpretation of IHC presents challenges.Objective: This study aimed to analyses whether the MammaTyper® platform improves the classification of HER2-equivocal breast cancer cases and to assess its concordance with FISH-based methods.Methods: We analysed 93 breast cancer patients. Estrogen receptor (ER) and progesterone receptor (PR) status, along with Ki67 index, were determined by IHC. HER2 status was initially assessed by IHC and FISH. MammaTyper® was then performed to classify intrinsic subtypes and determine HER2 status based on ERBB2 mRNA levels. Concordance between MammaTyper®, IHC, and FISH was evaluated.Results: Of the 93 patients, 83 (89%) were ER-positive, 42 (45%) PR-positive, and 51 (55%) had a high Ki67 index by IHC. Forty-two cases (45%) were classified as HER2-positive by IHC and FISH. MammaTyper® confirmed HER2-positivity in 25 (60%) of these cases. Among the 51 HER2-negative cases by IHC and FISH, MammaTyper® was positive in only 8 (15%). The overall concordance between MammaTyper® and the IHC/FISH combined assessment was 73%. Specifically, concordance was 60% for IHC/FISH-positive cases and 84% for IHC/FISH-negative cases. Of the 32 cases classified as HER2-positive by MammaTyper®, 23 (71%) had a HER2 FISH ratio of ≥2. Conversely, among the 55 cases classified as HER2-negative by MammaTyper®, 17 (31%) had a HER2 FISH ratio of ≥2. MammaTyper® showed a 72% concordance with IHC for HER2 phenotype determination, identifying a lower proportion of HER2-positive tumors (34%) compared to IHC (45%). This concordance was notably higher (84%) in HER2-negative cases.Conclusion: MammaTyper® demonstrates substantial concordance with IHC and FISH for HER2 status determination, particularly in HER2-negative cases. It identifies a smaller subset of HER2-positive tumors compared to IHC, suggesting its potential to refine the selection of patients for HER2-targeted therapies. Further clinical utility should be assessed by correlating MammaTyper® results with treatment response and survival outcomes.
Presentation numberPS4-04-03
Analytical concordance of NGS-based tests for ESR1 mutation detection in plasma
Ana Vivancos, Vall d’Hebron Institute of Oncology, Barcelona, Spain
A. Yarunin1, A. Martin2, M. Gomez-Rey2, A. Baizan3, M. Sesé4, S. Clavé5, L. Camacho5, C. Saura6, J. Longshore1, J. Hernández-Losa7, B. Bellosillo5, A. Vivancos2; 1Global Oncology Diagnostics, AstraZeneca UK Limited, Cambridge, UNITED KINGDOM, 2Cancer Genomics, Vall d’Hebron Institute of Oncology, Barcelona, SPAIN, 3Breast Cancer Unit, Vall d’Hebron Institute of Oncology, Barcelona, SPAIN, 4Pathology, Hospital Universitari Vall d’Hebron, Barcelona, SPAIN, 5Pathology, Hospital del Mar, Barcelona, SPAIN, 6Oncology, Hospital Universitari Vall d’Hebron (HUVH), Barcelona, SPAIN, 7Pathology, Hospital Universitari Vall d’Hebron (HUVH), Barcelona, SPAIN.
Background: Activating mutations in the ESR1 gene represent a well-known mechanism of acquired resistance to aromatase inhibitors (AIs) in patients with hormone receptor-positive (HR+) metastatic breast cancer. Liquid biopsy, through the analysis of circulating free DNA (cfDNA), provides a non-invasive and dynamic approach to detect ESR1 mutations, enabling real-time monitoring of tumor evolution. This is especially relevant in the context of novel oral selective estrogen receptor degraders (SERDs), where ESR1 mutation profiling will inform treatment selection and act as a biomarker of resistance and response. As these agents become part of routine clinical care, the ability to accurately detect and monitor ESR1 mutations in plasma is critical. As several next-generation sequencing (NGS)-based assays are commercially available, studies comparing their analytical concordance in detecting ESR1 mutations in real-life plasma samples are needed. Methods: This study aims to assess the analytical concordance between two amplicon-based NGS assays (Oncomine™ Precision Assay and Pillar Biosciences™ OncoReveal Essential LBx) and a hybrid-capture based NGS test, VHIO360 (tech transfer to VHIO of the Guardant360® assay) for the detection of ESR1 mutations in cfDNA. VHIO360, with established clinical validation is considered as the reference gold standard. One hundred plasma samples from patients with known ESR1 mutation genotypes as per VHIO360 testing (from the VHIO Molecular Prescreening Program), containing 64 pathogenic/ likely pathogenic variants in ESR1 (59 corresponding to hotspot Y537S/N/C, D538G, and E380Q/K) will be additionally tested with the aforementioned amplicon-seq based approaches. Mutant variant frequencies in the sample set are present at a wide range (0.02-69%). Concordance is measured using metrics such as overall agreement, positive and negative percent agreement, and analytical sensitivity across variant allele frequencies. Discordant results will be further investigated to identify assay-specific limitations or systematic biases, including variant dropout or differential coverage. Results: Results provide a comparative assessment of analytical performance across platforms, focusing on concordance in detection of clinically relevant ESR1 mutations and determination of assay-specific limits of detection. Particular attention will be paid to samples with low variant allele frequency (VAF) mutations, where differences in platform sensitivity may impact clinical interpretation. Conclusions: This study will provide critical evidence for the analytical performance and clinical reliability of commonly used NGS-based assays for detecting ESR1 mutations in plasma. As SERDs and other ESR1-targeted therapies become integrated into standard-of-care for HR+ metastatic breast cancer, accurate and validated mutation testing is essential in order to guide clinicians in selecting appropriate local testing approaches.
Presentation numberPS4-04-04
High RICTOR / Low RPTOR Gene‐Expression Signature as a Predictive Biomarker for Intravenous Everolimus Nanoparticle (Sapu003): Rationale for the First‐in‐Human Trial
Cynthia Lee, Sapu Bioscience, LLC, San Diego, CA
C. Lee1, T. Hoque2, S. Qazi2, S. saund3, C. Hsiao2, T. Joh2; 1R&D, Sapu Bioscience, LLC, San Diego, CA, 2Clinical, Sapu Bioscience, LLC, San Diego, CA, 3Manufacturing, Sapu Bioscience, LLC, San Diego, CA.
Background Everolimus (an mTORC1 inhibitor) packaged as Sapu003, an injectable deciparticle formulation, has shown superior cytotoxic activity compared with paclitaxel in glycolysis-addicted tumors. Leveraging retrospective pan-cancer transcriptomic datasets, we explored whether expression of core mTOR-complex scaffolds and metabolic modulators defines a subgroup most likely to benefit from Sapu003 and complementary metabolic therapy. Methods Publicly available survival cohorts (METABRIC, KM-Plotter, TCGA) were mined for overall survival (OS) and relapse-free survival (RFS) according to mRNA quartiles of mTORC1 (RPTOR, AKT1S1) and mTORC2 (RICTOR, MAPKAP1) components plus downstream metabolic genes (SKP2, LDHA, MK, STK11). Differential survival was validated across >9,000 samples spanning breast, lung, gastric, ovarian, RCC and AML. A composite biomarker was derived by recursive partitioning. Results High RICTOR expression—reflecting low baseline mTORC1 activity—was independently associated with prolonged OS (110 months vs 66 months) and RFS (48 vs 31 months) in breast cancer (P = 0.0029 and 4.7 × 10⁻⁷, respectively). Low RPTOR amplified OS gains conferred by low-AKT3, low-TSC1 or low-RHEB status in lung adenocarcinoma (median 118-139 vs 57-69 months; P < 10⁻⁵). Across seven tumour types, simultaneous low expression of ≥3 scaffold genes (RPTOR, MLST8, AKT1S1, MAPKAP1, PRR5L, RICTOR, PRR5, DEPTOR) predicted a ≥1.8-fold improvement in overall survival (OS). In glycolysis-addicted U-87 MG xenografts, Sapu003 achieved durable tumour regression with no weight loss. Conclusions A High RICTOR / Low RPTOR ± metabolic-gene-low signature enriches for tumors with intrinsic dependence on adaptive metabolism and may predict enhanced responsiveness to Sapu003. A Phase I/Ib study will prospectively enroll biomarker-positive patients (high-RICTOR / low-RPTOR breast and non-breast solid tumors) and incorporate pharmacodynamic confirmation of mTORC1 blockade and metabolic rewiring.
Presentation numberPS4-04-05
Construction and validation of a clinical model predicting pathological complete response after neoadjuvant immunotherapy in breast cancer
Min Lin, Sun Yat-sen University Cancer Center, Guangzhou, China
M. Lin1, T. Du2, J. Tang2; 1Department of ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 2Department of breast oncology, Sun Yat-sen University Cancer Center, Guangzhou, CHINA.
Background: Neoadjuvant immunotherapy combined with chemotherapy has shown promising potential in breast cancer management, but predicting pathological complete response (pCR) remains challenging. This study aimed to develop and validate a predictive model for pCR in breast cancer patients receiving neoadjuvant immunotherapy. Methods: We retrospectively analyzed breast cancer patients treated with neoadjuvant immunotherapy at Sun Yat-sen University Cancer Center (SYSUCC) between November 2019 and April 2025. pCR was defined as Miller-Payne grade 5, while grades 1-4 were classified as non-pCR. Clinical, pathological, and laboratory variables—including age, body mass index (BMI), menopausal status, TNM stage, histological type, Ki67 expression, chemotherapy regimen, immune checkpoint inhibitor type, immunotherapy cycles, treatment pattern, neutrophil-to-lymphocyte ratio (NLR), albumin, C-reactive protein, and serum amyloid A—were analyzed. Univariate logistic regression identified variables significantly associated with pCR (p < 0.05), which were further analyzed by multivariate logistic regression. Variables with p < 0.1 in the multivariate model were included in the final predictive model. The cohort was randomly divided (2:1) into a training and validation set. Model performance was assessed using receiver operating characteristic (ROC) curves, calibration plots, and decision curve analysis (DCA). Results: A total of 233 patients were included, and 52.8% achieved pCR. The final model incorporated pretreatment clinical T stage, pretreatment Ki67 expression, pretreatment NLR, chemotherapy regimens, and number of immunotherapy cycles. The model demonstrated good discrimination, with an AUC of 0.782 in the training set and 0.818 in the validation set. Calibration curves indicated good agreement between predicted and observed pCR probabilities. DCA showed the model provided a net clinical benefit over “treat all” and “treat none” strategies across a range of threshold probabilities. Conclusions: We developed and validated a practical predictive model for pCR in breast cancer patients receiving neoadjuvant immunotherapy. The model may assist clinicians in individualizing treatment strategies and optimizing patient outcomes. Prospective validation is warranted.
|
Characteristics |
Odds ratio |
95% CI |
P value |
|
Pretreatment clinical T stage |
|||
| T1 | Ref | – | – |
| T2 | 0.61 | 0.20-1.73 | 0.367 |
| T3 | 0.12 | 0.03-0.45 | 0.002 |
| T4 | 0.24 | 0.05-1.07 | 0.067 |
| Pretreatment Ki67 | 20.50 | 4.00-115.64 | <0.001 |
|
Chemotherapy regimens |
|||
| TCb-AC | Ref | – | – |
| TCb | 2.37 | 0.98-5.92 | 0.058 |
| AC-T | 0.43 | 0.18-1.05 | 0.065 |
| Others | 0.30 | 0.06-1.25 | 0.113 |
|
Immunotherapy cycle |
1.34 | 1.06-1.72 | 0.016 |
|
Concurrent initiation of immunotherapy and chemotherapy |
|||
| Yes | Ref | – | – |
| No | 1.74 | 0.65-4.92 | 0.283 |
| Pretreatment NLR | 0.80 | 0.60-0.99 | 0.055 |
Presentation numberPS4-04-06
Tumor-on-chip as a personalized platform for rapid drug testing in breast cancer
Clara Helal, Department of Medical Oncology, Institut Curie-Institut of Women’Cancers, Paris, France
C. Helal1, N. Schintu2, J. Jin2, L. Pinon2, E. Montaudon3, L. Sourd3, H. Derrien3, M. Nurmik4, M. Simon2, M. Parrini4, L. Cabel1, S. Descroix2; 1Paris, Department of Medical Oncology, Institut Curie-Institut of Women’Cancers, Paris, FRANCE, 2Paris, Institut Curie, CNRS UMR168, Laboratoire Physico Chimie Curie, Institut Pierre-Gilles de Gennes, PSL Research University, Paris, FRANCE, 3Paris, Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, Paris, FRANCE, 4Paris, Institut Curie, Inserm U1339, CNRS UMR3666 CNRS, PSL Research University, Paris, FRANCE.
Background: Metastatic breast cancer (mBC) is the leading cause of cancer mortality in women, yet treatment selection in later lines remains empirical. Patient-derived xenografts (PDX) and organoids (PDO) have been shown to predict patient’s drug response, however their clinical utility for real-time clinical decision-making is limited by lengthy turnaround times and a low tumor take for luminal breast cancers, and rapid functional assays are urgently needed. We developed a microfluidic tumor‑on‑chip (ToC) for rapid exvivo chemosensitivity profiling to demonstrate the potential of ToC using cancer cells from BC PDX and a fresh patient’s tumor sample. Methods: After tumor dissociation, tumor cells were isolated and embedded in 3D in collagen type I inside microfluidic device chips (AIMBiotech). Viability of tumor cells was assessed using a live/dead assay after several days of drug exposure (carboplatin, paclitaxel, 5‑fluorouracil, and trastuzumab‑deruxtecan if applicable) and compared to tumor growth measured invivo in the corresponding PDX model using a caliper. A panel of 6 PDX models was selected to reflect distinct BC subtypes and chemosensitivity profiles: 4 TNBC, 1 HER2 3+ and 1 ER+. To mimic clinical biopsies, we engineered a miniaturized chip by casting PDMS in 3D-printed silanized molds optimized for low cell input, and evaluated its performance using simulated core needle biopsies (18G) on PDX tumors and a patient sample. Results: We first investigated the response of ToC derived from fresh PDX samples. Differential drug responses were observed after 4 days of on-chip culture, suggesting that this duration is required to discriminate between sensitive and resistant models. The #152 PDX model (TNBC) showed a strong invivo response to carboplatin and paclitaxel, mirrored by a significant drop in cell viability on the ToC at D4. In contrast, neither mouse nor ToC responded to 5-FU. We performed live imaging to validate our endpoint measurement method, and comparable patterns of drug sensitivity were obtained. Overall, when a drug was identified as effective in the PDX model, we observed ToC sensitivity at D4 in 78 % of cases (in 7 cases out of 9, sensitivity was correctly detected on ToC in 6 PDX models using 4 drugs). Conversely, when the drug was resistant in PDX, we confirmed resistance in 100% of cases (11/11 using 3 drugs). Altogether, these findings highlight the correlation between the two models and demonstrate the ability of ToC platform to deliver rapid functional readouts within a clinically actionable timeframe of 4 days. Next, we successfully generated a functional ToC model using a fresh human primary tumor of TNBC; exposure to paclitaxel suggested tumor sensitivity. We finally developed an innovative microfluidic device compatible with patient biopsy samples. From three 18G core needle biopsies per tumor, we consistently isolated 100,000 to 200,000 viable tumor cells, allowing the generation of up to 12 individualized ToC units and demonstrating the feasibility of establishing ToC from limited biopsy material. Conclusions: Microfluidic ToC replicates PDX drug responses within 4 days using minimal tissue, a timeframe compatible with clinical decision-making. Optimization for cell use and ease of automation supports future clinical-scale implementation and high-throughput workflows, addressing a key gap in functional precision oncology. Prospective studies are warranted to validate ToC-guided therapy in BC.
Presentation numberPS4-04-07
Improved Diagnostic Accuracy for Breast Implant Rupture Through Strain Elastography – Results of a User-Based Evaluation Using Standardized Image Sets
Hanna Fritsch, St. Franziskus Hospital Münster, Münster, Germany
H. Fritsch1, L. Borchert1, L. Paul2, K. Hide-Moser2, C. Vogel-Minea3, C. Eichler1; 1Breast Cancer Centre, St. Franziskus Hospital Münster, Münster, GERMANY, 2Breast Cancer Centre, University Hospital Cologne, Cologne, GERMANY, 3Breast Cancer Centre, Rottal Inn Kliniken, Eggenfelden, GERMANY.
Improved Diagnostic Accuracy for Breast Implant Rupture Through Strain Elastography – Results of a User-Based Evaluation Using Standardized Image Sets Background: Ultrasound is a widely used tool for evaluating breast implant integrity, but its diagnostic accuracy remains limited by operator dependency. This study investigated whether the addition of strain elastography can improve diagnostic sensitivity and specificity in detecting implant rupture. Methods: Based on a standardized animal model, 15 silicone breast implants (intact and ruptured) were scanned using both B-mode ultrasound and strain elastography. From these scans, 30 paired images were selected for an online evaluation. In total, 99 participants reviewed the image sets and were asked to determine the presence or absence of implant rupture — first using B-mode images alone, and then with the corresponding elastography images. Results: With B-mode alone, participants achieved a diagnostic sensitivity of 81.4% and specificity of 71.0%. When strain elastography was added, sensitivity rose to 90.9%, and specificity increased to 87.6%.Conclusion: The integration of strain elastography significantly enhances diagnostic performance in assessing breast implant rupture. In particular, it reduces false-positive rates while increasing detection of true ruptures. Using standardized image data from an animal model enables reproducible evaluation and may serve as a basis for future training and quality assurance in breast imaging.
Presentation numberPS4-04-08
Radioimmunotherapy using <sup>64/67</sup>Copper labelled trastuzumab in mice bearing HER2-positve xenografts as a model for breast cancer therapy
Erica Sztangret, Clarity Pharmaceuticals, Eveleigh, Australia
S. E. Rudd1, J. Van Zuylekom2, B. Blyth2, M. Harris3, E. Sztangret4, P. S. Donnelly1; 1School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, AUSTRALIA, 2Models of Cancer Translational Research Centre, Peter MacCallum Cancer Centre, Melbourne, AUSTRALIA, 3R&D, Clarity Pharmaceuticals, Eveleigh, AUSTRALIA, 4Medical Affairs, Clarity Pharmaceuticals, Eveleigh, AUSTRALIA.
Background and Purpose: Human epidermal growth factor receptor 2 (HER2) overexpression occurs in several cancers including breast cancer. HER2 targeted therapies such as trastuzumab have significantly improved outcomes for HER2-positive breast cancer patients, however resistance and recurrence remain challenging. Radioimmunotherapy (RIT) utilizing trastuzumab could provide an additional treatment option for HER2+ breast cancer patients. We produced the theranostic pair 64/67Cu-SAR-trastuzumab and assessed the PET imaging and therapeutic potential of these agents in a HER2-posive mouse model. Methods: A bifunctional variant of the macrobicyclic cage amine sarcophagine (SAR) was conjugated to the monoclonal antibody trastuzumab. Proof of concept studies were performed using SKOV-3 tumour bearing BALB/c nu/nu mice, intravenously injected with a single dose of 64Cu-SAR-trastuzumab or 67Cu-SAR-trastuzumab to compare biodistribution of the theranostic pair. Tumor uptake was assessed using PET/CT imaging and/or ex vivo biodistribution studies. In therapy studies, a single dose of saline, cold SAR-trastuzumab, or 67Cu-SAR-trastuzumab was intravenously administered and endpoints assessed. Results: PET/CT imaging 48 hours after administration with 64Cu-SAR-trastuzumab demonstrated excellent tumor uptake, with mean SUVmax = 21.0 ± 2.5. 64Cu-SAR-trastuzumab and 67Cu-SAR-trastuzumab showed comparable high tumour-specific uptake in biodistribution studies, with tumour uptake at 48 hours of 61 ± 6 % IA/g for 64Cu-SAR-trastuzumab and 59 ± 7 % IA/g for 67Cu-SAR-trastuzumab. Uptake of ≤10 % IA/g was observed for both agents in all other organs measured. Mice treated with 67Cu-SAR-trastuzumab tolerated the treatment well with limited side effects or AEs. A transient reduction in animal body weight (≤7 % of baseline) was observed in all cohorts but this did not differ significantly from the vehicle control. Treated mice also showed significant tumor growth inhibition and extended survival in a dose dependent manner compared with the control groups. The median survival was 32 days in the control groups and 102 days in the 9 MBq group. Conclusion: These results demonstrate the potential of 64/67Cu-SAR-trastuzumab as a new theranostic approach for the treatment of HER2-positive cancer. This theranostic pair exhibit similar biodistribution in the same model, with the high tumor uptake of 64Cu-SAR-trastuzumab resulting in high quality PET images. 67Cu-SAR-trastuzumab effectively increased the survival of all treated groups. The outstanding therapeutic outcomes demonstrate the potential of 67Cu-SAR-trastuzumab as an RIT candidate for translation to clinical trials.
Presentation numberPS4-04-09
Activity of capivasertib in a panel of ER+ breast XPDX models harboring PI3-K/Akt pathway and other known mutations
Christopher Nelson, The START Center for Cancer Research, San Antonio, TX
C. Nelson, J. Flores, M. Lopez, A. Simonson, A. Nunez, S. Johnson, A. Rojas, A. Stackpole, J. Lund, P. Forofontov, L. Wilson, J. Garcia, I. Sturgill, A. Cunningham, M. Wick; XenoSTART, The START Center for Cancer Research, San Antonio, TX.
Background: Therapies targeting the PI3-K/Akt pathway are an active field of research. Capivasertib in combination with fulvestrant was recently approved in endocrine therapy-resistant, advanced or metastatic ER+/HER2- breast cancer patients harboring hotspot mutations in AKT, PIK3CA or PTEN genes. However, sensitivity to capivasertib therapy in ER+ breast cancers without these specific mutations is unclear. To this end, we established a panel of ER+ breast XPDX models with or without PI3-K/Akt pathway mutations and evaluated each in vivo with capivasertib alone or in combination with fulvestrant. Methods: Fifty-six previously developed XPDX models representing ER+ breast cancer were evaluated in this study. Models were grown subcutaneously in female athymic nude mice supplemented with estradiol in drinking water when necessary and ER expression confirmed at multiple passes; mutations in and out of the PI3-K/Akt pathway were determined by WES and RNAseq. For in vivo studies, capivasertib was administered twice daily by oral gavage at 100 mg/kg/dose on a four-days on/ three-days off cycle through study completion. Endpoints included tumor volume and time from treatment initiation with %T/C values and tumor regression reported at study completion; a T/C of ≤ 20% versus control was considered sensitive. Tumor regression (%T/C<0%) versus Day 0 tumor volume was also reported. Results: Identified PI3-K/Akt pathway mutations included AKT1E17K (N=3), PIK3CAE542X/E545X/H1047X (N=17) and other PIK3CA variants (N=8); alterations in PTEN were also reported in some models. In this study, capivasertib was found active in 12/56 models, including partial or complete tumor regressions in 3/12 and tumor stasis in 2/12 models. 1/12 models with AKT1E17K, 5/17 models with PIK3CAE542X/E545X/H1047X and 3/8 models with other PIK3CA variants were found sensitive to single agent capivasertib. Interestingly, the most sensitive model tested harbored only a PTEN deletion in the PI3-K/Akt pathway and 2/3 of the most sensitive models were established from HER2+ patients. In addition, co-treatment with fulvestrant antagonized efficacy in several of the sensitive models. Conclusion: We have benchmarked a panel of ER+ breast XPDX models with capivasertib alone and in combination with fulvestrant and compared activity of models based on mutations in and out of the PI3-K/Akt pathway. This data is a valuable tool in developing PI3-K/Akt pathway therapies in ER+ breast cancer.
Presentation numberPS4-04-10
Preclinical Evaluation of <sup>64</sup>Cu-Labeled Cetuximab Immuno-PET for Detecting Sentinel Lymph Node Metastasis and Assessing Therapeutic Potential in EGFR-Positive Breast Cancer
Takeshi Usui, The University of Osaka, Suita, Japan
T. Usui1, T. Miyake1, T. Watabe2, K. Hiroki3, K. Abe1, T. Ryu1, S. Yasufumi1, N. Masunaga1, C. Mishima1, M. Tsukabe1, Y. Sota1, T. Yoshinami1, T. Tanei1, K. Shimazu1; 1Breast and endocrine surgery, The University of Osaka, Suita, JAPAN, 2Nuclear Medicine and Tracer Kinetics, The University of Osaka, Suita, JAPAN, 3Advanced Radioisotope Medicine, Institute for Radiation Sciences, The University of Osaka, Suita, JAPAN.
Background: Accurate detection of sentinel lymph node (SLN) metastasis remains a diagnostic challenge in breast cancer. This study aimed to evaluate the feasibility of immuno-positron emission tomography (PET) using 64Cu-labeled cetuximab for detecting SLN metastasis and to assess its preliminary therapeutic effect in a preclinical model of epidermal growth factor receptor (EGFR)-positive breast cancer. Methods: The SLN metastasis model was established using EGFR-overexpressing MDA-MB-468 breast cancer cell line. [64Cu]Cu-PCTA-cetuximab was administered either intravenously (IV; 5.8 ± 0.9 MBq; n=12) or intradermally / subdermally (ID/SD; 4.3 ± 0.4 MBq; n=11) into the parapapillary region of the tumor-containing mammary gland. PET/computed tomography scans were performed 24 h after tracer injection, with delayed imaging at 48 h to evaluate retention and contrast. For comparison, 18F-FDG PET was also performed via IV (9.1 ± 1.4 MBq, n = 4) or ID/SD (5.4 ± 2.2 MBq, n = 3) routes in the same cohort. SLNs were identified using blue dye, and were pathologicaly evaluated for metastases. To evaluate therapeutic efficacy, additional mice received IV [64Cu]Cu-PCTA-cetuximab at 9, 18, or 37 MBq (n = 3 per group), and tumor diameters and body weight were monitored over time in comparison to untreated controls. Results: After intravenous administration of [64Cu]Cu-PCTA-cetuximab (n=12), radiotracer accumulation was detected in the primary tumor in all mice and in the axilla of eight mice (67%, SUVmax 1.24 ± 0.51), all of which were found to have SLNs with histologically confirmed metastasis. The sensitivity, specificity, accuracy, and negative and positive predictive values for PET imaging in this group were 89%, 100%, 92%, 75%, and 100%, respectively. In the ID/SD group , all 11 mice showed high tracer accumulation in both the primary tumor and axillary nodes (SUVmax 4.28 ± 1.19); however, only six mice (55%, SUVmax 5.01 ± 1.12) had histologically confirmed SLN metastasis. The sensitivity, specificity, accuracy, and positive predictive values for this route were 100%, 0%, 55% and 55%, respectively. SLN metastasis was not detectable by 18F-FDG PET regardless of the administration route. Regarding therapeutic evaluation, all IV-treated groups showed tumor growth suppression compared to controls. The 37 MBq group exhibited the most pronounced tumor reduction but also showed a marked decrease in body weight, indicating potential systemic toxicity at higher doses. Conclusions: Immuno-PET with IV-administered [64Cu]Cu-PCTA-cetuximab demonstrated high precision for diagnosis of SLN metastasis and showed promising therapeutic efficacy in an EGFR-positive breast cancer model. While ID/SD administration showed lower diagnostic specificity, its strong accumulation in SLNs indicates potential utility for localized therapy. These findings support further exploration of 64Cu-labeled cetuximab for theranostic applications in breast cancer.
Presentation numberPS4-04-11
Statins enhance trastuzumab deruxtecan efficacy: Preclinical synergy in HER2-negative models and clinical benefit in HER2-positive metastatic breast cancer
Taha Koray Sahin, Hacettepe University, Ankara, Turkey
T. Sahin1, C. Orhan2, I. Ozercan3, H. Muglu4, B. Er5, A. Akyildiz1, S. Tunbekici6, A. Oruc7, M. Aykan8, B. Koylu9, O. Akdogan10, I. Deliktas Onur11, O. Ates11, O. Yazici10, F. Selcukbiricik9, N. Karadurmus8, M. Araz7, D. Erdem12, E. Goker6, A. Bilici4, S. Aksoy1, D. Guven1; 1Department of Medical Oncology, Hacettepe University, Ankara, TURKEY, 2Department of Animal Nutrition, Fırat University Faculty of Veterinary Medicine, Elazig, TURKEY, 3Department of Medical Pathology, Fırat University Faculty of Medicine, Elazig, TURKEY, 4Department of Medical Oncology, Istanbul Medipol University, Istanbul, TURKEY, 5Department of Biology, Faculty of Science, Firat University, Elazig, TURKEY, 6Department of Medical Oncology, Ege University, Izmir, TURKEY, 7Department of Medical Oncology, Necmettin Erbakan University, Konya, TURKEY, 8Department of Medical Oncology, Health Science University, Gulhane School of Medicine, Ankara, TURKEY, 9Department of Medical Oncology, Koç University School of Medicine, Istanbul, TURKEY, 10Department of Medical Oncology, Gazi University, Ankara, TURKEY, 11Department of Medical Oncology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research, Ankara, TURKEY, 12Department of Medical Oncology, Samsun Medical Park Hospital, Samsun, TURKEY.
Background: Trastuzumab deruxtecan (T-DXd) has demonstrated significant clinical efficacy in metastatic breast cancer (mBC) with varying HER2 expression, including HER2-low and HER2-ultralow. Statins have been shown to increase HER2 membrane localization and stability, potentially enhancing responsiveness to HER2-targeted therapies. Given this mechanistic rationale, we investigated the potential of statins to enhance T-DXd activity through a translational approach combining a preclinical HER2-negative breast cancer model and a real-world clinical cohort of patients with HER2-positive mBC treated with T-DXd. Methods: In the preclinical arm, 60 female Wistar albino rats were induced with hormone receptor-positive, HER2-negative mammary tumors using N-methyl-N-nitrosourea (MNU). Animals were randomized into five groups: Control, MNU-only, MNU+T-DXd, MNU+statin, and MNU+T-DXd+statin. Tumor volume, number, and survival were monitored. HER2 expression, apoptotic markers (Bax, Bcl-2, Caspase-3/9), and proliferative signaling pathways (p-AKT, p-ERK) were assessed via immunohistochemistry (IHC) and Western blotting. In the clinical arm, 109 patients with HER2-positive mBC who received T-DXd were retrospectively analyzed. Patients were stratified based on concomitant statin use. Progression-free survival (PFS) and overall survival (OS) were evaluated using Kaplan-Meier analysis and log-rank testing. Results: The combination of T-DXd and statin significantly reduced tumor volume compared to either monotherapy (p < 0.0001), with a strong trend toward reduced tumor count (p = 0.051). HER2 protein levels were elevated in the statin and combination groups, as demonstrated by both IHC and Western blot analyses. The combination group exhibited significantly higher levels of pro-apoptotic markers (↑Bax, ↑Caspase-3), decreased anti-apoptotic Bcl-2, and stronger suppression of p-AKT and p-ERK (p < 0.0001 for all vs. T-DXd). In a real-world cohort of 109 heavily pretreated patients with HER2-positive mBC receiving T-DXd, concomitant statin use was associated with significantly improved mPFS (21.83 months vs. 14.02 months; p = 0.049) and median OS (not reached vs. 19.68 months; p = 0.023) compared to non-users. Conclusions: Our findings revealed that statin use significantly enhanced the efficacy of T-DXd in both preclinical and clinical settings. These results support the potential role of statins as cost-effective and widely available agents to potentiate HER2-targeted therapies. Prospective clinical trials are warranted to validate these observations.
Presentation numberPS4-04-12
Adaptive selection of p53 mutation metaplastic phenotypes in estrogen-independent progression of ER+ tumors: A mechanism for acquired resistance to hormonal therapy.
Alexander D Borowsky, UC DAVIS, SACRAMENTO, CA
H. Mori1, H. Moon2, Q. Chen1, A. Patino3, K. Ramamurthy3, J. Choi4, J. D. McPherson5, R. Cardiff1, J. Snyder3, A. D. Borowsky1; 1PATHOLOGY, UC DAVIS, SACRAMENTO, CA, 2Research, Eli Lilly, South San Francisco, CA, 3Cell and Molecular Biology, Duke University, Durham, NC, 4Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, KOREA, REPUBLIC OF, 5Biochemistyr, UC DAVIS, SACRAMENTO, CA.
Introduction: Estrogen receptor positive (ER+) subtypes of mammary adenocarcinoma comprise 79% of all breast cancer diagnosis and 67% of all breast cancer mortality. The paucity of models of ER+ mammary cancer that mimic human disease and response to treatment has limited critical preclinical study of mechanisms and new therapies for ER+ breast cancer. The Stat1 knockout, 129S6/SvEvTac-Stat1tm1Rds (Stat1-/-), females develop luminal type FoxA1+, ER+, and PR+ mammary carcinomas after prolonged latencies. Initial studies showed that a cell line derived from a Stat1-/- mammary carcinoma was tumorigenic in syngeneic mice, but non-tumorigenic in ovariectomized (Ovx) mice. Methods: Syngeneic tumor transplants, either from the SSM2 cell line established as a primary culture from a single mammary tumor in a Stat1-/- female, or from direct biopsy transplants of two different mammary tumors in Stat1-/- female mice, were allowed to establish growth under intact ovary syngeneic (129SvEv) female mice. After 3 weeks, these tumor bearing mice were Ovx’ed and growth was monitored, and tumor tissues harvested for analysis.Results: Ovx performed after SSM2 tumors establish growth results in ovarian hormone independent growth. The viable post-Ovx tumors were primarily composed of metaplastic CK14+ basal type cells with a high percentage p53 immunohistochemistry (IHC) positive “mutation pattern”, rather than the original luminal type tumors with low percent “wild type” pattern p53. Comparing whole exome sequences of ER+ Stat1-/- mammary tumors before and after Ovx, revealed basal keratins, mesenchymal (EMT) phenotypes, and unique mutation profiles in genes, including Trp53 and Prlr, in the estrogen-independent tumors (Prlr mutations were seen in both estrogen dependant and independant tumors). Our experimental findings are consistent with the clinical evidence of tumor heterogeneity of ER+ breast cancers in patients in recent whole genome sequencing studies. Similarly, spontaneous Stat1-/- tumors with high percentage p53 “mutation pattern” were more basaloid and grew rapidly after Ovx, while retaining high expression of ER and FoxA1. Conclusions: This study demonstrates that the STAT1-/-, ER+ estrogen dependent breast cancers can become resistant to through clonal selection of mammary cells comprised of metaplastic p53+/CK14+ basaloid cells. This implies that p53 mutation in human breast cancers may be a gain-of-function with respect to driving a more basal and/or metaplastic phenotype contributing to this highest mortality subtype of breast cancers (high grade/ Luminal B-type ER+ carcinomas).
Presentation numberPS4-04-13
Murine models for triple-negative breast cancer with differential responsiveness to immunotherapy
Thomas John Kalantzakos, UT Health San Antonio, San Antonio, TX
T. J. Kalantzakos1, Y. Zhou1, X. Liu1, J. Proehl1, C. Durfee1, I. Tamayo1, B. Troness1, A. Soni1, H. B. Gupta1, R. S. Harris2; 1Biochemistry & Structural Biology, UT Health San Antonio, San Antonio, TX, 2Biochemistry & Structural Biology, Howard Hughes Medical Institute, UT Health San Antonio, San Antonio, TX.
Breast cancer is the most common cancer diagnosis in women. Clinical studies with triple-negative breast cancer (TNBC) are encouraging for immunotherapy (αPD-1) combined with chemotherapy (paclitaxel and/or carboplatin). However, additional clinical advances could be pursued more rapidly with preclinical TNBC models including syngeneic mammary tumor cell lines. Here, we generate two mammary tumor cell lines and show differential responsiveness to immunotherapy in vivo. Spontaneous mammary tumors from C57BL/6J MMTV-Cre Tp53fl/+ animals were passaged serially in cell culture and in vivo in the mammary fat pad of fully wildtype animals. The resulting lines, MM001i and MM008i, lost Trp53 and formed 1000 mm3 tumors in the mammary fat pad within 21-28 days. Despite originating from the same genetic background, MM001i and MM008i exhibit differential responses to immunotherapy. For αPD-1 immunotherapy, MM001i is poorly responsive and MM008i is strongly responsive with near-complete tumor regression. In comparison, both MM001i and MM008i respond rapidly to αCTLA-4 therapy. Both models express unique tumor antigens as evidenced by immunity to subsequent engraftment. Primary MM008i tumors exhibit greater T cell infiltration, and CD8-positive T lymphocytes are required for αPD-1 responses. These TNBC models are promising for further mechanistic studies and testing future single and combinatorial therapies.
Presentation numberPS4-04-14
Evaluation of TNBC-PDX Models for Personalized Medicine
Shotaro Inoue, Kobe University Graduate School of Medicine, kobe, Japan
S. Inoue1, M. Miki1, M. Yamamoto2, H. Tanino3, T. Kunihisa1, S. Inubushi1; 1Division of Breast Surgery, Kobe University Graduate School of Medicine, kobe, JAPAN, 2Department of Breast and Endocrine Surgery, Kobe University Hospital, kobe, JAPAN, 3Division of Breast Surgery, Naga Municipal Hospital, Wakayama, Japan, Kinokawa, JAPAN.
Background: Triple-negative breast cancer (TNBC) is known to be highly heterogeneous compared to other subtypes. Establishing reliable preclinical models is essential for personalized treatment strategies, such as selecting effective drugs for each patient. Patient-derived xenograft (PDX) models, created by transplanting patient tumors into immunodeficient nude mice, are considered to more accurately replicate tumor diversity and drug responses. This study aimed to evaluate the retention of clinically relevant biomarkers in TNBC-PDX models.Methods: With approval from the Kobe University Institutional Review Board (Approval number: B200006), tumor tissues were collected from TNBC patients who met the inclusion criteria and provided informed consent. The tissues were implanted into nude mice to establish TNBC-PDX models. Expression of key biomarkers—EGFR, AR, CK5/6, HER2, and Trop-2—was assessed in both the original patient tumors and corresponding PDX tumors using immunohistochemistry.Results: EGFR and Trop-2 expression was retained in all models. However, AR expression was not preserved. CK5/6 and HER2 showed variable expression across cases.Conclusion: TNBC-PDX models are promising tools for drug evaluation in personalized medicine. However, since some biomarkers, such as AR and CK5/6, may not be consistently retained, prior validation of biomarker expression is essential for accurate therapeutic evaluation.
Presentation numberPS4-04-15
Mechanomedicine targeting of MT1-MMP in TNBC spheroids
Jaemoon Yang, Yonsei University, Seoul, Korea, Republic of
J. Yang, M. Ku; Department of Radiology, Yonsei University, Seoul, KOREA, REPUBLIC OF.
With breast cancer incidence rising worldwide—including in younger Korean women in their 40s—there is an urgent need for advanced disease models that capture mechanobiological features unique to this population. Post-surgical hormonal alterations and the high prevalence of bone metastases further complicate outcomes, underscoring the need for strategies addressing both primary tumor biomechanics and skeletal microenvironments. To meet this need, we developed a three-dimensional (3D) breast cancer spheroid model using biomimetic extracellular matrix materials to replicate native tissue mechanics and architecture, supported by collagen-producing stem cells. High-resolution confocal microscopy, immunofluorescent labeling of F-actin, β-Tubulin, and Vimentin, and computational analysis enabled detailed visualization of cytoskeletal organization and intracellular changes. Mechanobiology-driven evaluation identified membrane-type 1 matrix metalloproteinase (MT1-MMP) as a key regulator of invasive behavior in TNBC spheroids, mediating actin-based protrusions such as filopodia and lamellipodia. Functional assays showed that targeted MT1-MMP inhibition altered cellular mechanics and significantly enhanced chemotherapy efficacy in the 3D model.To validate this platform, dose-dependent accumulation and spatial distribution of doxorubicin were analyzed using confocal Z-stack imaging. Quantitative radial intensity profiling showed approximately 20% decrease in fluorescence signal from the spheroid edge to core at higher concentrations, indicating measurable penetration barriers. Channel-to-channel distance analyses confirmed heterogeneous but quantifiable drug distribution patterns. We also assessed drug distribution and viability using Calcein AM and fluorescence imaging. MT1-MMP inhibitor alone maintained high viability (~80% live-cell area) with spheroid diameters of ~180-200 µm and minimal drug uptake. Chemotherapy monotherapy reduced viability to ~45% with high drug uptake and decreased spheroid integrity (~160-180 µm). Simultaneous combination improved uptake but reduced viability to ~40%, with similar spheroid size. Notably, sequential MT1-MMP inhibitor pre-treatment preserved higher viability (~15%) while maintaining comparable drug uptake and spheroid size (~180 µm), indicating improved penetration with moderated cytotoxicity.To further characterize the model, XF Seahorse extracellular flux analysis was used to evaluate metabolic responses under different treatment conditions. By quantifying glycolytic activity and oxidative phosphorylation (OXPHOS), we identified shifts in ATP production pathways across monotherapy, combination, and sequential regimens. Sequential MT1-MMP inhibitor pre-treatment induced a more balanced metabolic profile, confirming that MT1-MMP inhibition can modulate and enhance control over OXPHOS activity. Protein marker analysis further supported the mechanism, showing that MT1-MMP-centered mechano-regulation can modulate chemosensitivity in this 3D spheroid system.Therefore, this advanced 3D spheroid system represents an essential platform for Mechanomedicine research and evaluation, closely replicating clinical tumor architecture and mechanical microenvironments. It enables precise study of mechanobiological drivers of metastasis and drug response, providing a foundation for identifying and validating Mechanomedicine targets like MT1-MMP and developing Mechanomedicine candidate therapeutics to improve precision oncology outcomes in breast cancer.
Presentation numberPS4-04-16
Discovering potential drug targets for estrogen receptor-positive breast cancer through Mendelian randomization
Chen Wei, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
C. Wei, L. Mengbo, Z. Hui; Department of Breast Surgery, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, CHINA.
Background: Estrogen receptor-positive (ER+) breast cancer (BC) is the most common BC subtype, which is prevalent in women and seriously threatens their health. However, the present chemotherapy for ER+ BC exists many challenges, suggesting that there is an urgent need for novel drug targets to ER+ BC.Methods: The protein quantitative trait loci (pQTLs) data of plasma proteins and genome-wide association study (GWAS) summary data of ER+ BC were searched from the Integrative Epidemiology Unit (IEU) OpenGWAS database, which were included in Mendelian randomization (MR) analysis. Five methods were employed to perform the causal inference between plasma proteins and ER+ BC, including MR-Egger, Weighted median, Simple mode, Weighted mode, and Inverse variance weighted (IVW). Moreover, the sensitivity analysis, steiger test and co-localization were carried out to further reinforce the causality between plasma proteins and ER+ BC. Finally, the phenotype scanning was implemented to search for potential diseases associated with druggable genes, and the molecular docking was also proceeded to ascertain the potential drugs for druggable genes.Results: Based on IVW, a total of 31 plasma proteins were significantly causally effected on ER+ BC, such as VEGFR-3, IL3RA, ISLR2, VEGFR-2 and MICB. The reliability of their causal relationships was validated using sensitivity analysis and steiger test. Specifically, VEGFR-3 (rs635634), IL3RA (rs115478735), ISLR2 (rs115478735), VEGFR-2 (rs635634) and MICB (rs34158769) shared the same variant with ER+ BC (rs532436) with the help of co-localization analysis (coloc.abf-PPH4>95%). In addition, only three symbol genes (FLT4, KDR and IL3RA) corresponding to above exposure factors existed potential drugs, such as both of FLT4 and KDR binding to Axitinib. With respect to phenotype scanning, we discovered that VEGFR-3, IL3RA and VEGFR-2 as protective factors were causally related to nausea and vomiting, BC and diabetes mellitus, respectively. Whhereas IL3RA was a risk factor for lung cancer, as well as VEGFR-2 for colorectal cancer.Conclusion: Our study supported that plasma proteins were causally connected with ER+ BC risk, especially VEGFR-3, IL3RA, ISLR2, VEGFR-2 and MICB, which might be promising drug targets for ER+ BC.
Presentation numberPS4-04-18
Direct targeting of amplified HER2 gene activates immune signaling through DNA damage response
Adam Krysztofiak, Yale School of Medicine, New Haven, CT
A. Krysztofiak, A. Brown, A. Minnah, F. Rogers; Therapeutic Radiology, Yale School of Medicine, New Haven, CT.
Human epidermal growth factor receptor 2 (HER2), which gene is often amplified in breast cancer (BC), promotes metastatic phenotype and confers aggressive tumor behavior. Progress made in recent years by therapeutic strategies using monoclonal antibodies has increased survival rates of HER2-positive BC patients. However, current clinical trials in BC combining immunotherapy (inhibition of PD-1/PD-L1) with anti-HER2 antibodies, resulted only in a relatively modest overall response rate. This underlines that rise of resistance, recurrence, and metastatic processes remain the main hindrance for successful therapy and complete cancer remissions. Thus, there is an urgent need to bridge a therapeutic gap for the development of targeted therapies, especially for HER2-positive metastatic BC. Direct targeting of amplified cancer genes to invoke apoptosis through the activation of DNA damage response offers an alternative mechanism to induce tumor-specific cell death. The technology recently developed in our laboratory utilizes triplex-forming oligonucleotides (TFOs) to form triplex structures at amplified oncogenic loci. Excessive distortion to the helical DNA structure caused by the TFOs leads to immoderate DNA damage and apoptosis, but only in response to the formation of multiple triplex structures. As a result, normal cells having only two copies of the gene can efficiently handle a low level of triplex-induced DNA damage by employing nucleotide excision repair (NER). This phenomenon provides an opportunity to specifically target and induce apoptosis in BC cells with amplifications of the HER2 gene. Intriguingly, recent studies provide strong evidence that genomic DNA damage leads to activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway and facilitates antitumor immune responses. HER2 also directly participates in a disruption of STING signaling, suppressing antitumor immunity and provides a survival advantage, conferring resistance to trastuzumab. Therefore, the crosstalk between stimulation of innate immune signaling and DNA damage response opens an interesting perspective of combination therapy strategies focusing on both DNA-damaging and immune system-activating therapies in BC. In this work, we discovered that TFO-induced DNA damage leads to activation of innate immune signaling in BC cell line models. First, we characterized HER2 amplification and expression of cGAS-STING pathway in numerous BC cell lines and assessed levels of triplex-induced DNA damage and apoptosis. Next, we evaluated activation of the cGAS-STING pathway in BC cell lines by analyzing STING downstream targets following TFO treatment. We discovered that TFO-induced DNA damage response in BC cells had limited effect on canonical STING signaling as measured by stimulation of TBK1-IRF3 downstream targets. However, TFO treatment induced phosphorylation of NF-κB p65 transcription factor. We validated STING-dependency in the stimulation of innate immune system following TFO treatment and confirmed that this activation can also be achieved with TFOs targeting various genomic regions including exons and introns within the HER2 gene. To reveal mechanistic landscape of the TFO-induced activation of innate immune signaling we performed analysis of gene expression using bulk RNA sequencing. In summary, we have validated that HER2-targeted TFOs can induce activation of the innate immune system signaling in BC cell lines. Our work contributes to understanding of the crosstalk between DNA repair, apoptosis, and innate immune signaling. This will aid in the design of novel combinations of therapeutic approaches to increase responsiveness of BCs to immune-targeted treatments through activation of DNA damage response, ultimately overcoming therapy resistance mechanisms.
Presentation numberPS4-04-19
A nucleic acid sensing system for selective therapeutic protein expression in breast cancer
Alinés Lebrón-Torres, Broad Institute of MIT & Harvard, Cambridge, MA
N. Priedigkeit1, J. Koob2, A. Lebrón-Torres3, J. Liao3, Y. Wang3, M. E. Hughes4, D. L. Abravanel5, S. M. Tolaney5, N. U. Lin5, S. Oesterreich6, A. V. Lee7, F. Chen8, T. R. Golub3; 1Medical Oncology, Dana-Farber Cancer Institute / Broad Institute, Boston, MA, 2Molecular and Cellular Biology, Broad Institute of MIT & Harvard, Cambridge, MA, 3Cancer Program, Broad Institute of MIT & Harvard, Cambridge, MA, 4Yale Cancer Center, Yale, New Haven, CT, 5Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 6Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 7Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 8Department of Stem Cell and Regenerative Biology, Broad Institute of MIT & Harvard, Cambridge, MA.
Background: The challenge of cancer heterogeneity and the limitations of protein-targeting therapeutics underscore an urgent need for innovative, patient-scalable strategies that can precisely eliminate tumor cells while sparing healthy tissues. Towards this aim, breast cancer is dominated by genomic structural variation-which generates hundreds of tumor-specific nucleic acid sequences per cancer. We have previously shown that up to one-third of metastatic breast cancers harbor highly expressed fusion RNAs as a result of these genomic aberrations (Priedigkeit, SABCS 2023). Although fusions have been successfully targeted at the protein-level such as NTRK and FGFR-family fusions, many remain undruggable with conventional therapies. We hypothesized that advances in nucleic acid technology could be applied to therapeutically exploit these cancer-specific biomolecules. Methods: To test this hypothesis, we applied a modular nucleic acid targeting approach (RADARS, Jiang, Koob et al 2023) that utilizes engineered nucleic acid “sensors” designed to selectively hybridize to target RNA sequences. Upon hybridization, the sensors create a double stranded RNA duplex and recruit an endogenous protein called ADAR, which deaminates an intentional A:C mismatch in the sensor-triggering translation of a linked protein cargo only in the presence of a target RNA. We designed and tested 91 sensors against cancer fusion RNA targets with two broad designs—linear sensors that hybridize directly across the fusion RNA breakpoint sequence and gap sensors that target distinct regions of the 5′ and 3′ fusion partners on the same guide. We tested the selective expression of protein payloads using both designs against two clinically relevant fusion RNAs: (1) an ESR1 fusion and (2) an endogenous fusion RNA in MCF7 cells. Sensor activity was assessed with microscopy for fluorescent protein payloads, and cell viability readouts including CellTiter-Glo, protein quantification of apoptosis markers, and live-cell imaging for cell death payloads. Results: We first confirmed that outlier-expressed fusion RNAs are prevalent across luminal, HER2+, and basal-like cell models—mirroring our prior data on human tumors. We then tested sensors targeting an ESR1::SOX9 fusion RNA. In cells co-transfected with the sensors and fusion RNA, we observed robust mNeon expression, whereas cells without the fusion showed only minimal background fluorescence (mean 25-fold increase vs. no target controls, p < 0.0001), validating the sensors’ specificity. Next, we targeted an endogenous ARFGEF2::SULF2 fusion RNA in the MCF7 breast cancer cell line, deploying sensors with iCaspase9; a pro-apoptotic cargo. Transfection of both linear and gap fusion-targeting sensors into MCF7 cells and subsequent treatment with iCaspase9 inducer drug AP1903 resulted in a significant reduction in cell viability (up to 84% 36-hours post sensor transfection vs. non-targeting controls, p < 0.0001). Live-cell imaging and probing for markers of apoptosis confirmed robust sensor-induced cell death in MCF7 cells, while fusion-negative cells exhibited no toxicity when treated with the same fusion-targeting sensors, demonstrating high selectivity. Conclusions: This study demonstrates the successful design and in vitro proof-of-principle of a biomolecule sensing system that selectively triggers a therapeutic protein effector in the presence of a cancer-specific nucleic acid. These findings have the potential to establish a novel therapeutic paradigm-in which an RNA medicine can repurpose cancer-defining nucleic acid sequences into a targeted, modular, and precise cell-killing mechanism. Our ongoing efforts focus on optimizing sensor design, interrogating mechanisms of sensor-mediated cell killing with diverse protein payloads, and addressing challenges of in vivo delivery.
Presentation numberPS4-04-20
Ralbp1 depletion downregulates DNA repair and potentiates PARP inhibitor activity in BRCA wild type and mutated breast cancer cell lines
Chhanda Bose, TTUHSC, Lubbock, TX
C. Bose1, S. Awasthi2, T. Hutson2, F. Sardela de Miranda3, M. F. Mahecha3, M. W. Melkus3, R. Layeequr Rahman3, S. P. Singh2; 1Neuropharmacology, TTUHSC, Lubbock, TX, 2Internal Medicine, TTUHSC, Lubbock, TX, 3Surgery, TTUHSC, Lubbock, TX.
Background: Rlip (aka RLIP76 protein encoded by RALBP1) is a Ral-regulated, membrane-associated ATPase enzyme that facilitates Phase 2 detoxification, plays a role in clathrin-dependent endocytosis (CDE), and is vital for cancer cell formation and survival. DNA repair is essential for both normal and cancer cell survival, as it protects cancer cells from DNA damage caused by chemotherapy or radiation. Poly (ADP-ribose) polymerases (PARPs) play a crucial role in the repair of DNA damage. By targeting these repair mechanisms in cancer cells, it is possible to improve the effectiveness of cancer treatments. A notable example is dysfunctional homology-directed repair (HDR), which results from inactivating mutations in the BRCA1 and BRCA2 genes, contributing to radiation resistance in human cancer cells. Therefore, identifying new targets within double-strand break (DSB) repair pathways is crucial. A core concept in using PARP inhibitors (PARPi) for treating patients with BRCA1/2 mutations is the concept of synthetic lethality. Our research found that Rlip depletion or inhibition, combined with PARPi, works synergistically in both BRCA1/2-mutated and BRCA1/2-wild-type breast cancer cells. Methods: Two PARP inhibitors, AZD2281 (Olaparib) and AZD2461, were tested on the proliferation and survival of different cancer cell lines, including MDA-MB-231 (triple negative), MCF-7 (ER+), SKBR-3, AU565 (HER-positive), and MDA-MB-436 (BRCA1 mutated triple negative), with and without Rlip depletion using an antisense locked nucleic acid (LNA). For all experiments, 2.5 µM AZD2461 and 10 µM AZD2281 were used for 24 hours in complete growth medium. Cell cycle analysis was performed by flow cytometry. Expression levels of γH2AX and Rad51 proteins were assessed via Western blotting and immunofluorescence staining. The comet assay was used to measure DNA damage and fragmentation. Results: Rlip depletion significantly increased the reduction in cell survival caused by PARPi treatments across all tested cell lines. Combination index (CI) values were calculated using the Hill equation and Chou-Talalay’s method with CompuSyn software. The effects were highly synergistic (CI 1), but the doses of both inhibitors were significantly reduced (p < 0.001) after Rlip depletion. Cell cycle analysis with Rlip depletion showed either an increase in the sub-G1 population (cell death) or G2/M phase arrest, suggesting cells attempt to undergo mitosis with unrepaired DNA damage, leading to elevated DNA double-strand break signaling, cell cycle arrest, and apoptosis. Alkaline comet assay confirmed more DNA damage with Rlip depletion compared to treatments with the inhibitor alone. Western blot analysis for Rad51 and γ-H2AX showed defects in protein levels following Rlip depletion. The number of γ-H2AX foci, characteristic of DNA damage in nuclei, increased significantly (p < 0.01) after Rlip depletion compared to inhibitor treatment alone. Conclusion: Our findings suggest that drugs that inhibit or deplete Rlip could help sensitize breast cancer cells, regardless of their BRCA status or whether they are wild-type ER+ or triple-negative breast cancers, to PARP inhibitors. The immunogenicity of HRD-associated malignancies and the immunomodulatory effects of PARP inhibition could be combined with Rlip inhibitors to treat HRD cancers. These findings clarify the underlying mechanisms and are expected to lead to the development of first-in-class therapies.
Presentation numberPS4-04-21
Deciparticle™ Everolimus (Sapu003): From Cytostasis to Cytotoxicity via a Single mPEG Polymer and Clinic-Ready Manufacturing
Sheng-Hao Min, Sapu Bioscience, LLC, San Diego, CA
S. Min1, K. Forero1, J. Anderson2, W. Putnam2, C. Evizi2, C. McCallum2, R. Hoff1, C. Hsieh2, K. Ho2; 1Formulation, Sapu Bioscience, LLC, San Diego, CA, 2Manufacturing, Sapu Bioscience, LLC, San Diego, CA.
Background: Everolimus is approved for advanced renal-cell carcinoma following VEGF-targeted therapy, for metastatic HR-positive/HER2-negative breast cancer in combination with exemestane, and for other oncology indications. However, everolimus lacks an intravenous option due to its extreme hydrophobicity, despite its low oral bioavailability of only 15-20%. Here, we describe the successful development of an intravenous formulation of Everolimus using our Deciparticle™ platform. Formulation: A 38-member mPEG-block polymer library was screened as potential candidates for encapsulating Everolimus. Nine polymers demonstrated the ability to form stable Deciparticles™. Among these candidates, VT111 was selected based on its favorable biocompatibility. VT111 and its variants were able to formulate both peptides and polyketides, including Everolimus, Sirolimus, Tacrolimus, and Cyclosporine. Manufacturing: Clinical lots were produced using a 7-day process at our cGMP facility. The manufacturing process began with the dissolution of VT111 and Everolimus in ethanol, which was subsequently solubilized into lactose and Water for Injection to form Deciparticles™. The process was conducted under yellow light and utilized amber vials to minimize Everolimus degradation. The bulk drug was filter sterilized. Fill/finish operations were in a temperature-controlled environment with ≥98% filling weight accuracy. The drug product was reconstituted to a final concentration of 4 mg/mL for intravenous administration. Upon reconstitution, the product yielded monodisperse Deciparticles™ that met sterility and particle-size specifications and remained within specifications for at least one month at 5 °C storage and for at least 24 hours of in-use stability at 25 °C. The same process has been successfully scaled to over 10 g, sufficient for Phase 1 clinical supply. In Vivo Activity: Sapu003 exhibited potent antitumor activity in vivo (IV, BIW × 4) and demonstrated tumor growth inhibition (TGI) percentages of: U-87 MG glioblastoma (TGI 97-98%), HT-29 colorectal cancer (71-73%), OVCAR-3 ovarian cancer (66%), and MDA-MB-231 breast cancer (48%). No phlebitis was observed during repeated tail-vein administrations. Sapu003 sensitivity correlated with glycolytic dependence (U-87 ≫ HT-29 > OVCAR-3 ≈ MDA-MB-231). In the glycolysis-dependent U-87 model, Sapu003 was cytotoxic and outperformed paclitaxel, without inducing body weight loss. Conclusions: We screened a 38-member mPEG-block polymer library for self-assembling capacity and identified a leading candidate, VT111, which reproducibly formed stable Deciparticles™ under 20 nm in mean particle diameter and demonstrated potent antitumor activity across multiple xenograft models. VT111 uniquely defines the chemical space for Deciparticle™ technology and enables a reproducible cGMP process yielding a long-term stable drug product. These data could support the ongoing first-in-human study of weekly Sapu003 and open the Deciparticle™ platform to other water-insoluble oncology agents.
Presentation numberPS4-04-22
Bacterial-tumor antigen cross reactive T-cells are enriched in tumors of newly diagnosed breast cancer patients.
Denise Cecil, University of Washington Medicine, Seattle, WA
D. Cecil, J. Childs, M. Disis; Cancer Vaccine Insitute, University of Washington Medicine, Seattle, WA.
Background: The breast tumor immune microenvironment is characterized by a Type II signature which is an early event in breast cancer and portends more aggressive disease. We have previously demonstrated that bacteria-specific T-cells, derived from the gut, also recognize tumor antigens with which they share a high degree of homology at the level of Class II epitopes presented in MHC molecules. These peripheralized bacterial-tumor antigen cross reactive T-cells are tolerogenic, secrete high levels of IL-10 and, in mouse models, when adoptively transferred, traffic specifically to breast tumors and accelerate tumor growth. We questioned if the Type II signature present in human breast cancer is due to these bacterial-tumor antigen (BAC-TA) cross reactive T-cells. Methods: PBMC and stool were collected from newly diagnosed, treatment naive breast cancer patients (n= 56) or volunteer aged matched donors (n=14). PBMC were screened with a panel of nine previously identified BAC-TA epitopes by IL-10 ELISPOT. Tumor biopsies were collected from three patients at the time of surgery. BAC-TA-specific T-cell lines were generated from PBMC from these three patients that responded with high magnitude IL-10 in the initial screen to BIRC5-p13-30 for Donor 1 (mean, 178±59 SPW / 106; p=0.0002) and Donor 2 (mean, 1612±234 SPW / 106; p=0.0011) and PRL3-p104-118 for Donor 3 (mean, 365±100 SPW / 106 T-cells; p=0.03). Extracted DNA from the PBMC, T-cell lines and tumor biopsy was subjected to TCRb sequencing. Metagenomic sequencing was performed on stool samples. Results: A higher precursor frequency of BAC-TA cross reactive IL-10-secreting T-cells were observed in 60% of breast cancer patients (median, 1:160,000 PBMC) as compared to 28% of the volunteer donors (median 50% homology to BIRC5-p13-30 were enriched in the stool of Donor 1 as compared to the levels in the stool of the 56 breast cancer patients evaluated in this study (L. lactis; enriched by 186-fold and S. oralis and P. fluorescens; both enriched by 3-fold). Similar trends were observed in the other two donors. Eleven BIRC5-p13-specific T-cell clones from Donor 2 were found in the matching tumor biopsy, with enrichment of two bacterial species with high BIRC5-p13-30 sequence homology (K. pneumoniae; 256-fold increase and P. distasonis; 4-fold increase). 25 PRL3-p104-specific T-cell clones in Donor 3 were found to be identical between the tumor biopsy and T-cell line. The stool of Donor 3 demonstrated a 15-fold and 3.5-fold increase in P. copri and P. fluorescens, respectively, two bacteria with at least 50% homology to PRL3-p104-118. Conclusion: IL-10-secreting BAC-TA T-cells can be found in high numbers in the peripheral blood of of newly diagnosed breast cancer patients and can be identified infiltrating the tumor. IL-10 is a key immunosuppressive cytokine that plays a central role in dampening immune responses and maintaining immune homeostasis. IL-10 inhibits the production of pro-inflammatory cytokines such as TNF-α and IFN-γ and can limit antigen-presenting cell function, leading to the reduction of Th1, Th17 and cytotoxic CD8+ T-cell activation. These data lay the foundation for a study that will follow newly diagnosed breast cancer patients correlating levels of BAC-TA cross reactive T-cells in blood with prognosis or response to therapy.
Presentation numberPS4-04-23
Pelp1 inhibition disrupts dna repair and enhances topoisomerase inhibitor efficacy in triple negative breast cancer
Khaled Mohamed Nassar, UT Health SanAntonio, SAN ANTONIO, TX
K. Nassar1, J. Sanchez1, D. Panneerdoss1, E. Behnam1, X. Yang1, U. Pratap1, M. Mahajan1, S. Alejo1, P. Subbarayalu2, D. Zhou3, G. Sareddy1, M. Rao2, S. Viswanadhapalli1, R. K. Vadlamudi1; 1ObGyn, UT Health SanAntonio, SAN ANTONIO, TX, 2GCCRI, UT Health SanAntonio, SAN ANTONIO, TX, 3Department of Biochemistry & Structural Biology, UT Health SanAntonio, SAN ANTONIO, TX.
Background: Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype associated with poor prognosis and high rates of metastasis, contributing to 15-24% of breast cancer-related deaths. The oncogene proline-, glutamic acid-, and leucine-rich protein 1 (PELP1) is frequently overexpressed in TNBC and is associated with poor survival. This study aimed to elucidate the critical role of PELP1 in DNA repair mechanisms of TNBC and to evaluate a novel therapeutic strategy using a PELP1 small-molecule inhibitor, SMIP34, in combination with topoisomerase inhibitors (TIs). Methods: PELP1-inducible knockdown (iKD) TNBC models (MDA-MB-231, BT-549, HCC-1806, SUM-149) were generated via lentiviral transduction using IPTG-inducible shRNA constructs. PELP1 signaling was inhibited pharmacologically using SMIP34. A drug screen of 140 FDA-approved compounds was performed using MTT assays to identify synergistic agents with SMIP34. Functional assays, including colony formation and Annexin V/PI apoptosis assays, were used to assess therapeutic efficacy. Mechanistic studies utilized RT-qPCR, Western blotting, comet assays, and DNA Damage Response Phosphorylation Arrays. Combination treatments were validated in cell line-derived xenografts , patient-derived organoids , and patient-derived xenografts . Results: Western blot analyses confirmed that IPTG induction resulted in a dose-dependent suppression of PELP1 expression in all four PELP1-iKD TNBC cell lines. PELP1 knockdown significantly impaired cell proliferation and colony formation while inducing apoptosis. In parallel, pharmacological inhibition using SMIP34 produced similar phenotypic outcomes, validating the functional relevance of PELP1 in TNBC cell survival. Importantly, PELP1 suppression led to a delayed DNA damage response (DDR), marked by elevated γ-H2AX and activation of key DDR kinases including p-ATM, p-ATR, and DNA-PKcs. SMIP34 treatment replicated these effects, confirming that both genetic and pharmacologic inhibition of PELP1 compromises DNA repair efficiency. A high-throughput MTT-based screen of 140 FDA-approved agents identified topoisomerase inhibitors (TIs) as potent synergistic agent with SMIP34. Co-treatment with SMIP34 and TIs resulted in significantly enhanced growth inhibition and apoptosis compared to monotherapy. This synergy was confirmed in PELP1-iKD cells, where knockdown further sensitized cells to TIs, providing genetic validation of PELP1’s role in TI resistance. Mechanistic studies showed that combination treatments (PELP1-iKD+TIs or SMIP34+TIs) markedly increased DNA damage and apoptotic signaling. Western blotting and phospho-antibody arrays revealed upregulation of DDR and checkpoint proteins, including γ-H2AX, p-CHK2, p-ATR, p-CHK1, p-BRCA1, and p21, indicating enhanced replication stress and cell cycle arrest. Comet assays demonstrated significantly greater DNA fragmentation with combination therapy compared to single agents. In preclinical models, SMIP34+TI treatment significantly suppressed organoid growth in patient-derived organoid cultures. In vivo, this combination led to marked tumor growth inhibition in both cell line-derived xenografts and patient-derived xenografts, outperforming either agent alone. No significant toxicity was observed, supporting the therapeutic potential of this approach. Conclusion: These findings highlight PELP1 as a critical modulator of DNA damage response in TNBC and support the development of combination strategies using SMIP34 and topoisomerase inhibitors. This approach offers a promising therapeutic avenue for improving outcomes in patients with TNBC.
Presentation numberPS4-04-24
Pharmacological Repurposing of Natural Products for Triple-Negative Breast Cancer
Marelly A Perez, University of the Incarnate Word, San Antonio, TX
M. A. Perez1, C. S. Fermaintt1, A. Risinger2; 1Department of Chemistry and Biochemistry, University of the Incarnate Word, San Antonio, TX, 2Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX.
Breast cancer affects approximately 1 in 8 women in the United States, with triple-negativebreast cancer (TNBC), representing 10–15% of all cases. TNBC is characterized by lowexpression of estrogen, progesterone, and HER2 receptors, which limits the efficacy ofapproved therapeutics targeted against these receptors. The molecular subtyping of TNBCshas been valuable in the search for new genetic and pharmacological vulnerabilities against thisheterogeneous disease and includes basal-like 1 (BL1), basal-like 2 (BL2), mesenchymal (M),and luminal androgen receptor (LAR) subtypes. We previously screened natural productextracts for selective cytotoxic efficacy among TNBC cell lines representing each of thesesubtypes, which led to the identification of the diterpenoid yuanhuacine as highly selectiveagainst the BL2 subtype, both in vitro and in vivo antitumor studies. This is notable as BL2tumors represent 13% of TNBCs and have the lowest pathological complete response rate toneoadjuvant chemotherapy among these molecular subtypes at 0%. Further studies havedemonstrated that structurally and functionally related ingenane diterpenoids in clinicaldevelopment for the topical treatment of skin malignancies, including tigilanol tiglate andingenol-3-angelate, retain BL2 subtype-selective activity with over one-thousand-fold selectivity,providing a unique opportunity for drug repurposing.To support these efforts, we have taken a multipronged approach to identify the mechanism ofselectivity of these compounds against BL2 TNBC cells, perform structure-activity relationship(SAR) and pharmacokinetic studies to inform on repurposing this drug class for systemicadministration, and determine their efficacy in combination with standard of care chemotherapy.We have demonstrated that the selective activity of yuanhuacine and ingenane diterpenoids isdue to activation of PKCβ, which is elevated in cell lines representing the BL2 TNBC subtype.The evaluation of sixteen structurally related compounds with potency in BL2 cell lines rangingfrom 0.07 – 50,000 nM with 3 – 300,00-fold selectivity for this subtype has informed on the SARfor BL2-specific cytotoxicity. In vitro pharmacokinetic studies have further demonstratedpotential liabilities of this drug class that need to be taken into account with systemicadministration that will be used in combination with SAR studies to inform on medicinalchemistry efforts to prioritize compounds for antitumor efficacy studies in BL2 tumor models anddevelop compounds with a greater therapeutic index than yuanhuacine. We have also identifiedoptimal combinations of these highly BL2-selective compounds with standard of carechemotherapeutics to identify drug pairings that would have optimal efficacy in heterogenousBL2-enriched TNBC tumors. Finally, we have identified additional tumor types that expressPKCβ, including pediatric cancers, and determined the potential for ingenane diterpenoids forrepurposing in a broader group of cancers through molecular profiling. Together, these studiesinform on the potential to repurpose a class of natural products developed for skin cancer for thetreatment of a molecularly defined subtype of TNBC to improve targeted treatment options forthese patients.
Presentation numberPS4-04-25
Selective Cytotoxicity of Natural Products in Triple-Negative Breast Cancer Mediated by PKCβ Activation
Sofia C Stanfield, University of the Incarnate Word, San Antonio, TX
S. C. Stanfield1, C. S. Fermaintt2, A. Risinger3; 1Biology, University of the Incarnate Word, San Antonio, TX, 2Chemistry and Biochemistry, University of the Incarnate Word, San Antonio, TX, 3Pharmacology, University of Texas Health San Antonio, San Antonio, TX.
Triple-negative breast cancer (TNBC) accounts for approximately 15–20% of all breast cancercases and disproportionately affects younger women. Characterized by the absence of estrogen(ER), progesterone (PR), and HER2 receptors, TNBC lacks the molecular targets utilized in mostcurrent therapies, resulting in limited treatment options and a heavy reliance on cytotoxicchemotherapy. TNBC is associated with a more aggressive clinical course, early metastasis, andpoor overall survival as compared to other types of breast cancer, highlighting the urgent needfor novel and effective therapeutic strategies. To identify new therapeutic targets, TNBC hasbeen classified into four molecular subtypes: basal-like 1 (BL1), basal-like 2 (BL2),mesenchymal (M), and luminal androgen receptor (LAR). Among these, the BL2 subtypeexhibits particularly poor prognosis and resistance to standard of care treatments. In our study,we identified yuanhuacine and other related diterpenoid natural products as compounds withpotent (0.07 – 7.7 nM) cytotoxicity against BL2 TNBC cells. These compounds are over one-thousand-fold more selective for the BL2 subtype of TNBC as compared to other TNBCsubtypes or immortalized normal breast lines supporting the identification of a novel targetablevulnerability in this breast cancer subtype. Most notably, we demonstrated this exquisite potencyand selectivity for BL2 TNBC is also a feature of compounds of this class that are undergoingclinical development for the topical treatment of skin cancers demonstrating an opportunity fordrug repurposing.Mechanistic studies revealed that the efficacy of these compounds is mediated by activation ofProtein Kinase C beta (PKCβ), which is elevated in BL2 TNBC cells. Pharmacologic inhibitionof PKCβ significantly reduced the potency of yuanhuacine and ingenol-3-angelate in BL2 TNBCcells, indicating that PKCβ activation is necessary for their selective activity. Geneticexperiments are being conducted to interrogate whether PKCβ expression is necessary andsufficient for sensitivity to yuanhuacine with reduced potency expected in in BL2 lines uponPKCβ knockdown and sensitization of non-BL2 cell lines when ectopically expressed. Cellbiological experiments demonstrated treatment of BL2 cells with yuanhuacine promotesphosphorylation of PKCβ at Ser660 and activation of PKC substrates within one hour oftreatment, suggesting rapid activation and engagement of downstream signaling. Additionalimmunofluorescence studies demonstrate the effect of this activation on the intracellularlocalization and degradation of PKCβ as well as induction of apoptosis.Our findings demonstrate that yuanhuacine and other ingenane diterpenoids, includingcompounds in clinical development for other indications, have the potential for the targetedtreatment of the BL2 subtype of TNBC. Additionally, the identification of PKCβ expression as abiomarker of sensitivity to this class of drugs opens the potential for efficacy in other cancerswhere PKCβ is expressed. Given the lack of targeted therapies for BL2 TNBC, the developmentof PKCβ-activating compounds like yuanhuacine represents a promising and urgently neededadvancement in personalized oncology.
Presentation numberPS4-04-26
Anti-tumor activity of ATNM-400, a first-in-class Actinium-225 antibody radioconjugate, in tamoxifen and trastuzumab resistant breast cancer models
Adeela Kamal, Actinium Pharmaceuticals, New York, NY
A. Kamal1, A. S. Chin1, S. Mukherjee1, J. Li1, K. Peregrina1, D. Lewis1, H. Sethi1, L. Xu1, D. Patel2, M. Roy1, A. Bardia3; 1R&D, Actinium Pharmaceuticals, New York, NY, 2Clinical, Actinium Pharmaceuticals, New York, NY, 3Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA.
Background: Hormone receptor positive (HR+) is the most common subtype of breast cancer (BC) and comprises >70% of BC diagnosis. Tamoxifen is the most widely prescribed endocrine therapy (ET) for modulating estrogen receptors, however, ~20-30% of BC patients have disease recurrence despite ET. Similarly, resistance to HER2-targeted therapy trastuzumab (Herceptin) can develop in HER2+ BC. While antibody drug conjugates (ADCs) like trastuzumab deruxtecan (Enhertu) show strong efficacy in HER2+ and HER2-low BC, their use is limited by interstitial lung disease toxicities. In contrast, Actinium-225-based radioconjugates may provide potent tumor killing with less off-target lung toxicity. Here, we report the development of ATNM-400, a novel antibody radioconjugate using Actinium-225 to target a protein that is overexpressed in BC. The target is linked to disease progression and metastasis, with overexpression correlating with poor clinical outcomes in BC patients. Furthermore, target expression is elevated in patients who develop ET and HER2 therapy resistance. Here, we evaluated the anti-tumor efficacy of ATNM-400 in preclinical BC models and evaluated ATNM-400’s potential to overcome current therapeutic limitations and improve outcomes beyond what is achievable with tamoxifen or trastuzumab. Methods: ATNM-400 was synthesized by conjugating ATNM400 antibody with p-SCN-Bn-DOTA, followed by radiolabeling with Ac-225 and characterized by radio-iTLC, HPLC and mass spectrometry. Target expression, binding affinity, and internalization were assessed using flow cytometry and radioactivity quantification in parental and drug-resistant BC cell lines, including MCF-7 (HR+), MCF-7 Tam1 (tamoxifen-resistant), BT-474 (HER2+), BT-474 Clone 5 (trastuzumab-resistant), and MDA-MB-468 (triple-negative). In vivo biodistribution and efficacy were evaluated in murine xenograft models. Efficacy of ATNM-400 alone and in combination with standard-of-care (SOC) therapies was assessed in both in vitro and in vivo resistance models. Results: ATNM-400 demonstrated over 98% radiochemical purity. ATNM-400 exhibited strong and specific binding and internalized in target-positive BC cell lines. Target expression was significantly elevated in tamoxifen- and trastuzumab-resistant lines, supporting its mechanistic link to resistance. In vitro, ATNM-400 induced potent, dose-dependent cytotoxicity as monotherapy, which was further enhanced when combined with SOC agents. In vivo, ATNM-400 achieved tumor growth inhibition (TGI) of 76.4% and 111.5% in MCF-7 xenografts at 20 and 40 μCi/kg, respectively, with the lower dose being well tolerated. In MDA-MB-468 xenografts, TGI reached 66.3% and 103.4% at corresponding doses, both of which were well tolerated. Notably, ATNM-400 retained efficacy in both tamoxifen- and trastuzumab-resistant models, demonstrating its potential to overcome resistance mechanisms. Conclusions: ATNM-400 shows strong anti-tumor efficacy and favorable tolerability across multiple BC subtypes, including models resistant to tamoxifen and trastuzumab. These findings support further development of ATNM-400 as a novel therapeutic approach for patients with limited options following endocrine or HER2-targeted therapy failure-both as monotherapy and rational combination regimens.
Presentation numberPS4-04-27
Slc39a7/zip7as a tumor-specific antigen enabling precision immunotherapy in breast and prostate cancers
John S Manavalan, Cancer Antibodies Inc, Longwood, FL
J. S. Manavalan1, A. Ahmad2, D. Mor1, J. Davis1, R. Chakrabarti3, A. Pollack2, E. Davis1; 1Oncotope Platform, Cancer Antibodies Inc, Longwood, FL, 2Radiation Oncology, University of Miami, Miller School of Medicine, Miami, FL, 3Surgery, University of Miami, Miller School of Medicine, Miami, FL.
Background: Antibody-drug conjugates (ADCs), bispecific antibodies, and CAR-T cells offer tumor-selective cytotoxicity with reduced off-target toxicity. Their success depends on identifying cancer-specific surface antigens absent in normal tissues. The Oncotope Platform, developed by Cancer Antibodies Inc., enables high-throughput antigen discovery through rabbit immunization with cancer cell lines, filtration to remove cross-reactive antibodies, and protein microarray screening of >20,000 human proteins. This platform shortened discovery timelines and demonstrated selectivity by generating antibodies that killed breast cancer cells (Hs578T) while sparing matched normal cells (Hs578Bst). One lead antigen was SLC39A7 (ZIP7), a zinc transporter normally confined to the ER/Golgi but aberrantly expressed on the surface of malignant cells. In contrast to PSMA, which is expressed in salivary glands and kidneys and associated with significant toxicities in radioligand trials (e.g., 81% salivary, 25% renal), ZIP7 showed restricted expression in normal tissues, making it a promising target for safer therapeutics in breast and prostate cancer, including castration-resistant disease. Methods: ZIP7 was validated using in vitro and in situ approaches. Breast cancer cell lines were evaluated by flow cytometry for ZIP7 surface expression and by cytotoxicity assays for selective tumor killing. ZIP7 mRNA expression was assessed in 10 breast cancer cell lines representing Luminal A, HER2+, and TNBC subtypes. Immunofluorescence (IF) was performed on TNBC samples (n=5; grades 2-3) and prostate cancer samples (n=6; Gleason 3+4, 4+3, including CRPC). Each case included adjacent benign tissue as control. CyTOF imaging analyzed co-localization of ZIP7 with CD4+ and CD8+ T cells. IHC-DAB was used to evaluate ZIP7 expression in PSMA-negative (PC3) and PSMA-positive (LNCaP) xenografts and in normal kidney, spleen, and brain tissues. Results: In vitro, filtered antibodies killed ZIP7-expressing Hs578T cells while sparing Hs578Bst cells. ZIP7 was detected in 8/10 breast cancer cell lines, and ZIP7-specific antibodies bound selectively and induced cytotoxicity. Flow cytometry confirmed surface expression. In FFPE samples, IF showed ZIP7 staining in all TNBC tumors (5/5), absent in adjacent normal epithelium. In prostate cancer samples, ZIP7 expression was observed across all grades and CRPC, with enrichment at invasive margins and immune-infiltrated areas. CyTOF showed ZIP7+ cells in proximity to CD4+/CD8+ T cells. IHC-DAB confirmed ZIP7 in both PC3 and LNCaP xenografts. No ZIP7 expression was detected in normal kidney, spleen, or brain tissues. Conclusions: ZIP7 is a tumor-selective surface antigen with aberrant expression in breast and prostate cancers, including resistant subtypes. Its absence from essential normal tissues supports its safety as a therapeutic target. The Oncotope Platform enabled the rapid discovery and validation of ZIP7, demonstrating its power to uncover high-value, tumor-specific targets for antibody-based cancer therapies.
Presentation numberPS4-04-28
Multifunctional lipid nanoparticles remodeling tumor immune microenvironment for breast cancer chemio-immunotherapy
Huiying Fang, Chongqing University Cancer Hospital, Chongqing, China
H. Fang1, W. Jiang2; 1Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, CHINA, 2Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, CHINA.
AbstractBackground: Breast cancer is one of the most common malignant tumors worldwide, and its treatment faces challenges such as the complexity of the immune microenvironment and the side effects associated with conventional therapies. Doxorubicin (DOX) is often confronted with issues of resistance and significant side effects, while Resiquimod (R848) has been shown to enhance anti-tumor immune responses. This study presents a liposome-based nanodrug delivery platform that combines the advantages of liposomes with the protective properties of chitosan, aiming to co-deliver DOX and R848 to target tumor cells and tumor-associated macrophages (TAMs), thereby improving the therapeutic efficacy in breast cancer treatment.Methods: In this study, folic acid (FA) and chitosan (CS) were conjugated to form an FA-CS complex for incorporation into liposomes, enhancing their stability and targeting ability. FA-CS-R848/DOX@Lip liposomes were prepared via the rotary evaporation method, and their morphology, particle size, and zeta potential were characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). The drug encapsulation efficiency, loading capacity, and in vitro release were assessed. In vivo, a mouse breast cancer xenograft model was established to evaluate the anti-tumor efficacy and biocompatibility of the drug delivery system. Flow cytometry was used to quantify immune cell populations in the tumor immune microenvironment, including CD8+ T cells, TAMs, regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSCs), to analyze changes in the immune response. Results: FA-CS-R848/DOX@Lip liposomes were successfully prepared with a particle size of approximately 211 nm and a zeta potential of 5.3 mV. The encapsulation efficiencies of R848 and DOX were found to exceed 80% and 70%, respectively, with drug loading capacities of 6.8% and 4.6%. Drug release studies indicated that the release profile of FA-CS-R848/DOX@Lip liposomes was relatively slow. Cellular uptake assays demonstrated significantly higher uptake efficiency in RAW 264.7 macrophages and EO771 tumor cells compared to non-targeted liposomes. Cytotoxicity evaluations showed that FA-CS-R848/DOX@Lip exhibited superior anti-tumor activity, effectively inducing tumor cell death while simultaneously stimulating immune responses. In vivo experiments revealed that FA-CS-R848/DOX@Lip significantly inhibited tumor growth, while maintaining favorable biocompatibility and safety profiles. Flow cytometry analysis indicated a notable increase in the proportion of CD8+ T cells with enhanced functionality, as well as a significant shift in the M1/M2 ratio of TAMs and a reduction in the numbers of Tregs and MDSCs. Collectively, these results suggest that this nanodelivery platform effectively activates immune responses and remodels the tumor immune microenvironment.Conclusion: The FA-CS-R848/DOX@Lip liposomes developed in this study synergistically combine the cytotoxic effects of DOX with the immune activation properties of R848, achieving a cooperative interaction between chemotherapy and immunotherapy. This delivery platform effectively targets tumor cells and TAMs, significantly enhancing anti-tumor immune responses while overcoming the limitations of conventional therapies. By optimizing the drug delivery system and enabling precise immune modulation, FA-CS-R848/DOX@Lip represents a novel and effective strategy for breast cancer treatment. This study provides a theoretical foundation for the combined application of chemotherapy and immunotherapy, offering promising prospects for advancing therapeutic approaches in other cancer types.
Presentation numberPS4-04-29
Antibody Inhibiting SCUBE3 for the Treatment of Triple Negative Breast Cancer
Deepika Singh, Greehey Children’s Cancer Research Institute, San Antonio, TX
D. Singh1, B. Onyeagucha2, D. Medina1, P. Subbarayalu1, R. Mojidra3, P. Venkata1, J. Huang1, J. Prochnau1, P. Do1, M. Rao1; 1Cell System & Anatomy, UTHSA, Greehey Children’s Cancer Research Institute, San Antonio, TX, 2Department of Science & Mathematics, Mississippi University for Women, Columbus, MS, 3Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX.
Introduction: Antibody-based therapy is one of the most successful strategies for treating patients with cancers. This is also true for breast cancers as trastuzumab (a monoclonal antibody against Her2) is used as the first line therapy for Her2+ breast cancer. Despite substantial progress, there remains a notable gap in effective targeted treatments for TNBC. In the landscape of cancer treatment, chemotherapy is the common treatment option for TNBC. However, resistance and tumor relapse remain the major obstacles hindering the effectiveness of chemotherapeutic agents in cancer patients. Therefore, the development of new therapeutics, especially targeted approaches like antibody-based therapies, holds immense significance. This study aims to identify and test the role of secretory protein/s that may support the growth and progression of TNBC cells. Additionally, it seeks to determine if targeting these oncogenic secretory proteins with antibodies can provide a novel therapeutic approach for TNBC patients. Our study identified secretory protein “Signal peptide CUB domain EGF-like 3 (SCUBE3)” as a critical factor that promotes survival and therapy resistance of TNBC cells. Importantly, using antibody-development platform, our study presents a novel therapeutic approach targeting SCUBE3 using anti-SCUBE3 monoclonal antibody, offering new hope for TNBC patients with limited treatment options. Methods: Unbiased high-throughput loss of function genomic screen was performed on TNBC cells in the presence or absence of doxorubicin. TNBC cells were transfected either with SCUBE3 overexpression plasmid or shRNA specific to SCUBE3 to perform functional assay, in vivo tumor xenograft assays, RNA sequencing and Mass spectrometry (MS). Syngeneic mouse model was used to study the effect of SCUBE3 in shaping tumor immune microenvironment of TNBCs. Hybridoma culture technique was used to generate anti-SCUBE3 monoclonal antibodies. Screening of top anti-SCUBE3 antibody producing clone was done based on ELISA, long-term and short-term colony formation assay. Therapeutic efficacy of antibodies was evaluated in tumor xenograft models. Result: Through a loss-of-function genomic screening approach, we identified secretory SCUBE3 as a critical player that promotes the survival, therapy resistance and metastasis of TNBC cells. Our MS data shows that SCUBE3 interacts with several oncogenic proteins including EFGR, TGFβR and mutant CALR. Transcriptomic analysis demonstrates that SCUBE3 depletion leads to inhibition of pro-tumorigenic signaling as well as activation of anti-tumor immunity pathways. Our results reveal that SCUBE3 regulates the expression of FOXR2 which in turn stabilizes Myc and recruits DNMT to form a repressor complex on the promoter of IRF1. Inactivation of IRF1 gene suppresses the expression of MHC class I and class II genes which leads to immune suppression. Further, our syngeneic mouse model study shows that suppression of SCUBE3 leads to increased infiltration of CD8+ T cells and NK cells into the tumor. Importantly, using multiple pre-clinical models including PDX models, we show that antibody targeting SCUBE3 blocks tumor growth. Conclusion: This study will set the stage for a new paradigm of treating TNBCs by inhibiting SCUBE3 activity. Our study is first to identify SCUBE3 as a novel ligand of EGFR1 and CALRmut. Notably, our study shows that SCUBE3 promotes pro-tumorigenic signaling cascade while inhibiting anti-tumor immunity to promote TNBC growth and render them resistant to therapy. Importantly, we provide compelling evidence that antibody targeting SCUBE3 may serve as a potent therapeutic for treating TNBCs.
Presentation numberPS4-04-30
Discovery of NKT5097: a first-in-class, highly potent and selective, orally bioavailable CDK2/4 dual degrader for cancer therapy
Ke Liu, NiKang Therapeutics Inc, Wilmington, DE
K. Liu, J. Geng, Z. Yu, Y. Yeh, H. Wei, W. Sun, W. Li, J. Lu, J. Deng, C. Yang, L. Geng, X. Luo, Y. Liang, Z. Liu, Z. Gao, Y. Lou; R&D Department, NiKang Therapeutics Inc, Wilmington, DE.
CDK4 is the pivotal driver of HR+HER2- breast cancer and CDK4/6 inhibitors in combination with endocrine therapy have revolutionized the treatment landscape. However, several challenges remain for the approved CDK4/6 inhibitors, including severe hematotoxicity associated with CDK6 inhibition and eventual resistance, often mediated by CDK2/cyclin E pathway activation. Therefore, simultaneously and selectively targeting CDK4 and CDK2 represents a promising therapeutic strategy. The high homology between CDK1 and CDK2, as well as between CDK4 and CDK6, poses a significant challenge for the discovery of selective CDK2/4 dual inhibitors. To overcome these limitations, we employed the targeted protein degradation approach and have discovered NKT5097, a first-in-class, selective, and orally bioavailable CDK2/4 dual degrader. Herein, we disclose the evaluation of NKT5097 activity across a panel of human cancer cell lines to assess its ability to induce CDK2 and CDK4 protein degradation. Its selectivity over other CDKs, especially CDK1 andCDK6, was confirmed through a series of cellular and biochemical assays. Additionally, anti-tumor efficacy and potential off-target effects of NKT5097 were assessed and benchmarked against CDK2 or CDK4 specific inhibitors currently in clinical development. NKT5097 degrades CDK2 and CDK4 rapidly and effectively (DC50: 0.6-15 nM), leading to potent inhibition of Rb phosphorylation and proliferation in CDK2- or CDK4-dependent cancer cells and has up to 50-fold greater potency than PF-07104091 (tagtociclib, a CDK2i) or PF-07220060 (atirmociclib, a CDK4i). In addition, NKT5097 demonstrates strong selectivity for CDK2/4 degradation over CDK1/6/7/9 in various cellular assays. Indeed, NKT5097 treatment predominantly arrests CDK2- and/or CDK4-dependent cells at G1 phase of the cell cycle and has minimal impact on G2 distribution, suggesting a lack of CDK1 inhibition. NKT5097 also demonstrates a 68-fold increase in selectivity for CDK2 over CDK1 in comparison to PF-07104091, and a 33-fold increase in selectivity for CDK4 over CDK6 compared to PF-07220060 in functional cellular assays. The selectivity of NKT5097 for CDK2/4 is further confirmed in a Kinome screen as well as by global proteomic profiling, in which CDK2 and CDK4 are the top hits among the >8,000 proteins quantified. Oral administration of NKT5097 dose-dependently degrades CDK2 and CDK4 and shows robust anti-tumor activities across multiple tumor xenograft models including HR+HER2- breast cancer and cyclin E1-high cancer models. Additionally, combination of NKT5097 with fulvestrant leads to deeper degradation of CDK2 and CDK4 in vitro, resulting in better tumor growth inhibition in HR+ breast cancer models. These findings establish NKT5097 as a potent, selective and orally bioavailable dual degrader of CDK2 and CDK4. NKT5097 offers distinct advantages over the combined use of CDK2 and CDK4 inhibitors, including more comprehensive pathway inhibition and a lower risk of overlapping toxicity, owing to its high potency against CDK2/4 and strong selectivity over CDK1/6. Therefore, as the first-in-class CDK2/4 dual degrader, NKT5097 presents a unique opportunity to serve as a backbone therapy for HR+HER2- breast cancer and other cancers with CDK2 and/or CDK4 dysregulation. NKT5097 is currently being investigated in a phase I clinical study.
Presentation numberPS4-06-02
Combinatory treatments of VRN101099 with antibody therapies for HER2-driven breast cancer
Hong-Ryul Jung, Voronoi Inc, Incheon, Korea, Republic of
H. Jung, S. Kim, S. Kim, Y. Lee, J. Yoo, J. Kwon, S. Han, D. Park, D. Shin, H. Kim, J. Kim, Y. Ko, S. Kim; R&D, Voronoi Inc, Incheon, KOREA, REPUBLIC OF.
Many systemic therapeutic options for advanced metastatic HER2-positive breast cancer have been approved, including antibody-based therapies, chemotherapies, and tyrosine kinase inhibitors (TKIs). However, brain metastasis and acquired resistance are still major unmet medical needs for HER2-driven metastatic breast cancer and advanced solid cancers. VRN101099 is an orally bioavailable, highly selective HER2 kinase inhibitor, demonstrating excellent specificity over other kinases, including EGFR. This compound potently inhibits HER2 by covalently binding to Cys805 within the ATP-binding pocket, leading to robust catalytic inhibition of HER2 kinase. In vitro studies show that VRN101099 exhibits potent anti-proliferative effects against HER2-driven breast cancer cell lines, such as BT-474 and SK-BR-3. Furthermore, VRN101099’s irreversible binding properties enable effective inhibition of various HER2 activating mutants, including L755S, V777L, D769H/Y and L869R. Notably, VRN101099 demonstrates brain penetrability. Given its distinct mechanism of action—kinase catalytic inhibition—which differs from that of antibody-based HER2 therapies (e.g., Trastuzumab, Pertuzumab, T-DM1, T-DXd, Zanidatamab), VRN101099 holds promise for synergistic therapeutic effects when combined with these agents, particularly in the treatment of metastatic HER2-positive breast cancer. Moreover, VRN101099 induces internalization of the HER2 receptor, promoting internalization of the antibody-drug conjugate. In BT474 or SKBR3, HER2 amplified breast cancer cells, xenograft models with subcutaneous or intracranial implantation, VRN101099 showed combinatory and synergistic anti-tumor effects with Trastuzumab, Pertuzumab, T-DM1, T-DXd, and Zanidatamab. The first-in-human dose escalation study of VRN101099 monotherapy will determine the maximal tolerable dose, and an optimal dose finding study for the combination will be followed.
Presentation numberPS4-06-04
First-in-human Phase 1 study of BTX-9341, a CDK4/6 bifunctional degrader, as monotherapy and in combination with fulvestrant in patients with advanced and/or metastatic HR+/HER2- breast cancer – first emerging data
Amita Patnaik, The START Center for Cancer Research, San Antonio, TX
A. Patnaik1, M. Block2, M. Cristofanilli3, Q. Liu4, H. Majeski4, A. Shakya4, P. Rajagopalan4, C. Deshpande4, T. Marmon4, S. Padam4, D. Powell4, A. H. Chourasia4, R. Urbanski4, M. Goetz5, R. M. Layman6; 1The START Center for Cancer Research, The START Center for Cancer Research, San Antonio, TX, 2Nebraska Cancer Specialists, Nebraska Cancer Specialists, Omaha, NE, 3Weill Cornell Medicine/New York-Presbyterian Hospital, Weill Cornell Medicine/New York-Presbyterian Hospital, New York, NY, 4Biotheryx, Biotheryx, San Diego, CA, 5Department of Oncology, Mayo Clinic, Rochester, MN, 6Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: BTX-9341 is a first-in-class bifunctional degrader of cyclin dependent kinase (CDK)4 and CDK6 that also inhibits the transcription of CDK2 and Cyclin E. Preclinical data highlight its superiority compared with approved CDK4/6 inhibitors (CDK4/6i) in inhibition of phosphorylation of retinoblastoma (p-RB), cell cycle arrest, and in vivo efficacy in breast cancer (BC) xenografts. BTX-9341 is active in CDK4/6i‑resistant models with p-RB half-maximal inhibitory concentrations (IC50s) of 1‑15 nM and can overcome key mechanisms that drive CDK4/6i resistance. Here we report early monotherapy data from the first-in-human study of BTX-9341 in patients (pts) with advanced and/or metastatic HR+/HER2- BC. Methods: BTX-9341-101 is a multicenter, nonrandomized, open-label trial to assess the safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), and preliminary efficacy of orally administered BTX-9341 in pts with RB1 wild type advanced and/or metastatic HR+/HER2- BC who received prior CDK4/6i therapy. An accelerated titration and Bayesian Optimal Interval design (BOIN) were used. The primary objective of dose escalation is to determine the maximum tolerated dose (MTD) or maximum evaluable dose (MED) of BTX-9341 monotherapy and in combination with fulvestrant. Secondary objectives include the characterization of BTX-9341 on PK and efficacy; exploratory objectives include the assessment of BTX-9341 on PD and PK/PD relationships. Results: As of June 9, 2025, 12 pts were evaluated at the following BTX-9341 once daily (QD) dose levels: 50 mg (dose level [DL]1), 100 mg (DL2), and 75 mg (DL3) monotherapy, and 50 mg in combination with fulvestrant (DLA). Pts received up to 4 lines of therapy in the metastatic setting prior to enrollment. No dose‑limiting toxicities (DLTs) were observed at any dose levels during the 28-day DLT period. The most common adverse events (AEs) during the DLT period were decreased neutrophil and white blood cell counts (Grades 1-4). There were no dose reductions in DL1. The starting dose was reduced in all pts in DL2; DL3 is currently enrolling. At least 2-fold accumulation was observed after QD dosing, and steady-state trough concentrations were in the range of in vitro IC50 values at DL1 and higher dose levels. Serum thymidine kinase activity (sTKa), a clinical biomarker for tumor response to CDK4/6 inhibition, showed >50% reduction at Cycle 2 Day 1 (C2D1) compared with baseline for 7 of 8 evaluable monotherapy pts, with 6 pts below the sTKa detectable level. “On target” PD reductions in CDK4, CDK6, CDK2, and Cyclin E levels were observed in peripheral blood mononuclear cells. Of the 8 evaluable monotherapy pts, the best overall responses in pts with measurable disease included 1 pt with a confirmed partial response (PR) and 3 pts with stable disease ([SD] first assessed at 8 weeks; i.e., the first time point for assessment in the protocol), and in pts with non-measurable disease, 2 pts had non-complete response (CR)/non‑progressive disease. For all pts, the longest duration on treatment was 10 cycles. A clinical benefit rate (defined as a best overall response of CR, PR, or SD for pts with measurable disease or non-CR/non-progressive disease for pts with non‑measurable disease, per RECIST v1.1 at Week 24) of 66.7% was observed in the completed cohorts (DL1 and DL2). Conclusion: BTX-9341 monotherapy shows a favorable safety profile, with the most common AEs being decreased neutrophil and white blood cell counts, at doses that show encouraging preliminary PK/PD and efficacy, enabling further evaluation of monotherapy and in combination with fulvestrant. The trial is actively enrolling with anticipated completion of dose escalation enrollment by the end of 2025 (Clinical trial: NCT06515470).
Presentation numberPS4-06-05
Sapu003: Everolimus for Injection — Pharmacokinetic Rationale for Phase I Evaluation in HR⁺/HER2⁻ Metastatic Breast Cancer
Wen-Han Chang, Sapu Bioscience, LLC, San Diego, CA
W. Chang1, E. Olivar1, G. Pai1, J. Low1, S. Min2, R. Hoff2, N. Chang3, T. Hoque3, A. Park3, C. Lee4; 1QC, Sapu Bioscience, LLC, San Diego, CA, 2Formulation, Sapu Bioscience, LLC, San Diego, CA, 3QA, Sapu Bioscience, LLC, San Diego, CA, 4R&D, Sapu Bioscience, LLC, San Diego, CA.
Background Oral everolimus (Afinitor) is an established therapy for hormone receptor-positive, HER2-negative breast cancer. However, its clinical utility is limited by low and variable oral bioavailability (~15%), gastrointestinal toxicity, and a narrow therapeutic window driven by first-pass gut extraction and CYP3A/P-gp interactions. Sapu003 is an intravenous Deciparticles™ formulation of everolimus designed to circumvent intestinal loss and toxicity while delivering consistent systemic exposure. Methods Pharmacokinetics (PK), tissue distribution, metabolism, and excretion of Sapu003 were characterized after a single 3 mg/kg IV bolus in Sprague-Dawley rats and a 0.6 mg/kg 30-minute IV infusion in beagle dogs, with oral everolimus (1-3 mg/kg) used as a comparator. Whole blood, organs, feces, and urine were quantified for everolimus by a validated LC-MS/MS method. In vitro plasma, microsome, and hepatocyte stability were assessed across multiple species, including mouse, rat, dog, monkey, and human. Results In rats, IV Sapu003 achieved a mean Cmax of 133 ng/mL and an AUC₀-∞ of 339 h·ng/mL, compared with 2.4 ng/mL and 52 h·ng/mL after a 3 mg/kg oral PEG-400 everolimus suspension (representing 55- and 6.5-fold increases, respectively). In dogs, the IV infusion yielded a Cmax of 51-76 ng/mL and an AUC₀-∞ of 203-395 h·ng/mL, compared with 8.4 ng/mL and 94-123 h·ng/mL after 1 mg/kg oral Afinitor (representing 6-9- and 2-4-fold increases, respectively), while maintaining comparable half-lives. Plasma, microsome, and hepatocyte stabilities were unchanged relative to everolimus. Sapu003 rapidly distributed to high-flow organs but, unlike oral everolimus, showed low deposition in the gastrointestinal tract. Elimination remained primarily biliary/fecal (>94% of the dose, completed within ≤48 h), with renal clearance accounting for <2%. No intestinal toxicity was observed in dogs administered Sapu003. Conclusions IV administration of Sapu003 overcomes the absorption bottleneck of oral everolimus, providing predictable exposure with a controllable Cmax while preserving the native metabolic fate. By bypassing the GI tract, Sapu003 exhibits consistent blood PK with no intestinal toxicity. These data support the ongoing Phase I evaluation of weekly 30-minute infusions of Sapu003 in metastatic breast cancer, aiming to enhance mTOR pathway suppression, including effects on mTORC2 targets.
Presentation numberPS4-06-06
Tfx06 dose expansion study showed favorable safety and efficacy in patients with er<sup>+</sup> /her2<sup>-</sup> breast cancer in a phase 2 study
Charles Z Ding, Yangli Pharmaceutical Technology, Hangzhou, China
Y. Chen1, J. Zhang1, H. Guo2, Y. Sun3, Y. Wang4, Z. Zhang5, T. Yao6, Y. Shi7, H. Zhang8, Y. Chen9, Y. Lu10, J. Song10, P. Li10, D. Fang10, W. Sha10, C. Z. Ding10; 1Breast Cancer, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 2Breast Cancer, Henan Cancer Hospital, Zhengzhou, CHINA, 3Breast Cancer, Shandong Cancer Hospital and Institute, Jinan, CHINA, 4Medical, Shandong Cancer Hospital and Institute, Jinan, CHINA, 5Breast Cancer, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, CHINA, 6Breast Cancer, Hospital of Bengbu Medical College, Bengbu Anhui, CHINA, 7Breast Cancer, Tianjin Clinical Research Center for Cancer, Tianjin, CHINA, 8Breast Cancer, Nanyang Central Hospital, Nanyang Henan, CHINA, 9Breast Cancer, Hospital of Zhejiang University School of Medicine, Hangzhou, CHINA, 10Medical, Yangli Pharmaceutical Technology, Hangzhou, CHINA.
Background: TFX06, a third-generation oral CERAN/SERD, was evaluated in a multicenter Phase 1a/b study (NCT05927779) in patients with ER+/HER2- locally advanced and/or metastatic breast cancer (Zhang J, et al., 2024 SABCS annual meeting). The recommended dose was established at 150mg QD. Dose expansion study at 150mg had been initiated, and this abstract summarizes our key findings. Patients and Methods: Eligible patients regardless of their menopausal status, who had received ≥1 lines of endocrine therapy (ET), prior treatment with CDK4/6i and fulvestrant (fulv); and ≤2 line of chemotherapy for metastatic disease were allowed to enroll. TFX06 tablets were orally administered 150mg once daily (QD) every 28 days of treatment cycle until disease progression or intolerable toxicity. As of cut-off date June 30, 2025, 57 patients received TFX06 monotherapy at 150mg dose, median age of 60 years (range 34-74) with ECOG score of 0-1. 78.9% of patients enrolled had visceral metastasis in liver, lung or both, and 14% of patients had brain metastasis. Median number of prior endocrine therapies were 2 (1-4) including fulvestrant (56.1%), CDK4/6i (61.4%) and combination of both (38.6%) and a median number of chemotherapies was 1 (0-3). Tumor assessments by investigators according to RECIST v1.1 were performed every eight weeks. Results: TFX06 was well tolerated. The most common (≥20% of patients) treatment-emergent adverse events (TEAEs) were hypertriglyceridemia, AST increased, anemia, lactate dehydrogenase increased and hypoalbuminaemia. The incidence of nausea, diarrhea and vomiting was low (7.0%, 5.3% and 1.8%, respectively). In addition, no ocular toxicity reported. Among 55 evaluable patients, there were 6 partial responses (ORR was 10.9%); clinical benefit rate (CBR) was 45.5% and mPFS for the overall population was 5.5 months. mPFS for ESR1m detectable and ESR1m-undetectable patients were 5.7 months and 4.6 months respectively. In addition, among the 13 patients who had prior CDK4/6i treatment and with detectable ESR1 mutation, the CBR was 46.2% and mPFS was 5.6 months. Among 21 patients who had prior fulvestrant+CDK4/6i combo treatment, CBR was 42.9% and mPFS was 3.7 months, while ESR1 detectable mutation patients, CBR was 55.6% and mPFS was 5.6 months. Notably, of four patients with measurable brain lesions, three were evaluable: two showed >70% brain lesion reduction (PR), one had stable disease, suggesting TFX06 had intracranial activity. Conclusions: TFX06 at 150 mg QD dose expansion trial enrolled endocrine therapy heavily treated patients, including prior treatment with combination of fulvestrant and CDK4/6 inhibitor progressed patients. TFX06 demonstrated favorable safety and clinical efficacy even in this hard-to-treat patient population.
Presentation numberPS4-06-07
Yes/src kinase inhibitor nxp900, currently in clinical development, combines synergistically with fulvestrant against luminal a breast cancer cell lines in vitro and in vivo
Asier Unciti-Broceta, University of Edinburgh, Edinburgh, United Kingdom
I. Lanzagorta-Calvillo1, B. Kaghazchi1, E. Poradosu2, N. O. Carragher1, A. Unciti-Broceta1; 1Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UNITED KINGDOM, 2R&D, Nuvectis Pharma Inc., Fort Lee, NY.
Background. Most breast cancer patients present tumors with high estrogen receptor levels (ER+), which make endocrine therapies (ET) the standard of care for this breast cancer subtype. However, resistance to treatment inevitably develops over time, which demands optimal combination strategies. The combination of CDK4/6 inhibitors with ET has emerged as the foremost therapeutic modality for hormone receptor-positive/HER2-negative advanced breast cancer. However, resistance to these therapies eventually develops with the clinical benefit of subsequent lines of therapy being limited and short lasting. Deregulation of the Hippo pathway leading to YAP1 nuclear translocation has been shown to promote resistance to CDK4/6 inhibitors. NXP900 (eCF506), is a novel, highly potent, YES/SRC kinase inhibitor with high selectivity that uniquely binds the kinase in the autoinhibited closed conformation, thereby blocking its catalytic and scaffolding properties [1]. Preclinical studies have demonstrated that NXP900 effectively inhibits YAP1 nuclear translocation and proliferation of FAT1 mutated cells [2]. In addition, NXP900 demonstrated potent inhibition of cell proliferation of induced endocrine resistance in vitro [3], thus, establishing the hypothesis that NXP900 may represent a therapeutic option for the treatment of endocrine and/or CDK4/6 resistance breast cancer. The aims of this project were to (i) investigate the sensitivity of luminal A cell lines to NXP900 as a single agent, and (ii) assessing NXP900’s efficacy in combination with endocrine therapy in vitro and in vivo in comparison to current approved combinations of endocrine therapy and CDK4/6 inhibitors. Methods. Cell proliferation and viability were determined by automated image analysis of confluence and Presto Blue assay, respectively. Dose response studies were performed for NXP900, dasatinib and fulvestrant in a library of ER+ luminal A cell lines. Western blot analysis was performed to study target inhibition. Dose-ratio combinations of; NXP900 and fulvestrant; dasatinib and fulvestrant, and CDK4/6 inhibitors and fulvestrant, were used to compare the effect of each combination. In vivo xenograft studies were performed for the combination of fulvestrant and NXP900. Results. NXP900 showed higher antiproliferative activity than dasatinib against the luminal A breast cancer cell lines tested. Notably, in these cell lines NXP900 demonstrated far superior SRC/YES inhibition than dasatinib, correlating with the cancer cell viability data. Dose ratio matrix drug combination analysis using SynergyFinder+ demonstrated significantly stronger synergistic interactions in ER+ cell lines treated with endocrine therapy in combination with NXP900, rather than in combination with dasatinib or CDK4/6 inhibitors. Conclusions. Our results demonstrate that luminal A cancer cell lines are generally sensitive to NXP900 treatment and that in combination with endocrine therapy it is more synergistic than the standard-of-care combination of fulvestrant and CDK4/6 inhibitors. This study supports the potential translation of NXP900 into breast cancer patients alongside endocrine therapies to improve the treatment of ER+ tumors. References [1] Temps et al. A Conformation Selective Mode of Inhibiting SRC Improves Drug Efficacy and Tolerability. Cancer Res. 2021, 81 (21), 5438-5450. [2] Kaghazchi et al. NXP900, a novel YES1/SRC kinase inhibitor currently in clinical development, blocks YAP1 signaling in NSCLC cell lines. Cancer Res. 2025, 85 (8, Suppl_1), 5599. [3] Lanzagorta-Calvillo et al. Endocrine therapy-resistant luminal A breast cancer cell lines are sensitive to the novel YES1/SRC tyrosine kinase inhibitor, NXP900. Cancer Res. 2025, 85 (8, Suppl_2), LB292.
Presentation numberPS4-06-08
A phase 1/2, first-in-human study of AVZO-021, a selective cyclin-dependent kinase 2 inhibitor (CDK2i), as a monotherapy and in combination for patients with advanced solid tumors, including hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer and cyclin E1 (CCNE1)‑amplified solid tumors: preliminary safety and efficacy results
Alberto J Montero, University Hospitals/Seidman Cancer Center, Case Western Reserve University, Cleveland, OH
A. J. Montero1, M. R. Patel2, A. I. Spira3, B. Bashir4, D. L. Richardson5, S. Thamake6, D. Trone6, B. Veresh6, P. LoRusso7; 1Department of Hematology and Oncology, University Hospitals/Seidman Cancer Center, Case Western Reserve University, Cleveland, OH, 2Drug Development, Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, FL, 3Medical Oncology, Clinical Research, NEXT Oncology-Virginia, Fairfax, VA, 4Department of Medical Oncology, Division of Solid Tumors, Sidney Kimmel Comprehensive Cancer Center at Thomas Jefferson University, Philadelphia, PA, 5Section of Gynecologic Oncology, Stephenson Cancer Center/Sarah Cannon Research Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 6Avenzo Therapeutics, Avenzo Therapeutics, San Diego, CA, 7Medical Oncology and Hematology, Yale Medicine, New Haven, CT.
Background: Resistance to CDK4/6 inhibitors (CDK4/6i) in HR+/ HER2− metastatic breast cancer (mBC) is frequently associated with CCNE1 overexpression and CDK2 activation. Targeting CDK2 may overcome this resistance and enhance the efficacy of CDK4/6i-based regimens. AVZO-021 is a potent CDK2i with high selectivity for CDK2 over CDK1, thus minimizing off-target toxicity. We present preliminary results from the Phase 1 segment of the ongoing first-in-human Phase 1/2 study of AVZO-021 (NCT05867251). Methods: The ongoing Phase 1 open-label, dose-escalation segment of this study is evaluating the safety, pharmacokinetics (PK), and preliminary efficacy of AVZO-021 alone and in combination with endocrine therapy [(ET) fulvestrant], CDK4/6i (ribociclib/abemaciclib) + ET (fulvestrant/letrozole), an antibody-drug conjugate (sacituzumab govitecan-hziy), or chemotherapy (carboplatin) in patients with advanced solid tumors, including HR+/HER2− mBC and CCNE1-amplified solid tumors. AVZO-021 is administered orally once daily (QD) across escalating dose levels (20 to 220 mg in monotherapy dose escalation). Results: As of May 14, 2025, 47 patients have received at least 1 dose of AVZO-021 (27 in monotherapy; 20 in combination). The median age was 63 years for patients who received AVZO‑021 in monotherapy and 59 years for patients who received AVZO‑021 in combination with ET ± CDK4/6i, sacituzumab govitecan-hziy, or chemotherapy. Overall, 96% of patients were female, 68% had breast cancer, 15% had ovarian/fallopian tube cancer, and 6% had endometrial/uterine cancer. Patients who received AVZO-021 in monotherapy had a median of 4 prior lines of systemic therapy, and those treated with combination therapy had a median of 6 prior lines. All patients with HR+/HER2− mBC (n=30) had received prior CDK4/6i and hormonal therapy. No dose-limiting toxicities (DLTs) or treatment discontinuations due to adverse events (AEs) were observed with AVZO-021 monotherapy up to a dose of 220 mg. The most common (>25%) all-grade treatment-emergent AEs (TEAEs) with monotherapy were fatigue (41%), nausea (37%), anemia (26%), and vomiting (26%); 2 patients required dose reductions due to AEs (Grade 2 thrombocytopenia [n=1, 180 mg dose level]; Grade 2 worsening fatigue [n=1, 120 mg dose level]). No new safety signals or DLTs were reported in the combination cohorts. AVZO-021 monotherapy demonstrated a favorable PK profile with a low peak-to-trough ratio, robust target coverage, and mean half-life of 14 hours. Updated clinical data will be presented at the conference, including ctDNA analysis and preliminary efficacy of monotherapy and combination therapy in HR+/HER2− mBC and CCNE1-amplified solid tumors. Conclusions: AVZO-021 is a novel CDK2i with high selectivity for CDK2 over CDK1 and a favorable preliminary clinical profile. Together with the mechanistic rationale, the emerging clinical data support the continued development of AVZO-021 as a monotherapy and in combinations, including with ET ± CDK4/6i and selective CDK4i.
Presentation numberPS4-06-09
Efficacy of a novel BCL-xL degrader, DT2216, in the treatment of triple-negative breast cancer
Yuxiang Lin, Baylor College of Medicine, Houston, TX
Y. Lin1, H. Sang1, A. Ku1, D. Thomas1, L. Jia1, A. Talab1, D. Zhou2, B. Lim3, Y. Li1; 1Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 2Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 3Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Triple-negative breast cancer (TNBC) accounts for approximately 10-15% of all breast cancer cases and has a poorer prognosis compared to other breast cancer subtypes. However, there are only limited therapy options for TNBC. The anti-apoptotic protein BCL-xL, a member of the BCL-2 family, has been implicated in promoting tumor cell survival and resistance to chemotherapeutic agents. However, clinical application of current small-molecule BCL-xL inhibitors has been hindered by dose-limiting toxicities, particularly thrombocytopenia resulting from on-target effects in platelets. DT2216 is a novel proteolysis-targeting chimera (PROTAC) designed to degrade BCL-xL in tumor cells while sparing platelets. Here, we aim to evaluate the potential use of DT2216 in the treatment of TNBC patients. Bioinformatics analysis revealed that the BCL-xL gene is highly expressed in TNBC and associated with poor clinical outcomes in TNBC patients. Consistently, BCL-xL expression was also elevated in TNBC cell lines relative to non-TNBC subtypes. BCL-xL knockdown significantly reduced viability of TNBC cell lines (MDA-MB-231 and HS578T) while showing little impact in estrogen receptor (ER)-positive breast cancer cell lines (MCF-7 and T47D), indicating a subtype-specific dependency on BCL-xL. In accordance with these genetic findings, DT2216 markedly inhibited growth of TNBC cell lines while exhibiting minimal effects on ER+ breast cancer cell lines. We further conducted a comprehensive evaluation of the combined effects of DT2216 and standard chemotherapy in TNBC models. DT2216 significantly potentiated the cytotoxic effects of two commonly used chemotherapeutic agents (paclitaxel and carboplatin) across a range of concentrations, producing a robust synergistic inhibition of TNBC cell proliferation. In vivo experiments validating these in vitro data are ongoing. Together, these data indicate that the selective degradation of BCL-xL by DT2216, either as monotherapy or in combination with standard-of-care chemotherapy, could offer a novel therapeutic strategy to improve outcomes for patients with TNBC.
Presentation numberPS4-06-10
A Promising Approach for Hard-to-Treat Breast Cancer: Photodynamic Therapy with ClAlPc Nanoemulsion Against Triple-Negative Cells
Elisângella de Paula Silveira Lacerda, Federal University of Goiás, Goiânia, Brazil
H. R. Luz1, L. P. Franchi2, A. C. Tedesco3, E. d. Lacerda4; 1Genetics, Federal University of Goiás, Goiânia, BRAZIL, 2Institute of Biological Sciences, Federal University of Goiás, Goiânia, BRAZIL, 3Department of Chemistry, University of São Paulo, Ribeirão Preto, BRAZIL, 4Center for Human Genetics, Institute of Biological Sciences, Federal University of Goiás, Goiânia, BRAZIL.
Triple-negative breast cancer is a highly aggressive subtype with the poorest prognosis among breast cancers. It is defined by the absence of estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER2), limiting the effectiveness of conventional hormone-based therapies. Around 20% of the 2.3 million annual breast cancer cases are triple-negative, and this subtype is linked to high metastasis and mortality rates. Its molecular heterogeneity and therapeutic resistance highlight the urgent need for alternative treatment strategies. Photodynamic therapy is a promising, minimally invasive technique already employed in Brazil’s public health system (SUS) for the treatment of basal cell carcinoma. It relies on the activation of a photosensitizer (PS) by a specific light source, leading to the generation of reactive oxygen species (ROS) that trigger oxidative stress, cell death, and immune response activation. This in vitro study assessed the cytotoxicity of photodynamic therapy using a nanoemulsion containing chloroaluminum phthalocyanine (ClAlPc/Ne) in breast cancer cell lines exposed to varying PS concentrations and light doses. The MCF-7 and MDA-MB-231 cell lines, representing luminal A and triple-negative breast cancer respectively, were incubated with ClAlPc/Ne for 24 hours before photoactivation, followed by a further 24-hour incubation before metabolic viability analysis using the MTT assay. Three experimental conditions were tested: (I) nanoemulsion without PS, (II) ClAlPc/Ne without irradiation, and (III) photoactivated ClAlPc/Ne. PS concentrations ranged from 0.01 µM to 2.0 µM, with light doses from 1 J/cm² to 16 J/cm². Absorbance was measured at 545 nm. Results showed selective phototoxicity of ClAlPc/Ne against the triple-negative breast cancer cell line MDA-MB-231, with IC50 (50% inhibitory concentration) values of 1.96 µM at 1 J/cm², 0.19 µM at 4 J/cm², and 2.0 µM at 1 J/cm², 0.14 µM at 4 J/cm², and <0.06 µM at 16 J/cm². These findings indicate that phototoxicity is dose-dependent and more pronounced in triple-negative breast cancer cells. The differential response between cell lines suggests that triple-negative breast cancer may be more sensitive to oxidative stress induced by photodynamic therapy. These findings reinforce the importance of molecular profiling in guiding treatment decisions and support the potential of photodynamic therapy as a targeted, less invasive approach for hard-to-treat breast cancer subtypes.
Presentation numberPS4-06-11
Initial Results from Correlative Analysis of a Single Arm Phase 2 Study of Peri-Operative Immune Checkpoint Inhibition and Cryoablation in Women with Hormone Receptor-Negative, HER2-Negative Early Stage/Resectable Breast Cancer
Shaan Bhandari, UT Southwestern Medical Center, Dallas, TX
S. Bhandari, A. Alhalwani, I. Terrazas, R. Sridharan, S. Rice, H. L. McArthur, I. S. Chan; Internal Medicine, UT Southwestern Medical Center, Dallas, TX.
Background: Triple negative breast cancer (TNBC) is a biologically distinct subtype with a high risk of early relapse, particularly in patients with residual disease following neoadjuvant chemotherapy (NAC). Failure to achieve a complete response may reflect underlying immune evasion mechanisms, including upregulation of immune checkpoints such as programmed cell death protein 1 (PD-1), which impair effective T cell-mediated antitumor responses. Cryoablation (cryo) can stimulate T-cell priming and reduce MDSCs to overcome resistance to immune checkpoint inhibition (ICI). Preclinical and early clinical data suggest that the combination of cryo with ICI may synergistically improve immune activation. We conducted a multi-center, single arm, phase 2 study to evaluate the impact of cryo with standard-of-care pembrolizumab (pembro) in women with residual TNBC after taxane-based neoadjuvant chemotherapy (NAC), a group at high risk of early relapse (NCT03546686). Methods: Eligible women are ≥18y, with ER < 10%, PR < 10%, HER2 negative (per ASCO/CAP definition), ≥ 1.0 cm, residual operable disease after taxane-based NAC. Patients underwent percutaneous, image-guided cryo with concurrent research core biopsy 7-10 days prior to surgery and receive peri-operative pembro per standard-of-care. Peripheral blood was collected pre- and post-cryo, and flow cytometry was performed on paired peripheral blood mononuclear cells (PBMCs) from an initial cohort (n=5) to identify changes in immune response. Results: Post-cryo, we observed an overall trend in the decrease of PD1-positive T cells and myeloid cells. PD1+CD8+ T cells decreased by 2.9-fold, PD1+CD4+ T cells decreased by 4.6-fold and PD1+CD3- decreased by 6.3-fold. Notably, we observed a decrease in CD11b+CD3- MDSCs by 14.92-fold. Additional exploratory correlative studies on tumor and blood, including tumor infiltrating lymphocytes and tumor PD-1 IHC, are ongoing to fully characterize the immunologic impact of the intervention and to explore predictors of efficacy. Conclusion: These initial results suggest that the addition of cryo to pembro may act synergistically to reduce the overall immunosuppressive immune environment, potentially resulting in improved immunity in patients with high-risk residual TNBC. Funding sources: Susan G. Komen, B-SEARCH, Conquer Cancer Foundation, Breast Cancer Research Foundation, Boston Scientific.
Presentation numberPS4-06-12
Dual-payload TROP2 targeted antibody drug conjugate: Multi-Payload Conjugates™ deliver orthogonal mechanisms of cell killing
Marco Lobba, CatenaBio, Berkeley, CA
M. Lobba1, D. Trinter1, M. Nguyen1, S. Brady1, C. Symister1, A. Lau1, D. Garcia-Almedina1, S. Johri1, F. Matthew2, R. Kendall1; 1Research and Development, CatenaBio, Berkeley, CA, 2Dept. of Chemistry, University of California at Berkeley, Berkeley, CA.
Introduction: Antibody-Drug Conjugates (ADCs) have a tremendous impact on patient outcomes in breast and other cancers. Dato-DXd, for example, is second-line therapy for stage IV, HR positive/HER2 negative metastatic breast cancer. Sacituzumab govitecan (SG), a TROP2 targeted ADC, is approved as a 3rd line therapy in locally advanced or metastatic HER2 negative breast cancer, and in combination with pembrolizumab, is likely to see approval in 1st line therapy for PD-L1 positive tumors. However, many patients fail to respond or relapse after treatment with ADC therapies due to tumor heterogeneity and eventual resistance to the ADC payload. Combination therapies have historically outperformed mono chemo approaches across most solid tumors to date, pointing to a potential avenue for improvement of ADC efficacy. We are developing next generation Multi-Payload Conjugates™ (MPCs™) that deliver targeted combination chemotherapies within a single molecule to address shortcomings in current ADCs. Method: CatenaBio has developed highly stable, dual-payload ADC combination therapies, with tunable payload ratios. Our selective conjugation platform allows the attachment of distinct payloads targeting different mechanisms of action at three unique sites on antibody scaffolds. Results: Catena’s lead TROP2 targeting MPC, CATB-101, features an optimized combination and ratio of tubulin and Topo1 inhibitors. CATB-101 demonstrates superior inhibition and excellent tolerability in mouse models of tumor growth in TROP2 expressing xenograft models including multiple CDX and PDX models of TNBC (triple negative breast cancer). In head-to-head comparisons with T-DXd, SG and Dato-DXd, MPCs out-perform the current approved ADCs in breast cancer and demonstrate full tumor elimination at low mg/kg doses. Furthermore, CATB-101 rescues CDX models after progression on SG treatment, demonstrating potential applications in late-line treatment. Non-GLP Non-Human Primate (NHP) toxicology trials with CATB101 demonstrate remarkable safety profile, surpassing benchmark mono-payload ADCs in tolerability. Conclusion: While advances have been made in the design of ADCs to expand to previously unaddressed populations, high patient relapse and the failure of recent mono-payload ADCs in late-stage trials indicate a need for novel multi-payload conjugates. Catena’s MPCs™ offer the next step in ADC design and allow for the targeted delivery of multiple mechanisms of action with a single MPC™ are highly efficacious at eliminating tumors across multiple CDX and PDX xenograft models with a range of target expression. Now validated in early NHP toxicology studies with a significantly enhanced therapeutic window, these molecules offer the potential to circumvent tumor resistance pathways to deliver deeper and more durable patient responses.
Presentation numberPS4-06-13
Pharmacokinetic Evaluation of MBQ-167 a Dual Rac/Cdc42 Inhibitor in Advanced Breast Cancer Patients
Jose F Rodriguez-Orengo, MBQ Pharma, Inc., San Juan, PR
J. F. Rodriguez-Orengo1, J. Duconge2, M. Acosta3, J. Wang4, D. Yardley5, S. Dharmawardhane6, N. Sankar1; 1Cancer Drug Development, MBQ Pharma, Inc., San Juan, PR, 2Pharmaceutical Sciences, University of Puerto Rico Medical Sciences Campus, San Juan, PR, 3Oncology, FDI Clinical Research, San Juan, PR, 4Cancer Treatment, Florida cancer Specialists and Research Institute, Tallahassee, FL, 5Oncology Therapy, SCRI Oncology Partners, Nashville, TN, 6Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, PR.
MBQ-167 is a first-in-class dual inhibitor of Rac and Cdc42 GTPases with potent preclinical anti-metastatic activity and is currently under evaluation in a Phase 1 clinical trial (NCT06075810) in patients with advanced breast cancer. This study aimed to characterize the pharmacokinetic (PK) profile of MBQ-167 and its active metabolite M6 in patients treated with 20 mg, 40 mg and 80 mg BID doses (cohorts 0, 1 and 2, respectively). Plasma concentration-time data were analyzed from eight patients using non-compartmental analysis. MBQ-167 was rapidly absorbed with a median Tmax of 1–4 hours and a Cmax ranging from 139 to 573 ng/mL (20 mg BID), 520 to 843 ng/mL (40 mg BID) and 578 to 688 ng/mL (80 mg BID). The elimination half-life (t½) of MBQ-167 was approximately 3–4.5 hours, and the mean residence time (MRT) was <7 hours. Dose-normalized systemic exposure (AUC/D) did not increase proportionally with dose, suggesting a possible reduction in bioavailability at higher doses, potentially due to enterohepatic recycling or saturation of absorption. Importantly, there was no evidence of systemic overexposure or meaningful accumulation (Rac < 2.0) of MBQ-167 or M6, indicating a favorable safety margin. Volume of distribution (Vdss) exceeded 30L for both compounds, reflecting extensive tissue distribution consistent with their lipophilicity. M6 demonstrated rapid formation and elimination with t½ values ranging from 3.1 to 4.4 hours, further supporting efficient metabolic conversion. These findings confirm that MBQ-167 exhibits linear PK at clinically relevant doses with manageable exposure and clearance. The observed pharmacokinetics support continued clinical development and provided essential dosing guidance for Phase 2 trials. Further analyses with additional cohorts will be presented up to 400 mg BID.
Presentation numberPS4-06-14
Tinengotinib in advanced or metastatic HR+/HER2- and TNBC: efficacy and biomarker correlative analysis from a phase Ib/II study
Sarina Piha-Paul, MD Anderson Cancer Center, Houston, TX
S. Piha-Paul1, S. Lavasani2, F. Dayyani3, S. Goel4, A. Tsimberidou1, N. Ibrahim1, C. Barcenas1, A. Ilheme1, J. Rodon1, E. E. Dumbrava1, K. Hennessy5, P. Peng6, C. Sun7, H. Wang7, R. Xu7, J. Fan5, F. Meric-Bernstam1; 1Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX, 2Medical Oncology, University of California, Irvine, Orange, CA, 3Medical Oncology, City of Hope National Medical Center, Duarte, CA, 4Oncology, Rutgers Cancer Center Institute of New Jersey, New Brunswick, NJ, 5Clinical Operations, TransThera Sciences (US), Inc, Gaithersburg, MD, 6Preclinical, TransThera Sciences (Nanjing), Inc., Nanjing, CHINA, 7Clinical Operations, TransThera Sciences (Nanjing), Inc., Nanjing, CHINA.
Background: Patients (pts) with advanced/metastatic hormone receptor positive /human epidermal growth factor receptor 2 negative (HR+/HER2-) breast cancer (BC) often develop resistance to endocrine therapy and CDK4/6 inhibitors, while those with triple-negative BC (TNBC) usually lack effective targeted therapies after progression on chemotherapy or antibody drug conjugates. Tinengotinib, a novel multi-kinase inhibitor targeting FGFRs, VEGFRs, Aurora A/B, JAK1/2, and CSF1R, demonstrated preclinical activity in BC models, including those resistant to standard therapies. Here we present the final efficacy, safety and correlative biomarker data of tinengotinib monotherapy in pts with HR+/HER2- and TNBC from the Phase Ib/II trial TT420X1103 (NCT04742959).Methods: Eligible pts with HR+/HER2- BC or TNBC with no effective standard therapeutic treatment options were enrolled. TNBC pts included acquired TNBC (aTNBC) disease, defined as pts initially diagnosed with HR+/HER2- BC but found to be transformed to TNBC at study entry. Tinengotinib was given as monotherapy at 10 mg or 12 mg daily dose (QD) dose levels, or 6 mg twice daily (BID) dose level . Response was assessed per RECIST v1.1, and safety was evaluated per CTCAE v5.0. Biomarker sampling for circulating tumor DNA (ctDNA) analysis was completed at baseline, cycle 3 day 1 (C3D1), the time of response, the time of progression and end of treatment (EOT). Results: As of 20Feb2024, 21 pts with advanced/metastatic BC were treated with tinengotinib monotherapy at 10 mg QD (n=3), 12 mg QD (n=16), and 6 mg BID (n=2) dose levels. All pts were female; median age 58 years (IQR 48-63), ECOG PS 1 in 100% of pts, 18 (85.7%) of pts had Stage IV disease, and 18 (85.7%) of pts had ≥3 lines of prior systemic therapy.Of 20 efficacy evaluable pts, objective response rate (ORR) was 30% (n = 6), duration of response was 4.81 months (mos) (95% CI 3.15, 6.01) and disease control rate (DCR) was 75% (n=16). Treatment related adverse events (TRAEs) were reported in 20 (95.2%) pts, 10 (47.6%) pts had G3/4 TRAEs, and no G5 TRAEs were reported. Eighteen pts were evaluable for ctDNA analysis. Among the most frequent alterations (TP53, DNMT3A, APC, PIK3CA, FGFR2 and PTEN), the incidence of ROS1 mutations was higher in pts with partial response (PR) (p=0.03), as well as in pts with PFS>3.5 mos (p=0.02). Of these 18 pts, 8 had TNBC and 6 had aTNBC. It was observed that pts with aTNBC had a longer PFS vs TNBC pts (4.3 vs 2.6 months, p=0.011); ORR was 67% vs 0% and DCR was 100% vs 50%. The incidence of ROS1 and DNMT3A mutations was higher in aTNBC vs TNBC (p=0.015 and p=0.026, respectively). Analysis showed a reduction in variant allele frequency (VAF) of 7 pts from baseline to C3D1 (p<0.0001), indicating a molecular response with treatment. One pt with progressive disease showed increased VAF in mutations of PIK3CA, PTEN, TP53, and RAD51D from baseline to EOT; 3 pts with acquired resistance to the therapy detected new gene mutations (DNMT3A, NF1, CTNNA1, ERBB3, MLL2, CDKN2A and PDGFRB) at EOT. Conclusions: Tinengotinib exhibited promising clinical benefit and manageable safety profile for the treatment of HR+/HER2- BC or TNBC. The biomarker correlative analysis revealed that ROS1 mutations could be a potential predictive biomarker. Early reduction in ctDNA VAF may serve as a pharmacodynamic biomarker. Baseline genomic alterations in PIK3CA, PTEN, TP53, and RAD51D may contribute to primary resistance, while acquired resistance may be associated with emergence of mutations in pathways of RAS/MAPK, PI3K/AKT, RTK, MET, and cell cycle regulation. The improved clinical outcomes and enrichment of ROS1/DNMT3A mutations in aTNBC pts define this transformed subtype as a subgroup warranting further investigation.
Presentation numberPS4-06-15
Safety Run-In Phase of the ATRiBRAVE trial: a Phase II Study Evaluating Ceralasertib Priming Followed by Durvalumab/Nab-Paclitaxel to Restore Immunotherapy Sensitivity in Advanced Triple Negative Breast Cancer (TNBC)
Valentina Guarneri, Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
V. Guarneri1, G. Griguolo2, C. Vernieri3, M. Dieci1, Y. Silvestri4, F. Giovanardi5, F. Ligorio3, G. Fotia6, S. Rapisarda4, A. Chirco7, A. Zambelli8, M. De Laurentiis9, A. Gennari10, S. Marsoni11, S. Piccolo12, M. Foiani13; 1Department of Surgery, Oncology and Gastroenterology – University of Padova, Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, ITALY, 2Department of Surgery, Oncology and Gastroenterology – University of Padova, Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, ITALY, 3Medical Oncology Department – Fondazione IRCCS Istituto Nazionale dei Tumori, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, ITALY, 4Precision Oncology Unit, IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, ITALY, 5Medical Oncology, Comprehensive Cancer Centre, AUSL-IRCS di Reggio Emilia, Reggio Emilia, ITALY, 6Medical Oncology Department – Fondazione IRCCS Istituto Nazionale dei Tumori, Oncology and Hematology-Oncology Department, University of Milan, Milan, ITALY, 7Medical Oncology Unit, ASST Papa Giovanni XXIII, Bergamo, ITALY, 8Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, ITALY, 9Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, ITALY, 10Department of Translational Medicine – University of Piemonte Orientale, Novara, IT, Division of Medical Oncology, Maggiore University Hospital, Novara, ITALY, 11Precision Oncology Unit, IFOM ETS, the AIRC Institute of Molecular Oncology, Padova, ITALY, 12Department of Molecular Medicine, University of Padova, IFOM ETS, the AIRC Institute of Molecular Oncology, Padova, ITALY, 13Istituto di Genetica Molecolare, CNR, IFOM ETS, the AIRC Institute of Molecular Oncology, Pavia, ITALY.
Background: Preclinical studies have reported that Ataxia telangiectasia and Rad3-related (ATR) inhibition results in increased nuclear envelope fragility and promotion of cGAS-STING activation, thus potentially resulting in an enhanced immune response. The phase II investigator-initiated ATRiBRAVE trial evaluates the efficacy of the ATR inhibitor ceralasertib priming followed by durvalumab-nab-paclitaxel in patients with TNBC relapsing after immune checkpoint inhibitors (ICIs) and chemotherapy (CT) treatment in the curative setting. This report presents results from the safety run-in phase (SRP) of the trial, representing the first assessment of the safety and feasibility of the triple combination of ceralasertib, durvalumab and nab-paclitaxel in human. Methods: This single-arm trial enrolls patients with unresectable/metastatic TNBC (ER<10% HER2-), previously treated with ICIs and CT in the early setting (adjuvant, neoadjuvant or both) and not previously treated in the metastatic setting. Treatment consists of: Ceralasertib 240mg orally BID (DL 0) for 7 days before cycle 1, then days 22-28 of each cycle (with planned dose de-escalation if needed), Durvalumab 1500 mg IV, day 1 (q28), Nab-paclitaxel 100mg/m2 IV, days 1, 8, 15 (q28). Treatment continues until disease progression or unacceptable toxicity. Primary endpoint of the study: Progression-free survival (PFS). Study sample size: 37 patients (34 evaluable patients + 10% drop-out) SRP design: a SRP with a 3+3 de-escalation schema was preplanned in the study to assess safety. If fewer than 2 dose-limiting toxicities (DLTs) occur in the first 6 patients at DL 0, SRP is successfully completed. Otherwise, ceralasertib will be de-escalated to 160 mg BID (DL-1) and, if necessary, to 120 mg BID (DL-2). Results: Up to date, 18 patients have been enrolled in the study, 9 of which were treated in the SRP (3+6). Among the first 3 patients, 2 DLTs (prolonged neutropenia leading to significant dose omissions) were observed, prompting ceralasertib reduction to 160 mg BID (DL-1). No DLTs were observed in the first 6 (3+3) patients at DL-1 (ceralasertib 160 mg BID for 7 days), thus successfully completing SRP. Among patients treated in the SRP, no grade 5 AEs occurred. All grade 3-4 AEs observed were hematologic but did not qualify as DLTs. No patients discontinued treatment due to toxicity during SRP. Durvalumab safety profile was consistent with previous reports. Conclusions: Ceralasertib priming at a 160 mg BID for 7 days, followed by durvalumab and nab-paclitaxel, is feasible and well tolerated. Nine additional patients have already been enrolled in the phase II portion of the trial (total enrolment: 18 patients) and patient enrolment is currently ongoing. Updated safety data for patients who completed treatment in the study will be presented at the meeting. This study was funded by Fondazione AIRC under 5 per mille 2019 (ID. 22759 program—group leader VG and SM). This study was conducted with the unconditional support of AstraZeneca.
Presentation numberPS4-06-17
First clinical data of DB-1310 (HER3-targeted antibody-drug-conjugate), in patients with pretreated hormone receptor-positive/HER2-negative breast cancer: efficacy and safety data from a phase 1/2a trial
Hua Mu, Dualitybiologics, Shanghai, China
Y. Yin1, A. Spira2, H. Lan3, M. Zhao4, Y. Mao5, W. Yao6, Y. Sun7, J. Wu8, X. Sun9, S. Liu9, H. Mu9, K. Wang9, Y. Qiu10, E. Hamilton11; 1Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, CHINA, 2Clinical Research, Next Oncology, Fairfax, VA, 3Department of Medical Oncology, Sichuan Provincial People’s Hospital, Sichuan Academy of Medical Sciences, School of Medicine UESTC, Chengdu, CHINA, 4Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, CHINA, 5Department of Oncology, The Affiliated Hospital of Jiangnan University, Jiangsu, CHINA, 6Department of Thoracic Medical Oncology, Sichuan Cancer Hospital, Chengdu, CHINA, 7Phase I Clinical Research Center, The Affiliated Cancer Hospital of Shandong First Medical University, Jinan, CHINA, 8Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou, CHINA, 9Clinical Development, Dualitybiologics, Shanghai, CHINA, 10R&D, Dualitybiologics, Shanghai, CHINA, 11Breast Cancer Research Program, Sarah Cannon Research Institute, Nashville, TN.
Background: HER3-targeted antibody-drug conjugates (ADCs) have shown benefits in breast cancer (BC). DB-1310 is a novel ADC comprised of a humanized anti-HER3 IgG1 monoclonal antibody with a highly internalizing unique Domain I epitope, a cleavable peptide linker, and a potent DNA topoisomerase I inhibitor P1021. Initial clinical data demonstrated a manageable safety profile and encouraging anti-tumor activity, particularly in patients with EGFR-mutant NSCLC (Lisberg ASCO 2025). Here, we present the efficacy and safety results of DB-1310 in pretreated hormone receptor-positive (HR+), HER2-negative (HER2-) BC patients. Methods: In the Phase 1 part of this trial (NCT05785741), patients with advanced solid tumors, including HR+/HER2- BC, received DB-1310 monotherapy intravenously every three weeks (Q3W). Safety was the primary endpoint, with efficacy endpoints including objective response rate (ORR), duration of response (DoR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) as secondary endpoints. Results: As of July 04, 2025, a total of 207 patients with solid tumors were enrolled and treated with DB-1310 monotherapy (1.5-6.5 mg/kg, Q3W) in Phase 1 (ECOG PS 1, 84.1%; White, 26.1%; Asian, 69.1%). Among these, 16 HR+/HER2- BC patients were enrolled (3.0-5.5 mg/kg, Q3W), with 15 of them being efficacy-evaluable, and a median prior line of systemic therapy was 2 (range, 1-7). Efficacy Results (N=15 HR+/HER2- BC patients from Phase 1): nine achieved a partial response (one pending confirmation), and four achieved stable disease, resulting in a confirmed ORR of 53.3% (95%CI: 26.59, 78.73) and a DCR of 86.7% (95%CI: 59.54, 98.34). Notably, all partial responses occurred in patients treated with 5-5.5 mg/kg (N=13), yielding an unconfirmed ORR of 69.2%, a confirmed ORR of 61.5%, and a DCR of 92.3% for this dose range. The median DoR is not reached. Median PFS is 14.78 months (95% CI: 11.60, not evaluable) and median OS is 14.69 months (95% CI: 11.89, not evaluable). Safety Results (N=207, all enrolled patients from Phase 1): The safety profile was assessed in the entire Phase 1 part. Treatment-related adverse events (TRAEs) of Grade (G) ≥ 3 occurred in 89 (43.0%) patients, and 25 patients (12.1%) had treatment-related serious adverse events. TRAEs led to dose reduction in 38 (18.4%) and discontinuation in 9 (4.3%) patients. No TRAE-related deaths were reported. Most common TRAEs (any grade/≥G3; >20% overall) were neutrophile count decreased (49.3%/ 26.1%), anemia (47.8%/ 6.8%), platelet count decreased (45.4%/ 15.9%), white blood cell count decreased (43.5%/ 11.1%), nausea (40.6%/ 1.0%), decreased appetite (29.5%/ 0.5%), aspartate aminotransferase increased (26.6%/ 0), alopecia (26.1%/ 0), vomiting (24.6%/ 0), and hypoalbuminemia (21.7%/ 0). Interstitial lung disease/pneumonitis occurred in 10 pts (4.8%; one Grade 2, all others G1). The safety profile in the 16 HR+/HER- BC patients was consistent with the overall Phase 1 part. Conclusion: DB-1310 showed promising anti-tumor activity in heavily pretreated HR+/HER2- BC patients. The manageable safety profile observed in the larger Phase 1 part, involving 207 patients, is consistent with that of the BC subpopulation, supporting its further development.
Presentation numberPS4-06-18
Early Results from a Phase 1 Clinical Study of a Cancer Vaccine Targeting Tumor Associated Antigen MUC1 in Ductal Carcinoma in Situ for Immune Interception of Breast Cancer
Emilia J Diego, University of Pittsburgh, Pittsburgh, PA
E. J. Diego1, J. A. Foldi2, P. F. McAuliffe1, Q. Sabih1, L. A. Emens3, A. Salazar4, O. J. Finn5; 1Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 2Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, 3Department of Internal Medicine, Kaiser Permanente South Sacramento, Sacramento, CA, 4Oncovir, Oncovir, Inc, Winchester, VA, 5Department of Immunology, University of Pittsburgh, Pittsburgh, PA.
Early Results from a Phase 1 Clinical Trial of a Cancer Vaccine Targeting Tumor Associated Antigen MUC1 for Immune Interception of Breast Cancer in Patients with Ductal Carcinoma in SituIntroductionHypoglycosylated tumor MUC1, a transmembrane glycoprotein, is recognized by human T-cells and antibodies as a tumor-associated antigen. It is overexpressed in premalignant precursor lesions, including ductal carcinoma in situ (DCIS), thereby serving as a potential potent DCIS rejection target. Vaccine-induced immune response to MUC1 may halt DCIS recurrence or progression to invasive disease and may offer a future strategy for primary disease prevention in high-risk individuals. We hypothesize that our MUC1 peptide vaccine will be both safe and immunogenic in patients with DCIS and that the elicited systemic immune response will affect changes in the microenvironment of the DCIS from pro- to anti-tumor. MethodsThis single institution, open label, randomized phase I clinical trial (NCT06218303) is designed for 50 post-menopausal women with previously untreated estrogen-receptor positive DCIS histologically confirmed on core needle biopsy (CNB). Patients with autoimmune diseases were excluded. Patients are randomized 2:1 to the vaccine group. The vaccine is composed of a 100aa long MUC1 peptide corresponding to 5 tandem repeats of 20 amino acids from the MUC1 variable number of tandem repeats region (VNTR), admixed with the poly-ICLC adjuvant Hiltonol. The vaccine is administered subcutaneously (SC) or intramuscularly (IM). The vaccine group receives the MUC1 peptide vaccine series pre-operatively (at 0, 2, and 10 weeks) with an optional aromatase inhibitor (AI). The control group receives only optional AI. All undergo surgery at 12 weeks. Research blood is drawn in the vaccine group at baseline and 2 weeks after each vaccine, and in the control group at baseline and at week 12. Tissue from the pre-treatment CNB and the post-treatment operation are also collected. An optional booster is available to vaccine responders 6 months post-surgery.The primary endpoint is vaccine immunogenicity, measured as anti-MUC1 IgG level. Vaccine responders are identified based on a > 2-fold increase in serum anti-MUC1 IgG from baseline, using enzyme-linked immunosorbent assay (ELISA). Adverse events are monitored per Common Terminology Criteria for Adverse Events (CTCAE) version 5.0.Fisher’s exact test will compare primary immunogenicity between the study arms with one-sided α=0.05. Correlative studies include extensive phenotyping of circulating as well as DCIS-infiltrating immune cells and increases in anti-MUC1 cellular immunity post vaccination. ResultsTo date,13 patients completed the study: 7 in the vaccine and 6 in the control group. Mean age is 66 years. There were no serious adverse events. The primary vaccine series elicited or boosted anti-MUC1 IgG in 6 of 7 vaccinated women, for a response rate of 86%. Only 1 patient to date was eligible to receive the booster vaccine and showed a strong immune memory response. There was no anti-MUC1 IgG present in the control group. Extensive phenotyping of peripheral blood mononuclear cell samples from both groups and evaluation of MUC-specific cellular responses are in progress, as are tissue analyses. ConclusionMUC1 peptide vaccine given in the premalignant DCIS setting is safe in the first 7 patients to receive the vaccine. The 86% immune response rate predicts a high rate of response, which will allow us to study immune mechanisms activated by the vaccine, including changes in the DCIS microenvironment.
Presentation numberPS4-06-19
Final results of a Phase I trial of an alpha-lactalbumin (aLA) vaccine for breast cancer
Justin M Johnson, Cleveland Clinic, Cleveland, OH
J. M. Johnson1, E. E. Rhoades2, H. B. Levengood1, A. Ali2, H. Gilmore3, M. L. Kruse2, E. E. Roesch2, T. Onger2, B. Elliott2, E. Haury2, C. Porvasnik2, T. Young4, T. Coutee5, J. A. Fitzgerald6, T. S. Stappenbeck1, G. T. Budd2; 1Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 2Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, 3Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, 4n/a, Previvorsandsurvivors.com, Inc., Delray Beach, FL, 5n/a, DiepCJourney Foundation, Duvall, WA, 6n/a, Sisters4Prevention, West Palm Beach, FL.
Background: α-Lactalbumin (aLA) is expressed in lactating breasts and 70% of triple-negative breast cancer (TNBC) but not at other times or in other tissues. Based on the “retired protein hypothesis” vaccination with aLA provided protection from development of autochthonous tumors in transgenic murine models of breast cancer and inhibited growth of established 4T1 transplantable breast tumors in BALB/c mice. Methods: We completed accrual to a Phase I trial of recombinant human aLA with GMP-grade zymosan adjuvant in Montanide ISA 51 VG vehicle in 3 cohorts of subjects: Ia) patients with high-risk TNBC who have completed all standard treatment; Ib) patients with BRCA1, BRCA2, or PALB2 mutations who are undergoing risk-reducing mastectomies; and Ic) patients with TNBC who have residual cancer after primary chemo-immunotherapy and are receiving post-operative treatment with pembrolizumab +/- capecitabine. Three vaccinations were given once every 2 weeks. Events of Common Terminology Criteria for Adverse Events (CTCAE) Grade ≥ 2 are considered dose-limiting toxicities (DLTs). Blood was drawn prior to therapy as well as 14, 28, and 56 days after the first vaccination to assess cellular response using enzyme-linked immunosorbent spot (ELISPOT) assays of interferon-gamma and interleukin-17 production in response to aLA and for humoral response by enzyme-linked immunosorbent assay (ELISA). The breast tissue of participants in the Phase Ib cohort were or will be examined for occult lactational foci and inflammatory changes. Results: We vaccinated 26 patients in Cohort Ia, 4 in Cohort Ib, and 5 in Cohort Ic. CTCAE toxicity by dose level (DL) is summarized in Table 1 by grade for each study cohort. All adverse events (AEs) were injection site reactions, with ulceration and need for incisional drainage representing the Grade 3 DLT events. 19 of 26 patients assayed to date across all cohorts met protocol specified definitions of an immune response based on ELISPOT assays that quantify frequencies of T cells producing IFNγ or IL-17 in response to recombinant aLA; these data included 4 of 6 subjects in Cohort Ia at DL1 and 6 of 11 subjects in all cohorts treated at DL1 and tested to date. ELISPOT and ELISA data for all subjects will be available in August 2025 and will be presented. Conclusions: DL1 is the maximum tolerated dose (MTD) by protocol definition, produced at worst Grade 1 toxicity in a total of 14 of 15 subjects, and produced an immune response in most patients, based on the criteria prospectively defined in the study protocol. DL1e was tolerable in 5 of 5 subjects but was not determined the MTD per our 3+3 trial design. Immune data for all subjects in the trial, including new data across all cohorts, will be reported and will inform the design of subsequent Phase II trials. Funding Source: Department of Defense (W81XWH-17-1-0592 and W81XWH-17-1-0593).
| Cohort | Dose Level | aLA Dose (mcg) | Zymosan Dose (mcg) | Total Enrolled Subjects | n Grade 1 | n Grade 2 | n Grade 3 | n Grade 4 | n Grade 5 |
| Ia | 1 | 10 | 10 | 6 | 6 | ||||
| 1b | 50 | 10 | 5 | 3 | 1 | 1 | |||
| 1e | 10 | 20 | 5 | 5 | |||||
| 2 | 100 | 10 | 6 | 5 | 1 | ||||
| 3 | 500 | 10 | 3 | 1 | 2 | ||||
| 2 (old) | 100 | 100 | 1 | 1 | |||||
| Ib | 1 | 10 | 10 | 4 | 4 | ||||
| Ic | 1 | 10 | 10 | 5 | 4 | 1* |
*Diagnosed while pregnant
Presentation numberPS4-07-01
Exploring the Impact of Circadian Timing and TIL Status on Immunotherapy Response in Breast Cancer
Kaho Nakamura, Mie University Hospital, Tsu, Japan
K. Nakamura1, K. Saito2, R. Ito1, M. Yoshida1, E. Hatakawa1, R. Yamakado1, M. Yoshikawa1, S. Watanabe1, N. Imai1, M. Shibusawa1, A. Noro1, Y. Kozuka3, K. Kawaguchi1; 1Breast Surgery, Mie University Hospital, Tsu, JAPAN, 2Medical Oncology, Mie University Hospital, Tsu, JAPAN, 3Pathology, Mie University Hospital, Tsu, JAPAN.
Background: Circadian rhythms regulate immune responses, but their clinical relevance in cancer immunotherapy remains underexplored. We investigated whether the timing of immune checkpoint inhibitor (ICI) administration, in conjunction with tumor-infiltrating lymphocyte (TIL) status, influences treatment efficacy in breast cancer. Methods: We conducted a retrospective analysis of two cohorts: a perioperative cohort receiving neoadjuvant ICI (n=22, early-stage) and a metastatic cohort treated with ICI in the advanced setting (n=18). For each patient, the median time of ICI administration was determined and dichotomized as morning (<12:00) or afternoon (≥12:00). In the perioperative cohort, pathological complete response (pCR) was assessed; in the metastatic cohort, progression-free survival (PFS) was analyzed. Pre-treatment stromal TIL levels were evaluated and stratified by median value (TIL-high vs. TIL-low). Associations between administration time, TIL status, and treatment outcomes were explored. Results: In the perioperative cohort, neither administration timing nor TIL level alone showed a significant association with pCR. However, when stratifying by both variables, an interaction emerged: among 9 patients with high TILs, those treated in the morning (n=4) achieved a 100% pCR rate, while those treated in the afternoon (n=5) had a 55.6% non-pCR rate. In contrast, patients with low TILs had high pCR rates regardless of administration time (morning: 83.3%, afternoon: 80.0%). In the metastatic cohort, PFS did not significantly differ between morning and afternoon administration, nor by PD-L1 or TIL status, although trends suggested numerically shorter PFS in afternoon-treated patients with high PD-L1 expression. Across both cohorts, no meaningful association was observed between time of administration and immune-related adverse events. Conclusion: While overall efficacy was not significantly influenced by ICI administration time, subgroup analysis revealed a paradoxical pattern among TIL-high patients in the perioperative setting: afternoon administration was associated with reduced pCR rates. This finding suggests that the functional phenotype of TILs—and possibly their interaction with circadian immune modulation—may shape response to immunotherapy. Further studies incorporating spatial and functional profiling of the tumor immune microenvironment are warranted to clarify the clinical implications of circadian timing in ICI treatment.
Presentation numberPS4-07-02
Determinants of early and late relapse in patients with early triple negative breast cancer from the prospective CANTO Cohort
Chiara Benvenuti, Gustave Roussy, Villejuif, France
C. Benvenuti1, L. Perotti2, A. Martin3, C. Gaudin3, C. Jouannaud4, M. Fournier5, C. Kaderbhaï6, A. Kieffer7, F. Cherifi8, M. Campone9, F. Lerebours10, P. Cottu11, F. André1, A. Bertaut12, D. Antonio1, S. Michiels2, B. Pistilli1, T. Grinda1; 1Medical Oncology, Gustave Roussy, Villejuif, FRANCE, 2Biostatistics and Epidemiology, Gustave Roussy, Villejuif, FRANCE, 3Data and Partnerships, UNICANCER, Paris, FRANCE, 4Medical Oncology, Institut Godinot, Reims, FRANCE, 5Medical Oncology, Institut Bergonié, Bordeaux, FRANCE, 6Medical Oncology, Centre Georges-François Leclerc, Dijon, FRANCE, 7Medical Oncology, Institut de cancérologie de Lorraine, Vandœuvre-lès-Nancy, FRANCE, 8Medical Oncology, Centre François Baclesse, Caen, FRANCE, 9Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 10Medical Oncology, Institut Curie, Saint-Cloud, FRANCE, 11Medical Oncology, Institut Curie, Paris, FRANCE, 12Medical Oncology, Centre Georges François Leclerc, Dijon, FRANCE.
Background: TNBC exhibits a high risk of early relapse, yet predictors of time to relapse remain poorly defined. Early relapses (while on or within 12 months after ending curative therapies) are associated with chemotherapy-resistant disease and poor prognosis (median overall survival <12 months; Grinda et al., Eur J Cancer 2023). Identifying key factors associated with early and late relapse may help guide therapeutic interventions. This study explores associations between clinical factors and timing of relapse in patients (pts) with TNBC from the prospective CANTO cohort (NCT01993498). Methods: Early-stage TNBC pts enrolled in the CANTO cohort between 2012 and 2023 were included. Descriptive statistics were used to summarize clinical variables. Invasive disease-free survival (IDFS) was estimated with Kaplan-Meier curves from the time of study entry. The proportional hazards assumption was assessed with Schoenfeld residuals and cumulative hazard plots. A time-varying fixed-effect multivariable Cox model was built using stratification at 24 months for distinguishing early and late relapses. To identify the best-fitting multivariable model for IDFS, stepwise selection (stepAIC) was applied starting from a priori covariates, including age, grade, surgery type, and TNM stage , with additional candidates tested based on univariate results and clinical relevance. Results: Among the 999 pts included, median age was 52.9 years (IQR: 45.1-61.9). Most pts presented with stage II disease (56.3%), followed by stage I (34.4%) and stage III (9.3%). Chemotherapy was administered in 93.5% of pts: adjuvant in 51.3% and neoadjuvant in 42.2% of cases. Breast-conserving surgery was performed in 71.7% pts, mastectomy in 28.3%. Around half of the pts underwent axillary dissection (47.6%).Median follow-up was 74.3 months (IQR: 51.2-99.4). During this period, 239 iDFS events occurred, 49% within the first 24 months. The best final time-varying fixed effect multivariate model for predicting iDFS included age, BMI at diagnosis, smoking, type of surgery, grade, stage and chemotherapy exposure. TNM stage showed a strong prognostic role across the entire follow-up, with stage III disease consistently associated with higher relapse risk compared to stage I (HR: 2.44; 95% CI: 1.44-4.14; p < 0.001). For early relapse, underweight status (BMI < 18.5 Kg/m2) was the strongest predictor (HR: 3.35; 95% CI: 1.58-7.11; p = 0.002). Axillary dissection (HR: 1.60; 95% CI: 1.00-2.55; p = 0.05) and receipt of (neo)adjuvant chemotherapy (HR: 1.51; 95% CI: 0.95-2.40; p = 0.078) trended toward a signifcant increased risk of early relapse. Despite adjustment for TNM stage to account for disease severity, these treatment-related variables may act as proxies for disease burden, and some residual counfounding cannot be excluded. Late relapse was associated with older age (HR: 1.03; 95% CI: 1.01-1.04; p = 0.003), while smokers had a borderline increased risk versus non-smokers (HR: 1.61; 95% CI: 0.98-2.63; p = 0.06). Conclusions: This large cohort study shows that early and late relapses in early-stage TNBC are driven by distinct clinical factors. Our findings integrate clinicopathological variables to support the development of models reflecting individual relapse risk and timing. Early relapses, accounting for half of the events, were strongly associated with underweight status, possibly reflecting patient frailty and comorbidities, and high disease burden at diagnosis, as indicated by stage III disease and high treatment intensity, including axillary dissection and chemotherapy. Late relapses were predicted by older age and stage III disease. These data support time-aware prognostic models to guide individual-risk adapted strategies and underscore the need for biological investigations to better understand early relapse in TNBC.
Presentation numberPS4-07-03
Adverse Events in Neoadjuvant Therapy for Triple-Negative Breast Cancer: A Systematic Review and Meta-Analysis of Phase III Trials
Marcelo Antonini, Hospital do Servidor Publico Estadual de São Paulo, São Paulo, Brazil
M. Antonini1, A. Mattar2, F. P. Cavalcante3, F. P. Zerwes4, E. C. Millen5, F. P. Brenelli6, A. L. Frasson7, M. D. Teixeira2, M. Madeira8, G. Facina9, H. L. Couto10, G. T. Tosello11, R. Arakelian12, A. D. Lima13, G. N. Garcia14, F. Bagnoli15, A. G. Amorim16, M. F. de Figueiredo2, L. R. Soares17, R. Freitas Júnior18, L. H. Gebrim19; 1Breast Surgery, Hospital do Servidor Publico Estadual de São Paulo, São Paulo, BRAZIL, 2Breast Surgery, Hospital da Mulher SP, São Paulo, BRAZIL, 3Breast Surgery, Hospital Geral de Fortaleza, Fortaleza, BRAZIL, 4Breast Surgery, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 5Breast Surgery, Americas Oncologia, Rio de Janeiro, BRAZIL, 6Breast Surgery, Universidade Estadual de Campinas, Campinas, BRAZIL, 7Breast Surgery, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 8Breast Surgery, Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, BRAZIL, 9Breast Surgery, Universidade Federal de São Paulo, São Paulo, BRAZIL, 10Breast Surgery, Redimama – Redimasto, Belo Horizonte, BRAZIL, 11Breast Surgery, Unoeste, Presidente Prudente, BRAZIL, 12Oncology, Hospital da Mulher SP, São Paulo, BRAZIL, 13Breast Surgery, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 14Breast Surgery, Oncoclínicas, São Paulo, BRAZIL, 15Breast Surgery, Santa casa de misericórdia de São Paulo, São Paulo, BRAZIL, 16Breast Surgery, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, São Paulo, BRAZIL, 17Breast Surgery, Universidade Federal de Goiás, São Paulo, BRAZIL, 18Breast Surgery, Universidade Federal de Goiás, Goiania, BRAZIL, 19Breast Surgery, Hospital Beneficiência Portuguesa de São Paulo, São Paulo, BRAZIL.
Background: Triple-negative breast cancer (TNBC) accounts for 15-20% of breast cancers and lacks expression of estrogen receptor, progesterone receptor, and HER2, limiting targeted options. Recent neoadjuvant therapy (NAT) advances include immune checkpoint inhibitors (ICIs) and PARP inhibitors, improving pathologic complete response (pCR) and survival but raising concerns about severe toxicity. Despite broader adoption of these regimens, a systematic analysis of Grade ≥3 adverse events (AEs) from contemporary phase III trials is lacking. Methods: We conducted a PRISMA-guided systematic review and meta-analysis (PROSPERO: CRD42024562785). MEDLINE, Embase, and Cochrane were searched for phase III randomized trials (Jan 2010-Jun 2025) evaluating NAT in non-metastatic TNBC and reporting Grade ≥3 AEs (CTCAE). One phase II trial (WSG-ADAPT-TN; n=336) with robust comparative design and AE reporting was also included. Pooled prevalence and risk ratios (RR) with 95% CI were calculated using DerSimonian-Laird random-effects models. Subgroup analyses included: Group A (PARP inhibitors), B (immunotherapy), and C (platinum-based regimens). Heterogeneity was assessed via I². Results: Six trials with 3,316 TNBC patients were included (BrighTNess, IMpassion031, KEYNOTE-522, NeoTRIP, PARTNER, WSG-ADAPT-TN). Neutropenia was the most common Grade ≥3 AE, highest with immunotherapy (33.5%), followed by PARP inhibitors (23.7%) and platinum regimens (16.0%) (Q=71.8, p<0.0001). PARP inhibitors significantly increased neutropenia risk (RR=1.84), while immunotherapy showed no increase (RR=1.00, p=0.94). Grade ≥3 anemia was higher with PARP inhibitors (20.6%) vs. immunotherapy (9.9%), with doubled risk vs. placebo (RR=2.08, p=0.02). Thrombocytopenia was most prevalent with immunotherapy (7.9%), but PARP inhibitors showed the highest risk (RR=3.56, p=0.0001). Febrile neutropenia was more common with ICIs (14.9%) than PARP inhibitors (3.0%). ALT elevation was more frequent in platinum regimens (6.4%), without significant differences across groups. Fatal AEs were <1% in all trials. Conclusions: Modern NAT regimens improve outcomes in TNBC but confer significant Grade ≥3 toxicity. PARP inhibitors pose the highest hematologic risk, while ICIs increase febrile neutropenia. Platinum regimens showed the most favorable toxicity profile. Findings highlight the need for biomarker-driven, individualized treatment and vigilant AE monitoring in early TNBC.
Presentation numberPS4-07-04
Rare but Resistant: Neoadjuvant Chemoimmunotherapy in Special Histological Subtypes of Triple-Negative Breast Cancer — Insights from the Neo-Real/GBECAM-0123 Study
Romualdo Barroso-Sousa, Brasilia Hospital, Oncologia Américas, Brasilia, Brazil
R. Barroso-Sousa1, L. Testa2, P. Mandó3, M. C. Tavares4, N. C. Nunes5, G. Cordoba3, F. Waisberg3, F. C. Balint4, I. M. de Sousa6, M. O. Andrade1, M. C. Gouveia7, F. Madasi8, J. Bines8, R. P. Ferreira9, D. D. Rosa9, C. L. Santos10, M. R. Monteiro11, Z. S. de Souza12, D. Assad-Suzuki12, C. dos Anjos13, D. M. Gagliato14, A. U. Gomes14, B. M. Zucchetti7, A. Ferrari15, M. L. de Brito16, M. F. Monteiro17, P. A. Signorini18, N. J. Gomes19, C. Gallina3, S. Sanches4, P. M. Hoff20, M. Estevez-Diz21, R. C. Bonadio20; 1Oncology, Brasilia Hospital, Oncologia Américas, Brasilia, BRAZIL, 2Oncology, Instituto D’Or de Pesquisa e Ensino, Brasilia, BRAZIL, 3Oncology, SUMA (Grupo Cooperativa Argentino para el estudio y la investigación del Cáncer de Mama), Buenos Aires, ARGENTINA, 4Oncology, A.C.Camargo Cancer Center, São Paulo, BRAZIL, 5Oncology, Grupo Américas, Rio de Janeiro, BRAZIL, 6Oncology, A.C.Camargo Cancer Center, Brasilia, BRAZIL, 7Oncology, Hospital 9 de Julho, Grupo Américas, São Paulo, BRAZIL, 8Oncology, Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, BRAZIL, 9Oncology, Hospital Moinhos de Vento, Porto Alegre, BRAZIL, 10Oncology, Instituto D’Or de Pesquisa e Ensino, Recife, BRAZIL, 11Oncology, Grupo Américas, Hospital Samaritano, São Paulo, BRAZIL, 12Oncology, Hospital Sírio-Libanês, Brasilia, BRAZIL, 13Oncology, Hospital Sírio-Libanês, São Paulo, BRAZIL, 14Oncology, Hospital Beneficência Portuguesa, São Paulo, BRAZIL, 15Oncology, Hospital Santa Paula, Grupo Américas, São Paulo, BRAZIL, 16Oncology, Clínica AMO, Salvador, BRAZIL, 17Oncology, Instituto do Câncer do Ceará, Fortaleza, BRAZIL, 18Oncology, Centro Integrado de Pesquisa da Amazônia (CINPAM), Manaus, BRAZIL, 19Oncology, Hospital São Domingos, DASA Oncologia, São Luiz, BRAZIL, 20Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 21Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRAZIL.
Rare but Resistant: Neoadjuvant Chemoimmunotherapy in Special Histological Subtypes of Triple-Negative Breast Cancer — Insights from the Neo-Real/GBECAM-0123 Study Background Special histological types (SHT) of triple-negative breast cancer (TNBC) exhibit distinct biological behavior and treatment responses. However, due to their rarity, patients with SHT are underrepresented in clinical trials. We aimed to evaluate the effectiveness of pembrolizumab (P) combined with neoadjuvant chemotherapy (NCT) in patients with SHT of TNBC. MethodsWe analyzed data from patients with TNBC and SHT enrolled in the Neo-Real/GBECAM-0123 study—a multicenter, real-world study including patients treated with neoadjuvant P+NCT across ten cancer centers since July 2020. Effectiveness outcomes included pathologic complete response (pCR) and event-free survival (EFS). ResultsOf the 727 patients included to date in the Neo-REAL study, 646 (88.9%) had TNBC of no special type (NST), 51 (7%) SHT, 4 (0.6%) undifferentiated, and 26 (3.6%) unknown histology. Among the SHT group, 20 metaplastic, 16 lobular, and 15 other subtypes (9 apocrine, 3 micropapillary, 2 neuroendocrine, and 1 medullary). Patients with lobular, metaplastic, and other SHT tumors were older than those with NST (median ages: 58, 51, 49, and 44 years, respectively; p = 0.001). Grade 3 tumors were more frequent in NST (76%) and metaplastic (84%) subtypes than in lobular (50%) and other SHT tumors (50%) (p = 0.019). A high Ki-67 index (≥50%) was also more common in NST tumors (76%) compared to metaplastic (52%), lobular (53%), and other SHT tumors (36%) (p < 0.001). Tumor stage distribution was similar across groups. Disease progression during P+NCT was significantly higher in the metaplastic group (33%) compared to NST (3%) and lobular tumors (0%) (p = 0.001). However, all but one patient with progression in the metaplastic group underwent surgery. pCR and EFS rates are presented in the Table. pCR rates were markedly lower in metaplastic tumors compared to NST and lobular subtypes. With a median follow-up of 22 months, 83 events of disease recurrence or death were recorded. Lobular and metaplastic tumors were associated with significantly lower 2-year EFS compared to NST. These differences remained significant in a multivariable Cox regression including tumor grade and stage. ConclusionPatients with SHT of TNBC had worse outcomes with neoadjuvant pembrolizumab plus chemotherapy compared to those with NST tumors. Metaplastic carcinoma was associated with lower pCR and a trend tower lower EFS rates. Although lobular carcinoma had pCR rates similar to NST, an unfavorable EFS was also observed in this group. These results underscore the urgent need for developing new tailored therapeutic strategies for these rare and aggressive subtypes of TNBC.
| pCR | OR (95% CI) | 2-year EFS | HR (95% CI) | |
| NST | 64.7% | ref | 88.0% | ref |
| Lobular | 69.2% | 1.22 (0.37-4.04) | 61.9% | 3.92 (1.58-9.76) |
| Metaplastic | 16.7% | 0.10 (0.031-0.38) | 77.7% | 2.63 (1.06-6.54) |
| Other SHT | 46.2% | 0.46 (0.15-1.41) | 78.3% | 2.23 (0.70-7.11) |
Presentation numberPS4-07-05
JNK1 and JNK2 isoforms promote triple-negative breast cancer aggressiveness by fostering an M2 macrophage-driven immunosuppressive tumor microenvironment
Xuemei Xie, University of Hawaii Cancer Center, Honolulu, HI
B. Kuntal1, X. Stern1, T. Semba2, K. Nakaoka1, M. Fujimoto1, M. William1, K. Leonard1, D. Rampa1, J. Lee1, N. Ueno3, X. Xie1; 1Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI, 2Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, JAPAN, 3Cancer Biology Program and Translational and Clinical Research, University of Hawaii Cancer Center, Honolulu, HI.
Background: Triple-negative breast cancer (TNBC) is characterized by an immunologicallycold tumor microenvironment (TME), which facilitates immune evasion and tumorprogression. Our recent study showed that c-Jun N-terminal kinase (JNK) signaling promotesan immunosuppressive TME via macrophage-derived CCL2, thereby driving TNBC growthand metastasis. JNK has three isoforms, among which JNK1 and JNK2 have been implicatedin the development of various cancers. However, the distinct roles of JNK1 and JNK2 inregulating the TME immune landscape and aggressiveness in TNBC remain poorly understood.Building on our previous findings, in this study, we tested our hypothesis that both JNK1 andJNK2 promote TNBC aggressiveness by fostering an M2 macrophage–drivenimmunosuppressive TME.Methods: The effect of JNK singling inhibition by the pan-JNK inhibitor JNK-IN-8 and theeffect of JNK1 or JNK2 knockout (KO) on the growth of murine PyMT-M TNBC cells wereassessed in vitro using CellTiter-Blue cell viability and clonogenic assays. The roles ofintratumoral JNK1 and JNK2 in regulating the TME immune landscape and tumor growth wereexamined using an immunocompetent syngeneic C57BL/6 mouse model bearing PyMT-Mtumors derived from JNK1 or JNK2 KO cells. The contributions of JNK1 and JNK2 in theTME was examined using jnk1-/- and jnk2-/- immunocompetent syngeneic C57BL/6 mousemodels bearing PyMT-M tumors. The effects of the loss of JNK1 and JNK2 in tumors or in theTME on immune cell tumor infiltration were analyzed by flow cytometry. Key cytokinesinvolved in JNK-mediated modulation of the TME immune landscape were identified bycytokine array analysis.Results: Inhibition of JNK signaling by JNK-IN-8 and KO of either JNK1 or JNK2significantly suppressed the proliferation of murine PyMT-M TNBC cells in vitro, suggestingthat both JNK1 and JNK2 are essential for TNBC cell growth. However, compared to the Cas9control cells, intratumoral KO of either JNK1 or JNK2 in PyMT-M tumor cells did not affecttumor initiation and growth or immune cell tumor infiltration in the wild-typeimmunocompetent syngeneic C57BL/6 mice, demonstrating that intratumoral JNK1 and JNK2are not required for PyMT-M tumor initiation and progression. In contrast, regardless of theabsence or presence of JNK1 and JNK2 in tumor cells, tumor initiation was markedly delayedand tumor growth was significantly reduced in jnk1-/- and jnk2-/- immunocompetent syngeneicC57BL/6 mice, with a more pronounced effect observed in jnk1-/- mice. This result highlightsthat both JNK1 and JNK2 in the TME play a critical role in PyMT-M tumor initiation andprogression. Interestingly, compared to tumors from wild-type mice, those from both jnk1-/-and jnk2-/- mice exhibited a marked increase in M1 macrophages and a reduction in M2macrophages, with a greater effect observed in jnk1-/- mice. This result indicates that JNKdeficiency in the TME induces the reprogramming of M2 macrophages toward an M1phenotype. Indeed, cytokine array analysis revealed elevated expression of CCL12, CCL5, andCXCL9 in JNK1 KO tumors from jnk1-/- mice, as well as increased CXCL9 expression in JNK2KO tumors from jnk2-/- mice. These cytokines play critical roles in macrophage recruitmentand polarization. Taken together, these findings support our hypothesis that both JNK1 andJNK2 promote TNBC aggressiveness via fostering an M2 macrophage–drivenimmunosuppressive TME.Conclusion: Our findings demonstrate that both JNK1 and JNK2 in the TME drive TNBCinitiation and progression via promoting an M2 macrophage–driven immunosuppressive TMEin the macrophage-enriched TNBC mouse model. These findings establish JNK1 and JNK2 askey regulators of the immunosuppressive TME in TNBC, highlighting their therapeuticimportance in treating TNBC.
Presentation numberPS4-07-06
Independent validation of TNBCDX, a 15-gene genomic risk stratification tool for early-stage triple-negative breast cancer
Fara Braso-Maristany, Fundació de Recerca Clínic Barcelona, Barcelona, Spain
F. Braso-Maristany1, E. Ciruelos2, E. Seguí3, J. Montes2, P. Tolosa2, M. Rey1, F. Pardo4, R. Sánchez-Bayona2, L. Lema2, M. Cobos2, P. Blasco1, P. Galván1, L. Parilla2, R. Moreno2, G. Riu5, I. Garcia-Fructuoso3, R. Gómez-Bravo3, M. Bergamino3, F. Schettini3, A. Madariaga2, B. Adamo3, M. Muñoz3, M. Marín-Aguilera4, L. Manso2, L. Paré4, T. Pascual3, M. Vidal3, A. Prat3, M. Alva2; 1Translational Genomics and targeted therapies in solid tumors lab, Fundació de Recerca Clínic Barcelona, Barcelona, SPAIN, 2Oncology, Hospital 12 de Octubre, Madrid, SPAIN, 3Oncology, Hospital Clínic Barcelona, Barcelona, SPAIN, 4Reveal Genomics, Reveal Genomics, Barcelona, SPAIN, 5Pharmacy, Hospital Clínic Barcelona, Barcelona, SPAIN.
Background: TNBCDX is a 15-gene composite biomarker (10-gene immune module, 4-gene proliferation signature, ERBB2 and clinical variables) for predicting pathological complete response (pCR) and recurrence risk in early-stage triple-negative breast cancer (TNBC). In Martin et al. (Ann Oncol 2024), TNBCDX was retrospectively validated in 527 patients (pts) from the WSG-ADAPT-TN, MMJ-CAR-2014-01 and NeoPACT cohorts—each receiving neoadjuvant taxane-based therapy without anthracycline/cyclophosphamide (AC)—where it independently stratified pCR rates, distant recurrence-free survival (DRFS) and overall survival (OS) after adjustment for pCR status and tumor-infiltrating lymphocytes (TILs). We now assess TNBCDX in 164 consecutive pts from two Spanish academic centers, 127 treated with sequential AC→taxane-carboplatin± pembrolizumab. Methods: Baseline pre-treatment formalin-fixed, paraffin-embedded tumor samples were collected retrospectively from pts with stage I-III TNBC treated at Clinic Barcelona Comprehensive Cancer Center (n=108) and Hospital 12 de Octubre, Madrid (n=56). Keynote-522 backbone regimen (AC-taxane-carboplatin) with and without pembrolizumab was used in 47 pts and 80 pts, respectively. Gene expression profiling was performed on the TNBCDX standardized platform. TNBCDX pCR and risk scores were calculated centrally, blinded to outcomes. Associations with DRFS, OS, and pCR were evaluated using Kaplan-Meier analyses and multivariable Cox and logistic regression models adjusted for age, stage, and TILs. Results: Median age was 56.2 years (range 27.3-84.9). Ductal morphology predominated (88.4%), followed by metaplastic (3.7%), apocrine (1.8%), and lobular (1.2%). Most tumors were high grade (68%), cT2 (59%), and cN0 (56%). Median TILs was 15% (range 0-90%). Nearly all pts (97.6%) received neoadjuvant taxane-carboplatin regimens—79.3% with AC and 36.6% with immunotherapy—achieving a 53% overall pCR rate. At a median follow-up of 44.5 months, there were 16 DRFS events and 15 deaths. Among the 127 pts treated with Keynote-522 backbone regimen, those receiving pembrolizumab (n=47) had a pCR rate of 53.7% versus 60.0% in those who did not (n=80) (p=0.562). The distribution of the TNBCDX pCR score was 32.9% low, 28.7% medium, and 38.4% high. High versus low TNBCDX pCR score was associated with higher pCR rates (65.5% vs 38.8%; odds ratio=6.51; 95% CI 1.45-3.41; p=0.019). The TNBCDX risk score classified 45.7% as low-risk and 54.3% as high-risk. High versus low TNBCDX risk score was independently associated with shorter DRFS (hazard ratio [HR]=5.12; 95% CI 1.58-16.50; p=0.006) and shorter OS (HR=5.56; 95% CI 1.51-20.5; p=0.010). Higher TILs predicted pCR (OR=1.03; 95% CI 1.01-1.05; p=0.005) but were not significantly associated with DRFS or OS (both p>0.05). Conclusions: In this independent cohort, TNBCDX pCR and risk scores were significantly associated with pCR and differential survival outcomes—independently of pCR status and systemic therapy. These data underscore the value of TNBCDX for tailoring systemic therapy intensity in early-stage TNBC.
Presentation numberPS4-07-07
Factors associated with pCR and distant recurrence in patients with early-stage TNBC treated with KN522
Rachel Jaber Chehayeb, Hospital of the University of Pennsylvania, Philadelphia, PA
R. Jaber Chehayeb1, P. Coutifaris2, F. White1, J. Heintz3, S. L. Jacob2, J. Q. Zhang4, O. M. Fayanju4, E. S. Lebrow2, N. Taunk5, K. Lee2, H. Knollman2, I. Makhlin2, A. Clark2, R. Jankowitz2, A. Nayak6, S. Domchek2, A. DeMichele2, P. D. Shah2; 1Perelman School of Medicine at the University of Pennsylvania, Hospital of the University of Pennsylvania, Philadelphia, PA, 2Division of Hematology Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, 3Biostatistics Analysis Center, Biostatistics Analysis Center, Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA, 4Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 5Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 6Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
Background: The KEYNOTE522 (KN522) regimen of neoadjuvant chemotherapy with pembrolizumab yields statistically significant and clinically meaningful improvements in pathologic complete response (pCR) rate, event-free and overall survival compared to chemotherapy alone in patients with early-stage triple-negative breast cancer (eTNBC). However, one-third of patients (pts) have residual disease, and of these, many develop distant recurrence that contributes to mortality; understanding characteristics of these high-risk pts is a critical unmet need. This study aimed to understand factors associated with (1) achieving pCR with KN522 and (2) in the absence of pCR, developing distant recurrence. Methods: We conducted a retrospective analysis of pts with eTNBC treated at the University of Pennsylvania with the KN522 regimen between September 2021 and December 2023. Multivariate logistic regression analysis was performed to identify variables associated with pCR using the generalized linear model function in R4.5.0. Given sample size restrictions, Fisher’s exact test was used to evaluate variables associated with recurrence. For these analyses, exact odds ratios and 95% confidence intervals are reported. Results: 225 pts with eTNBC initiated KN522 during the study period. 222 had surgery; 120 (54%) had pCR (no invasive cancer) and 102 (46%) had residual disease. At a median follow-up of 28 months, 26 pts (12%) including 2 with pCR developed distant recurrence. Factors significantly associated with achieving pCR were tumor grade 3 (vs 2; OR 3.42, 95% CI 1.31-9.84, p=0.016) and development of an immune-related adverse event (IRAE vs no IRAE, OR 2.26, 95% CI 1.05-5.07, p=0.041). Factors negatively associated with pCR included clinical stage III (vs I/II, OR 0.50, 95% CI 0.26-0.92, p=0.028) and older age at diagnosis (OR 0.97, 95% CI 0.94-0.99, p=0.047). Among patients who did not achieve pCR, factors associated with distant recurrence were pathologic (p)N stage (N1+ vs N0, OR 19.83, 95% CI 5.58 – 91.85, p<0.0001), pT stage (T2+ vs T0/T1, OR 4.50, 95% CI 1.54-13.53, p=0.003), and clinical stage III (vs I/II, OR 4.25, 95% CI 1.42-14.58, p=0.005). Conclusions: This real-world analysis reinforces established factors associated with pCR including tumor grade and clinical stage. Moreover, these results add to growing evidence across solid tumors that the development of IRAEs correlates with improved response. Among patients with residual disease after KN522, those with node positivity, pT2 or larger tumors, and stage III disease may represent target populations for studies investigating enhanced surveillance and treatment strategies, such as with circulating tumor DNA and antibody-drug conjugates.
| Variable | OR for pCR | 95% CI, lower | 95% CI, upper | p-value |
| Age at Diagnosis | 0.971 | 0.943 | 0.999 | 0.047 |
| Race (Non-White vs White ) | 1.562 | 0.828 | 2.979 | 0.171 |
| ECOG (1-2 vs 0) | 0.839 | 0.362 | 1.955 | 0.682 |
| Histology (IDC vs Other) | 2.110 | 0.800 | 5.829 | 0.137 |
| Tumor Grade (3 vs 2) | 3.423 | 1.312 | 9.838 | 0.016 |
| Pre-op ER % (≥1% vs <1%) | 0.892 | 0.311 | 2.666 | 0.834 |
| Clinical Stage (III vs I/II) | 0.496 | 0.262 | 0.922 | 0.028 |
| Time from biopsy to chemotherapy initiation | 1.006 | 0.999 | 1.014 | 0.088 |
| IRAE (vs no IRAE) | 2.263 | 1.051 | 5.068 | 0.041 |
| Steroid use during neoadjuvant treatment for IRAE or medical comorbidity (vs. no use) | 0.456 | 0.177 | 1.142 | 0.097 |
| Antibiotic use during neoadjuvant treatment (vs no antibiotics) | 1.693 | 0.647 | 4.610 | 0.289 |
| Chemotherapy dose reduction in neoadjuvant setting (vs no reduction) | 1.286 | 0.590 | 2.874 | 0.531 |
| Cycles of Taxol | 1.051 | 0.832 | 1.365 | 0.689 |
| Cycles of Carboplatin | 0.918 | 0.474 | 1.630 | 0.781 |
| Cycles of AC | 1.391 | 0.948 | 2.097 | 0.100 |
| Cycles of neoadjuvant pembrolizumab | 1.105 | 0.874 | 1.398 | 0.400 |
Presentation numberPS4-07-08
Spatial transcriptomics of TNBCs show an association between HOXB13 expression and formation of a plasmablast-rich neighborhood
Blake Flood, Massachusetts General Brigham, Boston, MA
B. Flood1, C. Nawrocki2, L. Neiman3, K. Treuner4, Y. Zhang4, D. Sgroi5; 1Pathology, Massachusetts General Brigham, Boston, MA, 2Krantz Family Cancer Center, Massachusetts General Hospital, Boston, MA, 3Krantz Family for Cancer Research, Massachusetts General Hospital, Boston, MA, 4Onocology, Biotheranostics Inc., a Hologic Company, San Diego, CA, 5Pathology, Massachusetts General Hospital, Boston, MA.
Background: HOXB13 gene expression has been extensively studies in hormone receptor positive (HR+) post-menopausal breast cancer patients. Recently, we demonstrated that HOXB13 is expressed in a subset of triple negative breast cancer (TNBC) patients and that ectopic expression of HOXB13 in a mouse mammary model of TNBC confers a tumoral growth advantage by impeding antitumor T-cell immunity. Here, we performed comparative spatial transcriptomic profiling of the immune microenvironment of HOXB13+ and HOXB13– human TNBCs. Methods: Using the Nanostring CosMx platform, we performed 1000-plex spatial transcriptomic profiling of 15 treatment-naïve human HOXB13+ TNBCs and 8 treatment-naïve HOXB13– TNBCs. Cells were segmented with Cellpose, then filtered based on RNA counts. Sparse areas of tissue were filtered entirely to retain structured tissue. Expression was normalized, log-transformed, and scaled for dimensionality reduction with PCA and UMAP. Cell-typing was performed using an immune-oncology reference profile. Cell types were validated via marker genes identified by differential expression. Differential expression analysis was conducted with generalized linear mixed modeling. Analytical neighborhood enrichment analysis for each cell type was conducted using a spatial neighborhood graph constructed by Delaunay triangulation.Results: Using spatial transcriptomics, we find two immune “neighborhoods” within TNBC patients denoted by an association of multiple cell types that preferentially are found adjacent to one another. The first “cytotoxic” neighborhood is associated with CD8+ T cells, antigen presenting cells, and CD4+ T cells all of which preferentially are found near one another but are not found in proximity to other immune cell types. The second “plasmablast-rich” neighborhood is comprised mostly of plasmablasts with B cells and plasma cells also present. HOXB13+ patients had significantly more plasmablasts (p= 0.017) than HOXB13– patients and these plasmablasts were typically found associated with B cells or plasma cells in a plasmablast-rich neighborhood and not associated with CD8+ T cells or any of the other cytotoxic neighborhood members.Conclusions: A plasmablast-rich immune neighborhood is more frequently seen in HOXB13+ rather than HOXB13– TNBCs, and such a neighborhood is not associated with CD8+ T cells. We have recently shown that HOXB13 expression in a preclinical model of TNBC impedes anti-tumor CD8 T-cell immunity. Our findings raise the possibility that plasmablasts may directly suppress CD8+ T cells or that the formation of these plasmablast-rich neighborhoods occupies a niche that comes at the cost of anti-tumor/cytotoxic neighborhoods. Understanding the molecular underpinnings of this process could provide for novel immunotherapeutic strategies.
Presentation numberPS4-07-09
Can high TILs compensate for incomplete neoadjuvant pembrolizumab in early TNBC? Insights from the PETRHA cohort
Daniela Vazquez-Juarez, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, Mexico
D. Vazquez-Juarez1, A. Aranda-Gutierrez2, Y. Chavarri-Guerra2, F. Petracci3, O. Peña-Curiel1, F. Acevedo4, E. Zamudio Lozoya5, L. Gonzalez Gonzalez5, W. Mantilla6, S. Franco7, C. Arce Salinas8, K. Centelles López8, H. Gomez9, P. Carreon10, E. Aguirre Alvarez11, A. Lopez-Galindo12, B. Martinez-Cannon2, M. Garcia Garces13, E. Korbenfeld14, A. Benitez-Cruz14, E. Willars15, M. Lema16, C. Lema17, C. Villarreal-Garza1; 1Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, MEXICO, 2Departamento de Hemato-Oncología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, MEXICO, 3Departamento de Oncología Clínica, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 4Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, CHILE, 5Unidad de Cancer de Mama, Centro Estatal de Cancerologia de Chihuahua, Chihuahua, MEXICO, 6Breast Cancer, Luis Carlos Sarmiento Angulo Center for Research and Cancer Treatment, Bogota, COLOMBIA, 7Breast Cancer, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Bogota, COLOMBIA, 8Unidad funcional de Cancer de mama, Instituto Nacional de Cancerología, Tlalpan, MEXICO, 9Oncology, Oncosalud-AUNA, Lima, PERU, 10Oncology, Oncosalud-AUNA, Lima, MEXICO, 11Breast Cancer Unit, SOHEC – Sociedad de Oncología y Hematología del Cesar, Valledupar, COLOMBIA, 12Oncology, Doctors Hospital-AUNA, Monterrey, MEXICO, 13Oncology, ISSEMYM Hospital Regional de Toluca, Toluca, MEXICO, 14Oncology, Hospital Británico Buenos Aires, Buenos Aires, ARGENTINA, 15Oncology, Centro Oncológico Lic. Salvador Chavarría Delgado, Saltillo, MEXICO, 16Oncology, Clínica de Oncología Astorga, Medellin, COLOMBIA, 17Oncology, Clínica de Oncología Astorga, San Pedro Garza Garcia, COLOMBIA.
Tumor-infiltrating lymphocytes (TILs) are an established predictive and prognostic biomarker in early triple-negative breast cancer (TNBC), associated with higher pathologic complete response (pCR) rates and favorable outcomes following chemoimmunotherapy. However, in real-world settings, access limitations, immune-related adverse events, and logistical challenges can result in early discontinuation of neoadjuvant immunotherapy. Previously, our collaborative group reported that an incomplete number of neoadjuvant pembrolizumab cycles was associated with a significantly lower pCR rate. Whether high baseline TIL levels can mitigate the impact of incomplete pembrolizumab exposure on pCR remains unknown. Patients with early TNBC who received neoadjuvant chemotherapy and pembrolizumab within the PETRHA retrospective cohort were included. Stromal TILs were assessed on pre-treatment core biopsies by local pathology teams and analyzed using two predefined cut-offs: >20% and >30%. Patients were stratified by pembrolizumab treatment completion: those who received all planned neoadjuvant cycles versus those who did not. The primary outcome was pCR rate, defined as ypT0/is ypN0. Descriptive statistics were used to summarize clinicopathological features. Associations between TILs levels, neoadjuvant pembrolizumab completion, and pCR rate were evaluated using chi-square or Fisher’s exact tests, with logistic regression used to explore associations and estimate odds ratios (OR) with 95% confidence intervals (CI). Of the 304 patients included in the PETRHA cohort, TILs data were available for 111 patients (36.5%). Among these, 59 patients (53.2%) had TILs <20% and 52 (46.8%) had TILs ≥20%, while 70 (63.1%) had TILs <30% and 41 (36.9%) had TILs >30%. Thirty-eight (34.2%) received an incomplete number of neoadjuvant pembrolizumab cycles, while 73 (65.8%) completed the regimen. Among patients who did not complete the neoadjuvant pembrolizumab cycles, those with higher TILs, whether defined as ≥20% or >30%, had significantly higher pCR rates (76.9%) compared to those with lower TILs (32.0%) (p=0.016 for both cutoffs). Similarly, among patients who completed all neoadjuvant cycles, higher TILs were again associated with increased pCR rates (>20% cut-off: 71.8% vs. 38.2%, p=0.004; and >30% cut-off: 85.7% vs. 37.8%, p<0.0001). In a multivariable logistic regression model including variables with a p-value <0.10 in univariate analysis, only TILs >30% (OR 12.74, 95% CI 2.68-60.49; p=0.001) and completion of neoadjuvant chemotherapy (OR 4.95, 95% CI 1.04-23.69; p=0.045) were independently associated with higher probability of achieving pCR. In contrast, TILs >20%, completion of neoadjuvant anthracyclines, and completion of neoadjuvant pembrolizumab were not significantly associated with pCR (p=0.633, p=0.318, and p=0.866, respectively). In our study, higher baseline TILs levels, particularly >30%, were associated with increased pCR rates, irrespective of neoadjuvant pembrolizumab completion. Notably, patients with TILs >30% who did not complete all pembrolizumab cycles had higher pCR rates than those with TILs <30% who completed the full neoadjuvant regimen (76.9% vs. 37.8%), suggesting that robust immune infiltration may partially mitigate reduced immunotherapy exposure. However, the highest pCR rate was observed in patients with both high TILs and complete neoadjuvant pembrolizumab treatment (85.7%), supporting a potential synergistic effect between host immune activation and full immunotherapy administration.
Presentation numberPS4-07-10
The Relationship Between Tumor-Infiltrating Lymphocytes (TILs), Pathologic Complete Response, and Event-Free Survival in Patients with Early-Stage Triple-Negative Breast Cancer Treated with KEYNOTE-522 regimen in a Real-World Scenario
Renata Colombo Bonadio, Instituto D’Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil
R. C. Bonadio1, M. C. Tavares2, F. C. Balint2, G. Ferreira3, C. dos Anjos4, D. Gagliato5, M. L. de Brito6, D. Assad-Suzuki7, D. D. Rosa8, N. J. Gomes9, N. C. Nunes10, L. Testa1, M. Rigesti3, V. Baro3, I. M. de Sousa2, M. O. Andrade11, M. Gouveia12, F. Madasi13, J. Bines13, R. P. Ferreira8, C. L. Santos14, M. Tavares15, M. Monteiro16, Z. S. de Souza17, A. U. Gomes5, B. M. Zucchetti12, A. Ferrari18, M. F. Monteiro19, P. A. Signorini20, A. Aguilar3, S. Sanches2, P. G. Hoff1, M. Estevez-Diz21, R. Barroso-Sousa11; 1Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), São Paulo, BRAZIL, 2Oncology, A.C.Camargo Cancer Center, São Paulo, BRAZIL, 3Oncology, SUMA (Grupo Cooperativa Argentino para el estudio y la investigación del Cáncer de Mama), Buenos Aires, ARGENTINA, 4Oncology, Hospital Sírio-Libanês, São Paulo, BRAZIL, 5Oncology, Hospital Beneficência Portuguesa, São Paulo, BRAZIL, 6Oncology, Clínica AMO, Salvador, BRAZIL, 7Oncology, Hospital Sírio-Libanês, Brasilia, BRAZIL, 8Oncology, Hospital Moinhos de Vento, Porto Alegre, BRAZIL, 9Oncology, Hospital São Domingos, DASA Oncologia, São Luiz, BRAZIL, 10Oncology, Grupo Américas, Rio de Janeiro, BRAZIL, 11Oncology, Brasilia Hospital, Oncologia Américas, Brasília, BRAZIL, 12Oncology, Hospital 9 de Julho, Grupo Américas, São Paulo, BRAZIL, 13Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, BRAZIL, 14Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), Recife, BRAZIL, 15Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), Salvador, BRAZIL, 16Oncology, Grupo Américas, Hospital Samaritano, São Paulo, BRAZIL, 17Oncology, Hospital Sírio-Libanês, Brasília, BRAZIL, 18Oncology, Hospital Santa Paula, Grupo Américas, São Paulo, BRAZIL, 19Oncology, Instituto do Câncer do Ceará, Fortaleza, BRAZIL, 20Oncology, Centro Integrado de Pesquisa da Amazônia (CINPAM), Manaus, BRAZIL, 21Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRAZIL.
Background: Tumor-infiltrating lymphocytes (TILs) are established prognostic and predictive biomarkers in early-stage triple-negative breast cancer (eTNBC). The addition of pembrolizumab to neoadjuvant chemotherapy (NAC) has demonstrated improved outcomes in this setting (KEYNOTE-522 trial). The role of TIL levels in this context needs to be further explored. Methods: The Neo-Real / GBECAM-0123 study is a retrospective multicenter cohort including patients (pts) with stage II-III TNBC treated with pembrolizumab plus NAC across institutions in Brazil and Argentina since 2020. Baseline stromal TILs were assessed on pre-treatment biopsies according to international guidelines and categorized as <30%, 30-49%, or ≥50%. Associations with pCR and event-free survival (EFS) were evaluated using logistic and Cox regression models, respectively. Results: A total of 248 pts were included (median age: 44 years; 70.8% with stage II disease). TILs <30%, 30-50%, and ≥50% were observed in 72.6%, 12.9%, and 14.5% of pts, respectively. In multivariable logistic regression including TILs, Ki-67 index, clinical stage, tumor grade, and number of neoadjuvant pembrolizumab cycles, both TILs ≥50% (OR 6.96, 95% CI 1.88-25.65, P=0.004) and Ki-67 ≥50% (OR 4.88, 95% CI 2.31-10.32, P<0.001) were strongly associated with pCR. Nearly all pts with both high TILs and high Ki-67 achieved pCR (Table).With a median follow-up of 24 months, pts with pCR had significantly higher 2-year EFS compared to those with residual disease (95.9% vs. 76.5%, P<0.001). In the multivariable Cox model, pCR and clinical stage remained independent predictors of EFS, whereas TILs were not. Pts who achieved pCR had an 80% lower risk of recurrence or death compared to those with residual disease (HR 0.20, 95% CI 0.07-0.54, P=0.002). Conversely, pts with stage III disease had significantly worse EFS compared to those with stage II (HR 3.68, 95% CI 1.58-8.53, P=0.002).Among pts with pCR, 2-year EFS was 97.0% in TILs < 30%, 95.2% in TILs 30-50% (HR 1.26, 95% CI 0.12-12.42, P=0.839), and 93.2% in TILs ≥50% (HR 1.74, 95% CI 0.28-10.53, P=0.544). Among those with residual disease, 2-year EFS was 73.3% in TILs < 30%, 90% in TILs 30-50% (HR 0.33, 95% CI 0.04-2.51, P=0.288), and 100% in TILs ≥50% (HR 1.39e-15, 95% CI NA, P=1.000; analysis limited by small number of pts in this group of TILs ≥50% with residual disease). Conclusion: In this real-world cohort of pts with eTNBC treated with the KEYNOTE-522 regimen, high baseline TILs and Ki-67 index were strongly associated with higher pCR rates. These findings support the role of TILs as a relevant biomarker for treatment response. Long-term outcomes were primarily driven by pathologic response and disease stage, underscoring the importance of achieving pCR.
| Any Ki67 | Ki67 < 50% | Ki67 ≥ 50% | |
| TILs < 30% |
59.0% (n=95/161) |
39.6% (n=19/48) |
67.6% (n=75/111) |
| TILs 30-50% |
65.6% (n=21/32) |
00.0% (n=0/1) |
67.7% (n=21/31) |
| TILs ≥50% |
91.4% (n=32/35) |
75.0% (n=6/8) |
96.3% (n=26/27) |
| P-value | 0.001 | 0.120 | 0.004 |
Presentation numberPS4-07-11
Analysis of patients with triple-negative breast cancer and germline BRCA mutation undergoing neoadjuvant treatment following the keynote 522 protocol in real-world. Insights from the Neo-Real/GBECAM-0123 Study
Monique C Tavares, AC Camargo, SAO PAULO, Brazil
M. C. Tavares1, F. C. Balint1, R. B. Barroso-Sousa2, S. Sanches1, L. Testa3, G. Colucci4, N. C. Nunes5, B. G. Giner4, M. Gill4, I. S. Martins1, M. Andrade6, M. Gouveia7, F. Madasi8, J. Bines8, R. Ferreira9, D. Rosa9, C. Santos10, M. Monteiro11, Z. S. Souza12, D. Assad-Suzuki12, C. Anjos13, D. M. Gagliato14, A. Gomes14, B. Zucchetti15, A. Ferrari10, M. Brito16, M. F. Monteiro17, P. A. Signorini18, N. J. Gomes19, S. Mazzotta4, P. M. Hoff20, M. Estevez-Diz21, R. R. Bonadio22; 1Oncology, AC Camargo, SAO PAULO, BRAZIL, 2Oncology, Brasilia Hospital, Oncologia Américas, Brasília, Brazil., SAO PAULO, BRAZIL, 3Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil, SAO PAULO, BRAZIL, 4Oncology, SUMA (Grupo Cooperativa Argentino para el estudio y la investigación del Cáncer de Mama), Buenos Aires, ARGENTINA, 5Oncology, Brasilia Hospital, Oncologia Américas, Brasília, Brazil., BRASILIA, BRAZIL, 6Oncology, Brasilia Hospital, Oncologia Américas, Brasília, Brazil, BRASILIA, BRAZIL, 7Oncology, Grupo Américas, Hospital 9 de Julho, São Paulo, Brazil., SAO PAULO, BRAZIL, 8Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil, RIO DE JANEIRO, BRAZIL, 9Oncology, Serviço de Oncologia, Hospital Moinhos de Vento, Porto Alegre, Brazil, PORTO ALEGRE, BRAZIL, 10Oncology, Grupo Américas, Hospital Santa Paula, São Paulo, Brazil., SAO PAULO, BRAZIL, 11Oncology, GRUPO AMERICAS, HOSPITAL SAMARITANO, SAO PAULO, BRAZIL, 12Oncology, Medical Oncology Department, Hospital Sírio-Libanês, Brasília, Brazil, BRASILIA, BRAZIL, 13Oncology, Medical Oncology Department, Hospital Sírio-Libanês, São Paulo, Brazil, SAO PAULO, BRAZIL, 14Oncology, Centro de Oncologia – Hospital Beneficência Portuguesa, São Paulo, Brazil, SAO PAULO, BRAZIL, 15Oncology, Grupo Américas, Hospital 9 de Julho, São Paulo, Brazil, SAO PAULO, BRAZIL, 16Oncology, DASA Oncology – Clínica AMO, Salvador, Brazil, SALVADOR, BRAZIL, 17Oncology, Instituto do Câncer do Ceará, Fortaleza, Brazil, SAO PAULO, BRAZIL, 18Oncology, Centro intergrado de pesquisa da Amazonia(CINPAM), MANAUS, BRAZIL, 19Oncology, Hospital São Domingos – DASA, SAO PAULO, BRAZIL, 20Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR), SAO PAULO, BRAZIL, 21Oncology, Instituto D’Or de Pesquisa e Ensino (IDOR),, SAO PAULO, BRAZIL, 22Oncology, Instiituto Rede d’or pesquisa e ensino Oncologia, SAO PAULO, BRAZIL.
Background The KEYNOTE-522 study established a new standard of care for early-stage triple-negative breast cancer (TNBC), demonstrating that the addition of pembrolizumab to neoadjuvant chemotherapy, followed by adjuvant pembrolizumab, significantly improves pathological complete response (PCR) rates, event-free survival (EFS), and overall survival (OS). BRCA1/2 mutations define a distinct molecular subgroup of TNBC, characterized by impaired DNA repair and potential differential sensitivity to platinum agents and immunotherapy. Real-world data on this subgroup remain limited. Methods Neo-Real/GBECAM-0123 is a multicenter, real-world study that included patients with early-stage TNBC treated with the KEYNOTE-522 regimen in Brazil and Argentina since July 2020. Patients were categorized as BRCA mutation carriers (mBRCA) or wild-type/unknown (wt/unkBRCA). Clinical, pathological, and treatment-related outcomes were compared using descriptive and inferential statistics. Results Among 726 patients, 104 (14.3%) were mBRCA and 622 (85.7%) were wt/unkBRCA (495 [84.2%] wild-type and 93 [15.8%] unknown). Patients with mBRCA were significantly younger at diagnosis (mean age: 40.2 vs. 45.3 years; p < 0.001). In both groups, the majority of patients had stage II (73.0% and 72.3%) and stage 3 (80.2% and 74.2%) tumors. The pCR rate was significantly higher in the mBRCA group (74.0% vs. 61.4%; p = 0.018). Mastectomy was performed more frequently in mBRCA patients (88.3% vs. 34.7%; p < 0.001), and radiation therapy was also administered less frequently (64.4% vs. 87.1%; p < 0.001). With a median follow-up of 22 months, 82 patients experienced disease recurrence or death. Two-year EFS rates were 92.3% in the mBRCA group and 86.0% in the wt/unkBRCA group (HR 0.51, 95% CI: 0.23-1.11; p = 0.091). Discussion This real-world analysis provides strong evidence that patients with triple-negative breast cancer (TNBC) with mBRCA experience distinct clinical characteristics and superior outcomes when treated with the KEYNOTE-522 regimen. The 12.3% absolute increase in the pCR rate translated into favorable EFS trends. These findings are clinically relevant and may guide future strategies for risk-adapted treatment de-escalation.
Presentation numberPS4-07-12
Real-world comparison of pembrolizumab dosing schedules (q3w vs q6w) in early TNBC: Insights from the PETRHA cohort
Alejandro Aranda-Gutierrez, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
A. Aranda-Gutierrez1, D. Vazquez-Juarez2, Y. Chavarri-Guerra1, F. Petracci3, O. Peña-Curiel2, F. Acevedo4, E. Zamudio Lozoya5, L. Gonzalez Gonzalez5, W. Mantilla6, S. Franco6, M. Bravo6, P. Herrera Ríos7, C. Arce Salinas8, K. Centelles López8, H. Gomez9, P. Carreon9, E. Aguirre Alvarez10, B. Martinez-Cannon1, A. Lopez-Galindo11, M. Garcia Garces12, E. Willars13, E. Korbenfeld14, A. Benitez-Cruz14, M. Lema15, C. Lema15, C. Villarreal-Garza2; 1Departamento de Hemato-Oncología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, MEXICO, 2Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, San Pedro Garza Garcia, MEXICO, 3Departamento de Oncología Clínica, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 4Hematology and Oncology, Pontificia Universidad Católica de Chile, Santiago, CHILE, 5Unidad de Cancer de Mama, Centro Estatal de Cancerologia de Chihuahua, Chihuahua, MEXICO, 6Breast Cancer, Luis Carlos Sarmiento Angulo Center for Research and Cancer Treatment, Bogota, COLOMBIA, 7-, UNEME-DEDICAM Servicios de Salud de Nuevo Leon, Guadalupe, Nuevo León, MEXICO, 8Unidad funcional de Cancer de mama, Instituto Nacional de Cancerología, Mexico City, MEXICO, 9Oncology, Oncosalud-AUNA, Lima, PERU, 10Breast Cancer Unit, SOHEC – Sociedad de Oncología y Hematología del Cesar, Valledupar, MEXICO, 11Oncology, Doctors Hospital-AUNA, Monterrey, MEXICO, 12Oncology, ISSEMYM Hospital Regional de Toluca, Toluca, MEXICO, 13Oncology, Centro Oncológico Lic. Salvador Chavarría Delgado, Saltillo, MEXICO, 14Oncology, Hospital Británico Buenos Aires, Buenos Aires, ARGENTINA, 15Oncology, Clínica de Oncología Astorga, Medellin, COLOMBIA.
Pembrolizumab combined with neoadjuvant chemotherapy has become the standard of care for most patients with early-stage triple-negative breast cancer (TNBC), typically administered at a dose of 200 mg every three weeks (q3w), as studied in the KEYNOTE-522 trial. A 400 mg every six weeks (q6w) dosing schedule was subsequently approved by regulatory agencies based on pharmacokinetic modeling and exposure-response analyses demonstrating comparable drug exposure and similar expected efficacy and safety to the standard q3w regimen. Although the q6w schedule was not evaluated in prospective neoadjuvant trials, its adoption has increased in real-world practice, particularly in settings with logistical constraints. However, comparative clinical data on the safety and efficacy of q6w versus q3w dosing in early TNBC remain limited. Patients with early TNBC who received neoadjuvant chemotherapy and pembrolizumab within the PETRHA retrospective cohort were included. Pembrolizumab was administered either 200 mg every three weeks (q3w) or 400 mg every six weeks (q6w), based on physicians’ choice and local practice. Patients receiving q3w and q6w dosing were compared in terms of treatment completion (defined as receipt of all planned neoadjuvant pembrolizumab cycles), immune-related adverse events (irAEs), and pathologic complete response (pCR) rates, defined as ypT0/is ypN0. Descriptive statistics were used to summarize baseline characteristics. Categorical variables were compared using chi-square or Fisher’s exact tests. A two-sided p-value <0.05 was considered statistically significant. A total of 304 patients were included, of whom 232 (76.3%) received pembrolizumab 200 mg q3w, and 72 (23.7%) received 400 mg q6w. Most patients had stage II (53.9%) or stage III (42.1%) disease, with a similar clinical stage distribution across dosing groups. The use of the q6w schedule was significantly more common in public healthcare settings, with only 0.6% of patients in private institutions receiving pembrolizumab q6w compared to 51.1% in public centers (p<0.001). Treatment completion rates were comparable, with 75.7% of patients in the q3w group and 76.4% in the q6w group completing all planned neoadjuvant pembrolizumab cycles (p=0.60). The pCR rate was higher in the q3w group (62.5%) compared to the q6w group (51.4%), although this difference did not reach statistical significance (p=0.093). irAEs occurred in 37.6% of patients receiving q3w and 25.0% of those receiving q6w dosing (p=0.051), with no statistically significant differences in grade 3 or higher irAEs (12.8% vs. 0%, p=0.205), In this real-world cohort of Hispano-American women with early TNBC, the use of a q6w neoadjuvant pembrolizumab dosing schedule was associated with similar treatment completion rates and a non-significantly lower pCR rate compared to the standard q3w neoadjuvant regimen. The numerically lower incidence and severity of irAEs with q6w dosing suggest a potentially favorable toxicity profile, although the difference was not statistically significant. These findings support the feasibility of q6w neoadjuvant pembrolizumab dosing in clinical practice.
Presentation numberPS4-07-13
Safety and Efficacy of Immune Checkpoint Inhibitors in Early Triple Negative Breast Cancer: A Systematic Review and Meta-Analysis of Real-World Evidence vs. Clinical Trial Data
Alessandra Longobardi, University of Naples “Federico II”, Naples, Italy
A. Longobardi1, R. Buonaiuto2, C. Calderaio2, A. Caltavituro1, V. Cantile1, G. Crimaldi1, F. Puglisi3, L. Del Mastro4, C. De Angelis5, M. De Laurentiis2, M. Giuliano1, G. Arpino1; 1Oncology Unit, Department of Clinical Medicine and Surgery,, University of Naples “Federico II”, Naples, ITALY, 2Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Naples, ITALY, 3Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy, Aviano, ITALY, 4Department of Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy., Genova, ITALY, 5Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, ITALY.
Background: Immune checkpoint inhibitors (ICIs) in combination with chemotherapy, have markedly transformed the therapeutic alogorithm of high risk early-stage triple-negative breast cancer (eTNBC). Although the safety and efficacy of ICIs have been extensively evaluated in randomized clinical trials (RCTs), their toxicity and effectiveness profiles may differ in real-world clinical practice. This systematic review and meta-analysis aimed to compare the incidence of all-grade adverse events (AEs), grade ≥3 AEs, treatment related discontinuation, and pathological complete response (pCR) rates across phase I-III RCTs and real-world evidence (RWE) in patients with eTNBC receiving neoadjuvant ICIs. Methods: Fourteen RCTs and ten RWE studies were included in the analysis. Incidence of all-grade AEs, grade ≥3 AEs, treatment related discontinuation and pCR rate were collected. Proportions were logit-transformed to stabilize variances and pooled using a random-effects model with Hartung-Knapp adjustment. Subgroup differences between RWE and RCTs were evaluated using the Q-test for subgroup analysis. Separate analyses were conducted for pCR, overall AEs, grade ≥3 AEs and treatment discontinuation to explore consistency of findings across different outcomes. Results: The pooled incidence of all-grade AEs was higher in RCTs compared to RWE, with estimates of 93% (95% CI: 78-98%) and 80% (95% CI: 27-98%), respectively. However, the difference between subgroups did not reach statistical significance in the random-effects model (Q = 1.18, p = 0.28). Similarly, the incidence of grade ≥3 AEs was 49% in RCTs (95% CI: 27-71%) versus 30% in RWE (95% CI: 14-52%), with no significant difference observed between groups (Q = 1.85, p = 0.17). Treatment discontinuation rates were consistent across study types, with both RCTs and RWE showing a pooled incidence of 19%, with no difference between subgroups (Q = 0.00, p = 0.97). Regarding pCR, the pooled rate was 58% (95% CI: 53-62%) in RCTs and 54% (95% CI: 43-65%) in RWD, with no statistically significant subgroup difference (Q = 0.46, p = 0.50). Conclusions: The incidence and severity of adverse events and pCR rates, were comparable between randomized clinical trials and real-world evidence, indicating that immunotherapy maintains consistent safety and effectiveness profiles across both contexts. These results reinforce the external validity of trial-derived data for application in routine clinical practice. Ongoing post-marketing surveillance and standardized reporting remain crucial to ensure patient safety, optimize outcomes, and inform evidence-based treatment decisions.
Presentation numberPS4-07-14
Germline BRCA Mutations and Chemo-Immunotherapy Predict Pathologic Complete Response in Triple-Negative Breast Cancer: Insights from a Resource-Limited Setting
Tamer Al-Batsh, King Hussein Cancer Center, Amman, Jordan
T. Al-Batsh, F. Tamimi, M. Horani, B. Sharaf, H. Bani Hani, L. El Saket, A. Issa, Z. Muhanna, O. Almuhaisen, O. Mahafdah, A. Shammout, M. El-Atrash, M. Abunasser, H. Abdel-Razeq; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Introduction: Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, often diagnosed at a younger age and associated with limited therapeutic options. Despite advances in systemic therapy, long-term outcomes remain suboptimal, particularly in resource-limited settings. This study aims to characterize the clinical features, germline mutation patterns, and survival outcomes of TNBC patients treated in Jordan, with a focus on real-world response to neoadjuvant chemo-immunotherapy and the prognostic value of germline genetic findings. Methods: This retrospective cohort study included patients diagnosed with TNBC between 2003 and 2023 at King Hussein Cancer Center. Demographic, clinical, pathological, and treatment-related data were extracted from medical records. All patients underwent germline genetic testing as part of their workup. Pathological complete response (pCR; defined as ypT0/is, ypN0), event-free survival (EFS), and overall survival (OS) were estimated using the Kaplan-Meier method. Multivariable logistic regression was performed to identify independent predictors of pCR. Results: A total of 526 patients with TNBC were included, predominantly Jordanian (473, 89.9%), with a median age at diagnosis of 45 years (range: 36-54). Notably, 165 patients (31.4%) were diagnosed before the age of 40, indicating a high proportion of young patients. A personal history of non-breast malignancy was reported in 35 patients (6.6%), and 11 (2.1%) had a prior diagnosis of breast cancer with a different hormone receptor profile. A positive family history of cancer was documented in 407 patients (77.4%). At presentation, 494 patients (94.0%) had non-metastatic disease, including 283 (52.7%) with locally advanced tumors. Histopathological analysis revealed invasive breast carcinoma of no special type (NST) in 434 patients (82.5%), with grade 3 histology in 422 cases (80.2%). HER2-low status was identified in 95 patients (18.1%). Germline genetic testing revealed pathogenic or likely pathogenic (P/LP) variants in 117 patients (22.2%), most commonly BRCA1 (77 patients, 65.8%) and BRCA2 (31 patients, 26.5%). Among the 494 non-metastatic patients, 337 (68.2%) received neoadjuvant therapy, including 74 (21.9%) who received chemo-immunotherapy. This was associated with a significantly higher pCR rate compared to chemotherapy alone (59.5% vs. 39.9%; OR: 2.2, p = 0.003). In multivariable analysis, both neoadjuvant immunotherapy (OR: 2.18, p = 0.006) and the presence of P/LP mutations (OR: 1.95, p= 0.017) were independently associated with increased odds of achieving pCR, while age, HER2 status, tumor grade, and clinical stage were not significant predictors. Mastectomy was performed in 281 patients (56.9%), and among those with P/LP variants, 60.8% underwent risk-reducing contralateral mastectomy and 51.6% had bilateral salpingo-oophorectomy. After a median follow-up of 55.8 months, the 5-year event-free survival (EFS) among patients who presented with non-metastatic disease was 74.2% (95% CI: 70.0%-78.7%). In subgroup analysis, 5-year EFS was similar in patients with P/LP variants and those without; 77.5% (95% CI: 68.6%-87.6%) compared to 73.1% (95% CI: 68.3%-78.2%), p= 0.205. Conclusion: This study highlights the clinicopathologic diversity and outcomes of TNBC patients in a Middle Eastern population. A notable proportion carried BRCA1/2 mutations, supporting routine genetic testing. Adding immunotherapy to neoadjuvant chemotherapy improved pCR rates. Long-term outcomes were favorable, with no survival differences by mutation status.
Presentation numberPS4-07-15
A single preoperative pembrolizumab dose plus a single subablative radiotherapy fraction (7 Gy) elicits anti-tumor immune response and increases stromal tumor infiltrating lymphocytes in triple negative breast cancer: a phase 1b/2 study
Julia Tchou, University of Pennsylvania, Philadelphia, PA
J. Tchou1, A. Nayak2, H. N. Xu3, J. Kollmar1, E. H. Smith1, M. Z. Mazur1, L. Keele1, A. Clark4, N. Taunk5, J. Fraietta6, on behalf of the BreastVax Study Team; 1Surgery, University of Pennsylvania, Philadelphia, PA, 2Pathology, University of Pennsylvania, Philadelphia, PA, 3Radiology, University of Pennsylvania, Philadelphia, PA, 4Medicine, University of Pennsylvania, Philadelphia, PA, 5Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 6Microbiology, University of Pennsylvania, Philadelphia, PA.
Background Neoadjuvant chemotherapy combined with immune checkpoint blockade (ICB) improves outcomes in high-risk TNBC, but this regimen is not approved and may be overtreatment for cT1N0 TNBC. Preclinical studies show that low dose radiotherapy (RT) can enhance antitumor immunity. In this study, we evaluated whether a neoadjuvant chemotherapy-free regimen of a single pembrolizumab (pembro) dose + a single subablative RT fraction (7Gy) may elicit antitumor immune response and increase stromal tumor infiltrating lymphocytes (sTILs) in TNBC to high levels, defined here as > 50%, which is a biomarker known to correlate with excellent outcomes without chemotherapy. Methods In this open-label phase 1b/2 trial (NCT04454528), participants with histologically confirmed early stage invasive breast cancer planned for standard of care (SOC) upfront surgery were enrolled. Each participant received a single preop dose of pembro either alone or in combination with RT given before or after pembro. The co-primary endpoints were demonstration of 1) feasibility of completing RT + pembro regimen within 21 days without delaying planned surgery > 14 days and 2) immune activation, defined as a) increase in % sTILs, and, b) increase in % proliferating (Ki67+) circulating T cells. Secondary objectives included assessment of tumor response by change in size from baseline. Responders are defined as those with tumor size reduction > 30% (TΔ30). Study sample size was powered for TΔ30. Assuming 40% of treated subjects achieve TΔ30 versus 0% in historical controls, 15 subjects yield 80% power at α=0.05. Exploratory analyses included immune profiling of peripheral blood mononuclear cells (PBMC) by flow cytometry for markers of T-cell activation, proliferation, differentiation and exhaustion, T-cell receptor repertoire (TCR) dynamics, and digital spatial profiling of the tumor immune microenvironment pre and post treatment. All participants received SOC adjuvant therapy after surgery. Findings Between January 6, 2021, and February 18, 2025, 30 participants (12 with HR+HER2-, 17 TNBC and one HER2+) enrolled and completed the preop regimen. All participants proceeded to surgery without delay. Median follow-up was 29 months (IQR 14 – 45). There were no adverse events (AEs) ≥ grade 2 which included one grade 2 immune-related AE (hypothyroidism). TΔ30 rate was significantly higher in RT + pembro arms (14 of 25, 56%, including 3 with pCR) compared to pembro only arm (0 of 5, 0%), p = 0.024. Post-treatment, sTILs shifted rightward from 20% (IQR 5 – 50 ) to 50% (IQR 10 – 60) overall and from 40% (IQR 20 – 55) to 60% (IQR 50 – 70) in TNBC. The proportion of TNBC with high sTILs (>50%) increased from 3 of 11 (27%) to 7 of 13 (54%) post treatment. Immune profiling of PBMC from responders demonstrated increased proportions of early-memory CD4⁺ and CD8⁺ T cells at baseline, marked expansion of activated, Ki67+ CD4+ and CD8⁺ T cells post treatment, and a higher and more diverse TCR repertoire both pre- and post-treatment. Spatial transcriptomic analyses demonstrated that tumor regions directly interacting with T-cells in responders were enriched for gene signatures of cGAS-STING-pathway activation, antigen processing and presentation, and response to ICB. Interpretation In this small single institution study, we demonstrate that a single pembro dose + a single sub-ablative RT fraction can elicit anti-tumor immune response and increase sTILs from low to high levels in TNBC. As only ~20% TNBC have high sTILs, this well tolerated neoadjuvant chemotherapy-free regimen may be evaluated in future studies as an immune induction strategy to expand the number of patients eligible to enroll in adjuvant chemotherapy omission trials such as the OPTImaL study.
Presentation numberPS4-07-16
Epigenetic Reactivation of estrogen receptor beta Enhances Antitumor Activity of a Novel estrogen receptor beta agonist CIDD-0149897 in Triple-Negative Breast Cancer
Uday Pratap, UT Health San Antonio, San Antonio, TX
U. Pratap1, M. Mahajan1, K. Nassar1, T. Adeniran1, N. Karinel2, G. Sareddy1, N. Mukherjee1, S. Viswanadhapalli1, S. McHardy2, A. Brenner1, R. Vadlamudi1; 1OBGYN, UT Health San Antonio, San Antonio, TX, 2OBGYN, UTSA, San Antonio, TX.
Background: Breast cancer (BC) is the most common cancer in women, with ~40,000 annual deaths in the U.S. Triple-negative breast cancer (TNBC), comprising 15-24% of cases, is aggressive and linked to high mortality. TNBC lacks ER alpha (ERa), PR, and HER2 but uniquely expresses ER beta (ERb), a tumor suppressor. However, ERb-targeted therapies remain underdeveloped due to limited selective agonists and epigenetic silencing in advanced disease. We identified CIDD-0149897, a novel ERb agonist, and evaluated its activity alone and with HDAC inhibitors (HDACi) to enhance ERb expression and therapeutic efficacy in TNBC. Methods: CIDD-0149897 was evaluated in seven TNBC cell lines (SUM159, BT549, MDA-MB-468, HCC1806, HCC70, MDA-MB-231, E0771). ERb expression was measured by RT-qPCR and Western blot. Functional assays included cell proliferation, colony formation, invasion, and apoptosis following treatment with CIDD-0149897, HDACi, or combination therapy. Mechanistic studies involved RNA sequencing, reporter assays, immunohistochemistry, and pathway analysis. In vivo efficacy was assessed using TNBC xenograft and syngeneic mouse models. Results: Expression analysis of ERb across seven TNBC cell lines (SUM159, BT549, MDA-MB-468, HCC1806, HCC70, MDA-MB-231, and E0771) revealed detectable but variable levels of ERb mRNA and protein. Treatment with the selective ERb agonist CIDD-0149897 led to a dose-dependent decrease in cell viability across multiple TNBC models. Treatment with CIDD-0149897 significantly induced apoptosis. ERb reporter assays confirmed increased transcriptional activity following CIDD-0149897 exposure. Overexpression of ERb further sensitized cells to treatment, while ERb knockout attenuated response. HDAC inhibitors (HDACi) upregulated ERb mRNA and protein expression, with chromatin immunoprecipitation confirming increased acetylation at the ERb promoter. Combination treatment with CIDD-0149897 and HDACi synergistically inhibited proliferation, reduced clonogenic survival, and suppressed invasion more effectively than either agent alone. RNA-seq and RT-qPCR showed enhanced activation of ERβ target genes involved in apoptosis and cell cycle arrest. In vivo, monotherapy with CIDD-0149897 or HDACi modestly inhibited tumor growth in HCC-1806 xenograft and E0771 syngeneic models. However, combination therapy significantly suppressed tumor volume over time, prolonged survival, and reduced tumor cell proliferation (Ki-67 staining). Immunohistochemical analysis revealed upregulation of ERb and increased cleaved caspase-3 in tumors treated with the combination therapy. In the immunocompetent E0771 model, combination therapy not only inhibited tumor progression but also enhanced infiltration of immune cells, suggesting favorable modulation of the tumor immune microenvironment. Conclusion: These findings demonstrate that CIDD-0149897, in combination with HDAC inhibition, effectively reactivates ERb signaling and modulates the tumor microenvironment. This combinatorial strategy offers promising therapeutic potential for TNBC and provides critical insights that may guide the development of more effective, targeted treatments.
Presentation numberPS4-07-17
Results of the Dose-Expansion Cohort of a Phase 1 Trial of Intratumoral HER2- and HER3-Primed Dendritic Cells Injections for the Treatment of Early-Stage TNBC and HR Low Positive Breast Cancer.</b>DecipHER trial</b>
Ricardo Costa, H. Lee Moffitt Cancer Center, New Tampa, FL
R. Costa, A. Soyano, A. Armaghani, N. Abdo, S. Wallace-Morrison, M. Al-Jumayli,, L. Loftus, E. Abraham, J. Whiting,, Q. Mo, Z. Jameel, T. O’Connor, K. Dvir, S. Hoover, J. Kiluk, M. Lee, C. Laronga, N. Khakpour, H. Han, H. Soliman, B. Czerniecki; Breast Oncology, H. Lee Moffitt Cancer Center, New Tampa, FL.
Title : Results of the Dose-Expansion Cohort of a Phase 1 Trial of Intratumoral HER2- and HER3-Primed Dendritic Cells Injections for the Treatment of Early-Stage TNBC and HR Low Positive Breast Cancer.DecipHER trial Authors : Ricardo L B Costa, Aixa E. Soyano, Avan Armaghani, Neveen Abdo, Shere Wallace-Morrison, Mohammed Al-Jumayli, Loretta Loftus, Edith Abraham, Junmin Whiting, Qianxing Mo, Zena Jameel, Tracey O’Connor, Kathrin Dvir, Susan Hoover, John Kiluk, Catherine Lee, Christine Laronga, Nazanin Khakpour, Hyo S. Han, Hatem H. Soliman, MD, Brian J. Czerniecki. Abstract Background : Patients (pts) with breast cancer (BC) harboring low expression of hormone receptors (HR) and human epidermal growth factor receptor-2 (HER2) have poorer outcomes compared to other subsets of BC. Results from the KEYNOTE-522 trial showed that activation of the immune system using a PD1/PD-L1-targeted approach leads to clinically meaningful improvement in the outcomes of patients with these high-risk tumors. Dendritic cells (DCs) are antigen presenting cells which are pivotal for robust cytotoxic responses via broader activation of the adaptive immune system against tumor-associated antigens. Methods : DecipHER is a dose-escalation, dose-expansion phase 1 trial designed to assess the safety and the preliminary efficacy of autologous, HER2- and HER3-primed DCs in combination with KEYNOTE 522 regimen. Pts with clinical stage cT1cN1/2 or cT2-4cN0-2, HR 20%, HER2-negative BCs are eligible. Patients with inflammatory BC and uncontrolled immune-mediated diseases are excluded. After collection through apheresis, autologous DCs are primed ex vivo against 6 HER2 and 8 HER3 immunogenic peptides. Pts receive alternating ultrasound-guided intratumoral HER2 and HER3 DCs injections administered twice a week for 8 doses starting 2 weeks prior to neoadjuvant therapy with KEYNOTE 522 regimen. The dose-escalation phase of the study had a classic 3+3 design (ie, DL1-3 [10-20, 30-50, 80-100 million], n=12). Additionally, 12 pts were treated at the maximum tolerated dose (MTD) of 80-100 million in the dose-expansion cohort. Endpoints included the absolute risk of adverse events, complete pathological response rate (PCR) and recurrence-free survival. Tumor tissue, blood and stool samples were collected for correlative analyses. Herein we report the safety results of the dose-expansion cohort ; 6 pts treated at DL3 were added for analysis of PCR. Results : A total 12 patients were enrolled into the dose-expansion cohort between 06/2024 and 09/2024. The median age in years was 53.5 (32-76) and 16.7% of pts were black; 33.3% had HER2-low, 16.7% had grade 3, 22.2% had T3 and 61.1% had N1 BCs.All patients received all planned vaccines. Grades 1 and 2 AEs that were at least possibly related to DCs (risk 10%) were fever (58.3%), chills (50%), pain (33.3%), headache (25%) fatigue (16.7%) and nausea (16.7%). No DC-associated grade ≥ 3 AEs were observed. None of the toxicities met the definition of DLT. Four pts had an SAE (i. bronchitis, ii. febrile neutropenia, iii. pulmonary embolism and iv. bilateral lower extremity weakness at weeks 13, 18, 22 and 14; respectively). One immune-related AEs was observed, hypothyroidism (1 pt). PCR was observed in 77.8% of the pts and it was observed at higher frequency among pts with T2 vs T3 tumors (92.3 vs 50%). Conclusion : Intratumoral DCs in combination with standard neoadjuvant chemotherapy and pembrolizumab showed preliminary efficacy and were well tolerated in pts with high-risk HR 20%, HER2-negative BC. This trial is ongoing to further assess the event-free survival of this novel treatment; correlative analyses will follow. The study is open at H. Lee Moffitt Cancer Center. Clinical trial information : NCT05504707 Funding : Shulas’ foundation.
Presentation numberPS4-07-18
Association of tumor-infiltrating lymphocytes (TILs) with outcomes in patients with early TNBC treated with neoadjuvant chemotherapy with or without pembrolizumab
Richard S Lee, Dana-Farber Cancer Institute, Boston, MA
R. S. Lee1, Q. Jin2, B. Binboga Kurt3, B. Koca1, J. Gomez Tejeda Zanudo1, A. Patel1, A. Barkell4, J. Baginska1, O. M. Cunningham1, C. E. Stever1, T. S. Parker1, T. Rahman1, M. Luo1, I. G. Martino1, B. M. Drummey1, S. A. Virani1, K. Santos1, J. Bsat1, C. Snow1, N. Tung5, S. Lo6, M. G. Faggen7, N. Sinclair1, N. Ahmad8, M. Constantinou9, S. Sinclair10, J. L. Meisel11, S. A. Kirschner4, T. A. King12, R. Salgado13, E. A. Mittendorf12, N. U. Lin1, N. Tayob2, E. P. Winer14, E. C. de Bruin4, S. M. Tolaney1, A. Moeini15, A. C. Garrido-Castro1; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Data Science, Dana-Farber Cancer Institute, Boston, MA, 3Pathology, Mass General Brigham, Boston, MA, 4Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UNITED KINGDOM, 5Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, MA, 6Medical Oncology, Stamford Health, Stamford, CT, 7Medicine/Oncology, Dana-Farber Brigham Cancer Center at South Shore Health, South Weymouth, MA, 8Phelps Cancer Center, Berkshire Health System, Pittsfield, MA, 9Rhode Island Hospital Cancer Institute, Brown University Health, Providence, RI, 10Medical Oncology, Northern Light Health, Brewer, ME, 11Hematology and Medical Oncology, Winship Cancer Institute at Emory University, Atlanta, GA, 12Surgery, Brigham and Women’s Hospital, Boston, MA, 13Pathology, ZAS Hospitals, Antwerp, BELGIUM, 14Medical Oncology, Yale Cancer Center, New Haven, CT, 15Translational Medicine, Oncology R&D, AstraZeneca, Barcelona, SPAIN.
Introduction: Neoadjuvant chemotherapy (CT) with pembrolizumab (P) is standard for most patients (pts) with high-risk early TNBC. Biomarkers for the addition of P to CT are lacking. The prognostic and predictive value of TILs in pts who receive neoadjuvant CT+P vs CT alone and the significance of change in TILs from pre- to post-treatment samples remains unclear. Methods: From the prospective DFCI Multicenter TNBC Registry, pts with stage I-III TNBC who received neoadjuvant CT or CT+P were identified. Stromal TILs were scored in tumor samples at baseline (BL) and surgery (if residual disease, RD). TILs were evaluated as a continuous and binary variable using a threshold selected based on BL distribution. Fisher’s exact test was used to compare TILs between CT and CT+P, and between RCB groups. Association with pathologic complete response (pCR) and with recurrence-free (RFS), distant recurrence-free (DRFS) and overall survival (OS) were evaluated using logistic regression and Cox proportional hazards models, respectively, adjusting for clinical stage and neoadjuvant treatment. All statistical tests were two-sided (significance p < 0.05). Results: Between 5/2019-1/2024, 121 TNBC pts were identified; 18 (14.9%) stage I, 49 (40.5%) stage II, 52 (43.0%) stage III, 2 (1.7%) unknown. 19 (15.7%) had germline BRCA1/2 mutations. 49 (40.5%) received CT and 72 (59.5%) CT+P, with anthracycline-based CT in 40 (81.6%) and 62 (86.1%) pts, respectively. The pCR rate was 51.0% (25/49; 95% CI 36.3-65.6%) with CT alone and 58.3% (42/72; 95% CI 46.1-69.8%) with CT+P. Median follow-up was 2.2 years (interquartile range [IQR] 1.4-3.9): CT, 4.2 years (IQR 3.4-5.1); CT+P, 1.7 years (IQR 1.2-2.2). 115 pts had TILs scored at BL and 41 at RD, of whom 35 had paired samples. BL TILs (range 0-90%) were categorized by quartiles (5%, 10%, 30%) with 10% used for primary analysis. Both BL and RD TILs did not differ between CT vs CT+P as a continuous variable (BL p=0.11; RD p=0.56) or ≥10% cutoff (BL p=0.62; RD p=0.56). High BL TILs (≥ 10%) were significantly associated with pCR and this did not differ by treatment (interaction p=0.36). No association was observed between TILs (at BL or RD) and RCB class (TILs as continuous variable: BL p=0.36, RD p=0.84; or 10% cutoff: BL p=0.31, RD p=0.86). High BL TILs (≥ 10%) were associated with longer RFS (aHR 0.15, p=0.01), DRFS (aHR 0.17, p=0.01) and OS (aHR 0.19, p=0.05; or as a continuous variable, aHR per 5-unit increase 0.46, p=0.05). Neither TILs in RD nor increase from BL to RD (in 14/35 pts, 40%) were associated with RFS, DRFS or OS. Conclusion: In this multicenter TNBC cohort, BL TILs were reinforced as a prognostic marker for pCR, regardless of the addition of P to CT. TILs at RD and changes during neoadjuvant therapy were not prognostic. Additional follow-up for survival outcomes is ongoing.
| Number of Events | aHR (≥10% vs. <10%) | CI (95%) | p-value | ||
| RFS | Baseline (n=114) | 14 | 0.15 | 0.04 – 0.63 | 0.009 |
| At Surgery (n=41) | 13 | 0.55 | 0.16 – 1.92 | 0.345 | |
| DRFS | Baseline (n=114) | 14 | 0.17 | 0.04 – 0.68 | 0.013 |
| At Surgery (n=41) | 13 | 0.55 | 0.16 – 1.92 | 0.345 | |
| OS | Baseline (n=114) | 10 | 0.19 | 0.04 – 1.00 | 0.050 |
| At Surgery (n=41) | 7 | 0.35 | 0.05 – 2.21 | 0.263 |
Presentation numberPS4-07-19
An agonist of mitochondrial CLPP, ONC206, overcomes mitochondrial dependencies and abates chemoresistance in triple negative breast cancer
Lily M Baek, Baylor college of medicine, Houston, TX
L. M. Baek1, A. S. Greer1, L. Audra1, J. T. Lei1, J. C. Yang2, L. E. Dobrolecki1, S. F. Faucher1, N. D. Griffith1, C. M. Sallas1, B. Lim3, A. L. Welm2, V. V. Prabhu4, M. T. Lewis1, G. V. Echeverria1; 1Breast Center, Baylor college of medicine, Houston, TX, 2Oncological Sciences, University of Utah, Salt Lake City, UT, 3Breast Medical Oncology, University of Texas, Houston, TX, 4R&D, Chimerix/Jazz Pharmaceuticals, Durham, NC.
Approximately 50% of triple-negative breast cancer (TNBC) patients treated with neoadjuvant chemotherapy such as platinums (e.g., carboplatin, CRB), taxanes (e.g., docetaxel, DTX), and anthracyclines administered alone, in combination, sequentially, or with or without anti-PD1 agents, retain residual cancer burden (RCB). RCB is strongly associated with rapid local and metastatic recurrence and mortality in TNBC. Thus, we conducted preclinical studies to address the urgent need to overcome chemoresistance. Mitochondrial function is known to promote chemoresistance and metastasis in TNBC and other cancers. We previously demonstrated that inhibiting mitochondrial oxidative phosphorylation (OXPHOS) or mitochondrial fusion could overcome chemoresistance in TNBC. In a recent preclinical trial using 50 orthotopic patient-derived xenograft (PDX) models of TNBC, multi-omic profiling revealed that mitochondrial pathways, particularly mitochondrial protein translation and OXPHOS, were significantly associated with resistance to DTX, CRB, or their combination. These findings are corroborated in human TNBC specimens (NCT02547987). Our recent work suggests that chemotherapy can induce the mitochondrial unfolded protein response (mtUPR), a retrograde signaling pathway that rewires nuclear transcription in response to mitochondrial proteotoxic stress. Caseinolytic peptidase P (CLPP), a mitochondrial matrix protease, plays a critical role in this response by targeting key proteins involved in mitochondrial respiration and homeostasis. In cancers, CLPP is essential for maintaining mitochondrial protein quality control. Both its inhibition, which causes protein accumulation, and its hyperactivation, which causes excessive proteolysis, can lead to mitochondrial dysfunction and cell death. Notably, high CLPP mRNA levels correlate with poor TNBC survival, while non-tumor cells are relatively insensitive to CLPP perturbation, suggesting a therapeutic window for targeting mtUPR in TNBC. ONC206, an analog of the first-in-class imipridone/dordaviprone/ONC201 with improved potency, is currently undergoing phase I clinical trials for CNS tumors (NCT04732065 and NCT04541082). We found that ONC206 exhibited strong single-agent activity and significantly enhanced chemosensitivity in select orthotopic PDX models, PDX-derived organoids (PDXOs), and human TNBC cell lines. Two PDX models, BCM-2665 (ONC206 responder) and BCM-0132 (ONC206 non-responder), were evaluated in combination chemotherapy trials. In BCM-2665, ONC206 induced complete regressions and suppressed long-term tumor regrowth when combined with CRB. One mouse showed no detectable tumor at euthanasia on day 225. No such effect was observed with DTX. In contrast, in BCM-0132, which did not respond to ONC206 alone, the addition of DTX but not CRB extended tumor regression. One mouse showed complete tumor clearance at day 190. ONC206 was tolerated in mouse trials even when combined with chemotherapy. To elucidate the mechanism of ONC206 activity, we knocked out CLPP, which completely abolished its efficacy. ONC206 treatment significantly reduced OXPHOS and levels of CLPP proteolytic substrates while elevating mtUPR-related proteins (PERK, GCN2, and phospho-eIF2α) in a CLPP-dependent manner. Moreover, ONC206 reversed chemotherapy-induced OXPHOS, suggesting that its combination with chemotherapy may further amplify mtUPR signaling and suppress OXPHOS to enhance therapeutic efficacy. Collectively, our findings provide evidence that ONC206 disrupts mitochondrial dependencies in TNBC by driving excessive CLPP-mediated proteolysis, impairing OXPHOS, and activating mtUPR. This represents a novel and potentially promising therapeutic strategy to enhance chemotherapy response in TNBC.
Presentation numberPS4-07-20
Mitomycin C plus carboplatin followed by paclitaxel vs. standard doxorubicin-cyclophosphamide followed by paclitaxel-carboplatin neoadjuvant therapy in BRCA1/2-associated breast cancer
Petr Vladimirovich Krivorotko, NMRC of Oncology named after N.N.Petrov of MoH of Russia, Saint Petersburg, Russian Federation
D. ?. Enaldieva, P. V. Krivorotko; Department of Breast Tumors, NMRC of Oncology named after N.N.Petrov of MoH of Russia, Saint Petersburg, RUSSIAN FEDERATION.
Mitomycin C plus carboplatin followed by paclitaxel vs standard doxorubicin-cyclophosphamide followed by paclitaxel-carboplatin neoadjuvant therapy in BRCA1/2-associated breast cancerKrivorotko P.V. ˡ, Imyanitov, E.N. ˡ, Enaldieva D.A. ˡ, Sokolenko A.P. ˡ, Zhiltsova E.K. ˡ, Gigolaeva L.P. ˡ, Tabagua T.T. ˡ, Komyakhov A.V. ˡ, Nikolaev K.S. ˡ, Pesotsky R.S. ˡ, Ulrikh D.G.ˡ, Amirov N.S. ˡ, Osipenko E. Yu. ˡ, Gorina A.O. ˡ, Semiglazov V.F. ˡˡ National Medical Research Center of Oncology named after. N.N. Petrova, St.Petersburg, Russia. BackgroundBRCA1/2-associated triple-negative breast cancer (TNBC) is an aggressive BC subtype, yet it exhibits high chemosensitivity. Emerging evidence suggest that mitomycin C, either as a monotherapy or combined with platinum agents, demonstrates efficacy in BRCA1/2-driven carcinomas. This clinical trial aimed at comparison of mitomycin C-based versus standard neoadjuvant chemotherapy (NACT) in BRCA1/2-associated TNBC. MethodThe study included 52 BRCA1/2 pathogenic allele carriers diagnosed with T1-3N0-3M0 TNBC. Patients were divided into two groups: an experimental group (n = 26) that received four cycles of mitomycin C 10 mg/m2 plus carboplatin (AUC 5) followed by 12 cycles of weekly paclitaxel 80 mg/m2 (MCbP-T) and a control group (n = 26) that received four cycles of doxorubicin 60 mg/m2 plus cyclophosphamide 600 mg/m2 followed by 12 cycles of weekly paclitaxel 80 mg/m2 plus carboplatin (AUC 2) (AC-TCbP). Therapy efficacy was evaluated based on RECIST criteria and the rate of complete pathological response (pCR) upon surgery. Results Objective responses were recorded in all patients involved in the study. Clinical complete response was achieved in 18/26 (69%) and 15/26 (58%) patients in the MCbP-T and AC-TCbP arms, respectively (p = 0.57, Fisher’s exact test). In the MCbP-T arm, the frequency of pCR was 21/26 (81%). This was numerically but statistically non-significantly higher than that in the AC-TCbP group (16/26; 62%; p = 0.2, Fisher’s exact test). In the AC-TCbP arm, grade III leukopenia and neutropenia occurred in 11/26 (42%) and 12/26 (46%) patients, respectively. In MCbP-T arm, grade III leukopenia and neutropenia were documented in 1/26 (4%; p = 0.002, Fisher’s exact test) and 2/26 (8%; p = 0.003, Fisher’s exact test) patients, respectively. The frequency of thrombocytopenia of all grades was non-significantly higher in MCbP-T group. All patients in the control group had alopecia grade II, whereas patients in the MCbP-T group didn’t report this side effect. ConclusionIn BRCA1/2-associated TNBC, the efficacy of MCbP-T regimen is comparable with standard anthracycline-based platinum-containing chemotherapy but demonstrates a significantly lower rate of adverse effects. Keywords: BRCA1 / 2 mutations, neoadjuvant chemotherapy, pathological complete response, triple-negative breast cancer, mitomycin C
Presentation numberPS4-07-21
Safety and Clinical Outcomes of Pembrolizumab Standard Dosing Versus Extended-Interval Dosing in Patients with Breast Cancer
Alexis LeVee, UCLA David Geffen School of Medicine, Los Angeles, CA
A. LeVee1, A. Kordic2, N. Ruel3, J. Mortimer2, I. Kang2, H. McArthur4, M. G. Lechner5, K. Tsai6; 1Department of Medicine, Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 2Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, 3Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, 4Department of Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, 5Department of Medicine, Division of Endocrinology, UCLA David Geffen School of Medicine, Los Angeles, CA, 6Department of Diabetes, Endocrinology and Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA.
Introduction: Extended-interval (EI) dosing of pembrolizumab (400 mg IV every 6 weeks) was approved across all solid tumors based on pharmacokinetic modeling and exposure-response analyses. Although EI dosing is more convenient for patients, whether the higher dose and extended dosing interval impacts immune-related adverse events (irAE) and clinical outcomes in patients with breast cancer is unknown. Methods: In this retrospective cohort study, patients with breast cancer treated with pembrolizumab between 2017 to 2024 were included. Safety (irAE) and clinical outcomes including event-free survival (EFS), progression-free survival (PFS) and overall survival (OS) were compared between patients who received only standard dosing (SD; 200 mg IV every 3 weeks) pembrolizumab and those who received ≥ 1 cycle of EI dosing. Results: Of the 355 patients included, 59 (17%) received ≥ 1 cycle of EI pembrolizumab, and 296 (83%) received ≥ 1 cycle of only SD. Of those who received ≥ 1 cycle of pembrolizumab EI, 27 (45.8%) started with 200 mg, 7 (11.9%) started with 400 mg, 9 (15.2%) received only 400 mg, and 16 (27.1%) switched between dosing schedules. The majority (71%) of patients had early-stage disease, and 92% had triple-negative breast cancer. Patients with early-stage disease who received ≥ 1 cycle of EI dosing had lower rates of grade 3 or higher irAE compared to those who received only SD (4% vs. 20%; p=0.01), while rates of any-grade irAE were similar (p=0.3). EFS and OS were similar between the two dosing regimens in patients with early-stage disease (p=0.8 and p=0.5, respectively). In patients with metastatic disease, any-grade irAE (p=0.5), grade 3 or higher irAE (p=0.1), PFS (p=0.8), and OS (p=0.5) were similar between the dosing regimens. Conclusion: This is the first study to demonstrate comparable rates of any-grade irAE and clinical outcomes in patients with breast cancer treated with pembrolizumab EI dosing compared to SD. A lower rate of grade 3 or higher irAE was observed in patients with early-stage breast cancer who received ≥ 1 cycle of EI dosing. As more patients are treated with EI dosing, this data provides new evidence that safety and efficacy are maintained while improving medication burden on patients.
Presentation numberPS4-07-22
Chemotherapy Decision Making and Outcomes in Very Early-Stage Triple Negative Breast Cancer
Elyse R. Lopez, MD Anderson Cancer Center, Houston, TX
E. R. Lopez1, A. Raghavendra2, S. Pasyar3, B. Lim2, K. Meghan2, V. Valero2, C. H. Barcenas2; 1Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 2Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Department of Biostatistics, MD Anderson Cancer Center, Houston, TX.
BACKGROUND The benefit of chemotherapy in very early-stage triple-negative breast cancer (TNBC) remains unclear, as these patients have historically been excluded from clinical trials. Guidelines recommend consideration of chemotherapy for T1b tumors (0.6-1 cm) and is not recommend for T1a tumors (≤0.5 cm). However, treatment utilization is heterogeneous, and the benefit remains poorly defined. We aim to evaluate recent trends in treatment patterns, outcomes, and provider decision-making for patients with stage IA TNBC. METHODS We conducted a single-institution retrospective study from Jan 2016-Jan 2025 of patients with TNBC, pathologic T1a (≤5 mm) or T1b (>5-10 mm), and pN0, pN0(i+), or pN1mi disease. Patients were stratified by pT stage and chemotherapy receipt. Kaplan-Meier curves assessed survival; logistic regression identified factors associated with chemotherapy use. RESULTS Among pT1a, 20.6% (n=13) received chemotherapy. Older age was associated with a significantly lower likelihood of chemotherapy use, while BRCA1(+) was associated with a significantly higher likelihood of receiving chemotherapy. Among pT1b, 67.6% (n=94) received chemotherapy. Older age and ECOG 1 (vs 0) were associated with a significantly lower likelihood of receiving chemotherapy; tumor grade III and invasive ductal carcinoma histology were associated with a significantly higher likelihood of chemotherapy. Recurrence and survival estimates are displayed in Table 1. Decision-making documentation was available for 191 (95%) cases. In pT1a, multifocal disease was the most frequently cited factor (33%) when chemotherapy was recommended. In pT1b, high comorbidities (30%), patient preference (20%), favorable histology (20%), and lack of data (20%) were the most cited reasons for recommending surveillance. Lack of high evidence data and departmental group discussions were cited in discussions across all subgroups. CONCLUSION Although 3-year OS and RFS were generally favorable, recurrences occurred in all groups except pT1a patients who received chemotherapy. This cohort represents a very important group of patients for whom oncologists must balance the risk of overtreatment with chemotherapy, while also ensuring high-risk patients receive appropriate care. Qualitative review of documentation revealed that clinical decision-making was often driven by nuanced considerations, including multifocal disease, histologic subtype, patient comorbidities, and patient preference; consistent references to lack of high-level evidence data and discussion at departmental meetings highlight the ambiguity and complexity in these cases. These findings underscore the need for prospective data and validated risk tools to guide chemotherapy decisions in T1a/b TNBC tumors.
| Characteristic | pT1a (n=63) | pT1b (n=139) |
| Median Age (years) | 60 | 62 |
| Received Chemotherapy, n (%) | 13 (20.6%) | 94 (67.6%) |
| Recommended Surveillance, n (%) | 40 (63.5%) | 20 (14.4%) |
| Offered both options, n (%) | 12 (19%) | 31 (22.3%) |
| Recommended Chemotherapy, n (%) | 9 (14.3%) | 79 (56.8%) |
| Oncologist’s recommendation not available for review, n (%) | 2 (3.2%) | 9 (6.5%) |
| BRCA1 Positive, n (%) | 10 (15.9%) | 11 (7.9%) |
| Grade III Tumor, n (%) | 34 (53.9%) | 96 (69.1%) |
| Invasive Ductal Histology, n (%) | 54 (85.7%) | 122 (87.8%) |
| ECOG 1 vs. 0, % | 6% vs 79% | 9% vs 72% |
| Recurrence in No Chemo Group, n (%) | 2 (4%) | 3 (6.8%) |
| Recurrence in Chemo Group, n (%) | 0 (0%) | 8 (8.5%) |
| 3-Year OS (%) | 97% | 94% |
| 3-Year RFS with chemotherapy (%) | 100% | 94% |
| 3-Year RFS without chemotherapy (%) | 97% | 96% |
Presentation numberPS4-07-23
Predictive Markers for Pathological Complete Response to Neoadjuvant Therapy in Early-Stage Triple-Negative Breast Cancer: A Single-Center Study in Vietnam.
Anh DINH, Vietnam National Cancer Hospital, Hanoi, Viet Nam
A. DINH1, Y. Le1, P. Han2, U. Le1, H. Hoang3, H. Duong3; 1Medical Oncology Department, Vietnam National Cancer Hospital, Hanoi, VIET NAM, 2Department of Pathology, Vietnam National Cancer Hospital, Hanoi, VIET NAM, 3Department of Radiology, Vietnam National Cancer Hospital, Hanoi, VIET NAM.
Predictive Markers for Pathological Complete Response to Neoadjuvant Therapy inEarly-Stage Triple-Negative Breast Cancer: A Single-Center Study in Vietnam.Anh Dinh1,3,4, Yen Le1,4, Han Pham2, Uyen Le2, Huong Hoang5, Huu Duong5.1Department of Pathology, Vietnam National Cancer Hospital. 2Medical Oncology Department,Vietnam National Cancer Hospital. 3Cancer Research and Clinical Trials Center. 4Hanoi MedicalUniversity. 5Department of Radiology, Vietnam National Cancer Hospital.Background: Pathological complete response (pCR) to neoadjuvant systemic therapy is a crucialprognostic indicator in early-stage triple negative breast cancer (eTNBC). In Vietnam, the immunecheckpoint inhibitor, pembrolizumab was approved for neoadjuvant regimens combined withchemotherapy for eTNBC in 2023. However, the lack of public health insurance coverage for thisnovel therapy creates a significant barrier, limiting access for the vast majority of Vietnamesepatients. This study aims to identify valuable factors that predict pCR achievement in eTNBCpatients receiving neoadjuvant treatment with pembrolizumab combined with chemotherapy inVietnam, thereby optimizing treatment strategies.Methods: This is a prospective, single-arm, single-center study conducted at the Vietnam NationalCancer Hospital from March 2023 to May 2025, involving 49 patients with early triple-negativebreast cancer (eTNBC) who received neoadjuvant chemotherapy with TC-AC combined withpembrolizumab. The primary endpoint was pathological complete response (pCR), which twoindependent pathologists assessed. Univariate and multivariate logistic regression analyses wereused to identify independent prognostic factors for pCR.Results: The majority of patients (98%) presented with Stage II or III disease. Median tumor sizewas 3.5 cm, and median tumor-infiltrating lymphocytes (TILs) were 35%. HER2-negative patientsconstituted 67.3% of the cohort, while 32.6% were HER2-low. The pCR rate in this study was67.3% (33/49). In multivariate logistic regression analysis, TILs percentage was identified as anindependent predictor of pCR achievement (OR: 1.14; 95% CI: 1.06–1.27; p=0.003). Whenstratified by TILs level, the pCR rate was 89.7% for patients with TILs > 30%, 50% for TILsbetween 20% and 30%, and 12.5% for TILs < 20%. HER2 expression status also significantlyinfluenced pCR achievement, with HER2-low patients exhibiting a lower pCR rate compared toHER2-negative patients (p=0.049).Conclusion:This study identifies TILs and HER2 expression as predictive factors for pCR. TILs,specifically, demonstrate potential as a biomarker to guide individualized treatment in eTNBC.This is particularly important as ICIs gain approval for eTNBC treatment in developing countrieslike Vietnam, underscoring the critical need for reliable predictive tools to optimize patientoutcomes.Keyworlds: Triple-Negative Breast Cancer, Neoadjuvant Treatment, Predictive Biomarker.
Presentation numberPS4-07-24
Phase 3 study of neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab for early-stage TNBC: KEYNOTE-522 magnetic resonance imaging (MRI) subgroup analysis
Javier Cortés, Quironsalud Group, and IOB Madrid, Hospital Beata Maria Ana, and Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
J. Cortés1, R. Dent2, H. McArthur3, L. Pusztai4, S. Kümmel5, C. Denkert6, J. O’Shaughnessy7, P. A. Fasching8, M. Untch9, R. Tarnawaski10, M. Mouret-Reynier11, S. M. Stemmer12, T. Foukakis13, J. Boileau14, C. Chung15, M. Fernandez16, J. A. Mejia17, F. Beca17, S. Hou18, P. Schmid19; 1International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, and IOB Madrid, Hospital Beata Maria Ana, and Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, SPAIN, 2Department of Medical Oncology, National Cancer Center Singapore, Duke – National University of Singapore Medical School, Singapore, SINGAPORE, 3Oncology Department, University of Texas Southwestern Medical Center, Dallas, TX, 4Yale Cancer Center, Yale University School of Medicine, New Haven, CT, 5Department of Gynecology with Breast Center, Breast Unit, Kliniken Essen-Mitte, Essen, Germany and Charité – Universitätsmedizin Berlin, Department of Gynecology with Breast Center, Berlin, GERMANY, 6Institute of Pathology, Philipps University Marburg, Marburg, GERMANY, 7Baylor University Medical Center, Texas Oncology, Dallas, TX, USA and Sarah Cannon Research Institute, Dallas, TX, 8University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Erlangen, GERMANY, 9Department of Obstetrics and Gynecology, Interdisciplinary Breast Cancer Center, Medical School Berlin, Helios Kliniken Berlin-Buch, Berlin, GERMANY, 10Department of IIIrd Radiotherapy and Chemotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, POLAND, 11Medical Oncology, Jean Perrin, Centre de Lutte contre le Cancer, Clermont-Ferrand F-63000, FRANCE, 12Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel, Tel-Aviv University, Tel Aviv, ISRAEL, 13Department of Oncology-Pathology, Karolinska Institutet and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Solna, SWEDEN, 14McGill University, Jewish General Hospital Segal Cancer Centre, Montréal, QC, CANADA, 15Oncology Department, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, TAIWAN, 16Hemato Oncólogo, IMAT-Oncomedica, Montería, COLOMBIA, 17Global Clinical Development, Merck & Co., Inc., Rahway, NJ, 18Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ, 19Centre for Experimental Cancer Medicine; Barts Cancer Institute, Queen Mary University London, London, UNITED KINGDOM.
Background: In the KEYNOTE-522 study (NCT03036488), neoadjuvant pembrolizumab (pembro) plus chemotherapy (chemo) followed by adjuvant pembro plus chemo significantly improved pathological complete response (pCR), event-free survival, and OS compared with placebo (pbo) plus chemo in participants (pts) with early-stage TNBC. Following neoadjuvant therapy, the estimated treatment difference in pCR for pembro plus chemo vs pbo plus chemo was 13.6% (95% CI, 5.4%‒21.8%; P < 0.001) at the first interim analysis (N=602). Although pCR provides a definitive indicator of neoadjuvant treatment success, other measures, such as RECIST v1.1 and functional tumor volume (FTV), may offer an earlier signal of therapeutic activity. This prespecified exploratory analysis evaluated associations of pCR with ORR at different time points per MRI by RECIST v1.1 and FTV by blinded independent central review. Methods: Pts with previously untreated TNBC (stage T1c N1-N2 or T2-T4 N0-N2) were randomized 2:1 to neoadjuvant pembro 200 mg Q3W or pbo, each with 4 cycles of paclitaxel plus carboplatin (treatment 1) then with 4 cycles of doxorubicin or epirubicin plus cyclophosphamide (treatment 2). After definitive surgery, pts received adjuvant pembro or pbo for 9 cycles or until recurrence/unacceptable toxicity. Breast MRI was performed for consenting pts at screening and after neoadjuvant treatments 1 and 2. Responders were defined as pts who achieved CR or PR per RECIST v1.1 by blinded independent central radiology review. The subgroup analyses population included the randomized pts who signed MRI consent and had baseline values by central radiology review. Results: At data cutoff (March 23, 2021), MRI subgroup analyses included 162 pts (pembro plus chemo, n = 97; pbo plus chemo, n = 65). After treatment 1, ORR per RECIST v1.1 was 91.8% for pembro plus chemo vs 84.6% for pbo plus chemo, while ORR per MRI FTV was 96.9% vs 93.8%. After treatment 2, ORR per RECIST v1.1 was 82.5% vs 90.8%, and ORR per MRI FTV was 84.5% vs 92.3% for pembro plus chemo and pbo plus chemo, respectively. In the post hoc exploratory analyses for pCR in the MRI subgroup, pCR rate (ypT0/Tis ypN0; 95% CI) was 62.9% (52.5%-72.5%) with pembro plus chemo vs 52.3% (39.5%-64.9%) with pbo plus chemo. Odds ratios (OR) for achieving pCR among pts with ORR per RECIST v1.1 or FTV across all patients (pembro plus chemo and pbo plus chemo combined) are shown in the Table. Conclusions: Across all pts (ie, for pembro plus chemo and pbo plus chemo combined), the odds of achieving pCR were higher among pts who had an objective response per either RECIST v1.1 or FTV.
|
Achieved pCR (n = 95) |
Did Not Achieve pCR (n = 67) |
Association Between pCR and ORR, Odds Ratio (95% CI) | |
| Neoadjuvant treatment 1 | |||
| ORR per RECIST v1.1a | |||
| Responder | 93 (57.4) | 51 (31.5) | 14.6 (3.2–66.0) |
| Nonresponder | 2 (1.2) | 16 (9.9) | |
| ORR per FTVb | |||
| Responder | 94 (58.0) | 61 (37.7) | 9.2 (1.1–78.7) |
| Nonresponder | 1 (0.6) | 6 (3.7) | |
| Neoadjuvant treatment 2 | |||
| ORR per RECIST v1.1a | |||
| Responder | 90 (55.6) | 49 (30.3) | 6.6 (2.3–18.9) |
|
Nonresponder |
5 (3.1) | 18 (11.1) | |
| ORR per FTVb | |||
| Responder | 91 (56.2) | 51 (31.5) | |
| Nonresponder | 4 (2.5) | 16 (9.9) | 7.1 (2.3–22.5) |
|
aProportion of pts with PR/CR by RECIST v1.1 (≥30% decrease in sum of target lesion diameters). bProportion of pts with ≥30% decrease in volume of tumor tissue showing contrast enhancement. |
Presentation numberPS4-07-25
Real World Analysis of Efficacy, Toxicity, and Treatment Patterns of Pembrolizumab-Containing Regimens for Older Adults with Early-Stage Triple Negative Breast Cancer
Claire Smith, Dana Farber Cancer Institute, Boston, MA
C. Smith1, A. Rami2, T. Li3, P. Patel1, M. Ling4, R. Freedman1; 1Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 2Internal Medicine, Brigham and Women’s Hospital, Boston, MA, 3Biostatistics, Dana Farber Cancer Institute, Boston, MA, 4Medical Oncology, AU/UGA Medical Partnership; DFCI, Athens, GA.
Real World Analysis of Efficacy, Toxicity, and Treatment Patterns of Pembrolizumab-Containing Regimens for Older Adults with Early-Stage Triple Negative Breast Cancer Background: Older adults are underrepresented in clinical trials of neoadjuvant chemotherapy and pembrolizumab for early-stage triple negative breast cancer (TNBC), limiting evidence to guide treatment in this population. Given the potential toxicity of multi-agent regimens, we examined treatment patterns, efficacy, and safety outcomes for patients ≥ 65 years who received neoadjuvant pembrolizumab containing treatments for early-stage TNBC. Methods: Through medical review at a National Cancer Institute-designated comprehensive cancer center, we abstracted records for those aged ≥ 65 years at the time of a stage I-III TNBC diagnosis during 08/2021-05/2024. Patients were included if they received a pembrolizumab-containing regimen as neoadjuvant therapy. The primary endpoint was relative dose intensity (RDI), defined as the average percent of planned neoadjuvant treatment received across agents. We also ascertained records for hospitalizations, early treatment discontinuation, immune related adverse events (IRAE), and pathological complete response (pCR) status at surgery. We examined associations between age (65-70) vs (>70) and occurrence of these events. In addition, we examined if treatment-related factors including RDI, duration of neoadjuvant therapy, anthracycline receipt, and age were associated with pCR. We used Kruskal-Wallis rank sum test for continuous variables and chi-square test for categorical variables.Results: Overall, 85 women were included in analyses; median age was 70. At the time of diagnosis, n=13 (15%) had heart disease, n=4 (5%) lung disease, n=6 (7%) chronic kidney disease, and n=1 (1%) cognitive impairment. For treatment regimens, 42% (n=36) received KEYNOTE 522 (carboplatin + paclitaxel + doxorubicin + cyclophosphamide + pembrolizumab), 41% (n=35) received carboplatin + paclitaxel + pembrolizumab, 4 (5%) doxorubicin + cyclophosphamide, + taxane + pembrolizuamb, and 4 (5%) docetaxel + carboplatin + pembrolizumab. Across regimens, median RDI was 90% (range 20-100%), and 55% (n=47) receieved ≥85% RDI. During neoadjuvant therapy, 41% (n=35) of patients were hospitalized at least once, 6% of all patients (n=5) required ICU admission, and 41% (n=35) discontinued treatment early due to adverse events. Overall, 39% (n=33) experienced an IRAE, of which 49% (n=16) required steroids. There were 3 deaths attributed to neoadjuvant treatment, specifically pneumonitis, pneumonia/septic shock, and multi-organ failure. Patients age >70, when compared to ages 65-70, were more likely to experience IRAE (OR 2.8, p=0.024) and receive lower RDI (OR 0.3, p=0.013). Rates of hospitalization, early treatment discontinuation, and path CR did not differ by age. The overall pCR rate was 43%. Factors including RDI, treatment duration, anthracycline, hospitalizations, immune related adverse events, and age were not associated with pCR. Conclusions: In this real-world cohort, half of older adults with early-stage TNBC received a non-anthracycline containing chemotherapy regimen with pembrolizumab, most commonly carboplatin and paclitaxel. Early discontinuation of neoadjuvant therapy was frequent with nearly 45% not receiving ≥85% RDI. Rates of hospitalization, immune related adverse events, and death were high, while pCR rates were lower than reported in clincal trials. Safer, more effective treatments are needed for older adults with early-stage TNBC.
Presentation numberPS4-07-26
Safety and efficacy of neoadjuvant cyclophosphamide, methotrexate, fluorouracil (CMF) and pembrolizumab in older adults with early triple negative breast cancer (TNBC)
Rima Patel, Icahn School of Medicine at Mount Sinai, New York, NY
R. Patel, J. Anderson, R. Farley, A. Tiersten; Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: The current treatment of early-stage, locally advanced TNBC consists of neoadjuvant pembrolizumab, and a four-drug chemotherapy backbone, including paclitaxel, carboplatin, doxorubicin and cyclophosphamide, based on the KEYNOTE-522 trial. Many older adults are unable to receive this regimen due to comorbidities or performance status that increase their risk of toxicities. CMF is a chemotherapy regimen that is used in TNBC and safe with little long-term toxicity even in older patients. However, it has not been studied in combination with pembrolizumab, which was approved for early-stage TNBC in 2021. The goal of this study was to assess the safety and efficacy of neoadjuvant CMF and pembrolizumab in patients with early TNBC. Methods: We performed a retrospective chart review of all patients treated with neoadjuvant CMF and pembrolizumab for clinical Stage II-III TNBC from 2021-2024 at our institution’s health system. Demographics, tumor characteristics and treatment information were extracted from the electronic medical record. Toxicity of the regimen was measured by the percentage of patients requiring early discontinuation of one or more drugs, dose reductions, dose delays, hospitalizations, and/or emergency department (ED) visits due to treatment-related adverse events (TRAEs). Efficacy of the regimen was measured by the rate of pathologic complete response, defined as the absence of invasive disease in the breast and axilla at time of definitive surgery, in the overall population. Results: We identified 10 patients with early-stage, locally advanced TNBC who were treated with neoadjuvant CMF and pembrolizumab and included in the analysis. These patients were not eligible for the KEYNOTE-522 regimen due to performance status and/or comorbidities. The age at diagnosis of the patients ranged from 62 to 92 years with a median of 81 years. The median ECOG performance status was 1 (range 0-2) and 60% (N=6) had one or more comorbidities. Forty percent (N=4) of patients were Black. Most patients had Stage II disease (60%, N=6) and lymph node involvement (60%, N=6). The median number of cycles of CMF and pembrolizumab administered were 8 and 7, respectively. In terms of treatment toxicity, only one patient required a dose reduction in CMF due to neutropenic fever, discontinuation of pembrolizumab due to Grade 3 colitis, and hospitalization for these TRAEs. Twenty percent (N=2) had treatment delays due to Grade 2 adverse events but did not require dose reductions. There were no other drug discontinuations, hospitalizations or ED visits due to TRAEs. Of the 10 patients, 20% (N=2) have not undergone surgery yet due to comorbidities and insurance issues but both patients clinically had a treatment response based on exam. Of the remaining 8 patients with surgical information available, the pathologic complete response rate was 50%. Conclusions: The combination of neoadjuvant CMF and pembrolizumab was found to be well tolerated and effective in older patients with early TNBC who were ineligible to receive the KEYNOTE-522 regimen. While the findings are limited by their retrospective nature and small patient cohort, the preliminary safety and efficacy in a predominantly older population with comorbidities are promising and warrant further evaluation. A prospective phase II trial evaluating the combination of CMF and pembrolizumab is currently being planned.
Presentation numberPS4-07-27
Pembrolizumab + chemotherapy vs placebo + chemotherapy as neoadjuvant treatment, followed by pembrolizumab vs placebo as adjuvant treatment for early-stage TNBC: a post hoc analysis of participants included and separately of those not included in the first interim analysis for pCR in the KEYNOTE-522 study
Peter Schmid, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, United Kingdom
P. Schmid1, J. Cortes2, L. Pusztai3, H. McArthur4, S. Kümmel5, C. Denkert6, Y. H. Park7, R. Hui8, N. Harbeck9, M. Takahashi10, T. Foukakis11, P. A. Fasching12, F. Cardoso13, M. Untch14, Y. Ding15, F. Beca16, J. A. Mejia16, R. Dent17, J. O’Shaughnessy18; 1Barts Cancer Institute, Queen Mary University London, Charterhouse Square, London EC1M 6BQ, UNITED KINGDOM, 2International Breast Cancer Center (IBCC), Pangaea Oncology, Quironsalud Group, Marquesa de Vilallonga 12, Clinics 18.19, Barcelona 08017, Spain and Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, C. Tajo, s/n, Villaviciosa de Odón, Madrid 28670, SPAIN, 3Yale School of Medicine, Yale Cancer Center, 35 Park Street, New Haven, CT, 4Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 5Breast Unit, Kliniken Essen-Mitte, Henricistraße 40, Essen 45136, Germany and Department of Gynecology with Breast Center, Charité – Universitätsmedizin Berlin, Charitéplatz 1, Berlin 10117, GERMANY, 6Institute of Pathology, Philipps-University Marburg and University Hospital Marburg, Biegenstraße 10, Marburg 35037, GERMANY, 7Department of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro Gangnam-gu., Seoul 06351, KOREA, REPUBLIC OF, 8Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital and University of Sydney, Hawkesbury Rd, Westmead, Sydney, NSW 2145, AUSTRALIA, 9Department of OB&GYN, Breast Center, LMU University Hospital, Marchioninistraße 15, Munich 81377, GERMANY, 10Department of Breast Surgery, Hokkaido University Hospital, Kita14, Nishi5, Kita-Ku, Sapporo, Hokkaido 060-8648, JAPAN, 11Department of Oncology-Pathology, Karolinska Institutet, 171 77 Stockholm, and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Karolinska Comprehensive Cancer Center, Solna, SE-171 76 Stockholm, SWEDEN, 12University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Bavarian Cancer Research Center (BZKF), Maximiliansplatz 2, Erlangen 91054, GERMANY, 13Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation and ABC Global Alliance, Av. De Brasília s/n, Lisbon 1400-038, PORTUGAL, 14Department of Obstetrics and Gynecology, Interdisciplinary Breast Cancer Center, Medical School Berlin, Helios Kliniken Berlin-Buch, Berlin 13125, GERMANY, 15Biostatistics and Research Decision Sciences, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ, 16Global Clinical Development, Merck & Co., Inc., 126 East Lincoln Avenue, Rahway, NJ, 17Department of Medical Oncology, National Cancer Center Singapore, Duke – National University of Singapore Medical School, 30 Hospital Boulevard, Singapore 168583, SINGAPORE, 18Baylor University Medical Center, Texas Oncology, US Oncology, 3500 Gaston Ave, Dallas, TX.
Background: In the phase 3 KEYNOTE-522 (NCT03036488) study in participants (pts) with early-stage TNBC, addition of pembrolizumab (pembro) to chemotherapy (chemo) led to statistically significant and clinically meaningful improvements in pCR, EFS, and OS at interim analysis 1 (IA1), IA4, and IA7, respectively. At the prespecified IA1 (per approved statistical analysis plan), pembro + chemo showed statistically significant pCR when compared to chemo alone in the first 602 pts who underwent randomization. However, the difference in pCR rates between treatment arms decreased at IA4, which was evaluated in the overall population. A post hoc subgroup analysis was conducted to separately evaluate outcomes (EFS, OS) in the group of pts who were included in the IA1 pCR analysis and pCR, EFS, and OS in the group of remaining pts who were not. Methods: Eligible pts had previously untreated, high-risk, early-stage TNBC. Pts were randomized 2:1 to receive neoadjuvant pembro + chemo followed by adjuvant pembro (pembro arm) or neoadjuvant placebo + chemo followed by adjuvant placebo (placebo arm). Dual primary endpoints were pCR (ypT0/Tis ypN0) per local pathologist and EFS per investigator; OS was a key secondary endpoint. Data cutoff dates were March 23, 2021 (IA4) for pCR and March 22, 2024 (IA7) for EFS and OS. Results: The first 602 pts who underwent randomization were included in the IA1 pCR analysis and the remaining 572 pts were not included as per statistical analysis plan. Baseline pt characteristics were generally balanced between the treatment arms in both groups (included in IA1: 99.8% female; median age, 49 years; 64% White; 23% Asian; 75% with stage II disease; 74% with tumor size T1/T2; 52% with positive nodal status; 50% with PD-L1 CPS ≥10; not included in IA1: 100% female; median age, 48 years; 63% White; 18% Asian; 75% with stage II disease; 75% with tumor size T1/T2; 51% with positive nodal status; 47% with PD-L1 CPS ≥10). Efficacy outcomes are shown in the Table. HRs for EFS and OS were similar and favored the pembro arm versus the placebo arm in both groups. Conclusions: In this post hoc analysis of the KEYNOTE-522 study, the pCR rate in the group of pts who were not included in IA1 for pCR was found to be similar between the treatment arms. EFS and OS outcomes favored the pembro arm versus the placebo arm with similar HRs in both groups and were consistent with those in the overall study population. These data support the use of pembro in combination with neoadjuvant chemo, then continued as adjuvant monotherapy after surgery for high-risk, early-stage TNBC.
Presentation numberPS4-07-29
Evaluating the tolerability of the combination of adjuvant pembrolizumab and capecitabine in a diverse real-world cohort of patients with early-stage TNBC
Prathyusha Pandu, Mount Sinai West, New York, NY
J. Anderson1, E. Baldwin2, J. Dejesus2, G. Van Hyfte3, P. Pandu4, N. Krishnamurthy5, M. Rattu1, R. Farley1, R. Patel1, A. Tiersten1; 1Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 2Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 3Tisch Cancer Institute – Biostatistics and Clinical Informatics, Icahn School of Medicine at Mount Sinai, New York, NY, 4Department of Internal Medicine, Mount Sinai West, New York, NY, 5Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: Although the KEYNOTE-522 trial demonstrated a significant pCR following neoadjuvant chemoimmunotherapy, 35.2% of patients had evidence of residual disease following surgery, a population who may be recommended to receive adjuvant capecitabine per the CREATE-X trial. However, there is a paucity of real-world data evaluating the tolerability of adjuvant pembrolizumab, per KEYNOTE-522, when combined with adjuvant capecitabine, per CREATE-X. Our study examines the tolerability of the adjuvant combination in a real-world, diverse, urban population at a large tertiary referral center. Methods: We performed a retrospective chart review of patients (pts) with stage II-III TNBC who received the KEYNOTE-522 regimen within our institution’s health system from August 2021 to February 2025. Pts who received >/= 1 cycle of adjuvant pembrolizumab were included in our analysis. Pts were stratified based on receipt of adjuvant pembrolizumab alone versus combination of pembrolizumab and capecitabine. Demographics, adjuvant toxicity data including rates of treatment delays, dose holds, and treatment discontinuations were extracted from the medical record. Descriptive statistics were used to describe patient data, and non-parametric tests were utilized in the stratified analyses. Due to the exploratory nature of our analysis, p-values are reported unadjusted. Results: 83 pts received adjuvant pembrolizumab and were included in this analysis. Overall, 64% (N=53) were on adjuvant pembrolizumab alone and 36% (N=30) on the combination of adjuvant pembrolizumab and capecitabine. Overall, 83% had Stage II disease and 45% with lymph node involvement. 28% of pts (N=23) identified as White, 28% (N=23) Black, 16% (N=13) Asian, 19% (N=16) Hispanic, and 9% (N=8) as Other. The early discontinuation rates (prior to 9 cycles) of adjuvant pembrolizumab in the pembrolizumab cohort and combination cohort were 17% (N=9) and 13% (N=4), respectively (p=0.761). Pembrolizumab was discontinued most frequently due to irAEs including thyroiditis, colitis, pneumonitis and myocarditis. In the combination cohort of 30 pts, 47% (N=14) required a capecitabine dose reduction, 27% (N=8) required a dose hold, and 30% (N=9) discontinued capecitabine prior to 6 cycles (Table 1). Capecitabine was dose reduced or held most frequently for non-hematologic reasons including hand-foot syndrome, GI toxicity, and mucositis. Conclusion: The adjuvant combination of capecitabine and pembrolizumab appeared to be well tolerated. Treatment was held, dose reduced or discontinued more commonly from known side effects of capecitabine or pembrolizumab, but not as the result of worsening toxicity from combination therapy. Overall, the combination of capecitabine and pembrolizumab warrants further study for pts with residual disease after neoadjuvant KEYNOTE-522.
| Toxicity Measure | N=30 |
| Pembrolizumab | |
| Dose hold | 3.3% (1) |
| Early discontinuation (prior to 9 cycles) | 13% (4) |
| Capecitabine | |
| Dose reduction | 47% (14) |
| Dose hold | 27% (8) |
| Early discontinuation (prior to 6 cycles) | 30% (9) |
Presentation numberPS4-07-30
A Phase II Trial of Ivonescimab in Combination with Carboplatin and Docetaxel in Patients with Early Stage Triple Negative Breast Cancer (TNBC)
Yuan Yuan, Cedars-Sinai Medical Center, Los Angeles, CA
Y. Yuan, M. Tighiouart, A. Tan, A. Giuliano, S. Karlan, M. Boyle, C. Dang, A. Chung, F. Amersi, P. McAndrew, M. El-Masry, S. Sikaria, D. Chan, S. Valdez, N. Tank, T. Shaw, T. Wang, S. Shiao, S. Merlo, M. Walker, D. Calhoun, G. Park, J. Shen, A. Polverini, A. Terando, D. Lin, Y. Choi, J. Bitar; Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, CA.
Background: Immune checkpoint inhibitor pembrolizumab in combination with chemotherapy has shown promising pathological complete response rate (pCR) in early stage TNBC and currently FDA approved for stage II-III TNBC. Ivonescimab, a tetravalent bispecific antibody targeting PD-1 and VEGF, has the potential to produce synergistic anti-tumor effects through both pathways via cooperative binding. Ivonescimab showed promising progression free survival (PFS) in combination with paclitaxel or nab-paclitaxel as first line therapy, including PD-L1 negative TNBC. This phase 2 trial is designed to evaluate the efficacy and safety of ivonescimab in combination with carboplatin and docetaxel in early stage TNBC. Methods: This is a single center phase II study in patients (pts) with newly diagnosed early stage TNBC. Pts will receive neoadjuvant ivonescimab at 20 mg/kg, carboplatin AUC6 and docetaxel 75 mg/m2 every 3 weeks for a total of 6 cycles followed by breast surgery. Key eligibility criteria include: female or male ≥18; ECOG 0-1; newly diagnosed high-risk early stage TNBC, defined by ER≤10%, PR≤10% and HER2 negative per ASCO/CAP guideline; clinically ≥ T1cN0, or any T, N1-2 ; plan to receive neoadjuvant chemotherapy and immunotherapy as standard-of-care treatment; willing to undergo tumor biopsy prior to planned neoadjuvant systemic therapy; willing to provide post-treatment residual tumor at time of surgery. The primary endpoint is pCR. The secondary endpoints are event free survival (EFS), overall survival (OS), patient’s quality of life (QOL) by EORTC QLQ-C30. Peripheral blood, tumor tissue and stool sample will be collected at baseline, cycle 4 day 1, time of surgery for exploratory analysis: peripheral blood immune profiling, tumor immune profiles, stool microbiomes associated with the efficacy of the combination therapy. Statistical Design: We hypothesize that the addition of ivonescimab will increase the pCR rate to 78%. We will test the null hypothesis H0: θ < 0.58 versus the alternative hypothesis H1: θ > 0.58. The decision rule is to reject the null hypothesis if the posterior probability that θ > 0.58 given the data is sufficiently high, i.e, reject H0 if P (θ > 0.58 | data) > 0.94. A non-informative Jeffreys prior distribution beta (0.5,0.5) is placed on the parameter θ. One interim analysis for futility is planned using the Bayesian predictive distribution. With a sample size of 34 patients with one interim analysis for futility, the power to detect an absolute increase in pCR of 20% is 0.80. If the true value of θ is 0.58, the one-sided type I error is 0.046. The trial is projected to start patient enrollment in July 2025. Clinical trial information: NCT07017673
Presentation numberPS4-10-01
Neoadjuvant immune checkpoint inhibitors plus chemotherapy in early-stage triple-negative breast cancer: A systematic review and meta-analysis.
Emmanouil Saloustros, University Hospital of Larissa, Larissa, Greece
S. Alexiou1, M. Georgios2, E. Saloustros3; 1Internal Medicine, General Hospital of Larissa, Larissa, GREECE, 2Emergency Medicine, University Hospital of Larissa, Larissa, GREECE, 3Oncology, University Hospital of Larissa, Larissa, GREECE.
Background Triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and poor prognosis. Immune checkpoint inhibitors (ICIs) have shown efficacy in the early-stage setting when combined with chemotherapy. We conducted a meta-analysis to evaluate their efficacy in terms of event-free survival (EFS), overall survival (OS), and pathological complete response (pCR). Methods We performed a systematic literature search of PubMed, Scopus, and ClinicalTrials.gov to identify randomized controlled trials comparing neoadjuvant or perioperative chemotherapy with or without ICIs in early-stage TNBC. Eligible trials reported on at least one of the following outcomes: EFS, OS, or pCR. Study selection, data extraction, and quality assessment were conducted independently by two reviewers, with discrepancies resolved by consensus. Hazard ratios (HRs) were pooled for EFS and OS using inverse-variance weighting. For pCR, both risk difference (RD) and risk ratio (RR) were calculated using Mantel-Haenszel methods. Heterogeneity was assessed using the I² statistic. Pre-specified subgroup and sensitivity analyses were conducted, and publication bias was evaluated via funnel plot asymmetry. The study protocol was registered in PROSPERO (ID: CRD420251082981). Results Six phase II-III trials (KEYNOTE-522, IMpassion031, NeoTRIP Michelangelo, GeparNuevo, GeparDouze, CamRelief) comprising 3.952 patients were included. ICIs significantly improved EFS (HR = 0.76; 95% CI: 0.65-0.89; p = 0.0005; I² = 3%) and OS (HR = 0.69; 95% CI: 0.57-0.84; p = 0.0002; I² = 49%). For pCR, the absolute increase was 8% (RD = 0.08; 95% CI: 0.05-0.11; p < 0.00001; I² = 0%), and the relative increase was 15% (RR = 1.15; 95% CI: 1.09-1.22; p < 0.00001; I² = 3%). Notably, OS was not available in NeoTRIP and CamRelief, and EFS was not available in GeparNuevo. Sensitivity analyses (e.g., model selection) confirmed robustness. No evidence of publication bias was detected. Conclusion ICIs combined with chemotherapy significantly improve EFS, OS, and pCR in early-stage TNBC. These data support their incorporation into standard neoadjuvant treatment. Further studies should refine predictive biomarkers and long-term safety monitoring.
Presentation numberPS4-10-02
A Real-World Correlation Analysis on the Age at Diagnosis, Pathologic Complete Response, Germline Mutations, and Toxicity in Early Triple-Negative Breast Cancer Patients Treated with the Neoadjuvant Therapy KEYNOTE-522
Stephanie Adel Haddad, UT Health San Antonio/Mays Cancer Center, San Antonio, TX
S. A. Haddad1, D. U. Portillo1, E. Kaser2, A. Baig2, C. F. Jones1, R. Banwait1, M. M. Canola1; 1Hematology & Medical Oncology, UT Health San Antonio/Mays Cancer Center, San Antonio, TX, 2Internal Medicine, UT Health San Antonio, San Antonio, TX.
Introduction: Age at diagnosis can significantly impact treatment outcomes and toxicity profiles in breast cancer. Pathologic complete response (pCR) following neoadjuvant therapy in triple-negative breast cancer (TNBC) is an established surrogate for long-term outcomes. While clinical and genetic factors have been explored as predictors of pCR, the relationship between age at diagnosis and pCR, especially in the context of immune checkpoint inhibitor-based therapy such as KEYNOTE-522 (KN522), remains unclear. The KEYNOTE-522 regimen (neoadjuvant pembrolizumab combined with a four-drug chemotherapy backbone, followed by adjuvant pembrolizumab) is a standard of care for stage II-III TNBC. We evaluated the correlations between age and pCR, germline mutation status, and treatment-related toxicities in a cohort of breast cancer patients treated with the neoadjuvant therapy, KN522 regimen. Methods: We evaluated 78 patients with early-stage TNBC enrolled in a clinical registry at this single institution. Of these, 66 (84.6%) received KN522-based neoadjuvant therapy, while others underwent chemotherapy (2.6%) or upfront surgery (12.8%). Age at diagnosis (range 23–85 years, mean 52.9 ± 14.9) was analyzed as a continuous variable, and pCR status (yes/no) was the binary outcome. We also evaluated key variables including germline testing, HER2 status, ECOG, BMI, and toxicity data. Pathologic response data was available for 72 patients (92.3%). Results: Pathologic complete response (pCR) was achieved in 51.4% (37/72) of evaluable patients. Age distribution did not differ significantly between those who achieved pCR and those who did not (median age: 52 vs. 53 years, respectively), and no significant linear correlation was observed between age and pCR (point biserial correlation r = –0.08, p > 0.3). Patients with germline BRCA1/2 mutations (13/70, 18.6%) demonstrated a numerically higher pCR rate, though statistical significance was limited by sample size. Older patients (>65 years) experienced higher rates of toxicity and treatment modifications; however, this was not associated with lower pCR rates. In the KN522 subgroup, pCR rates were slightly higher, aligning with existing trial data, but age remained a non-significant modifier of treatment response. The cohort was entirely female, 87% non-Hispanic white, and 65% Hispanic, with no significant correlation between age and race or ethnicity. Conclusion: In this real-world, retrospective cohort of early-stage TNBC patients receiving neoadjuvant therapy with KN522, pathologic complete response (pCR) was achieved in over half (51.4%) of evaluable patients with no significant association between age at diagnosis and pCR outcomes. Toxicities were common during KN522 treatment, with 100% of patients experiencing at least one toxicity. Although older patients experienced increased toxicity and treatment modifications, this did not translate into lower response rates. Older patients may experience greater rates of severe toxicity; however further prospective investigation should be considered. Germline BRCA1/2 mutation carriers showed a trend toward higher pCR, though statistical power was limited. These findings suggest age alone should not be a determining factor in predicting treatment response in early-stage breast cancer, consistent with existing KN522 trial data. These findings also emphasize the importance of incorporating age and genetic testing in personalized treatment planning.
Presentation numberPS4-10-03
Programmed cell death ligand 1 (PD-L1) expression profiles of early-stage triple-negative breast cancer (TNBC) in China: a retrospective observational study
Jiali Wang, MSD China, Shanghai, China
M. Shen1, L. Liu2, Y. Liu3, S. Zhou4, Y. Ding5, D. Lin3, R. Luo2, H. Bu1, J. Liu6, J. Wang6, L. Wang6, Y. Liu5, W. Yang4; 1Department of Pathology, West China Hospital, Sichuan University, Chengdu, CHINA, 2Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou City, CHINA, 3Department of Pathology, Beijing Cancer Hospital, Beijing, CHINA, 4Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 5Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, CHINA, 6Value & Implementation, Global Medical & Scientific Affairs, MSD China, Shanghai, CHINA.
Background The incidence and mortality of breast cancer (BC) in China are expected to rise to nearly 400,000 new cases and nearly 90,000 deaths annually by 2030, with TNBC accounting for 10-20%. Immune checkpoint inhibitors, including the programmed cell death 1 (PD-1) inhibitor, pembrolizumab, are approved for treating early TNBC (PD-L1 CPS ≥20) in China. However, data on the profiles of PD-L1 expression in Chinese patients (pts) with early-stage TNBC remain limited. Objectives To determine the prevalence of PD-L1 expression in Chinese pts with early-stage TNBC and to assess its correlation with clinicopathological characteristics. Methods This observational, retrospective study, conducted across five centers in China, included female adult pts with early stage (American Joint Committee on Cancer [AJCC] IIA-IIIB) TNBC. Eligible pts had a biopsy or surgical specimen available for PD-L1 testing, and had received no prior systemic therapy for TNBC, with no treatment received within the past 3 years. Newly obtained specimens were defined as tissue collected ≤90 days prior to PD-L1 testing; archival specimens were formalin fixed, paraffin embedded specimens collected by resection or core needle biopsy and were ≤3 years old. PD-L1 expression was evaluated at pathology laboratories of each hospital with the PD-L1 immunohistochemistry (IHC) 22C3 pharmDx assay (Agilent Technologies, CA, USA) performed on the Dako ASL48 platform. PD-L1 expression was reported as a combined positive score (CPS), whereby a score ≥1 was positive and <1 was negative. Data in medical records were used. For group comparisons (positive vs. negative), P values were calculated using a two-sided t-test or Wilcoxon rank sum test for continuous variables, a two-sided Pearson’s Chi-square test or Fisher’s exact test (if ≥25% cells had expected counts <5) for categorical variables, and a two-sided Wicoxon rank sum test for ordinal variables. 95% confidence intervals were calculated using the Clopper-Pearson method. Results In total, 300 patients were included, of whom 296 had evaluable samples. The median age at diagnosis was 54 years; 181 pts (61%) were postmenopausal. The majority of pts (n=255; 86%) had no family history of TNBC; family history was present in 13 (4%) and unknown in 28 (9%). Archival tissue was used in 292 (99%) cases, while newly obtained tissue was used in 4 (1%); specimens included resections (215 pts [73%]) and biopsies (81 pts [27%]). PD-L1 positivity was observed in 81% of evaluable pts, with CPS scores distributed as follows: <1 in 56 (19%), 1-<10 in 106 (36%), 10-<20 in 48 (16%), and ≥20 in 86 (29%). Compared with CPS-negative pts, CPS-positive pts had a younger age at diagnosis (median [Q1-Q3], 53 [45-61] vs 58 [49-66] years; p=0.0192) and had higher Ki-67 expression (median [Q1-Q3] expression, 70% [60%-80%] vs. 50% [25%-70%]; p=0.0001). No statistically significant difference between CPS-positive and -negative groups was found in menopausal status (pre/postmenopausal), family history, primary tumor site (left/right/bilateral), grade (well/moderately/poorly differentiated), stage at initial diagnosis, Eastern Cooperative Oncology Group (ECOG) status (0/1), specimen type (biopsy/resection), germline BRCA mutation (pathogenic/uncertain/benign). Conclusions This observational, retrospective study provides important data on the characteristics of pts with early stage TNBC in China, including the prevalence of PD-L1 expression, and its association with clinicopathological features.
Presentation numberPS4-10-04
Her2 expression is associated with pathologic complete response to neoadjuvant chemo-immunotherapy among patients with stage 1-3 triple negative breast cancer
Jennifer L Baker, UCLA, santa monica, CA
J. L. Baker1, S. M. Thomas2, V. Rey2, I. Eng1, D. M. Marzese3, M. Ensenyat-Mendez3, H. L. Siu-Yuan1, M. L. DiNome3; 1Surgery, UCLA, santa monica, CA, 2Department of Biostatistics & Bioinformatics, Duke University, Durham, NC, 3Surgery, Duke University, Durham, NC.
Introduction: The emergence of novel antibody-drug conjugates has brought significant attention to HER2-low breast cancer as a critical predictive biomarker. However, despite its therapeutic relevance, evidence supporting HER2-low breast cancer as a distinct biological subtype remains limited. We recently demonstrated that HER2-low triple-negative breast cancer (TNBC) exhibits a unique immune-evasive epigenetic and transcriptomic profile compared to HER2-zero, strongly suggesting that it represents a molecularly distinct entity. Based on these findings, this study aimed to assess the clinical response to standard of care neoadjuvant chemotherapy and immunotherapy (chemo-IO) in patients with early-stage TNBC based on HER2 status. Patients/Methods: We identified adult (age 18 and older) female patients with stage 1-3 TNBC treated with chemo-IO at two cancer centers over 3 years (6/2021-5/2024). HER2 status was stratified in two classification schemes: 1- Binary, HER2-zero (Immunohistochemistry [IHC] 0) versus HER2-low (IHC 1+ or 2+, FISH non-amplified); and 2- ordinal, based on IHC scores (0, 1+, or 2+). The primary endpoint was the pathologic complete response (pCR) rate by HER2 status. Univariate and multivariate logistic regression models were used to estimate the association between patient and tumor factors and pCR. To estimate the underlying population pCR rates more robustly and account for sampling variability, we conducted a bootstrap analysis (1,000 iterations), generating confidence intervals for the pCR rates in each subgroup. Results: We identified 165 patients (HER2-zero, n=50; HER2-low, n=115); those categorized as HER2-low were stratified into IHC 1+ (n=72) and IHC 2+ (n=43). The median age was 55 years [IQR 45-64]. Most patients were post-menopausal (60%), had clinical T2 or greater tumors (81%), or were node-positive (45%). No differences in patient or tumor characteristics were noted between groups except for race and ethnicity (p=0.001); patients of self-reported Black race had the highest proportion of HER2-low TNBC (88.4%) compared to White (66.7%), Asian (71.4%), and Hispanic (44.4%) patients. Rates of pCR notably differed by HER2 status with HER2-low TNBC tumors exhibiting lower rates of pCR (53.9%) compared to HER2-zero (66.0%). Increasing HER2 expression levels also correlated with lower rates of pCR (HER2 0: 66.0%, HER2 1+: 55.6%, HER2 2+: 51.2%). A bootstrap simulation confirmed the robustness of these estimates (HER2-zero: 65.6%, 95% CI 51.9-78.5%; HER2-1+: 55.6%, 95% CI 44.3-67.5%; HER2-2+: 51.0%, 95% CI 36.1-65.9%). Conclusions: While our analysis was underpowered to detect statistically significant differences due to the modest sample size, the observed 12.1% absolute decrease in pCR for HER2-low TNBC tumors suggests a clinically meaningful trend. Our results provide clinical evidence that HER2-low TNBC may be a distinct subtype less responsive to standard of care chemo-IO. These results underscore the need for expanded patient cohorts to further validate these associations and guide the development of tailored therapeutic strategies for this purported unique TNBC subgroup.
Presentation numberPS4-10-06
Analysis of Adverse Events and Outcomes of Adjuvant Therapies in TNBC as Monotherapy and Combination: Pembrolizumab, Capecitabine, and Olaparib
Rodolfo Garza-Morales, Mayo Clinic Arizona, Phoenix, AZ
R. Garza-Morales1, F. Raheem1, D. Alton2, S. Sharma2, B. Murad3, W. Harris4, E. Terwilliger2, F. Batalini1; 1Hematology & Oncology, Mayo Clinic Arizona, Phoenix, AZ, 2Mayo Clinic Alix School of Medicine, Mayo Clinic, Scottsdale, AZ, 3Departmento de Medicina, Faculdade de Medicina de Barbacena, Barbacena, BRAZIL, 4Internal Medicine, Mayo Clinic Arizona, Phoenix, AZ.
IntroductionTriple-negative breast cancer (TNBC) is an aggressive subtype lacking estrogen (ER), progesterone (PR), and HER2 expression. It accounts for ~15% of breast cancers and is more common in Black women, younger patients, and those with BRCA1 mutations. TNBC carries a poor prognosis with 5-year survival rates of 92% (localized), 66.8% (regional), and 14.3% (metastatic). Patients with high-risk of recurrence can be treated with different adjuvant therapies based on results of 3 trials (capecitabine, pembrolizumab, olaparib), but data on adverse events (AE) on their combination is limited.High-risk, stage II-III TNBC is treated with curative intent using neoadjuvant chemotherapy with pembrolizumab, followed by surgery and radiation. Pembrolizumab, an anti-PD-1 checkpoint inhibitor, was FDA-approved in 2021 based on KEYNOTE-522. This phase III trial showed that pembrolizumab plus chemotherapy significantly improved pathologic complete response (pCR) and event-free survival (EFS). 3-year EFS was 84.5% vs. 76.8% with placebo (HR 0.63); 5-year follow-up confirmed sustained benefit, with OS of 86.6%. Patients achieving pCR had better outcomes, while those with residual disease remained at high risk for recurrence, justifying further adjuvant therapy.In KEYNOTE-522, grade ≥3 treatment-related (AEs occurred in 77.1% receiving pembrolizumab-chemotherapy vs. 73.3% with placebo-chemotherapy. Pembrolizumab was linked to more serious AEs (34.1% vs. 20.1%) and higher discontinuation rates (23.3% vs. 12.3%). Before pembrolizumab, capecitabine was standard of care for TNBC with residual disease, based on the CREATE-X trial (HR 0.52 for OS benefit). Common AEs included hand-foot syndrome, fatigue, and liver enzyme elevation. Notably, KEYNOTE-522 excluded adjuvant capecitabine, and CREATE-X predated the use of pembrolizumab. Olaparib, a PARP inhibitor, was FDA-approved in 2022 for high-risk early TNBC with germline BRCA mutations, based on OlympiA. It improved invasive disease-free and OS. Grade ≥3 AEs included anemia, neutropenia, and fatigue. OlympiA did not include pembrolizumab or capecitabine, limiting insight into optimal sequencing or combination. Currently, no trials compare pembrolizumab, capecitabine, or olaparib directly, nor do they guide how to sequence or combine these therapies. Despite NCCN guidelines allowing combined/sequential use in high-risk patients, real-world safety and outcome data are lacking. This study aims to evaluate and compare the safety and effectiveness of capecitabine, olaparib, pembrolizumab, and their combinations. We will describe AE rates, onset, discontinuation, and dose modifications to inform clinical management and help reduce early termination of effective therapies.Methods We conducted a retrospective observational study across all Mayo Clinic sites. Eligible patients were ≥18 years with early-stage TNBC who initiated adjuvant therapy with capecitabine, pembrolizumab, olaparib, or combinations (capecitabine + pembrolizumab or olaparib + pembrolizumab) from October 2018 to September 2024. Patients with metastatic disease, other active malignancies, or receiving concurrent investigational therapy were excluded. The primary outcomes were incidence and timing of treatment-related AEs. Secondary outcomes included time to AE resolution, immune-related AEs, treatment interruptions/modifications, early discontinuation, and EFS. Time-to-event data will be analyzed using Kaplan-Meier and Cox proportional hazards models. Subgroup analyses by age, DPYD status, regimen, and dosing will be conducted. Data was abstracted from electronic health records and managed using REDCap.Results Data collection is completed. Final analyses are being conducted, and results will be presented at SABCS 2025.
Presentation numberPS4-10-07
Pathologic complete response rates in early-stage triple negative metaplastic breast cancer in the neoadjuvant immunotherapy era: a retrospective review
Naomi Dempsey, Miami Cancer Institute, Miami, FL
N. Dempsey1, L. Hodgson2, A. Sandoval-Leon1, L. Carcas1, M. Ahluwalia1, C. Wang3, H. Kim3, P. Advani4, R. Mahtani1; 1Medical Oncology, Miami Cancer Institute, Miami, FL, 2Research Administration, Miami Cancer Institute, Miami, FL, 3Data Analytics, COTA Healthcare, New York, NY, 4Medical Oncology, Mayo Clinic, Jacksonville, FL.
Introduction: Metaplastic breast cancer (MpBC) is a rare and histologically diverse subtype of breast cancer (BC) that is frequently triple negative (TN) and exhibits poor responsiveness to chemotherapy (CT). Our group previously reported a low pathologic complete response (pCR) rate among patients (pts) with MpBC treated with neoadjuvant CT in the pre-immunotherapy (IO) era. The standard neoadjuvant approach for pts with stage 2-3 TNBC has evolved with the results of the KEYNOTE-522 (KN522) trial, which demonstrated improved pCR rates, event-free survival (EFS), and overall survival (OS) with the addition of pembrolizumab to CT. However, pts with MpBC were not well-represented in KN522, and their response to IO remains poorly characterized. In this study, we examine the pCR rate with the addition of IO to neoadjuvant CT in pts with TN MpBC. Methods: Pts diagnosed with MpBC and treated at Miami Cancer Institute between 2017-2024 were retrospectively identified by COTA real-world Analytics® platform following IRB approval. Eligible pts had stage 2-3 ER-negative (defined as ER < 10%), PR-negative, HER2-negative MpBC, received neoadjuvant systemic therapy, and subsequently underwent definitive surgery. Demographic, clinical, treatment and outcome data were extracted from the electronic health record. pCR was defined as the absence of residual invasive cancer in the breast and axillary lymph nodes at the time of surgery. Kaplan-Meier method was used to estimate median disease-free survival (mDFS) and median OS (mOS). Comparison between treatment groups for pCR were performed using Fisher’s exact test. No multivariable or survival modeling was performed due to limited statistical power in this small cohort. All p values were two-sided. Results: A total of 27 pts were included in the analysis; 16 pts received CT (in the pre-IO era) with 19.2 months (m) median follow up and 11 pts received CT + IO with 15.5m median follow up. Spindle cell histology was reported in 18.5% of tumors, non-spindle cell in 59.3% and 22.2% were not further specified. Most patients (81.5%) had clinical stage 2 disease, 18.5% had stage 3; median tumor size was 3.2 cm. Anthracycline-based CT was administered to 81.5% of pts, and the remainder received non-anthracycline regimens. pCR was achieved in none of the 16 pts (0.0%) who received CT alone and in 3 of 11 pts (27.3%) who received CT + IO (p=0.046). Among the 3 pts who achieved pCR, 2 had non-spindle cell histology and 1 unspecified. All 3 pCR pts had stage 2 disease, received IO and anthracycline, and experienced immune-related adverse events. mDFS was 23.9m in the CT group and 14.6 m in the CT + IO group (p = 0.641), though follow-up duration differed between groups. Among all pts, mDFS was 15.3 m in those who did not achieve pCR and was not reached in those that did (p = 0.226). mOS was not reached in the CT group and was 20.7m in the CT + IO group (p = 0.192). Conclusion: In this small, retrospective real-world analysis of pts with early-stage TN MpBC, the addition of IO to neoadjuvant CT was associated with a higher pCR rate compared to CT alone. Although the observed pCR rate of 27.3% with CT+IO is lower than that reported in KN522 for unselected TNBC, this likely reflects the small sample size and the inherently CT-resistant biology of MpBC. None of the pts in the CT alone group achieved pCR, suggesting that IO may help to overcome CT resistance in MpBC. Differences in mDFS and mOS were not statistically significant, possibly due to limited follow up and sample size. Despite these limitations, our findings highlight the importance of incorporating IO into treatment for MpBC. Future multicenter analyses are ongoing to validate these results in a larger, more diverse population and to further explore the role of histologic subtypes and immune biomarkers in predicting response.
Presentation numberPS4-10-08
Impact of BRCA Mutation Status on Pathologic Complete Response and Treatment Patterns in Triple-Negative Breast Cancer Patients Treated with Chemoimmunotherapy: A Real-World Analysis
Mohammad F ALDAWOUD, King Fahad Medical City, Riyadh, Saudi Arabia
M. F. ALDAWOUD, F. T. FAQIHI, N. ALGAZLAN, M. M. ALHARBI, A. A. ALMAZYAD, A. R. ALTAMIMI, M. RUDAINEE, A. A. ALWOHAIBI, A. K. ALTWAIRGI; Adult medical oncology, King Fahad Medical City, Riyadh, SAUDI ARABIA.
Background Pathologic complete response (pCR) following neoadjuvant chemoimmunotherapy is an important prognostic marker in triple-negative breast cancer (TNBC). BRCA mutation carriers may exhibit distinct clinical features and treatment responses. We aimed to assess the impact of BRCA status on pCR and treatment patterns among TNBC patients receiving KN522 neoadjuvant regimens in a real-world setting. Methods We identified 45 BRCA-tested patients with stage I-III TNBC who received a KN522 chemoimmunotherapy regimen at King Fahad Medical City between January 2021 and December 2022. Clinicopathologic characteristics, treatment compliance, surgical approach, and pCR (defined as ypT0/is ypN0) were compared between BRCA-positive and BRCA-negative groups. Results Of the 45 patients, 11 (24.4%) were BRCA-positive and 34 (75.6%) were BRCA-negative. BRCA-positive patients were younger (median age 38 vs 46 years) and more likely to have bilateral breast cancer (9.1% vs 2.9%). Grade 3 tumors predominated in both groups (91%), and mastectomy was more frequent among BRCA-positive patients (81.8% vs 64.5%). The pCR rate was 81.8% in BRCA-positive patients (9/11) compared to 48.4% in BRCA-negative patients (15/31), showing a trend toward statistical significance (p = 0.08). Treatment compliance was high: 63.6% of BRCA-positive and 70.6% of BRCA-negative patients completed the full planned regimen. Early discontinuation occurred in 18.2% and 26.5% of patients, respectively. Adjuvant PARP inhibitors were given to 2 BRCA-positive patients (18.2%), while 8 BRCA-negative patients (23.5%) received adjuvant capecitabine. Conclusions In this real-world cohort of TNBC patients treated with neoadjuvant chemoimmunotherapy, BRCA-positive patients demonstrated a higher pCR rate and distinct clinicopathologic characteristics, including younger age and higher mastectomy rate. Treatment compliance and tolerability were acceptable across groups. These findings support the potential role of BRCA status in guiding neoadjuvant strategies in early-stage TNBC.
Presentation numberPS4-10-09
A cell-free DNA prognostic indicator for pathological response and recurrence in early triple negative breast cancer
Sabine Kasimir-Bauer, University Hospital Essen, Essen, Germany
S. Kasimir-Bauer1, C. H. Zuendorf1, A. Bittner1, O. Hoffmann1, R. Kimmig1, J. Siveke2, S. Lueong2; 1Department of Gynecology and Obstetrics, University Hospital Essen, Essen, GERMANY, 2Bridge Institute of Experimental Tumor Therapy (BIT), West German Cancer Center, University Hospital Essen, Essen, GERMANY.
Background: The addition of a checkpoint inhibitor to platinum containing neoadjuvant therapy (NACT) has significantly improved pathological complete response (pCR), progression-free (PFS) and overall survival (OS) in early triple negative breast cancer (eTNBC). However, clinical management of this breast cancer (BC) subtype remains challenging, as only between 40-50% of patients achieve a pCR and the rest witness rapid disease recurrence. Biomarkers to accurately predict pCR and recurrence, therefore, constitute unmet clinical needs. We herein deploy DNA-methylation patterns in tumor tissue as well as cell-free DNA (cfDNA) methylation before and after NACT for the identification of response and relapse biomarker in eTNBC. Patients and Methods: Blood and tumor tissue of 49 patients presenting with first diagnosis of eTNBC between Jan 2013 and Apr 2018 were evaluated for tumor DNA (n=27) and cfDNA methylation before (n=32) and after NACT (n=19). Paired plasma samples were available in 14 cases, paired tumor-plasma samples in 15 cases, respectively. Bead-based concentration and column isolation of cfDNA was achieved with the QiaAmp Mini Elute cfDNA Mini kit (Qiagen). Following fluorometric-based quantification, cfDNA methylation sequencing libraries were prepared with the Enzymatic Methylseq kit (New England Biolabs) followed by hybrid capture with an in-house panel. Libraries were sequenced on a Nextseq 2000 P4 flow cell and data analyzed with the Bismark suite. Missing values were imputed by K-Nearest Neighbors imputation (KNN). Differential methylation analyses was performed with the RnBeads suit and a penalized least absolute shrinkage and selection operator (LASSO) regression was used for biomarker identification. Maximal log-rank statistic was used for the determination of biomarker cut-off for all continuous variables. Results: Of all differentially methylated sites between pre- and post-therapy plasma samples, about 60% of sites showed a significant decrease in methylation levels. In paired analyses, tumor and plasma DNA methylation profiles mirrored each other for a substantial number of the selected markers. We identified sixteen differentially methylated sites associated with a pCR, most of which were located on chromosome 5. Single response marker analyses revealed markers with sensitivity and specificity higher than 0.8. Combination of at least two markers resulted in sensitivity and specificity over 0.9. The negative and positive predictive values were above 0.8 and 0.9 for single and combined markers, respectively. A prognostic index score combining all 16 response markers was significantly prognostic for OS (HR: 0.16, p value 1.25e-05). Furthermore, a 17 disease recurrence signature could be established with single markers attaining sensitivity and specificity of above 0.7 and 0.9, respectively, while combination of at least two markers reached 100% sensitivity and specificity. A recurrence risk signature combining all 17 recurrence markers was strongly prognostic for OS (HR: 0.16, p value 2.26e-05). Conclusion: Collectively, we have established cfDNA prognostic indicator signatures for pCR and recurrence in eTNBC. The validation of these signatures is ongoing in one of our independent validation cohorts and large multicentric cohorts could further foster clinical translation.
Presentation numberPS4-10-10
Trends in Neoadjuvant Pembrolizumab Use in Triple Negative Breast Cancer
Ellen Li, Cornell University, Ithaca, NY
E. Li1, S. R. Spierling Bagsic2, R. F. Belasco2, C. H. Nguyen3, S. Ali4, N. Liloia5, A. A. Aguayo6, D. Lindsay7, T. A. Buchholz8; 1Biology, Cornell University, Ithaca, NY, 2Research & Development, Scripps Health, San Diego, CA, 3Radiation Oncology, Scripps Cancer Center, San Diego, CA, 4Medical Oncology, Scripps Cancer Center, Scripps Clinic Medical Group, San Diego, CA, 5Neuroscience, Bucknell University, Lewisburg, PA, 6Biology, Charles R. Drew University of Medicine and Science, Los Angeles, CA, 7Director, Oncora, Philadelphia, PA, 8Medical Director, Scripps Cancer Center, San Diego, CA.
Introduction: Since its trial demonstrating efficacy in February 2020 and FDA approval for its use in the neoadjuvant setting occurred in July 2021, the integration of pembrolizumab with neoadjuvant chemotherapy (NAC) has significantly improved prognosis and treatment efficacy in triple-negative breast cancer (TNBC) (1,2). Our study investigated national rates of NAC-pembrolizumab use for TNBC, focusing on how patient factors and socioeconomic status affected adoption of pembrolizumab. Methods: We utilized the Epic Cosmos national database to analyze pembrolizumab use among patients with TNBC who received NAC between 1/1/2021 – 12/31/2024. We studied how race/ethnicity and the area of deprivation index (ADI), a standardized score highlighting the social economic designation of a geographic region derived from census variables (3), correlated with pembrolizumab use. Results: To define our population, we used breast cancer ICD-10 codes that included a new diagnosis of estrogen receptor (ER)-negative disease and/or triple negative disease. We excluded those with listing of ER-positive, progesterone receptor-positive, or HER2-positive disease, and those with Stage 0, I, or IV disease documented. In addition, we included only those patients who had an encounter with an oncologist and who received paclitaxel without trastuzumab as a component of neoadjuvant treatment prior to a breast surgical procedure. A total of 6,689 unique patients were identified. Overall rate of pembrolizumab use over the course of the study was 76.2%. This rate increased from 35.9% in 2021, 79.9% in 2022, 84.1% in 2023, and 87.1% in 2024. There were no differences in pembrolizumab according to race or ethnicity. Patients with the highest quartile (highest risk) of ADI scores had significantly lower rates of use compared to the rest of the population (77.4% vs. 74.0%, p=0.04). Investigating rates of adoption of pembrolizumab by year revealed a trend toward lower rate of use in Black patients compared to White patients in 2021 (32.1% vs, 36.9%, p=0.08), but these differences were negligible in subsequent years. Similarly, the differences in use according to ADI was more pronounced in the earlier years relative to the later. Conclusion: This study reveals high rates of pembrolizumab utilization within a year of FDA approval for its use with NCT treatment of triple negative breast cancer. Overall, we did not find significant differences in its use according to race and ethnicity. Patients who live in areas of higher social economic deprivation had lower rates of use. There was a suggestion that adoption rates were slower in those with higher ADI. References: 1. Schmid, P., Cortes, J., Pusztai, L., et al. Pembrolizumab for Early Triple-Negative Breast Cancer. New England Journal of Medicine, 2020. 382(9), 810-821. https://doi.org/10.1056/nejmoa1910549 2. Center for Drug Evaluation and Research. (2021, July 27). FDA approves pembrolizumab for high-risk early-stage triple-negative breast cancer. U.S. Food and Drug Administration. Retrieved from https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-high-risk-early-stage-triple-negative-breast-cancer. 3. Kind AJH, Buckingham W. Making Neighborhood Disadvantage Metrics Accessible: The Neighborhood Atlas. New England Journal of Medicine, 2018. 378: 2456-2458. DOI: 10.1056/NEJMp1802313. PMCID: PMC6051533.
Presentation numberPS4-10-11
Pre and Post Treatment Microbiota Profiling in Archival Tumour Tissue fromEarly-Stage Triple-Negative Breast Cancer
Emilene da Silva Morais, University College Cork, Cork, Ireland
E. da Silva Morais1, E. Lynch1, M. A. Hennessy2, C. P. Devoy1, G. Walsh3, E. Crowley4, C. Girleanu5, J. Barry1, J. Fay6, L. Young6, M. Tangney1, R. Connolly1; 1Cancer Research @ UCC, University College Cork, Cork, IRELAND, 2Dept Breast Medical Oncology, University of Chicago, Chicago, IL, 3Cancer Trials Cork, CUH/UCC Cancer Centre, Cork University Hospital, Cork, IRELAND, 4Clinical Research Facility-UCC, University College Cork, Cork, IRELAND, 5Department of Academic Pathology, Cork University Hospital, Cork, IRELAND, 6Department of Pathology, Royal College of Surgeons in Ireland, Dublin, IRELAND.
Background Emerging evidence highlights the role of the microbiota in breast cancer prognosis and treatment response, with tissue-resident bacteria potentially influencing the tumour microenvironment and immune modulation. Formalin-fixed paraffin-embedded (FFPE) tissue represents a vast, clinically annotated resource in pathology archives, offering a unique opportunity for retrospective microbiome studies. However, its suitability remains uncertain due to DNA fragmentation, low bacterial biomass, and background contamination. We accessed the feasibility of using FFPE breast tumour tissue for bacterial DNA analysis in early-stage triple-negative breast cancer (TNBC) patients receiving neoadjuvant chemotherapy. Methods Breast tumour specimens from 23 patients enrolled in an observational biobank protocol (NCT01840293) were obtained. A total of 38 samples were analysed: 18 baseline biopsies and 20 surgical resections. Paired baseline and surgical specimens were available for 70% (16/23) of patients. Median age at diagnosis was 42 years. All patients received standard-of-care neoadjuvant chemotherapy before primary surgery. 61% (14/23) of patients achieved a pathological complete response (pCR). DNA was extracted using a protocol optimised for FFPE tissue and assessed via Qubit fluorometry. Bacterial DNA was amplified using polymerase chain reaction (PCR) with primers targeting the 16S rRNA V1-V2 and V3-V4 regions. Quantification was performed using qPCR targeting the V6 region of the 16S rRNA gene, with a synthetic gene block as standard. Human DNA was quantified by qPCR targeting the RNase P gene, with human genomic DNA as standard. Water was used as a no-template control on each qPCR plate, alongside block-matched tissue-free FFPE shaves to monitor background contamination. Purified bacterial DNA was used as a positive control to validate PCR assays. Laboratory procedures were designed to minimise contamination, though archival FFPE specimens were not originally prepared for microbial analysis, limiting control over all sources of background DNA. Results Resection samples yielded average DNA concentrations of 85.75 µg/µl; biopsies averaged 95 µg/µl, consistent with FFPE expectations. Tissue-free negative controls yielded ~0.1 µg/µl, confirming minimal background DNA from the paraffin matrix. Despite adequate DNA yield, PCR targeting bacterial genes produced no visible amplicons. Positive controls yielded clear bands on agarose gel electrophoresis. qPCR detected similar levels of bacterial DNA in tumour samples and matched tissue-free negative controls, suggesting minimal genuine bacterial DNA and potential background amplification from paraffin contamination. Template-free negative controls showed no amplification. RNase P detection by qPCR confirmed amplifiable human DNA in tumour samples (resection and biopsy), with no amplification in negative controls. Conclusion Archived specimens collected without consideration for microbiota analysis pose challenges for microbial profiling. Despite careful optimisation and contamination-aware laboratory procedures, bacterial DNA signals in tumour samples were indistinguishable from tissue-free controls, likely due to contamination introduced during non-sterile sample collection and embedding. These findings highlight FFPE limitations for tumour microbiota analysis and have informed the refinement of our prospective protocol (NCT06709651), including a shift toward fresh-frozen sampling. While FFPE archives for microbial research would be transformative, background DNA from initial handling remains a major barrier. Funding Supported by the Breast Cancer Research Foundation, Health Research Board, Science Foundation Ireland and Breakthrough Cancer Research (Precision Oncology Ireland).
Presentation numberPS4-10-12
Real-world efficacy of non-anthracycline neoadjuvant chemo-immunotherapy in early-stage triple negative breast cancer: a retrospective two site cohort study
Amanda K Mennie, UNIV TX SW MEDICAL CTR, Dallas, TX
A. K. Mennie, J. Thomas, S. Kanjwal, S. Reddy, G. Delgado Ramos, B. Santos, N. Sadeghi, H. McArthur, N. Unni; Internal Medicine Division of Hematology-Oncology, UNIV TX SW MEDICAL CTR, Dallas, TX.
Background: The KEYNOTE-522 (K522) study demonstrated that the addition of pembrolizumab immune therapy to anthracycline- and taxane-based chemotherapy improves pathological complete response (pCR) rate, event-free survival, and overall survival in high-risk early TNBC. However, the optimal treatment strategy for patients with smaller node-negative tumors who do not meet K522 eligibility criteria is not known, and regimens that mitigate the risk of anthracycline-mediated toxicity in lower risk populations are being evaluated. To explore the efficacy of K522 versus a non-anthracycline approach, we conducted a retrospective cohort study comparing pCR rates in patients with lower risk early TNBC (T1-T2 and N0) who received K522 versus a non-anthracycline regimen.. We further investigated the role of the presence of Tumor Infiltrating Lymphocytes (TILs) and response to neoadjuvant chemoimmunotherapy between treatment groups, given earlier studies which have indicated their potential use as a predictive biomarker of treatment response. Methods: From institutional databases, we reviewed 271 cases of TNBC and identified 83 diagnosed with clinical T1-T2 N0 TNBC between June 2021 and December 2024 at two academic sites—UT Southwestern Medical Center (UTSW) and Parkland Memorial Hospital (PH). Imaging and pathology reports at diagnosis and at time of surgery were evaluated for all cases, as well as chemoimmunotherapy records. All cases had ER positive staining of ≤ 1%, PR staining of ≤5%, and HER2 staining of 0-2 with negative FISH results in those cases with 2+ staining. Presence of TILs within the tumor biopsy pathology report was considered positive, and its association with pCR was assessed. Standard statistical analyses were performed viaunivariate logistic regression and simple linear regression in Graphpad Prism. Results: Among the 83 patients evaluated, 51 (61.4%) achieved a pathologic complete response; 34 of the 59 (57.6%) patients who received the K522 regimen achieved pCR, while 17 of the 24 (70.8%) patients who received the non-anthracycline regimen achieved a pCR (p = 0.45). This was not due to differences in grade (p=.11), clinical stage (p=.36), or ki-67 (p=.54). TILs were reported in 33 patients (39.8%), of whom 25 received the entire K522 regimen (42.4% of this treatment group) and 8 received the non-anthracycline regimen (33.3% of this group). Presence of TILs was significantly associated with pCR (p = .032) with 75.8% of cases achieving pCR while only 52% achieved pCR in the negative TIL cases. This was not due to differences in grade (p=.54), clinical stage (p=.98), or ki-67 (p=.34). Conclusions: While this is a small retrospective study of 83 patients, our findings suggest that in selected patients with early-stage TNBC (T1-T2, N0), a neoadjuvant non-anthracycline regimen combining carboplatin, paclitaxel, and pembrolizumab can achieve pCR rates comparable to the full K522 regimen. While the pCR rates in this study was not-significant between the K522 and non-anthracycline groups, this may be due to small sample size, or to the comparable response of early TNBC to either regimen, and further study is warranted. These results are consistent with prior prospective studies, including NeoPACT and CALGB40603, that demonstrate comparable pCR rates with platinum- and taxane-based regimens in early TNBC. Furthermore, TILs continue to show promise as a predictive biomarker of treatment response and may play an important role in tailoring treatment recommendations for select patients. Further studies are warranted in order to delineate the most effective treatment approach in these patients.
Presentation numberPS4-10-13
Clinical Feasibility and Reproducibility of Tumor Infiltrating Lymphocyte (TIL) Reporting for Early-Stage Triple Negative Breast Cancer Patients at the University of Washington
Lynn Symonds, Fred Hutch Cancer Center, Seattle, WA
L. Symonds1, D. Reiter2, D. S. Hippe1, A. Kazerouni3, S. Mittal4, N. Hunter1, R. Yung1, W. R. Gwin III1, V. Nair1, N. Davidson1, H. Linden1, J. Specht1, M. Kilgore4; 1Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, 2Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, 3Department of Radiology, University of Washington School of Medicine, Seattle, WA, 4Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA.
Background: Tumor Infiltrating Lymphocytes (TILs) have prognostic and predictive value in triple negative breast cancer (TNBC). Higher TILs correlate with improved rates of pathologic complete response (pCR) following neoadjuvant chemotherapy and lower risk of recurrence and death. While TILs have emerged as an important biomarker, they are not routinely clinically reported. Incorporating TILs into pathology reports for patients with early-stage TNBC has the potential to impact patient care and support ongoing research efforts. Here we report our institutional experience standardizing TIL reporting to generate feasible, reproducible TIL evaluation for diagnostic breast biopsies for early-stage TNBC.Methods: Stage I-III TNBC cases were identified by breast pathologists at the University of Washington. Clinical characteristics and survival data was collected by chart review. TIL status was independently assessed by two pathologists according to the International TILs Working Group Guidelines: a complete assessment was done using at least a 4-5 um section x200-400 per patient and TILs (including all mononucelear cells) were reported for the stromal component within the border of the invasive tumor (TILs outside the tumor border and those in areas of necrosis/crush artifact were excluded). TIL status was categorized as A (low 40%). For cases where readers disagreed on the category, TILs were reassessed by both readers together. Read time was collected to assess impact on pathologist workload.Results: We evaluated TIL status in 56 patients with histologically confirmed TNBC between 2021-2023. Twenty-eight percent had clinical stage I disease, 51% were stage II, and 21% were stage III. The median TIL percentage was 25 (range 1-90) for reader 1 and 20 (range 1-90) for reader 2 (p=0.060). For reader 1, 41% were category A, 27% were category B, and 32% were category C. For reader 2, 36% were category A, 32% were category B, and 32% were category C. Percent agreement in TIL category was 80% (45/56, 95% CI: 68-90%) and unweighted Cohen’s kappa was 0.70 (95% CI: 0.55-0.86). The average time to review was 51 seconds (range 19-93 seconds) for reader 1 and 25 seconds for reader 2 (range 6-127 seconds). There was no significant difference in review times between cases where both readers agreed on category (n = 45) compared to when they disagreed (n = 11) (median time: 40 vs. 41 seconds, p = 0.17, Wilcoxon rank-sum test).Conclusions: Using the International TIL Working Group Guidelines, we found TIL reporting to be highly reproducible between pathologists with 80% concordance. TIL reporting was feasible and the impact on workload was manageable with an average added time of less than 1 minute per case. Standardization of reporting is crucial for ongoing research efforts and valuable for patient care.
| Variable | Reader 1 | Reader 2 | P-value* | Consensus | ||||||||||
| TIL % | 0.060 | |||||||||||||
| Mean ± SD | 26 ± 27 | 30 ± 28 | 29 ± 27 | |||||||||||
| Median (Range) | 25 (1 – 90) | 20 (1 – 90) | 20 (1 – 90) | |||||||||||
| TILs category | 0.39 | |||||||||||||
| A (<10%) | 23 (41%) | 20 (36%) | 20 (36%) | |||||||||||
| B (10-39%) | 15 (27%) | 18 (32%) | 14 (25%) | |||||||||||
| C (≥40%) | 18 (32%) | 18 (32%) | 21 (38%) | |||||||||||
| Time to review (seconds) | 51 (19 – 93) | 25 (6 – 127) | <0.001 | 45 (14 – 235) |
Presentation numberPS4-10-14
Prognostic and Predictive Role of Neutrophil-to-Lymphocyte and Platelet-to-Lymphocyte Ratios in Triple-Negative Early Breast Cancer Treated with Neoadjuvant Chemoimmunotherapy
Ana Godoy-Ortiz, Medical Oncology Unit.,Virgen de la Victoria University Hospital, Malaga, Spain
A. Godoy-Ortiz, A. Romanos-Nanclares, P. Vazquez-Rojas, B. Villaescusa-Gonzalez, A. Sanchez-Muñoz, E. Alba-Conejo; Medical Oncology, Medical Oncology Unit.,Virgen de la Victoria University Hospital, Malaga, SPAIN.
Authors: Godoy-Ortiz Ana1,2,3, Romanos-Monclares Alvaro1, Vazquez-Rojas Paula1, Villaescusa-Gonzalez Beatriz1, Sanchez-Muñoz Alfonso1,4,, Alba-Conejo Emilio,1,2,3,4 Affilations: 1. MEDICAL ONCOLOGY UNIT, VIRGEN DE LA VICTORIA UNIVERSITY HOSPITAL, MALAGA,SPAIN; IBIMA PLATAFORMA BIONAND, MáLAGA, SPAIN; 2. GEICAM; 3. CENTRO DE INVESTIGACIóN BIOMéDICA EN RED DE ONCOLOGíA, CIBERONC-ISCIII, MADRID, SPAIN; 4. UNIVERSITY OF MáLAGA, SPAIN Background: The integration of immune checkpoint inhibitors into neoadjuvant chemotherapy (NACT) has improved pathological complete response (pCR) rates in early-stage triple-negative breast cancer (eTNBC). However, reliable biomarkers of response are still lacking. Systemic inflammatory indices such as the neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) have emerged as potential prognostic and predictive markers in several solid tumors, included eTNBC. We evaluated the impact of baseline NLR and PLR in pathological response and survival outcomes in patients with eTNBC treated with neoadjuvant chemo-immunotherapy.Methods: A retrospective analysis was performed in patients with eTNBC treated with NACT combined with pembrolizumab. Baseline blood counts (pre-treatment) were used to calculate basal NLR and PLR. pCR was defined as no residual invasive cancer in breast and lymph nodes (RCB-0). Median values and interquartile ranges (IQRs) of NLR and PLR were compared between patients who achieved pCR and those with residual disease. Multivariate logistic regression assessed predictors of pCR. Cox proportional hazards models evaluated associations with invasive disease-free survival (iDFS) and overall survival (OS).Results: Seventy-nine patients were included; median age was 50 years (ED 10.4). The overall pCR rate was 64,2% (N:43). Baseline median NLR was 1.81 (IQR 1.37-2.55); PLR was 133.0 (IQR 104.5-164.9). No statistically significant differences in baseline NLR or PLR were observed between pCR and non-pCR groups (NLR: p=0.596; PLR: p=0.652). In multivariate analysis, baseline NLR >1.8 was not associated with pCR (OR 1.02; 95% CI 0.26-3.93; p=0.98). A non-significant trend toward lower pCR was observed among patients receiving antibiotics prior to treatment (OR 0.69; p=0.48). Regarding survival, pCR was associated with a favorable trend for both iDFS (p=0.19) and OS (p=0.082). Baseline NLR >1.8 showed a non-significant association with worse iDFS (coef. -2.21; p=0.19) and OS (coef. -1.79; p=0.30). Clinical T3-T4 tumors showed a trend toward poorer survival outcomes but did not reach significance.Conclusions: In this cohort of patients with localized TNBC treated with neoadjuvant chemoimmunotherapy, baseline NLR and PLR did not significantly predict pCR or survival outcomes. Although trends suggest that elevated NLR may correlate with poorer prognosis, these findings were not statistically significant, possibly due to limited sample size. Further validation in larger prospective cohorts is warranted.
Presentation numberPS4-10-16
Tumor Immune Microenvironment as a Predictive Marker of Neoadjuvant Pembrolizumab Combined with Chemotherapy (KEYNOTE-522 Regimen) Efficacy in Early Triple-Negative Breast Cancer
Nami Yamashita, Cancer Institute Hospital of JFCR, Koto-ku, Japan
N. Yamashita1, T. Osako2, M. Akiya2, Y. Aoyama3, Y. Ozaki3, M. Nishimura3, Y. Haruyama1, U. Nakadaira1, Y. Kimura1, A. lesato4, T. Maeda1, N. Uehiro1, T. Kobayashi3, T. Sakai1, S. Kitano5, T. Takano3, T. Ueno1; 1Breast Surgical Oncology, Cancer Institute Hospital of JFCR, Koto-ku, JAPAN, 2Pathology, Cancer Institute Hospital of JFCR, Koto-ku, JAPAN, 3Breast Medical Oncology, Cancer Institute Hospital of JFCR, Koto-ku, JAPAN, 4Next-ganken Program, Cancer Institute Hospital of JFCR, Koto-ku, JAPAN, 5Advanced Medical Development, Cancer Institute Hospital of JFCR, Koto-ku, JAPAN.
[Background] The current standard of care based on the KEYNOTE-522(KN522) trial for high-risk early triple-negative breast cancer (TNBC) is neoadjuvant pembrolizumab combined with chemotherapy. Tumor infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLS) have been identified in multiple solid tumors and are gaining attention as indicators of predictive biomarkers for immune check point inhibitors (ICIs). We report on the relationship between KN522 regimen treatment efficacy and immune microenvironment.[Methods] We retrospectively analyzed cStage II-III TNBC patients diagnosed from September 2022 to March 2024 who underwent surgery following neoadjuvant treatment with KN522 regimen. We investigated the TILs and TLS in baseline biopsy samples, histological treatment effect (Strict pCR; ypT0N0, pCR; ypT0/isN0), clinical overall response rate (ORR; CR+PR), and axillary lymph node conversion rate (cN+→ypN0). TILs assessment followed the International Immuno-Oncology Biomarker Working Group guidelines, with lymphocyte-predominant breast cancer (LPBC) defined as TILs occupying ≥50% of the area. TLS was defined as lymphoid follicles with germinal centers.[Results] A total of 66 patients were analyzed, with the median age at diagnosis was 51 (25-74) years. cStage II/III comprised 48 cases (73%)/18 cases (27%). TLS was observed in 13 cases (20%), and 19 cases (29%) were LPBC. Univariate analysis showed a mild association between TLS and TILs (p=0.08). Strict pCR/pCR was achieved in 36 cases (55%)/39 cases (59%). While Strict pCR/pCR showed no significant association with TLS (p=0.35/0.53), it was significantly associated with LPBC (p=0.0025/0.0002). The Strict pCR rates for TLS+/LPBC+, TLS-/LPBC+, TLS+/LPBC-, TLS-/LPBC- were 83%(5/6), 85%(11/13), 57%(4/7), and 40%(16/40) respectively. The pCR rates were 83%(5/6), 100%(13/13), 57%(4/7), and 43%(17/40); ORR was 100%(6/6), 100%(13/13), 86%(6/7), and 88%(35/40); and axillary lymph node conversion rates were 100%(3/3), 86%(6/7), 60%(3/5), and 48%(11/23).[Conclusion] Cases identified as LPBC in baseline biopsy samples showed high response rates to KN522 regimen. The immune microenvironment profile could be a potential predictive biomarker for ICI containing treatment.
Presentation numberPS4-10-17
The Impact of Pre-Treatment Clinical, Pathological, and Radiological Parameters on Pathological Complete Response and Prognosis Following Neoadjuvant Therapy in Patients with Triple-Negative Breast Cancer
Perya Abbasoglu, Istanbul Medipol University, Istanbul, Turkey
M. Zencıroglu1, P. Abbasoglu2, O. H. Topgul3, P. Basım2; 1General Surgery, T.C. Sultanbeyli State Hospital, Istanbul, TURKEY, 2General Surgery, Istanbul Medipol University, Istanbul, TURKEY, 3General Surgery, T.C. Sultangazı Haseki State and Research Hospital, Istanbul, TURKEY.
Triple-negative breast cancer (TNBC), accounting for approximately 15% of all breast cancers, is a subtype characterized by the absence of receptor expression, a high response rate to chemotherapy, and a tendency for early metastasis, leading to a generally poor prognosis. This study aims to evaluate the sociodemographic characteristics and clinical, pathological, and radiological features of patients diagnosed with TNBC; to assess their response to neoadjuvant chemotherapy (NACT), identify predictive factors associated with treatment outcomes, and evaluate their effect on disease-free survival (DFS) and overall survival (OS). Between December 2012 and September 2017, 90 female patients diagnosed with stage IIA-IIIB TNBC were retrospectively enrolled in the study from the Breast Surgery Clinic, Department of General Surgery, at İstanbul Medipol University.All patients received standardized treatment protocol beginning with NACT, followed by surgery. Inclusion criteria were confirmed TNBC diagnosis, female sex, age between 30-65 years, no history of prior malignancy, and full adherence to treatment and follow-up protocols. Exclusion criteria included: age outside the specified range, presence of distant metastasis at diagnosis, deviation from treatment or follow-up protocols, and non-cancer-related death during the study period. The data obtained during the study were examined in two separate sections. In the first section, patients were categorized into two subgroups based on whether they achieved a pCR following NACT, to assess the predictability of tumor response to treatment. This section focused on identifying factors associated with achieving pCR. In the second section, the impact of pCR on DFS and OS was examined in all patient- and disease-related variables.The continuous variables assessed using the Shapiro-Wilk test. Variables with normal distribution analyzed using the Student’s t-test, while non-normally distributed variables were analyzed with the Mann-Whitney U test. Categorical variables were evaluated using the Chi-square test, Fisher’s Exact test, and the Fisher-Freeman-Halton test. A total of 84 women with stage IIA-IIIB TNBC were included. The mean age was 49.1±11.5 years. pCR was achieved in 24 patients (28.6%). pCR was significantly associated with postmenopausal status (p = 0.04), smaller tumor size (p = 0.04), higher biopsy Ki-67 (p = 0.02), lower postoperative Ki-67 (p < 0.001), negative clinical and pathological nodal stage, low tumor grade, absence of lymphovascular invasion (LVI) and necrosis, and no axillary involvement at surgery (p < 0.05). MRI and PET-CT responses were strong predictors of pCR (p < 0.001)BRCA1/2 mutation status, tumor subtype, clinical T and N stage, pre-treatment tumor size, MRI and PET-CT response, residual tumor size, presence of LVI and necrosis, pCR status, and postoperative ki-67 level all associated with both DFS and OS. Biopsy ki-67 associated only with OS (p = 0.006). Notably, a postoperative ki-67 value of <10% was associated with significantly better survival outcomes.pCR and low postoperative Ki-67 levels were the strongest predictors of improved DFS and OS. Patients who were postmenopausal and had high biopsy Ki-67 values were more likely to achieve pCR. Although menopausal status and initial Ki-67 were not directly prognostic, their strong association with pCR indirectly linked them to better outcomes. These findings highlight the importance of treatment response in TNBC prognosis and support further multicenter studies to validate these markers in broader populations.
Presentation numberPS4-10-18
Tils as a predictor of response in triple-negative breast cancer: analysis of clinical and pathological outcomes in patients undergoing neoadjuvant chemotherapy based on the keynote-522 protocol in a brazilian cancer center
FLAVIA BALINT, Ac Camargo Cancer Center, Sao Paulo, Brazil
F. BALINT, G. de Almeida, N. Pandolfi, M. Cesca, L. Leite, S. Sanches, E. Santos, V. Cordeiro, M. Tavares; Oncology, Ac Camargo Cancer Center, Sao Paulo, BRAZIL.
Background: Triple-negative breast cancer (TNBC) tumors are aggressive, highly immunogenic, and often associated with early recurrence. Studies suggest that high levels of tumor-infiltrating lymphocytes (TILs) are associated with improved clinical outcomes, including overall survival (OS) and progression-free survival (PFS). We evaluated the presence or absence of complete pathological response (pCR) according to the TILs found in the tumor analysis of patients included in this cohort. Patients and Methods: A total of 121 patients diagnosed with clinical stage (CS) II-III TNBC who received treatment based on the KeyNote-522 protocol (pembrolizumab combined with neoadjuvant chemotherapy) from April 2022 to January 2025 were evaluated. Patients were divided into two groups with a TILs cutoff of >30 and ≤30. Subsequently, clinical outcomes of pCR, overall survival (OS), and progression-free survival (PFS) were assessed. Results: A total of 121 patients, with a median age of 44 years, were analyzed, with the majority (69%) classified as CS II and 63% being premenopausal. TILs analysis was performed in 104 patients, with 74 (71%) having TILs ≤30 and 30 (28.8%) having TILs >30.Regarding tumor characteristics, the predominant histological subtype was ductal in both subgroups (81.8%). The majority of the population had histological grade 3 (67.7%). Regarding pCR, we observed that in the TILs ≤30 subgroup, 37 patients (50%) achieved a complete response, while in the TILs >30 subgroup, 25 patients (83.3%) achieved a complete response. Patients with TILs greater than 30 had a higher likelihood of pCR (OR 0.24; p 30 subgroup, the median OS was 23 months, and the median PFS was 19 months. However, when analyzing the relationship between pCR and TILs, it was found that patients with TILs greater than 30 had a higher likelihood of pCR (OR 0.24; p < 0.01). Conclusion: The presence of TILs greater than 30 was associated with a significant increase in pCR. The data obtained in this study suggest that tumor lymphocytic infiltrate may serve as a prognostic biomarker for pCR in patients with TNBC. However, it should be considered that the sample size was small, and further studies in this area are needed.
Presentation numberPS4-10-19
Impact of Adjuvant Capecitabine in Triple Negative Breast Cancer Patients Undergoing Perioperative Pembrolizumab: A Real-World Brazilian Cohort.
Marcela O Sá, Materdei, Belo Horizonte, Brazil
M. O. Sá, P. C. Diniz, E. M. Lima, R. V. Guedes, V. A. Silva, I. E. Mendes, L. L. Campos, C. C. Avelar, C. B. Pirfo; Oncology department, Materdei, Belo Horizonte, BRAZIL.
Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive disease with limited effective treatment strategies. The addition of pembrolizumab to neoadjuvant chemotherapy (NACT) has reshaped standard care, as demonstrated in the KEYNOTE-522 trial. However, adjuvant capecitabine—previously shown to benefit patients with residual disease in the CREATE-X trial—was not permitted in KEYNOTE-522. This study aimed to evaluate the real-world impact of adjuvant capecitabine in TNBC patients receiving perioperative pembrolizumab in a resource-limited setting.We conducted a retrospective observational study using electronic medical records from a referral cancer center in Brazil. Eligible patients had non-metastatic triple-negative breast cancer (TNBC) with T≥2 cm tumors and/or axillary involvement, treated with neoadjuvant chemotherapy (NACT) between 2016 and 2024. Patients were grouped as follows: (1) NACT + pembrolizumab + adjuvant capecitabine (28.2%), (2) NACT + adjuvant capecitabine (33.3%), and (3) NACT alone (38.5%). The primary endpoint was event-free survival (EFS), defined as the time from diagnosis to recurrence or death. Cox regression was used for multivariable analysis. Intergroup comparisons were performed using appropriate statistical tests, and the Bonferroni correction was applied to adjust for multiple comparisons and reduce the risk of false-positive findings.In this cohort (n=39), the mean age was 46.6 years; 69.2% were premenopausal, and 92.3% had stage II-III disease. Invasive ductal carcinoma was the predominant histology. The most frequent regimen was dose-dense doxorubicin/cyclophosphamide followed by weekly paclitaxel and carboplatin (53.8%) regardless of treatment group. Clinical characteristics were well balanced. After a median follow-up of 58.6 months, pathologic complete response (pCR) was achieved in 51.4% of the cohort. Across all patients, no significant difference in EFS was observed between groups (p=0.316). However, among patients without pCR (Group 1: 44.4%, Group 2: 100%, Group 3: 6,7%) median EFS was notably higher in group 2 than group 1 (p=0.026). These real-world findings suggest that in TNBC patients with residual disease the benefit of perioperative immunotherapy may be mitigated by concurrent use of adjuvant capecitabine. This underscores the need for individualized post-neoadjuvant strategies, particularly in low-resource settings, where access to immunotherapy remains limited.
Presentation numberPS4-10-20
Exploring Potential Predictive Factors for Immunotherapy Response in Early-Stage TNBC: Insights from a Retrospective Analysis
Aroob Sweidan, Henry Ford Hospital, Detroit, MI
A. Sweidan1, J. Sherwood2, M. Ahmed3, S. Bai4, H. Ali1; 1Hematology and Oncology, Henry Ford Hospital, Detroit, MI, 2Internal Medicine, Henry Ford Hospital, Detroit, MI, 3Internal Medicine, Henry Ford Macomb Hospital, Clinton Township, MI, 4Internal Medicine, Henry Ford Jackson Hospital, Jackson, MI.
Background: Neoadjuvant chemoimmunotherapy (NACT-IO) has been the standard of care for high-risk early-stage triple-negative breast cancer (eTNBC) for over five years. However, data on predictive biomarkers and clinical factors associated with treatment response remain limited. This study evaluates the association of several factors with pathological complete response (pCR) following NACT-IO. BRCA1/2 mutations, present in ~15-20% of TNBC cases, are linked to higher tumor mutational burden (TMB), potentially enhancing immunotherapy response. Angiotensin-converting enzyme (ACE) is intrinsically expressed in immune cells and may influence anti-tumor immunity. Additionally, immune-related adverse events (irAEs) occur in ~30% of eTNBC patients receiving NACT-IO; while higher-grade irAEs have been linked to improved pCR, data on low-grade irAEs remain limited. Thus, we assessed how BRCA1/2 status, ACE inhibitor use, and irAE severity may impact response to NACT-IO. Methods: A retrospective analysis was conducted on eTNBC patients treated with NACT-IO at Henry Ford Hospital (Detroit, MI) between July 2021 and October 2024. Categorical variables were analyzed using Chi-square or Fisher exact tests; P < 0.05 was considered significant. Results: Among 173 eligible patients, 14 (8%) had pathogenic BRCA1/2 mutations, 18 (10%) were on ACE inhibitors, 7 (4%) had grade 1 irAEs, and 51 (29%) had grade ≥2 irAEs. pCR rates were significantly higher among those with grade ≥2 irAEs versus those with grade 1 or no irAEs. No significant differences in pCR rates were seen by BRCA status or ACE inhibitor use, though BRCA-mutated patients showed a numerical trend toward higher pCR rates. Table 1. Clinical Factors and Surgical Outcomes (C) = Chi-Square Test, (F) = Fisher Exact Test Conclusion: pCR rates were significantly higher in patients with grade ≥2 irAEs. The low number of grade 1 irAEs may reflect underreporting. While BRCA1/2 mutation carriers had numerically higher pCR rates, this was not statistically significant, suggesting a potential improvement in response that warrants further study. Notably, ~22% of patients lacked documented BRCA status, underscoring the need for universal genetic testing in TNBC. No significant association was found between ACE inhibitor use and surgical outcomes; however, larger studies are needed to clarify their potential impact on response to immunotherapy.
Presentation numberPS4-10-21
Clinicopathological Features and Outcomes of Apocrine Triple-Negative Breast Cancer: A Distinct Subtype with Favorable Prognosis
Thamilyn Saruwatari, AC camargo Cancer Center, São Paulo, Brazil
T. Saruwatari1, D. de Albuquerque1, V. Primo Basílio1, L. Rodrigues Soares1, M. De Brot2, S. Sanches1, V. Cordeiro1, E. Santana dos Santos1; 1Clinical Oncology, AC camargo Cancer Center, São Paulo, BRAZIL, 2Pathological Oncology, AC camargo Cancer Center, São Paulo, BRAZIL.
Introduction: Emerging evidence suggests that triple negative breast cancer (TNBC) is a heterogeneous group comprising biologically distinct subtypes with variable prognoses. Among these, apocrine TNBC represents a rare histological variant characterized by distinct morphological features and molecular profile that may confer a more indolent clinical course. Apocrine TNBC, as defined by the 2019 World Health Organization (WHO) classification, is a special subtype characterized by the absence of estrogen receptor (ER) and progesterone receptor (PR) expression and positivity for androgen receptor (AR). Retrospective cohort studies indicate that patients with apocrine TNBC, often older than 50 years, present with smaller, well- to moderately differentiated tumors compared to other TNBC subtypes. Breast cancer-specific survival (BCSS) and overall survival (OS) are significantly better for apocrine, adenoid cystic, and medullary subtypes compared to metaplastic and NST TNBC. However, data on the response to neoadjuvant or adjuvant chemotherapy in apocrine TNBC remain limited, and clinical decision-making often relies on guidelines developed for NST TNBC. This study aims to describe the clinicopathological characteristics and outcomes of patients with apocrine TNBC. Methods: This is a retrospective, single-center case series. We selected 13 patients with non metastatic apocrine TNBC, EC I, II and III, treated surgically at A.C. Camargo Cancer Center. All patients received neo or adjuvant Cht. Results: The mean age was 59.5 years (range: 35-71 years). All patients (100%) were negative for ER and PR, consistent with TNBC. T1 was the most common stage (N=7, 53.9%), follow by T2 (N=4, 30.7%) and T4 (N=1, 7.7%). The majory was node-negative (N0, N=11, 84.6%), and just one (7.7%) was N+ (N2). The mean Ki67 proliferation index was 33.5% (range: 10-90%). AR high expression in 8 patients (61.5%). 46.2% patient (N=6) had a lower proliferation ki67 of 1-20%, 30.8% patients (N=4) with ki64 between 21-50 and 15.4% (N=2) with ki67 >50%. Histologic grade (GH) was II in 7 patients (53.8%) and III in 4 patients (30.8%). Surgical interventions included breast-conserving surgery in 7 patients (53.8%) and mastectomy in 6 patients (46.2%). All pacients received neoadjuvant or adjuvant chemotherapy. Neodjuvant was administered in 3 patients. Among these, one achieved pCR, while the others were staged as ypT2ypN2 and ypT0ypN1. Local recurrence and distance metastasis occurred in 1 patient (7.7%), while 10 patients (76.9%) had no recurrence. The metastatic site was the contralateral breast, and both cases (local recurrence and contralateral breast) were treated with mastectomy. At the last follow-up, 12 patients (92.3%) were alive without disease, with 1 patient lost to follow-up (7.7%). Conclusion: This study describes the clinicopathological characteristics and outcomes of apocrine TNBC. The mean age of 59.5 years and predominance of T1 and N0 tumors and with a majority exhibiting low to moderate proliferation align with prior findings suggesting that apocrine TNBC occurs in older patients with smaller, well-differentiated tumors. The low rates of local recurrence and distant metastasis (7.7% each) and high rate of patients alive without disease (92.3%) support the hypothesis of a more favorable prognosis compared to NST TNBC. Th AR positivity in 84.6% of patients suggests potential for AR therapies, an area warranting further investigation. Limitations include the small sample size and retrospective design, which preclude robust inferential analyses. Future studies with larger cohorts and detailed molecular analyses are needed to confirm these findings and explore treatment response predictors.
Presentation numberPS4-10-22
Obesity and Triple Negative Breast Cancer: Untangling the Connection
Brusi Kola, Mitchell Cancer Institute, Mobile, AL
B. Kola, S. Kakkat, P. Suman, S. Atterberry, A. Richter, K. Sawrie, R. Gupta, A. Mahadevan, C. Nelson, D. Chakroborty, C. Sarkar; Pathology, Mitchell Cancer Institute, Mobile, AL.
Breast Cancer (BC) is the most commonly diagnosed cancer and the second leading cause of cancer-related deaths among U.S. women. In recent years, alongside with increased BC incidence, the increasing number of overweight and obese people is also a major health concern in the U.S. Obesity is a risk factor for BC and has been linked to high rates of BC recurrence and deaths, yet how it affects BC tumor growth and progression is not fully understood. Here, we analyzed the clinical information collected from 531 patients who received a BC diagnosis at the University of South Alabama Mitchell Cancer Institute Clinics between January 2024 and April 2025. Patients were categorized into three groups based on their body mass index (BMI): normal weight (BMI < 25), overweight (25 ≤ BMI < 30), and obese (BMI ≥ 30). BC patients were most commonly diagnosed with Luminal A BC (64%), followed by TNBC (18%), Luminal B BC (14%), and HER2-enriched BC (4%). Obese BC patients constituted the largest proportion of BC patients in all individual molecular BC subtypes. Interestingly, we observed that for patients with TNBC only, the prevalence of obesity was higher among individuals under 50 years of age (52%) compared to those aged 50 and over (44%). In addition, obese TNBC patients presented with grade 2 and 3 tumors that were characterized by higher Ki-67 levels compared to tumors from grade-matched normal-weight TNBC patients. These findings link obesity to increased TNBC incidence, particularly in young women aged under 50, and suggest an association between obesity and TNBC aggressiveness. To further study the impact of obesity on TNBC progression, we used a mouse model of diet-induced obesity where young female BALB/c mice were fed either a control or high-fat diet (60% kcals) for 12 weeks. To establish primary tumors, murine 4T1-luc2 TNBC cells were injected into the fourth mammary fat pads of lean or obese mice, and tumor growth and progression were monitored. Our results indicated that obese mice experienced an earlier onset of BC and accelerated tumor growth compared to their lean counterparts. One week after tumor transplantation, tumor incidence was found to be 83.3% in the obese group as opposed to 50% in the lean group. Moreover, our immunohistochemistry analysis showed that tumors that grew in obese mice exhibited increased cellular proliferation (Ki-67). In addition, angiogenesis and lymphangiogenesis, the two important processes that facilitate the metastatic dissemination of BC cells, were significantly upregulated in the tumors from obese mice compared to those from lean mice. Interestingly, our RNA-seq data also revealed significant differences in the expression of metastatic-related genes between the 4T1 tumors of lean and obese mice. Studies identifying the factors in the obese tumor microenvironment that make these tumors more aggressive and enhance their metastatic potential will open up exciting new avenues for BC treatment.
Presentation numberPS4-10-23
Uncovering Mechanisms Driving Phenotypic Distinctions between Breast Cancer Cells with Different p53 Mutations using a 3D Microfluidic Platform
Lydia Sakala, Arizona State University, Tempe, AZ
L. Sakala1, K. Ravi2, Y. Zhang1, E. Hurt2, J. G. Park1, M. Nikkhah2, J. LaBaer1; 1Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 2School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ.
TP53 is the most mutated gene in cancer, with alterations present in roughly 50% of all cancer cases and in approximately 80% of the aggressive Triple Negative Breast Cancer (TNBC). The wild type p53 protein plays a vital role in suppressing cancer by promoting DNA damage repair, regulating cell cycle arrest, inducing apoptosis, and inhibiting cell invasion among other functions. Most of these mutations are single amino acid substitutions, occurring in more than 130 forms and contributing to tumor heterogeneity with potential neomorphic activities. However, despite ongoing efforts of developing precision therapeutics for TNBC, the functional impacts of individual mutant p53 as distinct cancer drivers remain poorly understood. Previously, we created a cell line panel with ten p53 mutations common in breast cancer and reported that each mutation led to distinct molecular and phenotypic profiles. Our current study focuses on the R273C and Y234C mutants, which were the most and least invasive, respectively, to identify the underlying mechanisms of phenotypic heterogeneity and potential drug targets. We have employed a 3D tumor-on-chip technology, which offers detailed single-cell-level insights in more in-vivo-like conditions, and observed highly distinct cellular behaviors of R273C cells with mesenchymal morphology and reduced proliferation, compared to cells with wild-type p53 or Y234C mutant that formed large acini-like structures. Immunostaining and RNA-Seq analyses confirmed further that R273C strongly induced epithelial-to-mesenchymal transition (EMT) and gene expression profile changes similar to those of metastatic breast cancer cells such as MB-231. Using the RNA-Seq data, we have identified differentially regulated genes and pathways in R273C and Y234C. We are currently working on mechanistic investigation to understand how the R273C mutant induces EMT and identification/validation of the top candidates that can potentially intervene in TNBC with more aggressive p53 mutations.
Presentation numberPS4-10-24
Longitudinal trends in prognosis and post-treatment pregnancy in young woman with triple-negative breast cancer
Mikiko Kasahara, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
M. Kasahara1, A. Kataoka1, A. Kanazawa1, Y. Ito1, Y. Kimura1, N. Yoshida1, U. Nakadaira1, N. Uehiro1, C. Takahata1, Y. Ozaki1, M. Nishimura1, T. Takano1, T. Kogawa2, T. Sakai1, T. Ueno1; 1Breast Oncology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 2Department of Advanced Medical Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN.
Background: Triple-negative breast cancer (TNBC) in young women has traditionally been associated with poor prognosis. However, treatment outcomes are expected to improve with advances such as response-guided neoadjuvant chemotherapy, PARP inhibitors, and immune checkpoint inhibitors. For young women who have a desire for future pregnancies, whether pregnancy and childbirth are feasible after breast cancer treatment is a critical concern. Therefore, fertility preservation (FP) before cancer treatment and shared decision-making regarding post-treatment pregnancy are essential. Objective: To investigate treatment outcomes, temporal trends in FP, and pregnancy outcomes among young patients with TNBC. Methods: A retrospective analysis was conducted on 179 young patients under the age of 40 with stage I-III TNBC who underwent curative surgery at our institution between 2007 and 2022. We investigated treatment outcomes, FP rates, and subsequent pregnancy status. The primary endpoints were breast cancer recurrence, the development of secondary malignancies, and overall survival. Secondary endpoints were pregnancy and childbirth after breast cancer treatment. Results: The mean age at surgery was 34.4 years. Eighty-three patients (46.4%) had a history of childbirth. Among those tested, 41 patients (43.2%) carried pathogenic BRCA1/2 variants. The stage distribution was stage I in 42 patients (23.5%), stage II in 103 (57.5%), and stage III in 34 (18.0%). Chemotherapy was administered to 171 patients (95.5%), of whom 93 (52.0%) received neoadjuvant therapy. With a median follow-up of 5.8 years (range: 0.1-17.5), recurrence including contralateral breast cancer occurred in 14 patients (7.8%), secondary malignancies in 5 (2.8%)—of whom 3 had ovarian cancer—and 25 patients (14.0%) died. At the time of diagnosis, 61 patients (34.1%) had desire for future pregnancy. Among them, 26 (14.5%) underwent FP before chemotherapy, and 22 (12.3%) received LHRH analogs during chemotherapy for ovarian function protection. Comparing two time periods, 2007-2014 and 2015-2022, the 5-year overall survival significantly improved from 78.7% to 94.5% (p = 0.0088). FP implementation also increased over time, with the FP rate among those with desire for future pregnancies at the time of diagnosis rising from 12% to 58.3%. Seventeen patients conceived after treatment (6 spontaneously, 9 with assisted reproductive technology, 2 unknown), resulting in 23 pregnancies and 17 live births in 15 patients (8.4% of the total cohort, 24.6% of those who had desire for future pregnancies at the time of diagnosis). Among the patients who gave birth, one had been treated with immune checkpoint inhibitors. No serious perinatal complications were observed. All patients were disease-free at the time of attempting conception. No distant recurrence or breast cancer-related deaths occurred after childbirth. To date, none of the patients who treated with PARP inhibitor therapy became pregnant. Conclusion: Although numbers are limited, young patients with TNBC who attempted pregnancy after completing standard systemic therapy had favorable oncologic and perinatal outcomes. In this study, the treatment outcomes and FP implementation rate for young patients with TNBC improved over time. This real-world data may be useful in supporting shared decision-making for reproductive planning and treatment decisions for young patients with TNBC.
Presentation numberPS4-10-25
Intralesional Injection of IL-2 Prior to Surgery in Early-Stage Triple Negative Breast Cancer
Rachel Hemsworth, Izaak Walton Killam Hospital, Halifax, NS, Canada
R. Hemsworth1, G. Knapp2, A. Mayo1, H. Stewart1, A. Goyal3, G. Bethune4, K. Greenlaw5, Z. Kellow5, L. Helyer2, A. Drohan2; 1Breast Health Research Unit, Izaak Walton Killam Hospital, Halifax, NS, CANADA, 2Department of Surgery, Dalhousie University, Halifax, NS, CANADA, 3Research, Nova Scotia Health Authority, Halifax, NS, CANADA, 4Department of Pathology, Dalhousie University, Halifax, NS, CANADA, 5Department of Radiology, Dalhousie University, Halifax, NS, CANADA.
BACKGROUND: Triple negative breast cancer (TNBC) represents 15-20% of all newly diagnosed breast cancers. This form of the disease, which tends to have a higher mutational burden, responds favorably to modern systemic immunotherapy. As a systemic treatment, immunotherapy is associated with rare but potentially life-threatening side-effects. For local, early-stage disease there may be an opportunity to employ targeted intralesional immunotherapy. Preliminary data suggests that intralesional injection of interleukin 2 (IL-2) may be able to produce a favorable pathologic response in TNBC. However, there are currently no protocols for the study of intralesional IL-2 in early-stage TNBC breast cancer. METHODS: This is a single-center, single-arm, non-randomized experimental trial with an initial target enrollment of 10 patients. Women 18-80 years of age with a <2 cm TNBC confirmed on diagnostic imaging and core biopsy are eligible. Patients with major organ dysfunction, history of immune related disease or immunomodulating medication are excluded. In the window-of-opportunity between initial consultation and definitive surgery (<6 weeks), eligible women receive weekly (x4), image-guided (ultrasound), intralesional injections of IL-2. Dose calculation per injection is 500,000 IU (5 million IU/ml) per millimetre of tumor width at index diagnostic imaging to a maximum dose of 15 million IU per injection. The primary outcome is pathologic response as defined by the residual cancer burden (RCB) score on final pathology. Secondary outcomes include, protocol feasibility and adverse events (Common Terminology Criteria for Adverse Events). Final pathological specimens will be evaluated for tumor infiltrating lymphocytes (TILs) and PD-L1 expression, in addition to RCB score. EXPECTED OUTCOME: We expect four weekly injections of intralesional IL-2, at a dose of 500,000 IU per mm of maximum tumor width, to be safe and well tolerated in this cohort of patients. Moreover, we anticipate this protocol will demonstrate treatment response characterized by tumor cell death and increased TILs. FUTURE DIRECTIONS: This study will generate preliminary data necessary to support a larger, adequately powered phase II trial to explore treatment optimization and efficacy. Future studies may explore other forms of intralesional immunotherapy, such as PD-L1 inhibitors, and mechanisms to enhance treatment response.
Presentation numberPS4-10-27
Tolerability of the KEYNOTE-522 regimen in a diverse real-world cohort of patients with early triple negative breast cancer (TNBC)
Justine Anderson, Icahn School of Medicine at Mount Sinai, New York, NY
J. Anderson1, E. Baldwin2, J. Dejesus2, G. Van Hyfte3, P. Pandu4, N. Krishnamurthy5, M. Rattu1, R. Farley1, R. Patel1, A. Tiersten1; 1Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 2Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 3Tisch Cancer Institute – Biostatistics and Clinical Informatics, Icahn School of Medicine at Mount Sinai, New York, NY, 4Department of Internal Medicine, Mount Sinai West, New York, NY, 5Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: The KEYNOTE-522 trial demonstrated a significant improvement in efficacy with neoadjuvant chemotherapy (NACT) in combination with pembrolizumab compared to chemotherapy alone for patients (pts) with early TNBC. Real-world data on the toxicity of this regimen is limited, especially in racially diverse populations. Our study aims to describe the toxicity of the KEYNOTE-522 regimen in a diverse, urban population at a large tertiary referral center, and evaluate the impact of race/ethnicity and age on tolerability of the regimen. Methods: We performed a retrospective chart review of pts with anatomic Stage II-III TNBC who received the KEYNOTE-522 regimen within our health system from August 2021 to February 2025. Pts who received >/= 1 cycle of NACT and pembrolizumab were included. Toxicity was measured by the rates of dose reductions, drug discontinuations, treatment (tx) delays, hospitalizations, and ED visits due to treatment-related AEs (TRAEs). Descriptive statistics were used to describe patient data, and non-parametric tests were utilized in the stratified analyses by race/ethnicity and age. Results We included 111 pts who had a median age of 55 years. Most patients had Stage II (81%) and lymph node negative (52%) BC. Overall, 32% of pts were White, 26% Black, 14% Asian, 16% Hispanic, and 11% Other. Among all pts, 19% (N=21) had dose reductions, and 55% (N=61) had tx delays due to toxicity. 33% (N=36) of pts discontinued one or more agents early, with 14% (N=16) discontinuing pembrolizumab. Hospitalizations and ED visits due to AEs were reported in 33% (N=36) and 28% (N=31) of pts, respectively. There were no significant differences in the rates of dose reductions, drug discontinuations, tx delays, hospitalizations, or ED visits when stratified by race/ethnicity (Table 1). Asian (25%) and White (17%) pts had higher rates of pembrolizumab delays due to irAEs compared to Black (7%) and Hispanic (0%) pts, but this difference was not statistically significant (p=0.051). When stratified by age, 65 years (N=26), there were no significant differences in toxicity though pts 65 years (23%, p=0.036). Conclusion: In a diverse patient population, the observed drug discontinuation rate of the KEYNOTE-522 regimen was 33%, notably higher than the 23% reported in the KEYNOTE-522 trial. In our real-world study, one-third of pts were hospitalized and more than half had treatment delays due to TRAEs, highlighting the toxicity of the regimen. There were no significant differences in tolerability based on race/ethnicity. The higher rates of ED visits and hospitalizations due to irAEs in younger pts should be evaluated in future studies.
|
Overall Population 100% (N=111) |
White 32% (N=36) |
Black 26% (N=29) |
Asian 14% (N=16) |
Hispanic 16% (N=18) |
Other 11% (N=12) |
p-value | |
| Median number of neoadjuvant pembrolizumab cycles | 7.5 | 7 | 8 | 7 | 7.5 | 8 | 0.134 |
| Dose reduction due to AEs | 19% (21) | 17% (6) | 17% (5) | 13% (2) | 28% (5) | 27% (3) | 0.72 |
| Drug discontinuation due to AEs | 33% (36) | 37% (13) | 38% (11) | 25% (4) | 28% (5) | 25% (3) | 0.839 |
| Drug delays due to AEs | 55% (61) | 44% (16) | 62% (18) | 63% (10) | 67% (12) | 42% (5) | 0.403 |
| Pembrolizumab delays due to irAEs | 11% (12) | 17% (6) | 7% (2) | 25% (4) | 0% (0) | 0% (0) | 0.051 |
| Hospitalizations due to AEs | 33% (36) | 39% (14) | 28% (8) | 44% (7) | 22% (4) | 25% (3) | 0.579 |
| ED visits due to AEs | 28% (31) | 25% (9) | 28% (8) | 25% (4) | 44% (8) | 17% (2) | 0.574 |
Presentation numberPS4-10-28
Neoadjuvant Chemoimmunotherapy in Early-Stage TNBC: Real-World Analysis of Predictors of pCR and Survival
Ahmet Bilici, Istanbul Medipol University, Medical Faculty, Istanbul, Turkey
A. Bilici1, H. Muglu1, O. Ozcan2, E. Sunger1, T. Sahin3, B. Koylu4, K. Helvaci5, Y. Kemal6, U. Demirci5, F. Selcukbiricik4, G. Basaran7, B. Uluc7, O. Olmez1, S. Aksoy3, E. Gokmen2; 1Department of Medical Oncology, Istanbul Medipol University, Medical Faculty, Istanbul, TURKEY, 2Department of Medical Oncology, Ege University, Medical Faculty, Izmir, TURKEY, 3Department of Medical Oncology, Hacettepe University, Medical Faculty, Ankara, TURKEY, 4Department of Medical Oncology, Koc University, Medical Faculty, Istanbul, TURKEY, 5Department of Medical Oncology, Ankara Memorial Hospital, Ankara, TURKEY, 6Department of Medical Oncology, Samsun Medicalpark Hospital, Samsun, TURKEY, 7Department of Medical Oncology, Acibadem University, Medical Faculty, Istanbul, TURKEY.
Background: Triple-negative breast cancer (TNBC) is one of the most challenging breast cancer subtypes to manage due to its aggressive biology, high recurrence risk, and limited targeted treatment options. Neoadjuvant chemotherapy plus pembrolizumab (NACP) has become a new standard aimed at increasing pathological complete response (pCR) rates and improving long-term survival. This study aims to evaluate pCR and survival outcomes in early-stage TNBC patients receiving NACP based on real-world data, and to identify clinicopathological factors predictive of pCR and survivals.Methods: This multicenter retrospective study included 164 early-stage TNBC patients treated with NACP, followed by surgery. Demographic, clinical, radiological, and pathological data were analyzed. The primary endpoint was the pCR rate; secondary endpoints included disease-free survival (DFS) and overall survival (OS). Predictors of pCR and survivals were evaluated using multivariate logistic and Cox regression analyses, respectively.Results: Among 164 patients with a median follow-up of 49.2 months, the pCR rate was 60.4%. Recurrence and mortality rates were 18.3% and 3.0%, respectively. In the univariate analyses for DFS, radiological T stage at diagnosis (p=0.001), carboplatin administration schedule (p<0.001), radiological T response to treatment (p=0.036), overall radiological treatment response (p<0.001), and presence of pCR (p<0.001) were found to be statistically significant prognostic factors. In addition, ECOG PS (p=0.03), carboplatin administration schedule (p=0.043), radiological T response to treatment (p=0.003), adjuvant capecitabine administration (p=0.003) and radiological N response (p=0.04) were found to be statistically significant prognostic indicators for OS in univariate analysis. In the multivariate Cox regression analysis performed to evaluate variables affecting survival, carboplatin administration schedule (HR=5.137, p=0.001), age (HR=4.301, p=0.026), achievement of pCR (HR=0.138, p<0.001), and radiological treatment response (HR=0.197, p<0.001) for DFS and only pCR (HR = 0.10, 95% CI: 0.01-1.18, p = 0.004) for OS were found to be significant independent prognostic indicators. To identify predictors of achieving pCR, a multivariate logistic regression analysis was performed, and only carboplatin dosing schedule was significantly associated with pCR (OR=0.308; 95% CI: 0.128-0.740; p=0.008), with weekly dosing favoring higher pCR rates. These findings suggest that carboplatin scheduling may be an independent predictor of pCR, whereas other clinicopathological parameters showed no significant impact in this model.Conclusion: In real-world practice, NACP remains an effective treatment option for early-stage TNBC, yielding high pCR rates and survivals. pCR emerged as a strong predictor of survivals, while weekly carboplatin dosing may enhance treatment efficacy. Integrating clinical, radiological, and pathological factors may support personalized treatment decisions. Prospective studies with larger cohorts and biomarker analyses are warranted to validate these findings.
Presentation numberPS4-10-29
Diverse adjuvant treatment in triple-negative breast cancer patients with residual disease after neoadjuvant chemotherapy: A single center real-world study
Xi Chen, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, beijing, China
X. Chen, L. Ji, H. Lv, G. Song, Q. Li, J. Wang, Y. Fan, Y. Luo, B. Lan, S. Chen, R. Cai, F. Ma, B. Xu, P. Zhang; Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, beijing, CHINA.
Background: Triple-negative breast cancer (TNBC) patients exhibiting residual disease post-neoadjuvant chemotherapy are associated with a poor prognosis. The CREATE-X study indicates that capecitabine adjuvant therapy benefits these patients, particularly the TNBC subgroup, and is now guideline-recommended. However, there’s a lack of real-world data in China. This study aims to present the real-world adjuvant treatment status of TNBC patients with residual disease after neoadjuvant therapy. Methods:This study included TNBC patients with residual disease after neoadjuvant chemotherapy at the Cancer Hospital, Chinese Academy of Medical Sciences, from July 2008 and December 2024. Patients were categorized into capecitabine, intravenous chemotherapy, and no chemotherapy groups based on adjuvant treatment. Survival outcomes were compared among the groups, with a multivariate Cox model adjusting for confounders. Stratified analyses were performed using Neo-bioscore(NB) and residual disease burden. Results: This study included 340 patients, median age 47.4 years (20-76), 61.8% of patients were premenopausal. 120 received capecitabine-containing regimen, 118 had intravenous chemotherapy, 102 without adjuvant chemotherapy. The baseline characteristics were generally balanced among the three groups. In the capecitabine group, 63.3% (76/120) of patients received only capecitabine, and 36.7% (44/120) received intravenous chemotherapy followed by capecitabine. In the intravenous chemotherapy group, 39.8% (47/118) received the anthracycline plus cyclophosphamide (EC) regimen, 36.4% (43/118) the taxane plus carboplatin(TCb) regimen, and 11.0% (13/118) the anthracycline plus taxane(AT) regimen. The median follow-up time was 37 months. The 3-year DFS for the capecitabine group, intravenous chemotherapy group, and no chemotherapy group were 81.5%, 64.3%, and 70.5%, respectively, and the 3-year OS were 88.4%, 81.0%, and 74.5%, respectively. After adjustment by multivariate analysis, compared with the no chemotherapy group, the capecitabine group could significantly reduce the risk of disease recurrence and death (No chemotherapy vs. Capecitabine: DFS HR=1.92, P=0.032; OS HR=2.51, P=0.026); compared with the intravenous chemotherapy group, the capecitabine group could significantly reduce the risk of disease recurrence (Intravenous chemotherapy vs. Capecitabine: DFS HR=1.84, P=0.037). Stratified analysis for patients with NB≥5 showed the capecitabine group had better DFS than the no chemotherapy and adjuvant chemotherapy groups, with 3 – year DFS rates of 73.2%, 47.4%, and 46.5%, respectively(Capecitabine group vs. No chemotherapy group, Log-rank P=0.043; Capecitabine group vs. Intravenous chemotherapy group, Log-rank P=0.043), and the OS was also significantly better than that of the no chemotherapy group, with 3-year OS of 84.0% and 53.6%, respectively (Log-rank P=0.012). In patients with NB<5, there was no significant difference in DFS and OS among the three groups. In H-RD patients, the DFS and OS of the capecitabine group were better than those of the no chemotherapy group and the intravenous chemotherapy group, with 3-year DFS of 81.7%, 66.5%, and 61.1%, respectively, and 3-year OS of 90.3%, 70.2%, and 78.5%, respectively (All Log-rank P<0.05), while in near-pCR patients, there was no significant difference in DFS and OS among the three groups (All Log-rank P>0.05). Conclusion: The study presents the real world adjuvant therapy options for TNBC patients with residual disease after neoadjuvant chemotherapy. Adjuvant capecitabine significantly reduces recurrence and mortality risks compared to no adjuvant chemotherapy, especially in patients with NB≥5 or H-RD
Presentation numberPS4-10-30
Evaluating real world pathologic complete response (pCR) rates at an urban center following the KEYNOTE-522 regimen for early-stage TNBC
Justine Anderson, Icahn School of Medicine at Mount Sinai, New York, NY
J. Anderson1, J. Dejesus2, E. Baldwin2, G. Van Hyfte3, P. Pandu2, N. Krishnamurthy4, R. Farley1, M. Rattu1, R. Patel1, A. Tiersten1; 1Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 2Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 3Tisch Cancer Institute – Biostatistics and Clinical Informatics, Icahn School of Medicine at Mount Sinai, New York, NY, 4Department of Medical Education, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: Neoadjuvant chemoimmunotherapy has been the standard frontline treatment for early-stage TNBC since 2021, when the KEYNOTE-522 trial demonstrated a significantly higher pCR rate of 64.8% with chemotherapy and pembrolizumab compared to chemotherapy alone. Our study aims to determine the efficacy of the KEYNOTE-522 regimen, as measured by pCR, in a real-world, diverse, urban population at a large tertiary referral center and assess factors impacting the likelihood of pCR. Methods: We performed a retrospective chart review of patients (pts) with anatomic Stage II-III TNBC who received the KEYNOTE-522 regimen within our institution’s health system from August 2021 to February 2025. Pts who completed neoadjuvant chemotherapy and pembrolizumab and underwent surgery were included. Demographics, clinicopathologic characteristics, treatment sequence and schedule, surgery type, and pCR rates were extracted from the medical record. Descriptive statistics were used to describe patient data. Non-parametric tests were used to evaluate differences between clinical factors across continuous and categorical variables: Wilcoxon rank sum and Kruskal-Wallis rank sum for continuous variables, and chi square tests (or Fisher’s exact tests) for categorical variables. Due to the exploratory nature of our analysis, p-values are reported unadjusted. Results: We identified 104 pts who were treated with neoadjuvant KEYNOTE-522 and underwent surgery. Overall, 83% of patients were stage II and 43% had lymph node involvement. 34% of pts (N=35) identified as White, 25% (N=26) Black, 16% (N=17) Hispanic, 15% (N=16) Asian, and 9.6% (N=10) as Other. The median age of the cohort was 55. The pCR rate of our cohort was 60% (N=62). When stratified by race/ethnicity, Asian and Hispanic patients had higher pCR rates than White and Black patients, p=0.01 (Table 1). There was no statistically significant difference in pCR based on age. 32% (N=33) of pts 50+ years achieved pCR (p=0.063). 64% of pts (N=67) received chemotherapy with paclitaxel and carboplatin (TC) first, while 36% received doxorubicin and cyclophosphamide (AC) first. There was no significant difference in pCR rate (p=0.694) based on which regimen was given first; however, there was a statistically significant difference in pts who received dose-dense AC every 2 weeks (69%, N=44) vs. every 3 weeks (45%, N=18, (p=0.016). Although 47% (N=49) of patients did not complete 8 neoadjuvant cycles of pembrolizumab, this did not impact likelihood of pCR (p=0.315). Conclusion: In a diverse real-world population, pCR rates were similar to those in the KEYNOTE-522 trial. The higher pCR rates in Asian and Hispanic patients warrants further study. Dose dense AC administered every 2 weeks was associated with a higher pCR rate. Limitations of this analysis include its retrospective design and small sample size.
| Overall Population 100% (N=104) |
White 34% (N=35) |
Black 25%(N=26) |
Asian 15%(N=16) |
Hispanic 16%(N=17) |
Other 10%(N=10) |
p value | |
| pCR | 60% (62) | 49% (17) | 46% (12) | 75% (12) | 65% (11) | 100% (10) | 0.01 |
Presentation numberPS4-08-01
Overall Survival and Breast Cancer-Specific Survival in HER2-Low versus HER2-Zero Breast Cancer: A Single-Center Cohort Study
Min Jung Lee, National Cancer Center, Goyang, Korea, Republic of
M. Lee1, D. Choi2, H. Chae3, J. Kim1, J. Hong4, E. Lee1, J. Han1, S. Sim4, S. Lee1, K. Lee4, H. Kang1, H. Kim2, S. Jung1; 1Surgery, National Cancer Center, Goyang, KOREA, REPUBLIC OF, 2National Cancer Control Institute, National Cancer Center, Goyang, KOREA, REPUBLIC OF, 3Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, KOREA, REPUBLIC OF, 4Internal Medicine, National Cancer Center, Goyang, KOREA, REPUBLIC OF.
Background Human epidermal growth factor receptor 2 (HER2)-low breast cancer, defined as immunohistochemistry (IHC) 1+ or 2+ with negative in situ hybridization (ISH), represents approximately 40-50% of all breast cancers. Despite growing therapeutic interest, the prognostic significance compared to HER2-zero (IHC 0) tumors remains controversial. This study aimed to explore the clinicopathologic data and compare survival outcomes between HER2-low and HER2-zero breast cancers. Methods This retrospective cohort study analyzed data from breast cancer patients treated at the National Cancer Center in South Korea. From 13,785 patients diagnosed with breast cancer between 2005 and 2023, a total of 7,281 patients were included after excluding those with no surgery, distant metastasis at presentation, ductal carcinoma in situ (DCIS), follow-up duration <1 year, missing HER2 data, or HER2 IHC 3+ expression. HER2 status was categorized as HER2-low (IHC 1+ or 2+ with ISH-negative) or HER2-zero (IHC 0). Outcomes included overall survival (OS) and breast cancer-specific mortality (BCSM), analyzed using Kaplan-Meier curves and Cox proportional hazards models, stratified by hormone receptor (HR) status. Interaction effects with Ki-67 and HR status were evaluated using restricted cubic spline modeling. Results HER2-low tumors demonstrated more favorable tumor biology than HER2-zero tumors, with higher rates of low histologic grade, low nuclear grade, and lower Ki-67 index (all p<0.001). Overall mortality was significantly lower in the HER2-low group (6.8% vs. 8.3%, p=0.031), although breast cancer-specific mortality was comparable (5.3% vs. 6.0%, p=0.091). Kaplan-Meier analysis showed no significant difference in OS between HER2 groups in the overall population (p=0.88). In HR-positive patients, HER2-low tumors retained favorable features and had lower all-cause (6.0% vs. 7.6%, p=0.034) and breast-cancer-specific mortality (4.2% vs. 4.8%, p=0.035), although OS was not significantly different (p=0.73). Conversely, in HR-negative patients, HER2-low tumors showed no biologic advantage, higher (non-significant) mortality (13.1% vs. 10.4%, p=0.170), and significantly worse OS (p=0.02). Restricted cubic spline analysis showed rising mortality risk with increasing Ki-67 in the overall cohort. No interaction between HER2 status and Ki-67 was observed in the overall or HR-positive groups. However, in HR-negative patients, HER2-low tumors exhibited a U-shaped association with Ki-67, with elevated mortality at both low and high levels (p<0.01). Conclusion HER2-low status is not an independent prognostic factor in the overall or HR-positive breast cancer population but is associated with worse survival in HR-negative patients. Although Ki-67 is widely used to reflect tumor proliferation and prognosis, its predictive value may be limited in HR-negative, HER2-low tumors, suggesting the need for tailored prognostic strategies and potentially more aggressive treatment approaches in this challenging subgroup.
Presentation numberPS4-08-02
Prognosis and Management of DCIS with Microinvasion: Insights from SEER Database and a Chinese Cohort
Yaping Yang, sun yat-sen memorial hospital, Guangzhou, China
Y. Yang, X. Chen, q. Peng; Oncology, sun yat-sen memorial hospital, Guangzhou, CHINA.
ObjectiveDuctal carcinoma in situ with microinvasion (DCISM) is a subtype of DCIS characterized by microscopic invasive foci (≤1 mm). Representing about 1% of breast cancer cases and 5%-10% of DCIS diagnoses, DCISM has both in situ and invasive features, complicating its clinical management. This study compares the prognostic outcomes of DCISM with DCIS and evaluates therapeutic approaches to inform evidence-based decision-making and optimize patient care. MethodsThis retrospective cohort study analyzed data from two sources: the Surveillance, Epidemiology, and End Results (SEER) database (1998-2018), comprising 7,467 patients (4,458 with DCIS and 3,009 with DCISM), and a Chinese cohort of 1,132 patients (903 with DCIS and 229 with DCISM) from Sun Yat-sen Memorial Hospital and the Chinese Society of Clinical Oncology. To reduce confounding bias, risk regression analysis and propensity score matching (PSM) were applied, enabling a robust comparison of survival outcomes between DCIS and DCISM. Key variables included patient age, tumor size, molecular subtypes (estrogen receptor/progesterone receptor [ER/PR] status), and treatment modalities (surgical type, radiotherapy, chemotherapy, and endocrine therapy). Prognostic endpoints assessed were overall survival (OS), breast cancer-specific survival (BCSS), and disease-free survival (DFS). ResultsPatients with DCISM displayed more aggressive pathological characteristics than those with DCIS, including a higher prevalence of high-grade tumors (P < 0.001) and elevated HER2 expression (P < 0.001), alongside a greater likelihood of undergoing total mastectomy (P < 0.001). Despite these differences, no statistically significant disparities were observed in OS, BCSS, or DFS between the two groups, suggesting comparable long-term prognoses. Breast-conserving surgery (BCS) combined with radiotherapy emerged as the optimal treatment strategy for both DCISM and DCIS, significantly enhancing DFS compared to BCS alone or radical mastectomy (DCIS: BCS alone vs. BCS + radiotherapy, hazard ratio [HR] = 2.37, 95% confidence interval [CI]: 1.36-4.16, P = 0.003; DCISM: HR = 15.03, 95% CI: 1.68-134.60, P = 0.015). In DCISM, this approach also improved OS (HR = 3.85, 95% CI: 1.87-7.90, P 0.05). ConclusionDespite the more aggressive pathological profile of DCISM, its survival outcomes align closely with those of DCIS, highlighting the risk of overtreatment in clinical practice. BCS combined with radiotherapy is an effective therapeutic option for both conditions, significantly improving survival metrics. Chemotherapy appears contraindicated for DCISM, particularly in hormone receptor-positive cases, due to its adverse prognostic impact. Endocrine therapy is recommended for hormone receptor-positive DCIS patients, though its efficacy in DCISM remains unestablished. These findings underscore the need for tailored treatment strategies based on molecular profiles and recurrence risk, offering critical guidance for managing DCISM. Future studies should investigate long-term outcomes and molecular underpinnings to refine individualized therapeutic approaches.
Presentation numberPS4-08-03
An Exploratory Clinical Study for Neoadjuvant Treatment of HER2-low Expressing, Stage II-III Breast Cancer with Vedolizumab in Combination with Pembrolizumab
Ting Luo, West China Hospital, Sichuan University, China, Chengdu, China
T. Luo1, X. Liu2, Y. Song1, C. Zhuang1, P. He1, X. Zeng3, D. Zheng1, X. Yan1, X. Zhong1, T. Tian1, B. Wei1, 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, 2Cancer Center, Affiliated Hospital of Xuzhou Medical University, xuzhou, CHINA, 3Department of pathology, West China Hospital, Sichuan University, China, Chengdu, CHINA.
Background: HER2-low expressing breast cancer is a heterogenous subtype with limited targeted therapeutic options. Disitamab Vedotin, a novel HER2-targeted antibody-drug conjugate (ADC), and Penpulimab, an anti-PD-1 monoclonal antibody, have both shown promise in different contexts of cancer treatment. This study aims to evaluate the clinical efficacy and safety of Disitamab Vedotin in combination with Penpulimab as neoadjuvant therapy in patients with HER2-low expressing, stage II-III breast cancer.Methods: This single-center, prospective, non-randomized clinical trial included patients with newly diagnosed HER2-low expressing (IHC 1+ or 2+ without amplification by FISH), stage II-III breast cancer in West China Hospital. All patients will receive neoadjuvant treatment with Disitamab Vedotin (2.0 mg/kg IV) and Penpulimab (200 mg IV) every 3 weeks for a total of 6 cycles, followed by surgery. The primary endpoint was pathological complete response (pCR), and secondary endpoints included overall response rate (ORR) and treatment-related adverse events (AEs). As an exploratory biomarker analysis, tumor-infiltrating lymphocytes (TILs) were quantitatively assessed in pretreatment tumor biopsies.Results: A total of 20 patients were enrolled in the study from August 2023 to August 2024, with two patients withdrawn due to intolerance to adverse reactions and two others discontinued due to disease progression. At the end of the treatment, total pCR (ypT0/is ypN0) rate was 25.0% (4/16). ORR reached 56.3% (9/16) by the clinical response assessment at the end of neoadjuvant treatment. The PD-L1 positive subgroup demonstrated a higher tpCR rate compared to the PD-L1 negative subgroup (33.3% vs. 14.3%). Additionally, the IHC 1+ subgroup exhibited a lower tpCR rate than the IHC 2+ subgroup without amplification by FISH (12.5% vs. 37.5%). The most common AEs were alopecia (18/20, 90.0%) and pruritus (14/20, 70.0%), and no significant toxicities or treatment-related deaths observed. Pretreatment tumor immune analysis revealed significantly higher CD4+ TIL infiltration in patients achieving pCR (p= 0.0181). While CD8+ TILs showed a non-significant trend toward increased infiltration in the pCR group (p = 0.0676), overall immune infiltration was associated with pCR (p = 0.0451).Conclusion: Although the clinical benefit may be limited, the combination of Disitamab Vedotin and Penpulimab as neoadjuvant treatment for HER2-low expressing, stage II-III breast cancer shows manageable safety and potential for further refinement, warranting additional investigation to optimize treatment strategies. Pretreatment CD4+ TIL density and total immune infiltration may serve as predictive markers for neoadjuvant response.
|
Patient Characteristic |
N=20 |
|
Median age, years |
43.5 (33, 68) |
|
Female, n (%) |
20 (100) |
|
HER2, n (%) |
|
|
IHC 1+ |
10 (50.0) |
|
IHC 2+/FISH- |
10 (50.0) |
|
HR, n (%) |
|
|
ER+ |
18 (90.0) |
|
PR+ |
17 (85.0) |
|
T stage, n (%) |
|
|
T1 |
2 (10.0) |
|
T2 |
13 (74.5) |
|
T3 |
1(5.0) |
|
T4 |
4 (20.0) |
|
N stage, n (%) |
|
|
N0 |
3 (15.0) |
|
N1 |
14 (70.0) |
|
N2 |
3 (15.0) |
|
N3 |
0 |
|
Stage (cTNM classificationd), n (%) |
|
|
Ⅱ |
13 (65.0) |
|
Ⅲ |
7 (35.0) |
|
PDL1 , n (%) |
|
|
CPS>1 |
11 (55.0) |
|
CPS≤1 |
9 (45.0) |
Presentation numberPS4-08-04
Impact of adjuvant chemotherapy in the survival outcomes of early-stage hormone receptor (HR)-positive HER2-negative lobular versus ductal carcinoma patients
Arya Mariam Roy, The Ohio State University, Columbus, OH
A. Roy1, Y. Gokun2, B. Slover2, N. Lopetegui-Lia1, D. Quiroga1, G. Bader1, M. Cherian1, A. Davenport1, K. Johnson1, S. Sardesai1, R. Wesolowski1, S. Myers3, E. Burke3, M. Gatti-Mays1, D. Stover1, N. Williams1; 1Department of Medicine, The Ohio State University, Columbus, OH, 2Center for Biostatistics, The Ohio State University, Columbus, OH, 3Department of Surgery, The Ohio State University, Columbus, OH.
Background: Invasive lobular carcinoma (ILC) represents 10-15% of all breast cancers (BC) and differs biologically and clinically from invasive ductal carcinoma (IDC). Conflicting data exist with regard to variation in prognosis and benefit of chemotherapy (CT) between these histologic subtypes. Here, we compare the impact of adjuvant CT on survival outcomes in patients (pts) with early-stage (ES) HR+/HER2- ILC and IDC. Methods: We queried the National Cancer Database for pts who received upfront surgical resection for ES HR+ HER2- invasive BC between 2010 and 2021, classifying cohorts into ILC or IDC subtypes. Cox proportional hazards models assessed the relationship between overall survival (OS) and histology type adjusting for potential confounders including age, race, stage, grade, and treatment variables. Subgroup analyses were performed based on CT receipt and Oncotype DX recurrence score (RS), a genomic assay used to predict response to CT, categorized as (low (L) 0-15, intermediate (I) 16-25, high (H) >=26). Results: Of the 1,101,920 pts included, 14.7% (n= 162,430) had ILC. Compared to pts with IDC, the ILC cohort was comprised of a greater proportion of Whites (87.7 vs 85.4%), intermediate-grade (63.7 vs 48.8%), clinical T2 (26.5 vs 22.1%) and N0 disease (89.1 vs 88.3%) (all p<0.001). RS scores varied by histologic subtype with a greater proportion of ILC having L (17.9 vs 17.2%) and I RS (15.2 vs 11.9%) compared to IDC, in which H RS was more common (2.8 % ILC vs 5.8% IDC). Use of adjuvant CT (79.6 vs 77.3%) and hormonal therapy (89.3 vs 86.4%) was higher in ILC, while radiation use was lower (61.3 vs 64.5%) (all p<0.001). The 5-year (Y) OS of ILC was similar to IDC (89.5 (89.3-89.7) vs 90.4 (90.3-90.5) %), however 10Y OS was worse for ILC cohort (73.6 (73.3-74.0) vs 76.7 (76.6-76.9) %, p <0.001). Among pts who received adjuvant CT, at 5 Y, OS was nearly identical between ILC and IDC across all RS groups. However, at 10 Y, OS was consistently lower in ILC compared to IDC, with a more pronounced difference in the I and H RS groups (Table 1). While OS was lower in ILC pts who received CT compared to IDC pts that received CT in univariate analyses of the overall cohorts (HR: 1.16, p<0.001) and when stratified by RS (L – 1.07, p= 0.01; I – 1.18, p<0.001; H – 1.12, p = 0.02), these associations did not retain significance when adjusting for clinically relevant confounders (HRs: Overall – 1.01, p= 0.16; L – 0.98, p= 0.52; I – 0.95, p= 0.11; H – 0.97, p= 0.51). An interaction was observed between histology type and stage (p = 0.0026). Conclusion: Our study suggests that while 5-year survival may be comparable, ES ILC pts receiving adjuvant CT may experience worse long-term outcomes compared to IDC, particularly among those with higher genomic risk. Further research is needed to personalize treatment strategies for ILC by identifying underlying molecular differences that drive this disparity
| Group |
Time point (Year) |
ILC OS (%) (95% CI) | IDC OS (%) (95% CI) | P-value |
| Overall Cohort | 5 | 91.2 (91.1-91.4) | 92.2 (92.1-92.3) | <0.001 |
| 10 | 76.5 (76.2-76.9) | 79.6 (79.5-79.8) | ||
| Low RS | 5 | 96.5 (96.2-96.7) | 96.5 (96.4-96.7) | 0.01 |
| 10 | 88.1 (87.3-88.8) | 89.0 (88.7-89.2) | ||
| Intermediate RS | 5 | 95.5 (95.2-95.8) | 96.2 (96.1-96.3) | <0.001 |
| 10 | 85.5 (84.7-86.3) | 87.9 (87.5-88.2) | ||
| High RS | 5 | 92.1 (91.1-93.0) | 91.9 (91.7-92.2) | 0.02 |
| 10 | 78.0 (75.6-80.1) | 80.7 (80.1-81.2) |
Presentation numberPS4-08-05
Evaluating Idiopathic Granulomatous Mastitis Using the Inflammatory Breast Cancer Scoring System: A Comparative Study Highlighting the Role of Pain as a Diagnostic Criterion
Jacqueline Tsai, Stanford University, Stanford, CA
J. Tsai1, K. Stone1, I. Wapnir1, J. Hong2; 1Surgery, Stanford University, Stanford, CA, 2Medicine, Stanford University, Stanford, CA.
Evaluating Idiopathic Granulomatous Mastitis Using the Inflammatory Breast Cancer Scoring System: A Comparative Study Highlighting the Role of Pain as a Diagnostic CriterionBackground:Idiopathic granulomatous mastitis (IGM) is an inflammatory condition which has similar characteristics to inflammatory breast cancer (IBC). We sought to utilize the published IBC scoring system to compare scores between patients diagnosed with IBC and GM. Methods A retrospective chart review of patients with biopsy proven IGM and IBC from January 2014 to September 2024 was performed. Patient demographics, diagnostic imaging, clinical assessments, and treatments used (oral or intralesional steroids and immunomodulatory drugs) were reported from the EHR. The IBC scoring system1 was used, which included 6 of the 7 reported criteria: timing of symptoms, skin changes, breast swelling, erythema, nipple abnormalities, and breast imaging findings. Lymphatic tumor cell emboli biopsy results were excluded from GM patients, and the maximum modified score was 42. We separately compared scores with and without biopsy results. We then applied the proposed classifications of the DEFINITELY IBC (total score ≥ 42); STRONG POSSIBILITY (25-41); WEAK POSSIBILITY (14-24); and NOT IBC < 14). We then examined a separate pain score from 1-10 as a separate predictor. T-test was used for statistical analysis. Results 8 patients with IBC were identified and compared with 77 patients with GM. Patients with diagnosed IBC had a higher average score of 21.5 compared to 17.8 for those with diagnosed GM. However, at each score category there was no overall statical difference in average scores (Table1). When a separate pain scale score was evaluated, patients with GM presented with a significantly higher average pain reported than those with IBC (7.8 vs 2.3, p= .00008) ConclusionPatients with IGM present with similar characteristics to IBC. When utilizing the IBC scoring system for diagnosis, GM patients had similar scores as those with IBC. Most had scores in either weak or strong possibility of IBC categories. However, the most significant difference noted between these 2 groups is the maximum pain reported by the patients. Given this similarity, we propose a new scoring criterion with the inclusion of pain for prediction of GM.
Presentation numberPS4-08-06
Optimizing Postoperative Adjuvant Therapy for Early-Stage Breast Cancer by Combining Dynamic ctDNA MRD Monitoring with Clinical Risk Stratification: An Open-label, Multicenter, Prospective Study (AMENDER)
Fei Ma, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
F. Ma1, H. Ge1, H. Mo1, X. Sun2, K. Zhao3, L. Zhang3, Y. Zhang3, C. Li3, G. Wang3; 1Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, CHINA, 2Department of Medical Oncology, Huanxing Cancer Hospital, Beijing, CHINA, 3Medical&Marketing, Burning Rock Biotech, Guangzhou, CHINA.
Background: Adjuvant therapy for early-stage breast cancer is currently stratified based on clinical risk factors, enhancing prognosis but leaving some patients at risk of recurrence. Circulating tumor DNA (ctDNA)-based minimal residual disease (MRD) detection aids in identifying recurrence risks and predicting prognosis. However, its utility in guiding postoperative systemic treatment remains unclear. This study seeks to employ longitudinal MRD monitoring to optimize adjuvant therapy strategies for early-stage breast cancer. Methods: Early-stage breast cancer patients naive to systemic anti-tumor therapy were enrolled. A tumor-informed MRD testing approach (Burning Rock Biotech, Guangzhou, China) was applied, in which pre-systemic therapy tumor tissues underwent whole exome sequencing (WES) to create personalized ctDNA detection panels (≤50 variants). Blood samples were collected 2-6 weeks postoperatively and before adjuvant systemic therapy for MRD testing. Patients with post-operative clinical high risk (pT>5cm, or pN+, or non-pCR after neoadjuvant therapy) or positive MRD received intensive adjuvant therapy, while those with low risk and negative MRD received standard adjuvant therapy. After treatment initiation, MRD testing and routine imaging evaluations were conducted every 3 months for up to 2 years or until tumor recurrence. If MRD remains positive or turns positive again during follow-up, an alternative intensive regimen (if available) will be adopted. No more than two intensive treatments are allowed. The primary endpoint is the 3-disease-free survival (DFS) rate. Results: This study enrolled 126 patients (94 TNBC, 21 ER+HER2-, 11 HER2+), including 33 stage I, 67 stage II, and 26 stage III cases. Based on clinical risk stratification, 71 patients were classified as clinical low-risk and 55 as clinical high-risk. The median follow-up was 726 days. The post-operative MRD was detected in 12.8% cases (16/125; 1 test failure), comprising 3 low-risk and 13 high-risk patients. Among post-operative MRD-positive patients, 87.5% (14/16) received dynamically adjusted intensive treatment. Of these, 13 converted to MRD-negative after longitudinal monitoring and personalized treatment, while 1 patient remained persistently MRD-positive. During follow-up, 8.3% (9/109) patients converted from post-operative MRD-negative to positive (3 low-risk and 6 high-risk). Of these patients, 1 with clinical high risk did not receive post-operative intensive treatment; 1 turned negative after conventional treatment without intensive treatment; 3 experienced a recurrence before the initiation of dynamically adjusted intensive treatment; 3 reverted to MRD-negative after dynamically adjusted intensive treatment; and 1 was newly detected as MRD-positive. In addition, 25% (9/36) patients with clinical high-risk and persistently MRD-negative status did not receive intensive treatment. Conversely, 12.3% (8/65) patients with clinical low-risk and persistently MRD- negative status received intensive treatment. Overall, 105 cases were included in the per-protocol analysis set, with a recurrence rate of 2.9% (3/105). The 2-year DFS rate was 96.5% (92.7%-100%). Conclusion: Preliminary findings support the feasibility of tailoring postoperative adjuvant treatment based on clinical risk and dynamic MRD monitoring. The follow-up is still ongoing. Clinical trial registration: NCT05345860.
Presentation numberPS4-08-07
Predicting response and survival after neoadjuvant systemic treatment with on-treatment biopsies
Carsten Denkert, Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, Germany
C. Denkert1, A. Schneeweiss2, V. Schaser3, M. Untch4, M. Frank5, M. van Mackelenbergh6, J. Blohmer7, F. Holms8, V. Müller9, D. Zahm10, T. Karn11, P. Fasching12, E. Simon13, F. Marmé14, M. Darsow15, E. Stickeler16, S. Jud17, T. Fehm18, B. Heinrich19, C. Schem20, B. Felder3, J. Holtschmidt3, S. Loibl21, A. Noske22; 1Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, GERMANY, 2Division of Gynaecological Oncology, National Center for Tumor Diseases (NCT), University Hospital and German Cancer Research Center, Heidelberg, GERMANY, 3Medicine and Research Departement, GBG c/o GBG Forschungs GmbH, Neu-Isenburg, GERMANY, 4Department of Obstetrics and Gynecology, Interdisciplinary Breast Cancer Center, Medical School Berlin, Helios Kliniken Berlin-Buch, Berlin, GERMANY, 5Department of Obstetrics and Gynecology, Ortenau Klinikum Offenburg-Kehl, Offenburg, GERMANY, 6Department of Gynecology and Obstetrics, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, GERMANY, 7Department of Gynecology with Breast Center, Charité – Universitätsmedizin Berlin, Berlin, GERMANY, 8Department of Obstetrics and Gynecology, St. Barbara-Klinik Hamm-Heessen GmbH, Hamm, GERMANY, 9Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, GERMANY, 10Breast center, SRH Wald-Klinikum Gera GmbH, Gera, GERMANY, 11Department of Gynecology and Obstetrics, University of Frankfurt, UCT Frankfurt-Marburg, Frankfurt am Main, GERMANY, 12Department of Obstetrics and Gynecology, University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Erlangen, GERMANY, 13Department of Obstetrics and Gynecology, Kreiskrankenhaus Torgau, Torgau, GERMANY, 14University Medical Center Mannheim, University of Heidelberg, Mannheim, GERMANY, 15Competence Centre for Breast Surgery and Breast Health, Luisenkrankenhaus Düsseldorf, Düsseldorf, GERMANY, 16Breast Center, Department of Gynecology and Obstetrics, RWTH University Hospital Aachen, Achen, GERMANY, 17Department of Obstetrics and Gynecology, Klinikum Mutterhaus der Borromäerinnen, Trier, GERMANY, 18Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Centrum für Integrierte Onkologie (CIO) ABCD, Düsseldorf, GERMANY, 19Department of Internal Medicine, Hematology and Oncology, Haematologie-Onkologie im Zentrum MVZ GmbH, Augsburg, GERMANY, 20Mammazentrum Hamburg, am Krankenhaus Jerusalem, Hamburg, GERMANY, 21Medicine and Research Departement, GBG c/o GBG Forschungs GmbH, Neu Isenburg, Goethe University Frankfurt, Frankfurt am Main, GERMANY, 22Pathology Medica, Zurich, Switzerland, Institute of Pathology, School of Medicine and Health, TUM, Munich, GERMANY.
Background: Early prediction of response to neoadjuvant systemic treatment (NST) in breast cancer (BC) may guide treatment tailoring even before NST completion. Patients (pts) could avoid ineffective treatments and be assigned to non-cross-resistant therapies. Here, we analysed core needle on-treatment biopsies (OTB) taken after 2-4 cycles of NST for residual invasive cancer, Ki67, stromal tumor-infiltrating lymphocytes (sTILs) and their ability to predict treatment outcome. Study Design: Pts with available and evaluable OTBs (n=449) of neoadjuvant clinical trials GeparSixto (n=47), GeparSepto (n=194), GeparNuevo (n=99), GeparX (n=47), and GeparOLA (n=62) were included in the analysis. OTBs were retrospectively evaluated for the presence of tumorbeds, residual cancer defined as cellularity of viable invasive cancer cells >0 (OTB+) vs 0 (OTB-), Ki67, and sTILs, which was thereafter correlated with post-treatment response (pCR defined as ypT0/is ypN0) and time-to-event outcomes. Results: Most tumors were cT2 (55%), of no special type (88%), G3 (66.1%), and 69.6% of pts were clinically nodal negative. Most prevalent subtype at baseline was TNBC (n=257, 57.2%), followed by HR+/HER2- (n=108, 24.1%), and HRany/HER2+ BC (n=84, 18.7%). In 79.1% (355/449) a tumorbed in OTBs was identified. The remaining OTBs had unspecific (10%) or uncertain changes (6.2%) or no signs of tumorbed (4.7%). Pts with OTB+ were identified in 60% (213/355) and had a pCR rate of only 20.2% (43/213) after completion of NST, whereas pts with OTB- (40%, 142/355) had a pCR rate of 73.9% (105/142). In univariate logistic regression analysis, pts with OTB+ had a lower pCR rate (OR 0.091 CI 0.055-0.152, p<0.001). In multivariate analysis adjusting for other established prognostic factors like nodal stage, intrinsic subtype and grading, the presence of OTB+ still showed a statistically significant decrease in the odds for pCR (OR 0.111 CI 0.063-0.196, p<0.001). When analysed according to subtype, the pCR rate at surgery for pts with OTB+ was 48% for HER2+ (12/25), 21.6% for TNBC (24/111), and 9.1% for HR+/HER2- (7/77). Significantly lower chances for pCR in pts with OTB+ were consistent in all intrinsic subtypes (HRany/HER2+ OR 0.24 CI 0.07-0.80 p=0.02; HR+/HER2- OR 0.13 CI 0.03-0.52 p=0.004, TNBC OR 0.09 CI 0.05-0.18 p<0.001, interaction test p=0.468). OTB+ was linked to worse DFS and OS (DFS: HR 2.59 CI 1.72-3.91; OS: HR 3.99 CI 2.09-7.59, log-rank p<0.001 respectively), remaining significant in multivariate analysis (DFS: HR 2.37 CI 1.52-3.68; OS: HR 3.76 CI 1.91-7.40, p<0.001 respectively). In subgroup analysis, the overall results for DFS were confirmed for all intrinsic subtypes (HRany/HER2+ HR 2.77 CI 0.919-8.7 log-rank p=0.067; HR+/HER2- HR 3.82 CI 0.922-15.9, log-rank p=0.065; TNBC HR 2.19 CI 1.33-3.60 log-rank p=0.003, interaction test p=0.774). In multivariate analysis an impact on OS could only be demonstrated for TNBC (HR 4.17 CI 1.92-9.07, log-rank p<0.001), probably limited in other subtypes by paucity of OS events. Baseline Ki67 was lower in OTB+ (median 39%) vs OTB- (median 48%, p<0.001). Overall, Ki67 values decreased from baseline to OTB by a median absolute value of 30%. The highest decrease appeared in pts with high-grade TNBC (median absolute value of 37,5%, stratified signed rank test p=0.03). Pts with OTB+ had lower sTILs at baseline (median 10%) as compared to OTB- (median 20%, p<0.001). Pts with pCR had a median absolute increase of 5% in sTILs from baseline to OTB, whereas pts with no pCR showed no changes in sTILs (p-value 0.122). Conclusion: Our findings suggest that OTB are suitable to monitor neoadjuvant therapy response. Residual cancer cells have a prognostic impact, even if detected early during treatment, therefore OTB should be evaluated prospectively as biomarker to guide response adapted treatment in neoadjuvant clinical trials.
Presentation numberPS4-08-08
Comparison of localization techniques for non-palpable breast cancer: real world data
Kristin Lupinacci, University of Pittsburgh Medical Center, Pittsburgh, PA
B. Yigit1, M. Tokocin1, K. Lupinacci2, B. Demirors2, N. Zafer Utkan3, M. Masta3, E. Ozkurt4, U. Kesici5, A. Akan5, E. Sen6, A. Kamali POlat7, H. Karanlik8, B. Comcali9, A. Soyder10, L. Dogan11, A. Salamat12, M. Onur Kulturoglu13, K. Senol14, J. Alazhri15, A. Dag16, N. Cabioglu17, G. Maralcan18, H. Valiyeva19, M. Levhi Akin20, B. Balci Topuz21, B. Citgez22, D. Can Trabulus23, G. Ozan Kucuk24, S. Salimoglu25, B. Ozcinar17, O. Aytac26, B. Goktepe27, C. Mathelin28, V. Ozmen17, A. Soran2; 1Surgery, Bagcilar Training and Research Hospital, Istanbul, TURKEY, 2Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, 3Surgery, Koceli University, Kocaeli, TURKEY, 4Surgery, Istanbul Florence Nightingale Hospital, Istanbul, TURKEY, 5Surgery, Health Science University, Prof Dr. Cemil Tascioglu Training and Research Hospital, Istanbul, TURKEY, 6Surgery, Basaksehir Cam and Sakura City Hospital, University of Health Sciences, Istanbul, TURKEY, 7Surgery, Ondokuz Mayis University, Samsun, TURKEY, 8Surgery, Istanbul University, Institute of Oncology, Istanbul, TURKEY, 9Surgery, Ankara Bilkent City Hospital, Ankara, TURKEY, 10Surgery, Acibadem Mehmet Ali Aydinlar University, Altunizade Acibadem Hospital, Istanbul, TURKEY, 11Surgery, Ankara Etlik City Hospital, Ankara, TURKEY, 12Surgery, Optum Breast Care, Las Vegas, NV, 13Surgery, Ankara Etlik City Hospital, Surgical Oncology Clinic, Ankara, TURKEY, 14Surgery, Uludag University, Faculty of Medicine, Bursa, TURKEY, 15Breast and Endocrine Surgery, King Fahad Specialist Hospital, Dammam, SAUDI ARABIA, 16Surgery, Mersin University, Faculty of Medicine, Mersin, TURKEY, 17Surgery, Istanbul University, Faculty of Medicine, Istanbul, TURKEY, 18Surgery, Sanko University, Gaziantep, TURKEY, 19Surgery, Azerbaijan Medical University, Baku, AZERBAIJAN, 20Surgery, Liv Ulus Hospital, Istanbul, TURKEY, 21Radiation Oncology, Gaziantep City Hospital, Gaziantep, TURKEY, 22Surgery, Halic University, Memorial Atasehir Hospital, Istanbul, TURKEY, 23Surgery, Istanbul Bahcesehir University, Istanbul, TURKEY, 24Surgery, Samsun Training and Research Hospital, University of Health Sciences, Istanbul, TURKEY, 25Surgery, Izmir Tepecik Training and Research Hospital, Istanbul, TURKEY, 26Surgery, Baskent University, Adana, TURKEY, 27Surgery, Ege University, Faculty of Medicine, Izmir, TURKEY, 28Surgery, CANS and CHRU, Hopitaux Universitaires de Strasbourg, Strasbourg, FRANCE.
Background: Accurate tumor localization is critical in performing successful breast-conserving surgery (BCS). Multiple localization methods are available to identify and target non-palpable lesions. This multi-national study aims to compare various localization techniques, by evaluating the impact on margin status, re-operation and recurrence. Methods: We conducted a multi-center retrospective study of 1481 patients with Tis-1N0 breast cancer undergoing BCS from 2015-2024 across 28 international centers. Patients were grouped by localization technique: radioactive seed, RSL (n=711, 48%), wire-guided, WGL (n=561, 37.9%), ultrasound-guided, UGL (n=104, 7%), radio-guided occult lesion, ROLL (n=68, 4.6%), radar reflectors (n=22, 1.5%), magnetic marker (n=7, 0.5%), and radiofrequency identification RFID (n=8, 0.5%). Patients were then categorized by margin status as positive (n=54, 3.6%), <2 mm (n=102, 6.9%), or ≥2 mm (n=1325, 89.5%). Cross-tabulation and Pearson Chi-Square test assessed the association between techniques and margin status. Multivariate logistic regression analyzed re-operation and recurrence. Results: In 1266 cases (85.5%), surgeon preference was the primary factor guiding choice of localization technique, followed by availability of localization methods in 173 cases (11.7%). Tumor size was classified according to standard staging, pTis, pT1a, pT1b, and pT1c, with distributions of 305 (20.6%), 154 (10.4%), 383 (25.9%), and 639 (43.1%) cases respectively. The dominant histologic subtype was invasive ductal carcinoma (n=850, 57.4%). Molecular subtypes were predominantly luminal A (n=705, 47.6%) and luminal B (n=492, 33.2%). Re-operation was required in 100 (6.8%) patients. Local, regional, and systemic recurrence occurred in 13 (0.9%), 2 (0.1%) and 6 (0.4%) patients respectively. There was a significant association between localization technique and margin status (p < 0.001). WGL yielded the highest rate of positive margins (n=48, 8.6%), leading to additional surgeries. In contrast RSL had the lowest positive margin rate (n=2, 0.3%) and the highest proportion of margins ≥2 mm (n=705, 99.2%). Radar reflectors, magnetic markers, RFID, and ROLL had no positive margins, although ROLL showed a relatively high proportion of margins <2 mm (n=14, 20.6%), suggesting limitations in precision. The observation of a lower positive margin rate with UGL (n = 4, 3.8%) compared to WGL, with a substantially elevated re-operation risk (OR = 4.326, p < 0.001) indicates the impact of a low overall event rate and confounding variables. Among patients undergoing WGL, 100 (17.8%) had in situ disease, with 21 requiring re-operation. In the UGL group, only 4 (3.8%) patients had in situ disease and none underwent re-operation. This proportion of in situ disease may explain the higher re-operation rate observed with WGL, despite lower rate of positive margins, given current margin recommendation for in situ disease of ≥ 2mm. With median follow-up of 28.2 months (range 12.1-108.2 months) local recurrence was observed in 4 patients (5.9%) with ROLL, 6 patients (0.8%) with RSL and in 3 patients (0.5%) with WGL, while no local recurrence was reported with the other localization techniques, although logistic regression models were limited by low event rates and perfect separation. Conclusion: Our findings highlight significant differences in margin status and re-operation rates among localization techniques utilized in BCS for non-palpable breast cancer across diverse clinical practices. RSL demonstrated superior accuracy with the lowest positive margin and re-operation rates. Wide variability among localization techniques underscores the need to consider institutional factors including availability, cost and geographic standards in the selection of localization method to optimize surgical and oncologic outcomes.
Presentation numberPS4-08-09
Long-term outcomes and prognostic factors in metaplastic breast cancer treated with curative intent: a 22-year multicenter retrospective cohort study
Sze Wa Shirley Tse, Queen Elizabeth Hospital, Hong Kong, Hong Kong
S. S. Tse1, K. Bao1, K. Cheung1, J. Chow1, I. Wong2, C. Wong3, O. Mang3, H. Yiu1, C. Kwan2; 1Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, HONG KONG, 2Department of Oncology, United Christian Hospital, Hong Kong, HONG KONG, 3Hong Kong Cancer Registry, Hospital Authority, Hong Kong, HONG KONG.
Background:Metaplastic breast cancer (MpBC) is a rare and aggressive subtype of breast cancer with limited guidance on an optimal management approach in existing clinical guidelines. We evaluated long-term outcomes and treatment efficacy in early MpBC patients across major cancer centers in Hong Kong. Methods:Women with non-metastatic MpBC diagnosed from January 2001 to December 2023 at four Hong Kong hospitals were identified via Hong Kong Cancer Registry. Clinical data, treatment response, recurrence, and survival were analyzed by histological and molecular subtype according to current standards, based on review of original pathology reports. Survival analyses were conducted using the Kaplan-Meier method and compared using log-rank test. Factors associated with survival were evaluated by univariate and multivariate Cox regression. Results:In total, 145 consecutive patients with non-metastatic MpBC were included. The median age at diagnosis was 57 years (IQR 51-68 years), with 64.8% being postmenopausal. Most patients presented with a solitary tumor (94.5%), commonly in upper outer quadrant (58.6%). Squamous cell carcinoma (20.7%) and spindle cell carcinoma (13.1%) were the most common histological subtypes. Most cases were stage II (55.2%), with a median tumor size of 3.5 cm (IQR 2.4-6.0 cm), node-negative at diagnosis (66.2%), and grade 3 tumors (66.9%). Molecular subtyping showed that 49.7% of tumors were triple-negative, 45.5% were hormone receptor-positive only (≥1% ER and/or PR-positive tumor cells), 2.8% were HER2-positive only, and 2.1% were both ER/PR-positive and HER2-positive. 4.1% of tumors were ER low, and 37.9% HER2 low. Most patients underwent mastectomy (77.2%) with axillary dissection (58.6%), and 93.8% achieved clear margins. Neoadjuvant chemotherapy was given to 13.8% of patients, with a pathological complete response (pCR) in 5.0% and a partial response in 50.0%. Adjuvant chemotherapy and radiotherapy were given to 57.2% and 60.7% of patients, respectively. At a median follow-up of 112 months (IQR 28-165), the 5-year overall survival was 67%. Five-year locoregional and distant relapse-free survival rates were 90.4% and 72.1%, respectively. Locoregional recurrence occurred in 11%, and distant metastases in 27.6% of patients, mainly involving visceral sites (90%). Upon univariate analysis followed by multivariate analysis adjusted for grade, T stage, and molecular subtype, adjuvant chemotherapy was significantly associated with improved overall survival (HR 0.42, 95% CI 0.22-0.82, p=0.01), while adjuvant radiotherapy was not (HR 0.69, 95% CI 0.38-1.24, p=0.21). In T3/4 or node-positive subgroup, adjuvant chemotherapy remained significantly associated with improved overall survival (HR 0.32, 95% CI 0.12-0.83, p=0.02), whereas adjuvant radiotherapy did not show significance for overall survival (HR 0.65, 95% CI 0.29-1.45, p=0.29) or for locoregional relapse-free survival (HR 0.98, 95% CI 0.40-2.40, p=0.96). No significant differences in pCR rates following neoadjuvant chemotherapy were observed across molecular subtypes (p=0.67). Adjuvant endocrine therapy was not associated with overall survival in hormone receptor-positive cases (p=0.88), nor was targeted therapy in HER2-positive cases (p=0.47). Conclusion:This study showed that MpBC is a distinct subtype of breast cancer, characterized by unique clinicopathological characteristics and responsiveness to treatments. Adjuvant chemotherapy was associated with improved overall survival in non-metastatic MpBC, while adjuvant radiotherapy did not demonstrate a significant benefit. The low response rate to neoadjuvant chemotherapy highlights the urgent need to develop novel therapeutic strategies to enhance outcomes for this challenging patient population.
Presentation numberPS4-08-10
Clinical characteristics and survival outcomes of Pleomorphic Lobular Carcinoma: a comparative analysis with classical ILC and IDC
David Walji, University of Toronto, Toronto, ON, Canada
D. Walji1, V. Giannakeas2, D. W. Lim3; 1Temerty Faculty of Medicine, University of Toronto, Toronto, ON, CANADA, 2Women’s College Research and Innovation Institute, Women’s College Hospital, Toronto, ON, CANADA, 3Department of Surgery, Women’s College Hospital, Toronto, ON, CANADA.
Introduction: Pleomorphic lobular carcinoma (PLC) is an understudied, rare and aggressive form of invasive lobular carcinoma (ILC), distinct from both invasive ductal carcinoma (IDC) and classical ILC. Though histologically related to ILC, PLC clinically resembles high-grade IDC. We evaluated overall survival (OS) and disease-specific survival (DSS) in PLC relative to IDC and ILC, using robust causal inference methods to adjust for clinicopathologic and treatment differences. Methods: The SEER (Surveillance, Epidemiology, and End Results) 17 database was used to identify all adult female patients with primary invasive breast cancer between 2000 and 2022. Inverse probability of treatment weighting (IPTW) based on a multinomial propensity score was used to adjust for baseline differences in demographics (age, race, income, rurality), tumor characteristics (grade, AJCC stage, receptor subtype) and treatment variables (surgery, radiation, chemotherapy). Covariate balance was assessed using standardized mean differences (SMDs) via Love plots. Residual imbalances (SMDs >0.15) were addressed using multivariable Cox models for doubly robust estimation. Proportional hazards (PH) assumption was assessed via Schoenfeld residuals. If satisfied, IPTW-weighted Cox models were used; if violated, time-varying Cox models with log(time) interactions estimated dynamic hazard ratios. Analyses were conducted at all-time, 5- and 10-year follow-ups. Kaplan-Meier curves were used to visualize survival by subtype. LASSO-regularized logistic regression identified 5- and 10-year OS and DSS predictors among PLC patients. Statistical analysis was conducted in R using p < 0.05. Results: Of the 721,127 patients identified, those with PLC (n = 181) when compared with IDC (n = 631,198) and ILC (n = 75,653) presented with larger (PLC: 3.49 ± 2.57 cm, IDC: 2.06 ± 1.53 cm, ILC: 2.73 ± 1.94 cm), higher-grade (grade 3+; PLC: 51.4%, IDC: 35.3%, ILC: 8.1%) and more advanced-stage tumors (stage III; PLC: 23.2%, IDC: 9.8%, ILC: 14.2%). PLC tumors were less likely to be HR+ (ER+ PLC: 70.7%, IDC: 77.6%, ILC: 94.3%; PR+ PLC: 58.6%, IDC: 67.4%, ILC: 80.0%), less likely to be HER2- (PLC: 43.6%, IDC: 50%, ILC: 60%) and more frequently triple negative (PLC: 8.8%, IDC: 6.9%, ILC: 0.8%). PLC patients received more mastectomies (PLC: 55.2%, IDC: 37.9%, ILC: 48.7%) and chemotherapy (PLC: 56.9%, IDC: 43.4%, ILC: 31.9%) yet had comparatively worse unadjusted OS (PLC: 64.1%, IDC 75.9%, ILC: 74.2%) and DSS (PLC: 86.7%,IDC: 91.4%, ILC: 91.1%). PH assumptions were violated in PLC vs ILC comparisons at all-time and 10-year follow-ups. Time-varying models demonstrated a significant early survival advantage in ILC over PLC in both OS (HR = 0.12, 95% CI: 0.05-0.28, p < 0.0001) and DSS (HR = 0.05, 95% CI: 0.02-0.14, p < 0.0001), though this benefit diminished over time (OS HR(tt) = 1.58, 95% CI: 1.29-1.93, p < 0.0001; DSS HR(tt) = 1.88, 95% CI: 1.46-2.43, p < 0.0001). For IDC, a survival advantage was present against PLC in OS at all-time (HR: 0.72, 95% CI: 0.56-0.93, p < 0.05), 5-year (HR = 0.68, 95% CI: 0.49-0.95, p < 0.05) and 10-year follow-up (HR = 0.74, 95% CI: 0.55-0.98, p < 0.05). In the PLC-specific LASSO analysis, poorer outcomes were associated with larger tumors, more advanced stage tumors and identifying as Black, while improved outcomes were associated with lower grade, HR+/HER2+ status and receiving chemotherapy. Conclusion: PLC is associated with significantly worse survival outcomes compared to both IDC and ILC. While ILC confers an early survival benefit that diminishes over time, IDC maintains a consistent OS advantage. Our findings support PLC as an aggressive subtype of ILC warranting dedicated research to improve earlier detection and treatment.
Presentation numberPS4-08-11
Standardized Definitions for Efficacy End Points (STEEP, STEEP2) in the CCTG MA.32 randomized breast cancer trial of metformin versus placebo: Implications of the type of recurrence and new non-breast cancer primary on outcome assessment
Katarzyna Joanna Jerzak, University of Toronto, Toronto, ON, Canada
K. J. Jerzak1, W. R. Parulekar2, B. E. Chen2, A. E. Lohmann3, P. J. Goodwin1; 1Medicine, University of Toronto, Toronto, ON, CANADA, 2Canadian Cancer Trials Group, Queen’s University, Kingston, ON, CANADA, 3Medical Oncology, University of Western Ontario, London, ON, CANADA.
Background: STEEP criteria were developed to standardize interpretation of adjuvant breast cancer (BC) clinical trial outcomes. Invasive disease-free survival (IDFS), defined as time from randomization to the earliest occurrence of invasive local, regional, or distant recurrence, new primary invasive cancers (breast or non-breast), and death due to any cause, was initially recommended as a preferred optimal outcome in STEEP (Hudis JCO 2007). Non-breast primary cancers were excluded in Invasive BC-Free Survival (IBCFS) in STEEP 2 (Tolaney JCO 2021). We examine these STEEP outcomes, as well as Distant Recurrence-Free Survival (DRFS), BC Specific Survival (BCSS) and Overall Survival (OS) in CCTG MA.32. Methods: MA.32 (NCT01101438) evaluated the efficacy of metformin versus placebo among 3,649 non-diabetic women with high-risk early BC enrolled between 2010 and 2013. The previously reported analysis of the primary outcome measure (IDFS) and OS showed no metformin benefit on hormone receptor positive or negative BC (any HER2) but suggested a beneficial effect in HER2+ BC (Goodwin JAMA 2022). We summarized individual components of IDFS across BC subtypes- luminal (ER/PR+, HER2-), triple negative (TN; ER/PR/HER2-), HER2+ (any ER/PR) and analyzed treatment effect (metformin versus placebo) on IDFS, IBCFS, DRFS, BCSS and OS to determine whether conclusions regarding treatment efficacy differed by endpoint selection. Statistical methods included Cox regression models and log rank tests. All analyses were two-sided and statistical significance was defined at the p=0.05 level. Results: In the overall population, 710 patients experienced an invasive cancer event; 127 (17.9%) of these events were local/regional. The majority of events were distant recurrences (luminal 55.8%, TN 47.5%, HER2+ 54.5%) and a minority were new non-breast primary cancers (luminal 20.4%, TN 15.2%, HER2+ 20.2%). Within each BC subtype, there was little variation in the hazard ratios (HRs) comparing metformin to placebo and no difference in statistical significance across outcomes (IDFS, IBCFS, DRFS, BSCC, OS). The types of events and HRs for efficacy endpoints of interest are summarized by subtype in the Table. Conclusions: MA.32 enrolled patients with high risk breast cancer, resulting in a predominance of distant breast cancer recurrences. All endpoints provided similar information regarding metformin efficacy across BC subtypes regardless of whether metformin was ineffective (luminal, TN) or showed potential efficacy in an exploratory analysis (HER2+) in this trial where most events were distant recurrences.
| Subtype | Median follow-up, months | IDFS | IBCFS | DRFS | BCSS | OS | |||||||||||||||||||||
| Luminal n=2104 | 96.2 | 1.05 (0.87-1.28), p=0.61 | 1.03 (0.84-1.25), p=0.81 | 1.05 (0.84-1.31), p=0.69 | 1.09 (0.81-1.48), p=0.57 | 1.13 (0.87-1.47), p=0.37 | |||||||||||||||||||||
| TN n=925 | 94.5 | 1.18 (0.90-1.56), p=0.24 | 1.09 (0.81-1.47), p=0.58 | 1.07 (0.78-1.48), p=0.67 | 0.99 (0.68-1.44), p=0.95 | 1.08 (0.76-1.52), p=0.68 | |||||||||||||||||||||
| HER2+ n=620 | 95.2 | 0.64 (0.43-0.95), p=0.03 | 0.56 (0.36-0.80), p=0.013 | 0.55 (0.34-0.88), p=0.01 | 0.57 (0.29-1.07), p=0.08 | 0.54 (0.30-0.99), p=0.04 |
Presentation numberPS4-08-12
Adherence to Fasting Diet and Mediterranean Diet protocol in women with early stage breast cancer receiving chemotherapy; a randomized clinical trial
Dimitrios Tryfonopoulos, St Savas Oncology Hospital, Athens, Greece
N. Kontou1, D. Tryfonopoulos2, A. Apostolou2, N. Stavrou2, D. Panagiotakos3, O. Androutsos4; 1Department of Nutrition and Dietetics, St Savas Oncology Hospital, Athens, GREECE, 22nd Department of Medical Oncology, St Savas Oncology Hospital, Athens, GREECE, 3School of Health Sciences and Education, Department of Nutrition and Dietetics, Harokopio University, Athens, GREECE, 4Lab of Clinical Nutrition and Dietetics, Department of Nutrition and Dietetics, Scool of Physical Ed, University of Thessaly, Trikala, GREECE.
Background: Different types of fasting diet protocols have been associated with a protective effect towards chemotherapy side effects and quality of life. There are not any data of the implementation of a combined protocol including fasting diet against a Mediterranean-type diet in breast cancer patients. Study design: In this randomized clinical trial, 33 newly diagnosed women with early breast cancer were enrolled; Group A (n=16) received a 60 hours fasting diet (36 hours before and 24 hours after chemotherapy) and then guided to follow Mediterranean diet between the chemotherapy cycles, whereas Group B (n=17) received standard of care and no specific diet recommendation. The mean age of Group A and Group B was 45,56±8,23 and 47,35±10,08, respectively. Adherence to Mediterranean diet was assessed by MedDietScore. For each chemotherapy cycle participants were using a self-reported scale to assess adherence to fasting-diet or Mediterranean diet (range 1-10, 1 lower and 10 higher). Results: In total 30.3% (n=10) of the participants received 4 cycles of chemotherapy, 66,67% (n=12) received 6 cycles of chemotherapy and 33.33% (n=11) received 8 cycles of chemotherapy, with no differences between Groups. At baseline, MedDietScore for Group A was 31.47±4.42 and for Group B was 27.93±8.52 (p=0.163). In subgroup analysis for the first four cycles of chemotherapy mean adherence to fasting diet was 8.43/10, whereas adherence to the Mediterranean Diet was 7.79/10. Conclusions: This is the first study reporting data from breast cancer patients following a fasting diet in combination with the Mediterranean Diet. Adherence to both the fasting regimen and the Mediterranean diet was high, indicating that this combined approach is feasible for use in cancer clinical trials. However, its effectiveness in reducing side effects and improving quality of life remains to be established.
Presentation numberPS4-08-13
The intrinsic subtypes in a large cohort of pure DCIS – frequencies and associations with local recurrence
Michael T Hallett, Western University, London, ON, Canada
M. T. Hallett1, S. Nofech-Mozes2, E. J. Mucaki1, S. Trebinjac3, Y. Amemiya4, A. Seth4, E. Hahn5, L. Paszat6, E. Rakovitch6; 1Biochemistry, Western University, London, ON, CANADA, 2Department of Anatomic Pathology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON, CANADA, 3Evaluative Clinical Sciences, Sunnybrook Research Institute, Toronto, ON, CANADA, 4Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, CANADA, 5Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, CANADA, 6Department of Radiation Oncology, Sunnybrook Health Sciences Centre and University of Toronto, Toronto, ON, CANADA.
Background: Ductal carcinoma in situ (DCIS) is a non-invasive precursor lesion confined to the duct of the breast, and is diagnosed in ~54,000 women in the USA annually. Most are treated with breast-conserving surgery (BCS) with or without breast radiotherapy (RT). Randomized control trials found that the 10-year cumulative incidence of invasive local recurrence (ILR) is estimated to be up to 14% for women treated with BCS alone and 6.5% in women who also received RT. The development of ILR necessitates significant additional treatment, since it is associated with a 7-17 fold increased subsequence risk of breast cancer mortality. There remains a critical need to identify prognostic and predictive markers for DCIS which would allow for patient-specific targeted strategies for prevention of invasive breast cancer (BC) (in aggressive cases) and the reduction of overtreatment (in indolent cases). Invasive BC has long been recognized as a heterogeneous disease consisting of at least four intrinsic subtypes (luminal A and B, basal-like, HER2-enriched) which are distinct at the molecular level, have different treatment options, and have differential patient prognoses. Several efforts have established that DCIS samples also possess these subtypes, albeit with differences in expression of some marker genes and with significant differences in the relative frequencies of the four intrinsic subtypes e.g. the HER2-enriched subtype is estimated to be twice as frequent in DCIS than invasive BC. Hypotheses/Objectives: (i) To identify the intrinsic subtypes across our large Ontario population-based cohort of pure DCIS treated with BCS and/or RT; (ii) To compare the frequency of subtypes in our DCIS cohort against previous DCIS and IDC cohorts; (iii) To evaluate the prognostic capacity of the intrinsic subtypes with respect to ILRs; and (iv) To identify associations between intrinsic DCIS subtypes and clinicopathological factors including age and grade. Methods: Using a custom Nanostring nCounter panel, we evaluated the RNA expression of the 50 genes from the PAM50 intrinsic subtyping tool across N=976 pure DCIS samples, and performed unsupervised and supervised analysis to classify intrinsic subtype and to identify molecular components that differ from the canonical patterns found in invasive BC. Cox-based regression is used to estimate hazard ratios (HRs) for other variables with respect to ILR-free survival. Results: Our cohort’s increased size (in comparison to 8 previous studies) and the fact that it is close to a true population-based study provide for more robust estimates of intrinsic subtype frequencies. Within luminal samples, we see an increase in luminal B (18%) and a decrease in luminal A (40%) compared to previous findings. Surprisingly, we observe a significant reduction in the HER2-enriched subtype (16.6%) below previous reports (~25%), although this remains elevated from their observed frequency in invasive BC (~11.5%). We concur with previous findings that the basal-like subtype is molecularly more heterogeneous than their invasive counterparts. ILR was significantly elevated only in the HER2-enriched subtype (HR=1.69 p<0.036; baseline luminal A). The prognostic capacity of some clinicopathological factors were strongly dependent on subtype e.g. reduced HR for age only in the basal-like. Adjuvant RT reduced ILR across the entire cohort (HR=0.70; p<0.055), however it was most beneficial in HER2-enriched DCIS (HR=0.44; p<0.035). Significance: Since the molecular differences between subtype are so large, they must be considered when designing studies. Our results provide more robust, closer to population-based estimates for several important variables including intrinsic subtype and clinicopathological factors.
Presentation numberPS4-08-14
A Systematic Review and Extracted Individual Patient Data Meta-analysis of Long- Term Outcomes in Triple-Negative Breast Cancer after a Pathologic Complete Response: Does the Type of Neoadjuvant Therapy Matter?
Lis Victoria Ravani, University of Sao Paulo School of Medicine, São Paulo, Brazil
L. Ravani1, S. Wander2, M. Kok3, K. McCann4, J. Cortes5, R. Barroso-Sousa6, M. Lustberg7, J. Binez8, I. Michelon9, L. Testa10, M. Wang11, D. Deng12, R. Colombo Bonadio13; 1Medicine, University of Sao Paulo School of Medicine, São Paulo, BRAZIL, 2Hematology/Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, 3Medical Oncology, Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, NETHERLANDS, 4Oncology, University of California Los Angeles Medical Center, Los Angeles, CA, 5Oncology, International Breast Cancer Center, Barcelona, SPAIN, 6Oncology, DASA Oncology, Brasilia Hospital, DASA, Brasilia, BRAZIL, 7Medical Oncology, Yale Cancer Center, Yale University, São Paulo, CT, 8Oncology, Instituto Nacional de Cancer, Rio de Janeiro, BRAZIL, 9Hematology and Oncology, University of Virginia Comprehensive Cancer Center, Charlottesville, VA, 10Oncology, Instituto do Câncer do Estado de São Paulo (ICESP), São Paulo, BRAZIL, 11Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 12Department of Public Health Sciences, Penn State University College of Medicine, São Paulo, PA, 13Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL.
INTRODUCTION: Neoadjuvant chemotherapy is standard of care for stage IB-III triple-negative breast cancer (TNBC), with pathological complete response (pCR) strongly associated with survival. Although the escalation of neoadjuvant therapies with platinum and immune checkpoint inhibitors (ICI) improves pCR rates and long-term outcomes, patients with pCR in the control arms of pivotal trials also show favorable outcomes. Whether the type of neoadjuvant regimen leading to pCR impacts long-term survival differently is largely unknown.METHODS: We conducted a systematic review and meta-analysis, searching PubMed, Embase, Cochrane, and conference proceedings for phase II and III trials including patients with early-stage TNBC and pCR. A pooled analysis of Kaplan-Meier-derived individual patient data was performed for event-free survival (EFS) and overall survival (OS), with subgroup analyses by treatment regimens.RESULTS: Of 2,830 identified publications, 18 trials (16 randomized and 2 single-arm) comprising 3,430 TNBC patients with pCR were included. Neoadjuvant ICI with chemotherapy improved EFS (HR 0.67; 95%CI 0.50-0.89; p<0.01) compared with chemotherapy-only regimens, with no significant OS difference for patients with pCR (HR 0.84; 95%CI 0.50-1.41; p=0.51). In contrast, EFS and OS were not significantly different regardless of platinum use (HR 0.55; 95%CI 0.20-1.50; p=0.24 and HR 0.33; 95%CI 0.09-1.22; p=0.10, respectively). Similarly, anthracycline-containing regimens showed comparable EFS to anthracycline-free regimens (HR 0.86; 95%CI 0.51-1.45; p=0.58). For patients with pCR after ICI therapy, the benefit of adjuvant ICI for EFS or OS was not statistically significant (HR 1.16; 95%CI 0.55-2.44; p=0.70 and HR 2.91; 95%CI 0.40-21.37; p=0.29, respectively). No significant EFS difference was observed between neoadjuvant strategies using 6 or 8 chemotherapy cycles (HR 0.87; 95% CI 0.49-1.55; p=0.64).CONCLUSION: Neoadjuvant ICI-based regimens improve EFS in patients with early-stage TNBC beyond pCR. However, among patients who achieved pCR, EFS seems not to be impacted by the type of neoadjuvant chemotherapy used.
| Regimens | EFS | OS |
| ICI vs Non−ICI | 0.67 (0.50-0.89) | 0.84 (0.50-1.41) |
| Platinum vs Non−platinum | 0.55 (0.20- 1.50) | 0.33 (0.09-1.22) |
| Anthracycline vs Non−anthracycline | 0.86 (0.51-1.45) | 2.91 (0.40-21.37) |
| Adjuvant ICI vs Non−adjuvant ICI | 1.16 (0.55- 2.44) | |
| 8 vs 6 chemotherapy cycles | 0.87 (0.49-1.55) |
Presentation numberPS4-08-15
Towards Personalized Medicine – Patient-Derived Breast Tumor Grafts as Predictors of Relapse and Response to Therapy: Final Results
Christos Vaklavas, Huntsman Cancer Institute, Salt Lake City, UT
C. Vaklavas1, C. B. Matsen2, Z. Chu3, K. M. Boucher4, S. D. Scherer3, S. Pathi3, A. Beck1, K. E. Brownson2, S. S. Buys1, N. Chittoria1, E. D’Astous5, H. E. Gulbahce6, N. L. Henry1, S. Kimani1, J. M. Porretta2, R. Rosenthal2, J. Ward1, M. Wei1, B. E. Welm7, A. L. Welm3; 1Division of Oncology, Huntsman Cancer Institute, Salt Lake City, UT, 2Department of Surgery, Huntsman Cancer Institute, Salt Lake City, UT, 3Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, UT, 4Division of Epidemiology, Huntsman Cancer Institute, Salt Lake City, UT, 5Clinical Trials Office, Huntsman Cancer Institute, Salt Lake City, UT, 6Department of Pathology, Huntsman Cancer Institute, Salt Lake City, UT, 7Division of Surgical Oncology, Huntsman Cancer Institute, Salt Lake City, UT.
Background: Predicting the risk of relapse in patients with non-metastatic breast cancer is important for medical decisions. Pathologic response and especially pathologic complete response (pCR) after preoperative chemotherapy has been associated with risk of relapse; however this association is imperfect. Our work in patient-derived xenografts (PDX) indicated that tumor engraftment in mice correlated with risk of recurrence. To understand further the prognostic utility of PDX, we designed a prospective clinical trial to determine the correlation between PDX generation with residual disease and patient outcome following neoadjuvant chemotherapy. The secondary endpoint of disease-free survival (DFS) in 1 year has been published. We now present the final result of the primary endpoint, DFS in 3 years. Methods: Women with newly diagnosed non-metastatic hormone receptor low-positive (HR-low, ER and/or PR ≤ 10%) or negative breast cancer planned to receive systemic chemotherapy prior to definitive surgery were eligible. Tumor tissue at diagnosis was orthotopically implanted into NOD/SCID mice. The primary objective of the study was to correlate the ability of a tumor to engraft in mice with pathologic responses and clinical outcomes. Results: Between 12/2016 and 1/2021, 80 patients enrolled (triple negative breast cancer (TNBC), n=51; HR-low/Her2-, n=10; HR-low or negative/Her2+, n=18; mixed, n=1). Patients received uniform preoperative chemotherapy regimens per standard of care. PDXs were successfully established from 18 patients (PDX(+), 22.5%). 37 patients achieved pCR (46.3%). In the Her2- subgroup (n=62), 17 (27.4%) patients were PDX(+) and 23 (37.1%) achieved pCR. With a follow up of the last enrolled patient of 3.6 years, 14 (17.5%) patients relapsed, of whom 10 were PDX(+) and 4 PDX(-). Three patients who relapsed had achieved a pCR, all of whom were PDX(+). The PDX(+) patients who relapsed, invariably relapsed early (<1 year from definitive surgery) and their overall survival (OS) following relapse did not exceed 1 year. Among the 4 PDX(-) patients who relapsed, 1 relapsed early and died of recurrent disease; 1 relapsed 2.7 years following her definitive surgery and remains alive and off treatment 5.5 years following her recurrence. One PDX(-) patient with TNBC had a new diagnosis of stage 1 contralateral HR+/Her2- breast cancer 5.4 years following her definitive surgery. PDX engraftment remains significantly associated with relapse (Hazard ratio (HR) for relapse: 12.9 (95% CI, 4.02 – 41.4, p<0.001)) while the achievement of pCR has not reached statistical significance (HR for relapse: 3.5 (95% CI, 0.98 – 12.5, p 0.055)) even with longer follow-up. PDX engraftment remains significantly associated with OS and breast cancer specific survival (BCSS, HR for BCSS: 21.8 (95% CI, 4.69 – 102, p<0.001)) while achievement of pCR is not (HR: 2.43 (95% CI, 0.64 – 9.15, p 0.2)). This is particularly true for the Her2- subgroup: PDX engraftment remains significantly associated with relapse (HR: 13.6 (95% CI, 3.72 – 50.1, p<0.001)) while the achievement of pCR has not reached statistical significance (HR: 2.1 (95% CI, 0.58 – 7.64, p 0.3)). PDX engraftment remains significantly associated with OS and BCSS (HR for BCSS: 33.2 (95% CI, 4.19 – 263, p<0.001)) while achievement of pCR is not (HR: 1.42 (95% CI, 0.37 – 5.5, p 0.6)). Conclusion. Our functional studies identify not only a patient population with a high risk of relapse with greater precision than the achievement of pCR, but also patients whose relapse has a particularly aggressive natural history. We established patient derived models that recapitulate this aggressive disease and have used these models for medium throughput drug screens. Ongoing prospective studies evaluate the impact of these functional studies on treatment selection and patient outcomes.
Presentation numberPS4-08-16
Evaluation of Clinicopathological Features and Prognoses of Male Patients with Breast Lymphoma
Abdulmunir Azizy, Istanbul University Institute of Oncology, Istanbul, Turkey
A. Azizy1, I. Dogan2, M. Bozkurt3, O. Dülgeroglu4, S. Yücel5, I. Yıldız3, I. D. Subası6, A. Arican3, C. Uras7; 1Medical Oncology, Istanbul University Institute of Oncology, Istanbul, TURKEY, 2Medical Oncology, Acibadem Healthcare Group, Istanbul, TURKEY, 3Medical Oncology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 4Breast Surgery, Acibadem University Atakent Hospital, Istanbul, TURKEY, 5Radiation Oncology, Acibadem Research Institute of Senology, Istanbul, TURKEY, 6Breast Radiology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 7Breast Surgery, Acibadem Research Institute of Senology, Istanbul, TURKEY.
Background: Primary breast lymphoma is an extremely rare malignancy, and its occurrence in male patients is even less common, with limited published data available. Due to its rarity, the clinicopathological characteristics and prognostic factors in male breast lymphoma remain poorly understood. Better characterization of this unique patient group may contribute to more tailored diagnostic and therapeutic approaches. This study aimed to evaluate the clinicopathological features and prognoses of patients diagnosed with male breast lymphoma. Method: Data for this study were extracted from the Surveillance, Epidemiology, and End Results (SEER) database, covering cases of male breast lymphoma diagnosed between 2000 and 2021. Demographic, clinicopathological, and survival data were collected and analyzed. Survival outcomes were assessed using Kaplan-Meier curves with the log-rank test. Results: A total of 138 patients were included in the study. 86 (62.3%) of the patients were over 65 years of age. The race of 122 (88.4%) patients was white. The disease was detected in the right and left breast at a similar rate (Right- 48.6%, Left – 47.1%). The most common histopathological types were Diffuse Large B Cell Lymphoma (39.1%), MALT (mucosa-associated lymphoid tissue) lymphoma (23.9%), and Follicular lymphoma (17.4%), respectively. According to the Ann Arbor staging system, the valid percentage of Stage 1 disease was 48.8% and the rate of Stage 2 disease was 21.3%. Breast surgery was performed in 30.4% of the patients, as a valid percentage, the rate of patients who received radiotherapy was 31.2% and the rate of patients who received chemotherapy was 44.9%. B symptoms were present in 6.7% of the patients. The 5-year and 10-year survival rates were 71.7% and 55.7%, respectively. In the prognostic evaluation of the factors affecting survival; age (p<0.001), tumor side (p=0.727), race (p=0.539), origin (p=0.888), presence of B symptom (p=0.778), lymphoma type (p<0.001), stage (p=0.040), breast surgery (p=0.555), radiotherapy (p=0.033), chemotherapy (p=0.017) were determined. Conclusion: Male breast lymphoma is quite rare and is more common in the elderly population. Age, type of lymphoma, and tumor stage were identified as prognostic factors. Receiving chemotherapy and radiotherapy in treatment has a positive effect on prognosis.
Presentation numberPS4-08-17
Evaluation of Clinicopathological Features and Prognoses of Female Breast Sarcoma Patients
Abdulmunir Azizy, Istanbul University Institute of Oncology, Istanbul, Turkey
A. Azizy1, I. Dogan2, O. Dülgeroğlu3, S. Yücel4, M. Bozkurt5, I. Yıldız5, A. Arican5, I. D. Subası6, C. Uras7; 1Medical Oncology, Istanbul University Institute of Oncology, Istanbul, TURKEY, 2Medical Oncology, Acibadem Healthcare Group, Istanbul, TURKEY, 3Breast Surgery, Acibadem University Atakent Hospital, Istanbul, TURKEY, 4Radiation Oncology, Acibadem Research Institute of Senology, Istanbul, TURKEY, 5Medical Oncology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 6Breast Radiology, Acibadem University Atakent Hospital, Istanbul, TURKEY, 7Breast Surgery, Acibadem Research Institute of Senology, Istanbul, TURKEY.
Background: Primary breast sarcomas are extremely rare malignant tumors that account for less than 1% of all breast malignancies. These tumors differ significantly from common breast carcinomas in terms of histogenesis, clinical behavior, and treatment approaches. Due to their low incidence, comprehensive data on clinicopathological characteristics and prognostic factors in female patients are limited. In this study, we evaluated the characteristics and prognostic features of female breast sarcoma patients. Method: Data for this study were obtained from the Surveillance, Epidemiology, and End Results (SEER) database, including female patients diagnosed with primary breast sarcoma, between 2000 and 2021. Demographic, clinicopathological, and survival data were analyzed to identify prognostic factors. Survival outcomes were evaluated using Kaplan-Meier analysis and multivariate Cox regression models. Results: A total of 4389 female breast sarcoma patients were identified from the database. A total of 2613 (59.5%) patients were under the age of 60. The five most common sarcoma types were malignant phyllodes tumor (58.4%), hemangiosarcoma (16.3%), carcinosarcoma (6.2%), sarcoma not otherwise specified (NOS) (3.7%), and spindle cell sarcoma (2.3%), respectively. The disease stages at diagnosis were localized (80.3%), regional (15.2%), and metastatic (4.5%), respectively. Surgical breast surgery was performed in 95.1% of the patients, 23.5% of patients received radiotherapy, and 14.9% of patients received chemotherapy. The 3-year, 5-year, and 10-year cancer-free survival rates were 82.3%, 79.1%, and 76.7%, respectively. The factors affecting prognosis were evaluated and in univariate analysis; age at diagnosis (p<0.001), race (p=0.021), origin (p<0.001), tumor side (p=0.590), sarcoma type (p<0.001), disease stage (p<0.001), surgery (p<0.001), radiotherapy (p=0.003) and chemotherapy (p<0.001) were determined. In multivariate analysis, age (p < 0.001), stage at diagnosis (p < 0.001), sarcoma type (p < 0.001), and surgery (p < 0.001) were identified as prognostic factors in breast sarcoma patients. Conclusion: Breast sarcomas are quite rare in women. According to the results of this study, cancer-specific deaths generally occur within the first 3 years. Age at diagnosis, sarcoma type, initial stage, and surgical treatment were determined as prognostic factors in breast sarcoma patients.
Presentation numberPS4-08-18
Tailoring escalated adjuvant chemotherapy for multifocal and multicentric breast cancer with low tumor burden
Yizi Zheng, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
Y. Zheng1, Z. Zhan2, J. Ying1; 1Department of Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, CHINA, 2Department of Mathematics and Computer Science, Eindhoven University of Technology, Eindhoven, NETHERLANDS.
Objectives: Multifocal and multicentric breast cancer (MMBC) is generally defined as two or more malignant lesions in one breast, accounting for approximately 6% to 60% of breast cancers. MMBC may be more likely to spread to regional lymph nodes (LNs) and patients with MMBC are at higher risks of both distant metastasis and local recurrence than patients with unifocal breast cancer (UBC). Ongoing discussions persist concerning the prognosis of multifocal and multicentric breast cancer (MMBC) with low tumor burden, and pertinent adjuvant chemotherapy guidelines for these individuals are absent. Methods: Female patients diagnosed with cT1-T2 stage MMBC or unifocal breast cancer (UBC) were extracted from the Surveillance, Epidemiology and End Results database (2010-2020). Propensity score matching (PSM) was utilized to balance baseline characteristics. The log-rank test was used for each matched dataset to compare overall survival (OS) and BCSS between the pairs, respectively. The Kaplan-Meier curves for different groups were also provided. Additionally, univariate and multivariate Cox proportional hazards regression models were fitted to estimate the hazard ratios (HRs) and the 95% confidence intervals (CIs) for OS and breast cancer-specific survival (BCSS). Subgroups of different tumor sizes were defined based on typical cut-off values for chemotherapy in the NCCN guidelines, namely, less than 1 cm, between 1 and 2 cm, and greater or equal to 2 cm. Results: There were 9,751 pairs of MMBC and UBC patients effectively matched by PSM method. The survival analysis showed that the OS and BCSS of patients with MMBC were significantly worse than UBC (OS, p < 0.001; BCSS, p < 0.001, log-rank test). Multivariate analyses of the 9,751 cases revealed that MMBC patients who were under 50 years old, married, Caucasian, with low-grade tumor, had a smaller tumor size, had N0 stage disease, ER-positive subtype, PR-positive subtype, had fewer lesions, and received chemotherapy had better OS. Cox multivariate analyses of the 9,751 patients with MMBC showed that receiving chemotherapy was related to a better OS (hazard ratio, [HR] = 0.583, 95% confidence interval [CI], 0.522-0.652) and a better BCSS (HR = 0.830, 95%CI, 0.702-0.980). Given that chemotherapy has shown an effect of improving survival in multivariate analyses, we further analyzed the value of chemotherapy in the population with LN-negative MMBC according to different tumor sizes. Intriguingly, as were shown in KM survival curves, chemotherapy was associated with a significantly improved OS in LN-negative patients when tumor size ≥ 1 cm (tumor size < 1 cm, HR = 0.889, 95%CI, 0.549-1.438, p = 0.631; 1 cm ≤ tumor size < 2 cm, HR = 0.617, 95%CI, 0.443-0.861, p = 0.004; tumor size ≥ 2 cm, HR = 0.449, 95%CI 0.347-0.582, p < 0.001). Nevertheless, chemotherapy was not related to BCSS (tumor size < 1 cm, HR = 2.019, 95%CI, 0.839-4.857, p = 0.117; 1 cm ≤ tumor size < 2 cm, HR = 0.905, 95%CI, 0.528-1.55, p = 0.715; tumor size ≥ 2 cm, HR = 0.742, 95%CI, 0.510-1.081, p = 0.120). Conclusions: Multifocality and multicentricity were independent predictors for worse survival outcomes. Chemotherapy should be considered in patients with node-negative MMBC whose tumors are equal to or larger than 1 cm. Future prospective studies are expected to corroborate the findings.
Presentation numberPS4-08-19
Different gene expression between breast cancer and adjacent normal breast tissue according to breast cancer subtype
Ji Yeon Kim, Samsung Medical Center, Seoul, Korea, Republic of
J. Kim1, K. Park2, J. Shin1, H. Ahn1, J. Ahn1, S. Lee1, Y. Park1; 1Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 2Translational Genomics Center, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF.
Introduction: Through the remarkable advances in genomics, we have identified many of the genetic alterations causing breast cancer(BC) development. Moreover, recent research revealed that these genetic alterations have been continually occurring and accumulating long before cancer develops. However, the role of normal breast tissue in BC patients has been veiled. In this study, we aimed to determine gene expression affecting cancer development from normal breast tissue using paired breast normal-cancer tissues. Methods: We collected BC-normal paired tissues which harvested during curative BC surgery since August 2008 to February 2013. Using these samples, we performed RNASeq, nCounter customized gene expression analysis and WES. Results: Of 134 BC tissues, 121 tumor tissues and 51 normal tissues were analyzed using nCounter method. Among these samples, 50 were paired BC-normal tissues. In 51 normal breast tissues, there was all normal like intrinsic subtype. Further DEG analysis according to tumor subtype, there was no significant different in normal tissues among BC subtype. We also analyzed the different gene expression between tumor-normal tissues according to BC subtypes. In HR+HER2- BC, MMP11, BRIC5 and KNTC2 were significantly upregulated in tumor tissue compared to normal tissue (all adjusted P value 1). In HR+HER2+BC, only MMP11 was upregulated but SFRP1, ELF5 and FOXC1 expression decreased. TNBC had significantly different gene expression compared to normal breast tissue. Ten genes (ANLN, APOBEC3B, BIRC5, CDCA1,CEP55, EXO1, KIF2C, KNTC2, MYBL2 and ORM2) were overexpressed in tumor tissue whereas seven genes(AR, ESR1, ERBB4, MLPH, PGR, PIP and SCUBE2) expression was decreased. In HR-HER2+BC, ANLN overexpression but ESR1, MIA and KRT14 downregulation were observed. Further DEG analysis according to survival outcome, upregulation of MMP11 and MKI67 were associated to BC recurrence but BIRC5, CEP55M EXO1 and KNTC2 overexpression were associated to BC recurrence free survival regardless of BC subtypes (all adjusted P value 1). Conclusion: Although there were no different gene expression in normal breast tissues according to BC subtype, significant difference of gene expression between tumor and normal tissues were observed according to BC subtype. In particular, TNBC had the highest number of genes with differential expression in tumor and normal tissue. In addition, different expression of some genes was also associated to BC recurrence.
Presentation numberPS4-08-20
Immune-related adverse events and pathological complete response in early TNBC treated with neoadjuvant chemo-immunotherapy: a real-world analysis
Gonzalo Lendinez-Sanchez, Hospital Regional Universitario de Málaga, Málaga, Spain
G. Lendinez-Sanchez1, A. Godoy-Ortiz2, N. Bousdar-Ahmed1, P. Vazquez-Rojas2, B. Villaescusa-González2, A. Romanos-Nanclares2, J. Pascual2, F. Carabantes1, E. Alba2, E. Villar-Chamorro1, N. Ribelles2, A. Sánchez-Munoz2; 1Department of Medical Oncology, Hospital Regional Universitario de Málaga, Málaga, SPAIN, 2Department of Medical Oncology, Virgen de La Victoria University Hospital, Málaga, SPAIN.
Introduction:Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype. After the KEYNOTE-522 trial showed improved pathological complete response (pCR) rates, neoadjuvant chemo-immunotherapy (Ch-IT) became standard of care. This study evaluates the efficacy and safety of Ch-IT in early-stage TNBC, focusing on the association between irAEs and pCR.Methods:We conducted a retrospective analysis of medical records from 68 patients with early-stage TNBC >2 cm and/or lymph node positive who received neoadjuvant Ch-IT between January 2023 and November 2024. Endpoints were the rate of pCR (defined as absence of invasive cells in breast and lymph nodes), rate of treatment discontinuation prior to surgery, incidence of irAEs (graded according to CTCAE v5.0) and their association with pCR. Results:A pCR was achieved in 44 patients (64.7%). Baseline clinical and pathological characteristics stratified by pCR status are shown in Table 1. Neoadjuvant treatment was permanently discontinued before surgery in 15 patients (22.1%) all attributable to Ch-IT toxicity. A total of 32 patients (47.1%) experienced irAEs detailed in Table 2. No statistically significant association was observed between the occurrence of irAEs and achieving a pCR (χ² = 1.36; p = 0.24); however, 5 of the 6 patients who developed grade ≥3 irAEs achieved a pCR. Conclusions:In this real-world cohort of patients with early-stage TNBC, neoadjuvant Ch-IT demonstrated a pCR rate consistent with the KEYNOTE-522 trial. Although no statistically significant association was found between irAEs and pCR, a numerical trend was observed, particularly among patients with grade ≥3 events. Further studies with larger cohorts are warranted to elucidate the potential predictive role of irAEs in treatment response and long-term outcomes. Table 1 Table 2
Presentation numberPS4-08-21
Neoadjuvant breast cancer in a real life experience in Southern of Italy
Mariangela Ciccarese, Regional Agency for Social and Health development, Bari, Italy
M. Ciccarese1, L. Orlando2, F. Giotta3, M. Di Millo4, M. Morritti5, L. Ciuffreda5, S. Bambace6, F. Giuliani7, O. Custodero7, S. Bruno7, S. Stucci8, M. Moschetta8, S. Pisconti9, G. Palmiotti10, E. De Matteis11, G. Cairo11, S. Cinieri2; 1Regional cancer network, Regional Agency for Social and Health development, Bari, ITALY, 2Oncology Unit-Breast Unit, Ospedale “A. Perrino” Brindisi, Brindisi, ITALY, 3Oncology Unit-Breast Unit, IRCCS Giovanni Paolo II, Bari, ITALY, 4Surgery Unit-Breast Center, OO Riuniti Foggia, Bari, ITALY, 5Oncology Unit-Breast Unit, Casa Sollievo della Sofferenza, San Giovanni Rotondo, ITALY, 6Radiotherapy Unit-Breast Unit, Ospedale Di Miccoli Barletta, Bari, ITALY, 7Oncology Unit-Breast Unit, San Paolo Hospital Asl Bari, Bari, ITALY, 8Oncology Unit-Breast Unit, Aziena Ospedaliera Policlinico Bari, Bari, ITALY, 9Onclogy Unit-Breast Unit, Ospedale SS Annunziata Taranto, Taranto, ITALY, 10Breast Unit, GVM Breast Unit Santa Maria hospital, Bari, ITALY, 11Breast Unit, Ospedale Vito Fazzi-Lecce, Lecce, ITALY.
Background: Neoadjuvant therapy (NAT) is a cornerstone of breast cancer management (1). However, real-world criteria for patient selection remain poorly defined outside the context of clinical trials. A previous Italian study, which included southern regions, highlighted substantial variability in NAT utilization (2,3). According to EUSOMA, the use of NAT represents a key quality indicator within breast unit care pathways (4). This study aimed to investigate which centers in the Apulia region offer NAT, the criteria employed for patient selection, perceived barriers to its use, and the level of adherence to EUSOMA guidelines. Methods: Twelve regional Breast Units (BUs) and five of the seven spoke centers in Apulia participated in the survey (one BU did not respond). The survey was conducted between October and November 13, 2024, and analyzed data from the preceding six months. It consisted of three thematic sections: • C1: Organizational structure, multidisciplinary team (MDT) case volume, proportion of NAT-eligible cases, and the interval between the end of NAT and surgery. • C2: Proportion of NAT-eligible cases by tumor subtype (HER2+, ER/PgR+/HER2-, triple-negative). • C3: Subtype-specific barriers to NAT, with relative relevance ratings. Results: The results are described in table 1 Barriers to Neoadjuvant Therapy (NAT) Identified by Clinicians: Breast Units reports • HER2+ cases: 41% considered tumor size ≤2 cm and node-negative status (T≤2/N0) as a barrier to NAT. • ER/PgR+ cases: 100% identified T1-T2 and node-negative status (T1-T2/N0) as a limiting factor. • Triple-negative breast cancer (TNBC): 83% considered tumor size ≤1 cm as a barrier. Spokes reports: • 80% cited T≤1 cm and node-negative status (T≤1 cm/N0) as a barrier in HER2+ cases. • 100% cited T1-T2 with N0-N1 status (T1-T2/N0-1) as a limitation in ER/PgR+/HER2- cases. • 100% identified T≤1 cm/N0 as a barrier in TNBC cases. Table1
| Criteria for neoadj (C1-C2-C3) | BU (12/13 ) | Spoke (5/7 ) |
| Cluster 1 (C1) Overall pts in MTD | N=1546 | N=380 |
| (C1) neoadj pts/overall pts (%) | 31.9% ( 5-61) | 17% (65) |
| (C1) timing of surgery after neoadj | 35 days ( 30-50) | – |
| (C2) pts suitable neoadj Her-2 + | 22% | – |
| (C2) pts suitable neoadj ER/PGR+Her-2 – | 15% | – |
| (C2) pts suitable neoadj TN | 17% | – |
| (C3) Barriers for neoadj | BU (12/13 ) | Spoke (5/7 ) |
| Weighing Barriers Overtreatment (OT) Delay of surgery (DS) chemiosensibility (CS) | OT 45% DS 13% CS 34% | OT 50% DS 12% CS 73% |
Conclusions: Although limited to a six-month period, the findings reflect the heterogeneity previously reported in broader studies. NAT rates and surgery timing were consistent with guideline recommendations, despite an unselected patient population. Tumor and nodal size—particularly in small, high-risk subtypes—emerged as key limiting factors. Spoke centers expressed greater concern about the risk of over-treatment in low-volume, high-risk scenarios. Further analyses will investigate treatment decision-making and access to clinical trials.
Presentation numberPS4-08-22
Hypofractionated Whole Breast Irradiation with Concomitant Boost following Breast Conservation Surgery in Early-Stage Breast Cancer.
Pavnesh Kumar, The Ohio State University Wexner Medical Center, Columbus, OH
P. Kumar1, T. Jitwatcharakomol2, J. Schoenhals1, A. Crum1, S. Daniel1, M. Mestres-Villanueva1, G. Yevgeniya3, T. Pathmarajah4, T. Y. Andraos1, R. Young1, J. M. Eckstein1, J. R. White5, J. G. Bazan6, E. H. Healy7, S. Jhawar1, S. J. Beyer1; 1Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, 2Division of Radiation Oncology, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, THAILAND, 3Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 4Radiation Oncology, The Ohio State University College of Medicine, Columbus, OH, 5Radiation Oncology, The University of Kansas Cancer Center, Kansas City, KS, 6Radiation Oncology, City of Hope National Medical Center, Duarte, CA, 7Radiation Oncology, University of California, Irvine, Orange, CA.
Background: Hypo-fractionated radiation therapy (HFRT) for breast cancer has been shown to have comparable local control and toxicity to conventional fractionation (CF) whole breast irradiation (WBI). Tumor bed boost improves local tumor control and can be delivered either sequentially (SB) or concomitantly (CB) with WBI. Initial results from NRG RTOG 1005 reported CB with HFRT over 15 fractions (F) was non-inferior to SB after HFRT or CF WBI in terms of in-breast recurrence (IBR), adverse events (AE), and cosmesis, with the added benefit of reduction of overall treatment time. We report outcomes of HFRT with CB in patients with invasive breast cancer (IBC) and ductal carcinoma in situ (DCIS), with majority treated in prone position at our institution. Methods: We retrospectively identified patients with early-stage, clinically node-negative IBC and DCIS treated with HFRT WBI with CB to the lumpectomy cavity from 2013 to 2025. All underwent breast-conserving surgery followed by WBI 40 Gy, plus a CB to lumpectomy cavity of 48 Gy, delivered in 15 F over 3 weeks using 3D conformal (3DCRT). Patient demographics, clinical characteristics, and treatment details were collected from electronic medical records. Ipsilateral breast recurrence (IBR), Disease-free survival (DFS), overall survival (OS), breast cancer specific survival, radiation toxicities, and cosmesis outcomes were recorded. Radiation toxicities were graded using CTCAE v4.0, and physician-reported cosmetic outcomes were categorized as excellent/good versus fair/poor according to the NRG-RTOG Global Cosmetic Score. Results: A total of 407 patients (343 IBC, 64 DCIS) met inclusion criteria; 74 were <50 years of age. Median age was 60 years (IQR: 52-68) and median follow-up was 39 months. Higher risk clinical features included ER- (26.3%), T2 (20.4%), positive margins (3.2%), and grade 3 (43.8%). Most patients (95%) were treated in prone position. 39.6% patients received chemotherapy and 67.1%, endocrine therapy. The most common acute adverse events (AEs) were grade 1 radiation dermatitis (58%), fatigue (63.1%), and breast pain (28.3%). Most frequent late AEs were grade 1 fatigue (21%), breast edema (18.8%), hyperpigmentation (42.5%), fibrosis (20.4%), and atrophy (9.4%) at 6 months, and 15.1%, 14.9%, 28.4%, 21.7%, and 15.1%, respectively, at 1 year. No patients developed grade ≥3 toxicity. At 1 and 3 years, excellent/good cosmesis was observed in 96% and 89% of patients, and fair outcomes in 4% and 11%, respectively, among patients with available data. No poor cosmetic outcomes were observed. Acute and late adverse events, as well as cosmetic outcomes (p-value of 0.777 at 1 year and p-value of 0.364 at 3 years, respectively), were comparable between patients aged <50 and ≥50 years. Three-year IBR was 0.98%. Three and five-year DFS/OS were 98.3%/97.5% and 97.5%/94.7%, respectively. 5-year breast cancer specific survival was 98.9%. Conclusion: The combination of HFRT with concomitant boost offers a promising approach to treatment for early-stage invasive breast cancer and DCIS, providing excellent local control with minimal toxicity and favorable cosmetic outcomes. This study presents real-world experience from a single institution on HFRT with CB to lumpectomy cavity for breast radiation.
Presentation numberPS4-08-23
Reassessing Systemic Therapy in Breast Adenoid Cystic Carcinoma: Insights from a Large Retrospective Cohort
Alexandra Bili Newman, MD Anderson Cancer Center, Houston, TX
A. B. Newman1, F. Bonini2, C. Albarracin3, S. Damodaran4, L. Licitra5, R. Ferrarotto2; 1Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 2Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Department of Anatomical Pathology, MD Anderson Cancer Center, Houston, TX, 4Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 5Division of Cancer Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY.
Background: Adenoid cystic carcinoma (ACC) comprises 10% expression. Two patients received neoadjuvant chemotherapy; one had distant progression during treatment and one had distant recurrence <6 months after surgery. Among the 7 patients (15%) who received adjuvant systemic therapy– endocrine therapy alone (n=1), combined endocrine/chemotherapy (n=1), and chemotherapy alone (n=5), 5 developed recurrence. Adjuvant radiotherapy was administered to 51% of patients. Among patients with localized disease, 15 (32%) developed distant recurrence with recurrence rates similar across histologic subtypes (33% solid only, 47% non-solid only, 56% mixed). ER-positive tumors had numerically lower recurrence rate in this small sample size (11% vs 37% for ER-negative). Notably, 60% of patients with recurrence had received systemic therapy (either neoadjuvant or adjuvant). For localized disease, median time to recurrence was 35 months, with 1 and 5-year survival rates of 98% and 66%, respectively. Conclusions: This represents the largest single-institution cohort of breast ACC reported to date. Our findings highlight significant variability in management and relatively high recurrence rates despite systemic therapy, underscoring the absence of evidence-based treatment guidelines. While selection bias may contribute to these findings, limited efficacy of chemotherapy—consistent with ACC at other primary sites—suggests that systemic therapy in the curative-intent setting should be applied judiciously.
| Median Age at Diagnosis (min-max) | 55 (38-92) |
| Stage at Diagnosis | |
| T1 | 18(38) |
| T2 | 27(57) |
| T3 | 3(6) |
| N0 | 42(89) |
| N1 | 5(11) |
| N2/3 | 0(0) |
| Metastatic | 4(8) |
| Receptor Status | |
| ER positive (≥1%) | 9(18) |
| PR positive (≥1%) | 8(16) |
| HER2 positive | 0(0) |
| TNBC | 42(83) |
| Histologic Subtype | |
| Solid Only | 12(24) |
| Mixed Solid and Non-Solid | 9(18) |
| Non-Solid Only | 15(29) |
| Unknown | 15(29) |
| Treatment | |
| Segmental Mastectomy | 28(58) |
| Mastectomy | 20(42) |
| ALND | 2(4) |
| No Surgery | 4(8) |
Presentation numberPS4-08-24
The Mechanisms and Predictors of Aromatase Inhibitor-associated MusculoSkeletal Symptoms: The ZAP Trial
Jennifer Sheng, Johns Hopkins University, baltimore, MD
J. Sheng1, R. Chen1, J. Zacharski1, D. Tsang1, A. Yende1, H. Qi1, v. stearns2, D. sharma1, C. Santa-Maria1; 1Medical Oncology, Johns Hopkins University, baltimore, MD, 2Medical Oncology, Cornell, New York, NY.
Background: Patients receiving treatment with aromatase inhibitors (AIs) often suffer from Aromatase Inhibitor-Associated Musculoskeletal Symptoms (AIMSS), defined as myalgias, arthralgias, or joint stiffness. These often lead to discontinuation and could impact breast cancer outcomes. We conducted the prospective single arm Zoledronic Acid Prophylaxis (ZAP) trial, which demonstrated that zoledronic acid administered prior to initiating letrozole and after 6 months was associated with a reduced incidence of AIMSS compared to historical controls (BCRT 2018). Our correlative analysis aims to elucidate the mechanism by which AIs contribute to the development of AIMSS. We hypothesize that alterations in muscle injury, vitamin D deficiency, inflammation, and estrogen deprivation may be associated with AIMSS. Methods: Participants from ZAP were postmenopausal women with stage 0-III breast cancer receiving adjuvant letrozole, who also received zoledronic acid at study entry and 6-months. Blood samples were obtained at baseline and 6 months. Blood-based correlatives included serial assessments of 25-OH vitamin D, CK-MB, a multiplex cytokine array, and estradiol (ultrasensitive). At baseline we also collected whole blood for pharmacogenomic analysis SNPs in a target genes list. AIMSS was assessed using the Health Assessment Questionnaire-Disability Index (HAQ-DI) and Visual Analog Scale (VAS) at 6 months. AIMSS was defined as an increase of 0.22 in a scale of 0-3 in the HAQ-DI and/or an increase of 2.0 cm in a scale of 10 cm in the VAS. We also evaluated other patient reported outcomes (PROs): depression (CESD), anxiety and depression (HADS-A), hot flashes (HFRDI), menopausal symptoms (NSABP-MSQ), sleep quality (PSQI), and quality of life (EuroQol). ROC analysis evaluated the predictive accuracy of biomarker changes for AIMSS. Pearson correlation assessed inter-biomarker correlations. Results: From 2011 to 2013, 59 postmenopausal women enrolled in ZAP. Samples were collected in 59 and 49 participants at baseline and 6 months, respectively. Of the cohort included in this analysis, the median age was 59.0. Among 59 women, 5(8.5%) were Black, 59(100%) were non-Hispanic. The median BMI was 28.5 kg/m2 and baseline arthritis was present in 30 (50.8%). Decreases in TNF-b and MCP4 were significantly associated with cumulative incidences of AIMSS (p=0.023, p=0.03) where those who developed AIMSS had a 16% median decrease in TNF-b, 11% median decrease in MCP4 versus a 13% median increase in TNF-b and 12% median increase in MCP4 in those who did not develop AIMSS. Increase in VAS cumulative incidence was significantly associated with decreases in IL27 (p=0.025), IL21 (p=0.026), and VEGFA (p=0.034). Increase in HAQ was significantly associated with decrease in TNF-b (p=0.01), IL17B (p=0.03), but increase in IL1RA (p=0.043). ROC curve-based analyses show that TNF-b exhibits high negative predictive values (NPVs) in classifying AIMSS (0.824; 95%CI [0.566, 0.962]), HAQ (1; 95%CI [0.824, 1]), and VAS (0.824; 95%CI [0.566, 0.962]). IL1RA (0.97; 95%CI [0.82,1]) and IL17B (0.93; 95%CI [0.79, 0.98]) show high NPV for HAQ classification. IL27 (0.815; 95%CI [0.619, 0.937]), IL21 (0.821; 95%CI [0.631, 0.939]), VEGFA (0.8; 95%CI [0.593, 0.932]), and MCP4 (0.81; 95%CI [0.581, 0.946]) achieve similarly good discriminatory ability for VAS classification. Other cytokines and vitamin D levels were not significantly associated with AIMSS. We will report pharmacogenomic associations of genes and estradiol with AIMSS, and biomarker associations with PROs at the conference. Conclusions: Decreases in TNF-beta and MCP4 were associated with the reporting of AIMSS. Early changes in cytokines profile may identify those at greater risk for AIMSS and who may derive greater benefit from earlier symptom management to prevent discontinuation.
Presentation numberPS4-08-25
Trends and determinants of non-treatment in early-stage breast cancer (ESBC)
Marija Sullivan, The University of Texas MD Anderson Cancer Center, Houston, TX
M. Sullivan1, X. Lei2, I. Jackson1, S. H. Giordano3, M. Chavez-MacGregor3; 1Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Department of Health Services Research and Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: ESBC is curable, with established guidelines recommending timely multimodal management to optimize outcomes. Despite this, a subset of patients does not receive any form of cancer-directed therapy. Understanding the factors associated with non-treatment is essential. This study aims to evaluate contemporary, nationwide trends in non-treatment of ESBC and to identify factors associated with non-receipt of cancer care. Methods: We conducted a retrospective cohort study using data from the National Cancer Database (NCDB) to identify patients 18 years of age and older with clinical stage I-III breast cancer diagnosed between 2019 and 2022. We calculated annual rates of non-treatment (no surgery, chemotherapy, radiation, or hormone therapy for breast cancer) and assessed temporal trends across the study period. Univariate and multivariable logistic regression were used to identify clinical and sociodemographic factors associated with non-treatment, including year of diagnosis, age, race and ethnicity, clinical stage, breast cancer subtype, insurance status, among others. Results: Of the 579,005 patients with ESBC identified in the NCDB between 2019 and 2022, 13,631 (2.4%) patients received no cancer treatment. The annual rate of non-treatment was highest in 2019 (2.6%), but similar across 2020 (2.3%), 2021 (2.2%), and 2022 (2.3%). The mean age at diagnosis was 64 years for those patients who did not receive treatment and 61 years for those who did. Black (aOR=1.51, 95%CI 1.44-1.59) and Hispanic (aOR=1.18, 95%CI 1.10-1.26) patients were more likely to not receive treatment compared to White patients. Higher clinical stage (stage II-III vs. stage I), HER2-positive (aOR=1.24, 95%CI 1.18-1.31) and triple-negative breast cancer subtypes (vs. HR-positive/HER2-negative) (aOR=1.10, 95%CI 1.04-1.16) were associated with higher odds of non-treatment. Compared to those with private insurance, patients with Medicaid (aOR=1.31, 95%CI 1.23-1.40) or without insurance (aOR=2.70, 95%CI 2.45-2.97) were more likely to not receive treatment, while those with Medicare (aOR=0.90, 95%CI 0.86-0.95) were less likely. Higher education quartile and treatment in academic facilities were associated with lower odds of non-treatment. When compared to the lowest income quartile, those in the highest income quartile had similar odds of non-treatment (aOR=1.02, 95%CI 0.95-1.10). Conclusions: Despite overall low rates of non-treatment in ESBC, over 13,000 patients in our cohort did not receive any cancer-directed therapy, highlighting a significant opportunity to improve outcomes in patients with curable disease. Older age, Black and Hispanic race and ethnicity, lack of insurance, and treatment in non-academic centers were associated with higher odds of not receiving any cancer treatment. While these findings indicate persistent disparities in breast cancer care, further research is needed to better understand the extent to which non-treatment is driven by informed patient choice, clinical factors, or systemic barriers to accessing cancer care.
Presentation numberPS4-08-26
Weekly carboplatin and paclitaxel with trastuzumab and pertuzumab (HER2+) or bevacizumab (HER2-) in the neoadjuvant treatment of breast cancer: a phase II trial
Farah Shah, University of California Irvine, Orange, CA
F. Shah1, S. Gibson2, F. Luo2, M. Thomas2, N. N. Nafissi1, E. Vosoughi3, S. Lavasani1, K. T. Lane4, E. H. Lin4, K. J. Kansal4, H. M. Yong4, R. S. Mehta1, R. Parajuli1; 1Hematology/Oncology, University of California Irvine, Orange, CA, 2Internal Medicine, University of California Irvine, Orange, CA, 3Medical Oncology, University of California San Francisco, Burlingame, CA, 4Surgical Oncology, University of California Irvine, Orange, CA.
Background: Neoadjuvant chemotherapy (NAC) enables breast conservation and early assessment of treatment response in breast cancer. Anthracycline-based regimens have shown efficacy, but their use is limited by associated long-term toxicities, including cardiotoxicity and secondary leukemia. Weekly taxane-based regimens offer improved tolerability, and dual HER2-targeted therapy with trastuzumab and pertuzumab has demonstrated high pathologic complete response (pCR) rates. For HER2-negative disease, antiangiogenic therapy with bevacizumab may help improve response rates in certain subsets of breast cancer. Objective: To evaluate the efficacy, safety, and tolerability of an anthracycline-free neoadjuvant regimen of weekly carboplatin and paclitaxel with trastuzumab/pertuzumab (HER2+) or bevacizumab (HER2-). Methods: In this single-institution, open-label, phase II trial, patients with histologically confirmed breast cancer (≥1 cm or node-positive) were enrolled. HER2+ patients (Cohort A) received weekly trastuzumab (2 mg/kg; loading 4 mg/kg) and pertuzumab every 3 weeks (420 mg; loading 840 mg). HER2- patients (Cohort B) received bevacizumab 10 mg/kg every 2 weeks. All received paclitaxel (80 mg/m²) and carboplatin (AUC 2) weekly for 12 weeks. Primary endpoint was 2-year progression-free survival (PFS), and secondary endpoints included pCR (RECIST) and toxicity (CTCAE v4.0). Results: 117 patients were randomized (47 to Cohort A, 70 to Cohort B). Baseline characteristics included a median age of 51 years and 80% hormone receptor positivity (HR+). The majority were White (47%) or Asian (24%). Estimated 10-year PFS was 90.3% (95% CI: 81.1-99.5) in Cohort A—89.3% in HR+ (n = 32) and 93.3% in HR- patients (n = 15). In Cohort B, PFS was 68.2% (95% CI: 51.7-84.7), including 59.2% in HR+ (n = 58) and 100% in triple-negative (TNBC) patients (n = 12) (p = 0.0176). Pathologic complete response rates were higher in Cohort A (58.3%) vs. Cohort B (42.6%), especially in HR-/TNBC subsets (88.2% vs. 66.7%). Grade ≥3 treatment-related adverse events (AE) occurred in 28% (Cohort A) and 40% (Cohort B) of patients. Neutropenia was the most common AE in both groups. Neuropathy was reported in 30% (Cohort A) and 51% (Cohort B) of patients, mostly grade 1-2. No cardiotoxicity occurred in Cohort A. Proteinuria was observed in 8.5% of Cohort B patients without treatment delays. Conclusions: This anthracycline-free neoadjuvant chemotherapy (NAC) regimen demonstrated robust efficacy and favorable tolerability in patients with HER2-positive breast cancer, achieving high long-term progression-free survival (PFS) and response rates, particularly among hormone receptor-negative (HR-) subgroups. In HER2-negative disease, clinical outcomes were more heterogeneous, with particularly promising results observed in patients with TNBC. The 10-year follow-up provided critical insights, highlighting favorable outcomes across most subtypes, with the notable exception of HR+/HER2-negative patients. Integration of CDK4/6 inhibitors can help improve outcomes in this subset of patients. In TNBC patients with residual disease, the addition of bevacizumab and immunotherapy may offer therapeutic value. These findings underscore the potential for biomarker-guided treatment strategies and warrant further investigation in prospective clinical trials.
Presentation numberPS4-08-27
Risk of CNS relapse following pathological complete response to neoadjuvant chemotherapy
Luciana de Moura Leite, AC Camargo Cancer Center, Sao Paulo, Brazil
L. M. Leite1, G. R. de Almeida1, M. C. Tavares1, M. G. Cesca1, F. B. Campos1, F. A. Oliveira2, D. M. Dornellas3, E. F. Saldanha4, P. T. Guimarães1, D. D. Arruda1, M. S. Held1, R. L. Viana1, F. G. Moura1, S. K. Loose1, S. F. Silva1, R. Pirolli1, C. A. Fogassa1, B. R. Mattos1, S. M. Sanches1, V. C. Cordeiro de Lima1; 1Medical Oncology, AC Camargo Cancer Center, Sao Paulo, BRAZIL, 2Medical Oncology, Instituto do Câncer do Estado de São Paulo (ICESP), Sao Paulo, BRAZIL, 3Medical Oncology, Hospital Israelita Albert Einstein, Sao Paulo, BRAZIL, 4Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Sao Paulo, ON, CANADA.
Background: Pathological complete response (pCR) after neoadjuvant chemotherapy (NAC) improved breast cancer (BC) outcomes. However, systemic therapy has limited central nervous system (CNS) penetration, and pCR may not predict reduced risk of brain metastases. We evaluated CNS recurrence patterns in early-stage BC patients with or without pCR after NAC.Patients and Methods: All consecutive stage I-III BC patients treated with NAC and surgery between 2007 and 2018 at A.C. Camargo Cancer Center were analysed. BC subtypes were defined by hormone receptor (HR) and HER2 status from pretreatment biopsies and pCR was defined as ypT0/is ypN0. The primary objective was the impact of pCR on the risk of CNS recurrences across BC subtypes. The secondary objectives were patterns of CNS recurrence (brain/leptomeningeal only versus synchronous systemic and CNS metastasis), CNS recurrence-free survival (CNS-RFS) and overall survival after CNS metastasis. Fisher`s Exact test and Pearson’s Chi-square test were used to evaluate differences in CNS across pCR status. Kaplan-Meier and regression analyses were performed. Two-tailed p-values<0.05 were considered significant.Results: Among 1147 patients treated with NAC, 537 patients had HR-positive/HER2-negative, 301 HER2-positive, and 309 triple-negative (TNBC) breast cancer. The median age was 45 years, most had ductal histology (84.7%), grade II (48.6%) or III (42.9%), and stage III (59.2%). Ninety-five percent received anthracycline+taxane NAC and all HER2-positive patients received at least 1 dose of (neo)adjuvant trastuzumab. Three hundred sixty-five (31.8%) achieved pCR – 59/537(11%) HR-positive/HER2-negative, 158/301(52.3%) HER2-positive, 148/309(47.9%) TNBC. CNS recurrence occurred in 72 (6.2%) patients, with no difference between pCR and non-pCR (4.7% vs. 7.0%, p=0.15). Among subtypes, there was no difference for HR-positive/HER2-negative (3.4% vs. 4%,p=1.0) and TNBC (5.4% vs 9.3%,p=0.2), but there was a reduction in risk for HER2-positive (4.4% vs. 14.7%, p=0.003). Isolated CNS relapse was more common among pCR, particularly in HER2-positive tumors (Table). CNS-RFS was improved in HER2-positive patients with pCR but not in other subtypes. Median overall survival after CNS relapse was 12 months, highest in HER2-positive patients (31 months) and lowest in TNBC (8 months). Multivariable analysis identified HER2-positive and TNBC subtypes and cN2-3 status as independent predictors of CNS recurrence. Conclusion: Pathological complete response was not associated with reduced CNS recurrence overall, but was linked to a lower risk of CNS relapse in HER2-positive breast cancer. Isolated CNS relapse was the predominant pattern, suggesting a sanctuary effect.Tailored surveillance strategies are needed in high-risk subtypes, particularly patients HER2-positive and TNBC regardless of pCR status.
| Subtype | Isolated CNS recurrence (%) | CNS plus systemic recurrence (%) | Total CNS recurrences (%) | p-value |
| All Patients | ||||
| pCR | 14 (82.4) | 3 (17.6) | 17 (100) | 0.006 |
| non-pCR | 24 (43.6) | 31 (56.4) | 55 (100) | |
| HR+/HER2- | ||||
| pCR | 1 (50) | 1 (50) | 2 (100) | 1.0 |
| non-pCR | 10 (52.6) | 9 (47.4) | 19 (100) | |
| HER2+ | ||||
| pCR | 7 (100) | 0 (0) | 7 (100) | 0.007 |
| non-pCR | 8 (38.1) | 13 (61.9) | 21 (100) | |
| TNBC | ||||
| pCR | 6 (75) | 2 (25) | 8 (100) | 0.19 |
| non-pCR | 6 (40) | 9 (60) | 15 (100) |
Presentation numberPS4-08-28
Patient-reported outcomes of adjuvant therapy with CDK4/6 inhibitors for HR+/HER2- early breast cancer: a retrospective cohort study
Yongsheng Wang, Tumor Hospital Affiliated to Shandong First Medical University, Jinan, China
B. Cong1, C. Hou2, C. Shan3, S. Sun4, S. Sun5, Y. Wang1; 1Department of Breast Surgery, Tumor Hospital Affiliated to Shandong First Medical University, Jinan, CHINA, 2Breast Center, Tai’an Central Hospital, Tai’an, CHINA, 3Department of Breast and Thyroid Tumors, Affiliated Hospital of Jining Medical College, Jining, CHINA, 4Department of Breast Surgery, Linyi People’s Hospital, Jinan, CHINA, 5Department of Gland Surgery, Liaocheng People’s Hospital, Liaocheng, CHINA.
Objective To compare the patient-reported outcomes (PROs) and quality of life in patients with HR+/HER2- early breast cancer (EBC) treated with ribociclib (RIB) and abemciclib (ABE) as adjuvant therapy. Methods A multicenter, retrospective cohort study was conducted in Chinese patients receiving ribociclib or abemciclib as adjuvant therapy for HR+/HER2- EBC. PRO data were collected via an electronic questionnaire. The questionnaire was based on the PRO scale for breast cancer patients in China (NCC-BC-A scale), EQ-5D-5L and safety information of CDK4/6 inhibitors, including six dimensions of physiological function, psychological status, social support, treatment related symptoms, economic dimension and overall self-evaluation. The higher score of the questionnaire, the better quality of life. Results Between Mar 15, and May 12, 2025, 197 patients completed the survey, including 94 patients who received ribociclib and 103 patients who received abemciclib. Overall, 195 patients (99%) were female and two (1%) were male. No difference was observed in baseline characteristics such as age, employment status, medical insurance, and concomitant treatment. The total score of the PRO in ribociclib group was higher than that in abemciclib group (187.14±17.57 vs 180.66±22.26, p=0.024). In therapeutic domain, the score was significantly higher in ribociclib group (34.81±5.29 vs 32.77±5.51, p=0.009). Patients in ribociclib group were less affected by the treatment-related symptoms of diarrhea (3.88±0.53 vs 2.31±1.33, p<0.001), fatigue (2.29±0.91 vs 2.00±1.02, p=0.038) and hair loss (2.94±1.39 vs 2.49±1.24, p=0.018). The proportion of patients receiving ribociclib who discontinued due to the side effects of long-term treatment (28.6% vs 50%) or reduced the dose due to adverse events (22.2% vs 70%) was lower. The overall health status score of ribociclib group was higher (6.67±1.27 vs 5.83±1.45, p<0.001). Conclusion HR+HER2- EBC patients who received ribociclib had a better PRO score than those who received abemciclib. Patients in ribociclib group experienced less treatment-related adverse reactions, especially diarrhea, fatigue, and hair loss, and had a lower rate of discontinuation/reduction due to adverse events, a better self-evaluation of their overall health status.
Presentation numberPS4-08-29
Neoadjuvant antibody-drug conjugates for early-stage breast cancer: a systematic review and meta-analysis
Gabriela Barbosa Silva, Oncoclínicas&CO, Rio de Janeiro, Brazil
G. B. Silva1, A. R. Simões2, R. H. da Silva3, R. L. Nohmi4, G. O. Bretas1, M. O. Andrade5; 1Department of Medical Oncology, Oncoclínicas&CO, Rio de Janeiro, BRAZIL, 2Department of Internal Medicine, São Paulo Hospital, São Paulo, BRAZIL, 3Department of Internal Medicine, University Hospital of Maringá, Maringá, BRAZIL, 4Department of Internal Medicine, University of São Paulo, São Paulo, BRAZIL, 5Department of Medical Oncology, Brasília Hospital, Rede Americas, Brasília, BRAZIL.
Background: Pathologic complete response (pCR) is a key endpoint of neoadjuvant systemic therapy in early-stage breast cancer (EBC) and is associated with improved survival in triple-negative (TNBC) and HER2-positive subtypes. Several antibody-drug conjugates (ADCs) have shown efficacy in the metastatic setting and are now under investigation for use in localized disease. Preliminary clinical evidence points to promising pCR rates with ADCs in EBC, even among biologically aggressive subtypes. This study evaluates the efficacy and safety of neoadjuvant ADCs in EBC. Methods: We systematically searched PubMed, Embase, and Cochrane databases for studies including patients diagnosed with EBC who received neoadjuvant ADCs. All EBC subtypes were included, and ADCs could be administered as monotherapy or in combination with other systemic agents. Studies were excluded if they involved adjuvant therapy, metastatic disease, or lacked disaggregated data for the ADC arm. Meta-analyses were performed using a random-effects model, with heterogeneity assessed via the I² statistic and Cochran’s Q test. Sensitivity analyses were conducted using meta-analyses of proportions with subgroup analyses. Results: Among 2,809 screened studies, 23 met the inclusion criteria. A total of 1,762 patients treated with ADCs were included in the pCR analysis. Most of the included studies were phase I/II trials, evaluating nine distinct ADCs, directed against four molecular targets (HER2, TROP2, HER3, and LIV1) and utilizing six different payload types. The pooled pCR rate across all neoadjuvant ADC regimens was 36.94% (95% CI 27.44-47.58). A statistically significant difference (p<0.0001) was observed in the subgroup analysis by histological subtype, with the highest pCR rates seen in HER2-positive disease (52.51%; 95% CI 45.50-59.43), followed by TNBC (29.5%; 95% CI 22.4-37.7), and HR+/HER2- (4.05%; 95% CI 2.00-7.75). Meta-analysis of proportions revealed a pCR rate of 43.5% (95% CI 33.48-54.99) for HER2-targeted ADCs, and 41.4% (95% CI 28.7-55.4) for TROP2-targeted ADCs. In an analysis stratified by payload mechanism of action, topoisomerase I inhibitor-based ADCs demonstrated a pooled pCR rate of 29.91% (95% CI 14.16-52.47), while anti-microtubule-based ADCs showed pCR of 42.09% (95% CI 33.87-50.77) (p=0.3028). The most frequent treatment-related adverse events (any grade) included nausea (61.03%), diarrhea (49.40%), alopecia (40.68%), ALT elevation (39.39%), AST elevation (35.45%), neutropenia (30.24%), anemia (28.40%), vomiting (26.32%), and thrombocytopenia (17.46%). Grade ≥3 adverse events were less common, with neutropenia being the most frequent (18.25%), followed by ALT elevation (5.46%), thrombocytopenia (4.99%), and diarrhea (3.67%). Conclusion: Neoadjuvant ADCs demonstrate promising pCR rates in early-stage breast cancer, particularly in HER2-positive subtype. Treatment-related adverse events were frequent, but generally manageable. These findings support further investigation of ADCs in neoadjuvant treatment strategies.
Presentation numberPS4-08-30
Prognostic evaluation of HER2-low status in patients with early breast cancer who underwent neoadjuvant chemotherapy
Sofia Giusti Alves, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
S. Giusti Alves1, M. Silveira Graudenz2, G. Vasconcelos Alves1, C. Pimentel Oppermann3, J. Basso1, M. Morás1, L. Gonçalves de Oliveira1, G. Luís França Rêgo1, A. Lopes Silva1, E. de Farias Barra1, V. Cristina Rodrigues Borges1, A. Baggio Pereira1, D. Dornelles Rosa1; 1Oncology, Hospital de Clínicas de Porto Alegre, Porto Alegre, BRAZIL, 2Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, BRAZIL, 3Oncology, Grupo Hospitalar Conceição, Porto Alegre, BRAZIL.
Prognostic evaluation of HER2-low status in patients with early breast cancer who underwent neoadjuvant chemotherapy Introduction:In the last few years, several studies have highlighted the prognostic and therapeutic implications concerning the heterogeneity of HER2 expression in breast cancer (BC). In the metastatic setting, antibody-drug-conjugates (ADCs) targeting HER2 are approved for patients with low HER2 expression and showed overall survival benefit. In the early setting, however, the role of HER2 low is still under investigation. We aim to determine if HER2 low tumors show differences in pathological complete response rates (pCR) after neoadjuvant chemotherapy (NCT) and survival outcomes when compared to HER2 zero tumors. Methods:Retrospective cohort in Hospital de Clínicas de Porto Alegre, an academic hospital in the South of Brazil. Inclusion criteria: adult patients with stage I-III invasive BC who underwent neoadjuvant chemotherapy and surgery between 2010 and 2020. Patients with HER2 overexpression, unknown or inconclusive HER2 status were excluded. HER2 low was classified as 1+ on immunohistochemistry (IHC) or 2+ on IHC without amplification in the Silver-enhanced in situ hybridization (SISH), while HER2 negative was characterized as 0+ on IHC. Main outcomes: 1) pCR rates in the breast and axilla, comprising absence of invasive residual disease (in situ carcinoma allowed); 2) recurrence-free survival (RFS), defined as the time to distant or local recurrence; 3) overall survival, defined as the time to death due to any cause. Results:N = 173 female patients with a mean (standard deviation) age of 49.24 (10.41) years. Median (interquartile range) follow up: 5.69 (4.71) years. Menopausal status: 89 (51.44%) premenopausal. Stage at diagnosis: 55 (31.8%) stage II and 117 (67.63%) stage III. Hormonal receptors (HR) were positive in 121 (69.94%) patients. NCT comprised an anthracycline-based regimen in 167 (96,50%) patients. HER2 status: 88 (50.87%) HER2 low and 85 (49.13%) HER2 zero. HER2 low patients had a higher frequency of positive HR than HER2 zero patients (82.96 vs. 56.47% respectively, Chi square P < 0.001). pCR rates were 15.61% in the whole sample, 10.23% in HER2 low and 21.18% in HER2 zero patients; however, statistical significance was not reached (Chi square P = 0.076). RFS rate was 67.05% in 5 years and 61.27% in 10 years, with no difference in RFS between HER2 low and HER2 zero patients (Hazard ratio [HR] 0.91, 95% Confidence Interval [CI] 0.56 – 1.47, P = 0.702). OS rate was 70.52% in 5 years and 65.32% in 10 years, also with no difference between HER2 low and zero (HR 0.64, 95% CI 0.39 to 1.08, P = 0.092) Conclusion:In our study, HER2 low and zero patients appeared to have similar pCR and survival rates. More studies concerning HER2 status in the early BC setting should be conducted in order to verify possible differences in oncological outcomes which may change clinical practice. Furthermore, the definition of HER2 low is quite arbitrary and could benefit from revision.
Presentation numberPS4-11-01
Medicare Part B Drug Cost Burden of Pembrolizumab Based Triple Negative and Dual HER2 Triple Positive Regimens for Early Breast Cancer at 2025 Q3 Payment Rates.
Pragya Jain, Baptist Hospital of Southeast Texas, Beaumont, TX
P. Jain1, V. Majmundar2, C. Bhanushali3, N. Ganatra1, M. Patel4, R. Patel1, A. Kothari5, K. Patel1, T. Naqvi6; 1Internal medicine, Baptist Hospital of Southeast Texas, Beaumont, TX, 2Cardiology, University of Miami Miller School of Medicine, Miami, FL, 3Internal medicine, Saint Vincent Hospital, Worcester, MA, 4Surgical Oncology, University of Miami Miller School of Medicine, Miami, FL, 5Medicine, Smt. NHL Municipal Medical College, Ahmedabad, INDIA, 6Hematology-Oncology, Baptist Hospital of Southeast Texas, Beaumont, TX.
Background and Rationale : The direct cost of modern systemic therapy is a growing obstacle for United States patients who rely on Medicare Part B. Two biologic rich standards dominate early breast cancer care: an immunotherapy containing regimen for triple negative disease and a dual HER2 regimen for tumors that are hormone receptor positive and HER2 positive, often called triple positive. Up to date information on the absolute Medicare and patient out of pocket cost for each regimen is limited. Objective : To measure 1 year Medicare drug spending and beneficiary coinsurance for the immunotherapy regimen used in triple negative cancer and the dual HER2 regimen used in triple positive cancer, and to test how those totals change with body size and with 340B acquisition pricing. Methods : Average Sales Price files for Medicare Part B quarter 3 2025 supplied unit prices. Doses came from National Comprehensive Cancer Network Breast Guideline version 4 2025. Rules were applied to a reference woman who weighed 70 kg and had a body surface area (BSA) 1.82 m². For each drug the per dose rule was converted to milligrams, multiplied by cycle count, divided by the billing unit size listed in the Healthcare Common Procedure Coding System, rounded up, and multiplied by Average Sales Price plus 6 percent. Beneficiary liability was 20 percent coinsurance. Sensitivity runs repeated every calculation for 60 kg and 90 kg and for a 40 percent 340B discount. Results : For the 70 kg patient Medicare paid 212,008 USD for the immunotherapy regimen and 205,484 USD for the dual HER2 regimen. Coinsurance liabilities were 42,402 USD and 41,097 USD, respectively. Pembrolizumab accounted for more than 90 percent of triple negative cost, whereas trastuzumab plus pertuzumab accounted for more than 80 percent of triple positive cost. Changing weight from 60 kg to 90 kg moved annual immunotherapy cost by less than 0.2 percent, yet shifted dual HER2 cost from 196,539 USD to 223,302 USD (plus 13.6 percent). Coinsurance moved in parallel, from 39,308 USD to 44,660 USD. A 40 percent 340B discount lowered every total proportionally without changing the rank order. Conclusions : At 2025 quarter 3 prices Medicare pays about 200,000 USD for 1 year of either regimen and the beneficiary faces approximately 42,000 USD in coinsurance. Fixed dose pembrolizumab makes the triple negative regimen cost almost independent of body size, whereas weight based trastuzumab and pertuzumab make the triple positive regimen strongly size dependent. These updated figures quantify present financial burden and provide transparent inputs for future budget impact and cost effectiveness work.
Presentation numberPS4-11-02
Factors associated with lymph node metastasis and its prognostic impact in breast cancer patients with small tumors (≤2cm)
Janghee Lee, Ewha Womans University Mokdong Hospital, Seoul, Korea, Republic of
J. Lee1, K. Yeonjoo2, P. Youri2, L. Young Ah2, W. Joo Hyun1, L. Woo Sung1, K. Hee-Joon2, M. Byung-In1; 1Surgery, Ewha Womans University Mokdong Hospital, Seoul, KOREA, REPUBLIC OF, 2Surgery, Dongtan Sacred Heart Hospital, Dongtan, KOREA, REPUBLIC OF.
Introduction: Although previous randomized controlled trials have suggested that axillary surgery may be omitted in patients with clinically node-negative breast cancer with tumor ≤ 2cm (pT1), approximately 15% of these patients were found to have lymph node (LN) metastasis. Furthermore, while these studies demonstrated that axillary surgery, including sentinel LN biopsy, does not provide a therapeutic benefit, concern remain regarding the prognostic impact of occult LN metastasis. Therefore, the aim of our study was to identify factors associated with LN metastasis in small tumors (≤ 2cm) and to analyzed its impact on recurrence. Methods: We identified patients with pT1 primary breast cancer who underwent treatment at two hospitals between September 2003 and December 2019. To ensure accurate assessment of pathologic LN metastasis, patients who received neoadjuvant systemic therapy or did not undergo axillary surgery were excluded. Multivariate analysis was performed to identify factors associated with LN metastasis and to evaluate the impact of LN metastasis on recurrence-free survival (RFS). Results: A total of 1,745 patients were included in the study, of whom 384 (22.0%) had axillary LN metastasis. In the multivariate binary logistic regression model, LN metastasis was significantly more common in tumors larger than 1cm (odds ratio [OR], 2.94; 95% confidence intervals [CI], 0.15-4.08; P<0.001) and in the presence of lympho-vascular invasion (OR, 4.41; 95% CI, 3.37-5.78; P<0.001) while it was less frequent in tumors with histology other than ductal or lobular type (OR, 0.45; 95% CI, 0.22-0.89; P=0.022). In the multivariate Cox proportional hazard model, LN metastasis was identified as a significant risk factor related with worse RFS (hazard ratio, 1.99; 95% CI, 1.26-3.14; P=0.003). In subgroup analysis, LN metastasis was associated with poor prognosis across most subgroups, with its impact being particularly pronounced in patients with tumors>1cm, histologic grade I or II, and the HR+/HER2- subtype. Conclusion: Our study found the factors associated with a higher likelihood of LN metastasis in small (pT1) breast tumors and confirmed that LN metastasis is associated with wore prognosis. Because the presence of LN metastasis can increase the risk of recurrence, even in small tumors, careful consideration should be given to performing axillary surgery to avoid potential undertreatment in patients with a high risk of LN metastasis.
Presentation numberPS4-11-03
Prognostic impact of PET and/or breast MRI radiological response in early breast cancer patients undergoing neoadjuvant chemotherapy
Matilde Corianò, University Hospital of Parma, Medical Oncology and Breast Unit, Parma, Italy
M. Corianò1, C. Tommasi1, G. Reni2, A. Ingallinella3, C. Tornali1, F. Pratticò1, M. Baronchelli1, E. Cardinale1, M. Scarlattei4, C. Cidda4, E. Spaggiari2, E. Martella5, G. Di Maria1, A. Cosenza1, D. Boggiani1, D. Zanoni1, C. D’Aloia2, E. Silini5, L. Ruffini4, B. Pellegrino1; 1Medicine and Surgery, University Hospital of Parma, Medical Oncology and Breast Unit, Parma, ITALY, 2Diagnostic department, University Hospital of Parma, Section of Radiology and Breast Unit, Parma, ITALY, 3Medicine and Surgery, University Hospital of Parma, Parma, ITALY, 4Diagnostic department, Nuclear Medicine Unit, University Hospital of Parma, Parma, ITALY, 5Onco-ematologic department, Pathology Section, University Hospital of Parma, Parma, ITALY.
Purpose: Pathological complete response (pCR) to neoadjuvant chemotherapy (NACT) is the most relevant prognostic factor for patients with early breast cancer (eBC). However, the prognostic value of complete radiological response assessed by PET and/or breast MRI (bMRI) following NACT remains unclear. In this study, we investigated the potential prognostic significance of radiological complete response (rCR) in patients with eBC undergoing NACT. Methods: We retrospectively identified patients with eBC receiving NACT at the University Hospital of Parma between 2018 and 2022. Data about radiological response in terms of rCR, partial response and disease progression/stable disease and about pCR were collected. We used Kaplan-Meier curves to analyze relapse-free survival (RFS) and overall survival (OS) based on radiological response in the overall population and after excluding patients with Luminal eBC. Comparisons between groups were performed with log-rank test. Cox regression models were used to determine independent predictive role of rCR for RFS and OS, corrected by age, lymph nodes status and pCR. Results: Overall, 120 patients were included in the analysis. The mean age at diagnosis of the study population was 49.4 ± 9.84 years. 58 patients (48.3%) had positive lymph nodes at diagnosis. 38 patients (31.7%) had triple negative BC (TNBC), 47 patients (39.1%) had HER2-positive BC, 35 patients (29.2%) had Luminal BC. Median follow up time was 49 months (IQR 42.1-58.8). At 48 months-follow up, 63.5% of patients without any CR, 96.3% of patients who achieved CR on both PET and bMRI and 97.4% of patients with CR on at least PET or bMRI were alive. Moreover, 47.6% of patients without any CR, 96.3% of patients who achieved CR on both PET and bMRI and 97.4% of patients with CR on at least PET or bMRI were recurrence-free. rCR on at least one radiological assessment was independently associated with better RFS [aHR 0.24 (95% CI, 0.09-0.62, p = 0.003)] and OS [aHR 0.24 (95% CI, 0.06-0.91, p = 0.035)]. No patients who failed to achieve rCR on both PET and bMRI achieved pCR. In TNBC, 13 out of 19 patients (68.4%) with CR on both PET/bMRI obtained pCR, 3 out of 9 patients (33.3%) with CR on at least PET or bMRI obtained pCR (p = 0.002). In HER2-positive BC, 20 out of 27 patients (74.1%) with CR on both PET/bMRI obtained pCR, 7 out of 16 patients (43.8%) with CR on at least PET or bMRI obtained pCR (p = 0.008). In Luminal BC, 3 out of 8 patients (37.5%) with CR on both PET/bMRI obtained pCR, 2 out of 14 patients (14.3%) with CR on at least PET or bMRI obtained pCR (p = 0.0058). Considering only the 85 patients affected by TNBC and HER2-positive BC, RFS and OS were statistically significantly better in patients with rCR on at least one radiological assessment [aHR 0.23 (95% CI, 0.07-0.71, p = 0.011 and HR 0.16 (95% CI, 0.03-0.78, p = 0.024, respectively]. Conclusions: rCR on PET and/or bMRI following NACT is a strong prognostic marker for improved survival in eBC, particularly in TNBC and HER2-positive subtypes. This benefit appears to be independent of achieving pCR, highlighting the potential of imaging response as an additional tool for risk stratification and treatment decision-making in clinical practice.
Presentation numberPS4-11-05
Usefulness of axillary ultrasonography in selecting cases in which sentinel node biopsy can be omitted for early breast cancer
Wakako Tajiri, NHO Kyushu Cancer Center, Fukuoka, Japan
W. Tajiri, Y. Nakamura, Y. Koi, S. Akiyoshi, H. Ijichi, K. Chinami, E. Tokunaga; Department of Breast Oncology, NHO Kyushu Cancer Center, Fukuoka, JAPAN.
[Background]The SOUND and INSEMA trials demonstrated that the omission of sentinel lymph node biopsy (SLNB) is noninferior to axillary staging in patients with early-stage breast cancer (EBC) and negative axillary ultrasonography (AxUS). In addition, a Choosing Wisely guideline recommends omission of SLNB in low-risk elderly females with EBC. Thus, omission of SNB for EBC is on the uptrend. [Purpose]This study was conducted to identify the clinicopathological features and AxUS findings in predicting the negative sentinel nodes in our cohort. [Patients and Methods]Of the 1988 cases who underwent SNB without preoperative systemic therapy for their primary EBC from January 2016 to December 2023, 1395 cases with cT1-2N0 tumors and documented AxUS findings were included in this study. Relationships between the clinicopathological features or AxUS findings and the SNB results were evaluated. Cortical thickness greater than 2.5 mm or absence of fatty hilum and a maximum diameter greater than 1 cm were defined as findings of possible metastatic lymph nodes by AxUS. If none of these three findings were identified, the patient was defined as having no abnormal AxUS. [Result]Among 1395 cases, SN was negative in 1063 patients (76.2%). Any SLNB was positive in 332 (23.8%) patients and 83 cases (25.0%) underwent axillary dissection (ALND) and positive LN status was pN1mi(n=73,5.2%), pN1(n=213,15.3%), and >pN2(n=46, 3.3%). Older age (≥70 years), smaller clinical tumor size, and negative abnormal AxUS findings were significantly associated with SN negativity, while invasive lobular carcinoma (ILC) type was associated with SN positivity. Tumor subtypes defined by ER, PgR, and HER2 status were not associated with SN positivity. ILC type was also significantly associated ≥4 total positive lymph nodes. These findings were also true in HR+/HER2- subtypes (Table 1).When clinical tumor size was ≦1.5cm and AxUS findings were negative, SN was negative in 83.5 % and 88.2 % in all and HR+/HER2- subtype, respectively. Among these patients, moreover, in older (≥ 70) patients or patients with nuclear grade (NG)1 tumor, SN was negative in 88.2% and 92.9% in whole cohort and in 88.6% and 94.3% in HR+/HER2- subtype, respectively. [Conclusion]Older age (≥70 years), smaller clinical tumor size, and negative abnormal AxUS findings were significantly associated with SN negativity in whole cohort and HR+/HER2- subtype. This study suggests that evaluation of cortical thickness, absence of fatty hilum and maximum diameter of LNs in preoperative AxUS is very important in adapting the omission of SNB.
Presentation numberPS4-11-06
A comparison of mortality in SOUND matched population – A single institution experience
Chandana Gavisiddappa, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, United Kingdom
C. Gavisiddappa, R. Parmeshwar; Breast Surgery, University Hospitals of Morecambe Bay NHS Foundation Trust, Lancaster, UNITED KINGDOM.
A comparison of mortality in SOUND matched population – A single institution experience Introduction: Sentinel lymph node biopsy (SLNB) is the standard of care for axillary staging in early-stage breast cancer (EBC). Nodal positivity in T1 disease remains low. The early results of the SOUND trial have demonstrated that eliminating SLNB in patients with cT1 cN0 disease is not inferior and may potentially lead to further de-escalation of axillary surgery in EBC without compromising the oncological safety. This study aims to compare mortality in the given cohort. Methodology: A retrospective study using single unit data from October 2008 to August 2023. All invasive breast cancer up to 2 cm in diameter, with no involvement of the axillary nodes, and a planned breast-conserving surgery and radiation were included in the study in line with SOUND trial. Pre-operative axillary ultrasonography was performed in all these patients as part of their preoperative assessment to confirm cN0 status and any suspicious nodes were then subjected to FNA. Patients’ demographics, disease distribution, nodal positivity were studied along with breast cancer specific and overall mortality. Results: Out of 662 patients in the given period, 198(29.9%) had Grade 1 disease, 365(55.1%) had Grade 2 and 99(14.9%) Grade 3 disease. Mean age of the entire cohort was 61.1, the mean tumour size was 12.31mm. 65(9.8%) patients had positive sentinel nodes out of which 53(8%) had pN1 disease, 6(0.9%) had pN2 disease, 6 had micro-metastasis only. 41(63.07%) patients were screen detected cancers, and 21(32.3%) patients were symptomatic. 56(86.1%) patients were hormone receptor positive type, 3 (4.61%) patients had triple negative and 3(4.61%) had HER2 positive disease. Most common type of cancer noted was invasive ductal in 58(89.2%) patients and 3(4.61%) of them had invasive lobular carcinoma and remaining 1(1.53%) patient had tubular carcinoma. Mean Follow up of 66.3 months, while 3 out of 65 data lost in follow up. A total of 2 loco-regional recurrence (in breast), 1 had contralateral breast cancer and 2 had distant metastasis. The mortality rate in this subset of patients was 1.9% vs 2.6% in SOUND trial. Conclusion: Our study shows that nodal positivity 9.8% while 14% in SOUND trial with comparable N2 disease (0.9% vs 0.6%). SOUND had a lower proportion of micro-metastasis (5.1% vs 9.2%) than our study with comparable mortality rates (2.6% vs 1.9%). The mortality rates are similar in both node negative and node positive groups adding weight to the argument on further axillary de-escalation in EBC. Our study has limitation of small numbers but aligns well with SOUND and the long-term data of the trial will possibly make way for changes in current practice of routine SLNB in low risk EBC.
Presentation numberPS4-11-07
Residual axillary disease in Node-Positive Breast Cancer Patients After Neoadjuvant systemic therapy- A Single centre Retrospective Study
Qurratulain A Chougle, Guy’s & St Thomas NHS Trust, London, United Kingdom
Q. A. Chougle1, C. Kalyvioti1, A. Amylidi2, N. Alluhaydan3, A. Saleeb1, G. Bitsakou1, E. Shaari1, H. Hamed1, K. Cox1; 1Breast Surgery ( Surgical Oncology), Guy’s & St Thomas NHS Trust, London, UNITED KINGDOM, 2Medical Oncology (Breast), Guy’s & St Thomas NHS Trust, London, UNITED KINGDOM, 3Breast Radiology, Guy’s & St Thomas NHS Trust, London, UNITED KINGDOM.
BackgroundAxillary lymph node status is an integral prognostic factor in breast cancer treatment and steers the decisions on local, regional and systemic treatments. Advances in neoadjuvant chemotherapy has led to de-escalation of axillary surgery in patients with N0-N1 disease. Residual axillary lymph node (LN) metastasis after neoadjuvant chemotherapy (NACT) generally warrants axillary lymph node dissection (ALND). We investigated the risk of having additional metastases in the axilla when the lymph nodes are removed by targeted axillary dissection (TAD) harboured metastases after NACT. We aimed to identify subgroups suitable for de-escalated axillary treatment.MethodsWe conducted a retrospective audit of 82 consecutive patients that underwent targeted axillary dissection post neoadjuvant chemotherapy at our centre from January 2019 to December 2023. The data was extracted from patient electronic medical records following patient confidentiality and data protection guidelines. The study has been approved by the trusts governance team.ResultsEighty two patients were included in our retrospective study. The median age was 49 years (range 34-68). Among the 82 patients, there was only one failed identification of the clipped node leading to a primary ALND. Forty patients (49.4%) did not have involved nodes on the targeted axillary dissection (TAD). In our cohort 41 (50.6%) patients had a positive TAD. Thirty eight patients (46.3%) were hormone receptor positive and HER 2 negative. Triple negative breast cancers were found in 13 patients (15.9%) and 28 patients (34.2 %) had HER 2 positive disease. There was no statistically significant correlation with the cancer type or receptor status. Among 44 patients undergoing ALND after positive TAD, 13 (29.5%) had further residual disease in the axilla. Breast complete pathologic response did not correlate with the residual disease in the axilla, allowing for the small sample limitation.
| Histological Subtype | Total | TAD Positive | TAD Negative |
| Hormone + | 38 | 29 | 09 |
| TNBC | 13 | 02 | 11 |
| HER 2+ | 28 | 10 | 18 |
| Total | 81 | 41 | 40 |
Conclusion TAD lymph node positivity is independent of the receptor subtype as shown by our study. We conclude that TAD may be considered for de-escalation of axillary surgery in all histological receptor subtypes. Our study findings are consistent with other similar studies. However, further studies are required with a larger sample size to validate and confirm these findings.
Presentation numberPS4-11-08
Clinical characteristics and prognosis in interval detected breast cancer patients: a single institution cohort analysis
Yesim Eralp, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, Turkey
Y. Eralp1, S. Bayrakceken2, K. Turker Karayazi1, G. Esen Icten3, F. Tokat4, N. Bese5, T. Korkmaz1, O. Sonmez1, H. Kara2, N. Bakir2, C. Uras1; 1Medical Oncology, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, TURKEY, 2General Surgery, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, TURKEY, 3Radiology, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, TURKEY, 4Pathology, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, TURKEY, 5Radiation Oncology, Acibadem MAA University, Research Institute of Senology, Maslak Acibadem Hospital, Istanbul, TURKEY.
Aim: The aim of this study is to identify clinical characteristics and outcomes in a cohort of patients with interval-detected breast cancers. Methods: Interval-detected cancers were tumors that arose within a year of the last screening breast imaging that appeared normal, regardless of having a previous breast cancer diagnosis. Disease-free survival (DFS) was defined separately for primary and secondary cancers from the date of diagnosis of the interval tumor to the date of first recurrence or death. Results: We identified 43 patients who met the criteria for interval-detected breast cancer. Twelve patients had a previous diagnosis of breast cancer, 2 of whom had DCIS. Median follow-up was 24.2 mo ( 4.7-10.2.5). The median age of this cohort (second-primary interval cancer) was 49 (32-70); 10 (83.3%) were premenopausal. The mean time to the diagnosis of the new cancer was 28 months;14 mo for HER2 (+) and 38 mo for ER (+). The pathologic subtypes of invasive primaries were ER (+) in 6 and Her2 (+) in 4 (40%) patients. The interval tumor was detected in the ipsilateral breast in 8 patients, among whom 6 (75%) developed the second primary in the same localization of the primary tumor. Two of those (33%) had mastectomies as primary surgical treatment. There were 8 recurrences, among whom 5 were systemic and 4 were HER2 (+). Thirty-one patients had interval-detected primary breast cancers. The median follow up period was 25.4 mo (1-114.2). The median age was 51 (42-75) and 20 (64.5%) were premenopausal. Among them 2 were triple negative; 10 were Her2 (+); and 19 were ER (+). There were 7 recurrences; among whom 2 were systemic. In the whole cohort, 27 (62.8) patients had a family history of cancer. Among second-primary interval cancer patients, 8 of 12 patients had a family history. Among those 8 who had genetic testing, 5 had pathogenic variants (3:BRCA2; 1: PALB2;1:CHEK2)The DFS of the primary group and the secondary group were 96.8 (95% CI: 78,2-115.4) and 50.6 mo (95% CI: 35,1-98.6), respectively. There was a tendency for a worse outcome in the secondary interval cancer group (p:0.2), mostly driven by the HER2 subgroup. Conclusion:Interval cancers were associated with a high incidence of pre-menopausal status, family history or hereditary pathogenic variants. Secondary interval cancers were more likely to have HER2 (+) primary tumors, a shorter time to diagnosis of the interval cancer and a tendency to occur in the same site of the primary despite adequate local and systemic treatment. There was a tendency for a worse outcome in the secondary interval cancer group. Our data indicate closer follow-up for second primary interval cancers in HER2 (+) patients. These data should be interpreted with caution due to the retrospective nature; small sample size and should be validated in larger datasets.
Presentation numberPS4-11-09
Axillary Management Strategies in Breast Cancer Patients With ypN0 After Neoadjuvant Therapy
Qingyao Shang, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
Q. Shang1, Z. Yan2, L. Sheng3, W. Xin1; 1Breast Surgical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, CHINA, 2Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC, 3Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Beijing, NC.
Background: The optimal axillary management for breast cancer patients achieving nodal pathological complete response (ypN0) after neoadjuvant therapy (NAT) remains unclear, particularly regarding the use of sentinel lymph node biopsy (SLNB) versus axillary lymph node dissection (ALND). We aimed to compare survival outcomes associated with SLNB and ALND in ypN0 patients and evaluate the influence of clinical nodal stage and postoperative axillary radiotherapy. Methods: Using the National Cancer Database (2018-2022), we identified female patients with clinical stage I-III invasive breast cancer who received NAT and achieved ypN0. Patients were grouped based on receipt of SLNB or upfront ALND. Overlap weighting using propensity scores was applied to adjust for baseline differences. Overall survival (OS) was compared using Kaplan-Meier analysis and Cox proportional hazards modeling. Subgroup analyses were stratified by initial clinical nodal stage (cN0-cN3), and exploratory analyses evaluated the impact of axillary radiotherapy on OS. Results: Among 36,452 eligible ypN0 patients, 86.3% received SLNB and 13.7% underwent ALND. After weighting, covariate balance was achieved. SLNB was associated with improved OS compared to ALND (weighted HR: 1.33; 95% CI: 1.11-1.60; p=0.03). Survival benefit from SLNB was strongest in cN0 (HR: 0.65; 95% CI: 0.47-0.89) and cN1 (HR: 0.67; 95% CI: 0.52-0.87) subgroups. No significant OS difference was observed in cN2 or cN3 patients. Axillary radiotherapy did not significantly influence OS in either SLNB or ALND groups. Conclusions: In breast cancer patients achieving ypN0 after NAT, SLNB is associated with favorable survival compared to ALND, particularly in those with low baseline nodal burden (cN0-cN1). The lack of OS benefit from axillary radiotherapy suggests that de-escalation strategies may be safe and appropriate in selected patients. These findings support individualized, response-adapted axillary management after NAT.
Presentation numberPS4-11-10
Patient-centric development of shared decision-making aids: The ASSESS online personalized early breast cancer treatment decision support tool and resource website
Bethany A Kerr, Susan G. Komen, Dallas, TX
B. A. Kerr1, J. D. Alexander2, H. Zhao3, M. Marczyk4, L. C. Crocker2, A. A. Erwin1, E. Kuhn5, H. Baker6, S. Vaidhyanath6, T. Polischuk6, K. A. Sabelko1, L. Pusztai4, S. H. Giordano3, R. J. Volk2, G. M. Zinser1; 1Scientific Strategy and Programs, Susan G. Komen, Dallas, TX, 2Decision Science Core Facility, University of Texas MD Anderson Cancer Center, Houston, TX, 3Department of Health Services Research, University of Texas MD Anderson Cancer Center, Houston, TX, 4Medical Oncology and Hematology, Yale Cancer Center, New Haven, CT, 5Health Information and Publications, Susan G. Komen, Dallas, TX, 6Information Technology, Susan G. Komen, Dallas, TX.
People newly diagnosed with breast cancer spend a considerable amount of time researching their diagnosis, treatment options and associated side effects. Even with access to trusted evidence-based information, such as Susan G. Komen’s About Breast Cancer pages (https://www.komen.org/breast-cancer/), patients and doctors may struggle to account for the many factors in an individual’s unique diagnosis that influence treatment and outcomes, making it hard to balance the potential benefits of therapy with the risks of side effects on a personalized basis. The discontinuation of the popular Adjuvant! Online tool compounded this challenge, creating a need for a new, publicly available tool to provide survival estimates for the early-stage breast cancer population in the U.S. To address this, Komen collaborated with teams from MD Anderson and Yale Cancer Centers to develop a web-based decision support tool that provides personalized five-year survival estimates for women with early-stage breast cancer. The tool, ASSESS, uses an algorithm developed with U.S.-based SEER data to estimate outcomes for various systemic adjuvant therapies based on an individual’s clinical and pathological characteristics. The ASSESS online tool was designed and built by a multidisciplinary team of experts, including scientists, clinicians, website developers and marketing professionals, using an iterative approach to incorporate feedback from patient advocates and breast cancer clinicians. Initial design requirements for ASSESS included a simple and engaging user interface and being both public-facing and suitable for use in a clinic as a shared decision-making tool. It was designed to be responsive and support use on multiple devices, incorporating best practices for communicating risks, and offering a summary feature to save and print results.Cognitive interviews were conducted with patient advocates and breast cancer clinicians to inform the design of the tool, resulting in changes that made the interface and results output clearer and more accessible to a lay audience. Once finalized, additional usability testing was conducted with patient advocates and breast cancer medical oncologists to ensure the tool was deployed as intended. Komen hosts the ASSESS tool on komen.org and has developed supporting information to empower those newly diagnosed with breast cancer to make informed decisions by offering tailored treatment options, along with education on each treatment option and how treatment combinations may affect survival. Within the tool, links are provided to Komen-generated and expert-vetted information that describes the significance of each input requested (age at diagnosis, menopause status, molecular subtype, tumor size/grade, etc.). By integrating these resources, the tool empowers patients by providing sound and understandable information, enabling them to be actively involved in their treatment decisions. The ASSESS tool, publicly launching later this year, should help providers visually demonstrate different treatment option outcomes and provide links to accessible information that can be shared with patients for each treatment, including details on how it works, its side effects, and references to the associated expert clinical guidelines. Built using a patient-centered approach, the ASSESS tool supports shared decision-making for people with early-stage breast cancer, helping them make the best individual decisions for their treatment and care.
Presentation numberPS4-11-11
Germline Mutation Landscape in DCIS: Impact of Age and Family History in a Middle Eastern Cohort
Sarah Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
S. Abdel-Razeq, L. El Saket, F. Tamimi, H. Bani Hani, B. Sharaf, A. Al-Atary, M. El-Atrash, M. Horani, T. Al-Batsh, O. El Khatib, Y. Talab, Y. Al-Masri, M. Abunasser, H. Abdel-Razeq; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: Ductal carcinoma in situ (DCIS) represents a heterogeneous pre-invasive breast cancer entity, with genetic predisposition playing a potential role in disease development. This study aims to evaluate the frequency and spectrum of pathogenic germline variants among Jordanian patients with DCIS who underwent genetic testing. Methods: A retrospective review was conducted on clinical and genetic data on patients diagnosed with DCIS. Variables included age at diagnosis, hormonal receptor status (ER/PR/HER2), family history of cancer, and results of germline genetic testing. Variant classification was harmonized according to the American College of Medical Genetics and Genomics (ACMG) guidelines. Eligible patients, as per the National Comprehensive Cancer Network (NCCN) guidelines, underwent germline genetic testing (GGT) using an NGS-based multi-gene panel. Rates of GGT was stratified against known variables including age, family history and hormonal status. Results: Between July 2006 and September 2024, a total of 622 patients with DCIS were treated and followed at our institution. Germline genetic testing was performed on 271 (43.6%) patients. Except for one, all were female and median age (range) was 47 (20-90) years. Twenty patients (7.4%) were identified as harboring pathogenic or likely pathogenic (P/LP) variants. The most frequently mutated gene was BRCA2, detected in 9 (45.0%) patients, followed by CHEK2 (3 patients including one likely pathogenic), BRCA1 (2 patients), TP53 (2 patients), ATM (2 patients), and single cases involving NF1 and PALB2. Additionally, 4 (1.5%) patients carried variants classified as “Increased Risk Allele”; all were in the APC gene and were considered separately. A significant proportion of patients (n=94, 34.7%) had variants of uncertain significance (VUS), reflecting the challenges of interpretation in underrepresented populations. Patients with P/LP variants tended to be diagnosed about 5 years earlier (mean age: 43.7 years) compared to 48.9 years for those without. The prevalence was notably higher in patients aged ≤50 years (n=16, 9.5%) compared to those over 50 (n=4, 3.9%), though the difference did not reach statistical significance (p = 0.078). While there’s a clear numerical difference in rates among 229 patients with family history of cancer (n=18, 7.9%) versus (n=2, 4.8%) in 42 patients without, it didn’t reach statistical significance (p = 0.52), too. Hormone receptors (HR) positivity was high in both groups and did not significantly distinguish carriers from non-carriers. Conclusions: In this Arab DCIS cohort, the observed 7.4% prevalence of pathogenic germline variants supports routine genetic testing, particularly in patients with a family history or early-onset disease. However, the rate was significantly lower than the 11% we previously reported in over 6000 patients with invasive breast cancer. The predominance of BRCA2 mutation suggests a possible founder effect or population-specific enrichment, warranting further genomic studies. High rates of VUS further emphasize the need for regional variant annotation efforts. These findings advocate for integrating germline testing into the diagnostic workup of DCIS in similar populations to inform surveillance, risk-reduction strategies, and familial counseling.
Presentation numberPS4-11-12
Impact of the ESMO-MCBS v2.0 on Benefit-Risk Assessment of Emerging Therapies in Early Breast Cancer: A Comparison with the ASCO Value Framework
Vittoria Molinaro, University of Naples “Federico II”, Naples, Italy
A. Longobardi1, V. Molinaro1, V. Cantile1, R. Buonaiuto2, A. Caltavituro1, G. Crimaldi1, M. Giuliano1, G. Arpino1, C. De Angelis3; 1Oncology Unit, Department of Clinical Medicine and Surgery,, University of Naples “Federico II”, Naples, ITALY, 2Department of Breast and Thoracic Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy., Naples, ITALY, 3Clinical and Translational Oncology, Scuola Superiore Meridionale, Naples, ITALY.
Background: Balancing the clinical benefit and tolerability of adjuvant systemic therapies in patients with early breast cancer (eBC) remains challenging. Frameworks such as the American Society of Clinical Oncology Value Framework (ASCO VF) v2.0 and the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS v2.0) have been developed to help clinicians interpret trial results by weighing potential clinical benefit and toxicity. The European Society for Medical Oncology has recently revised ESMO-MCBS to version 2.0, which refines benefit thresholds and introduces explicit annotations for acute (AT) and persistent (PT) toxicity. This analysis applied these frameworks to evaluate the benefit-risk profiles of therapeutic strategies for high-risk eBC. Methods: Twelve phase II-III trials (KATHERINE, APHINITY, ExteNET, PEONY, NOAH, NeoSphere, KEYNOTE-522, OlympiA, CREATE-X, monarchE, NATALEE, and GIM2) were evaluated using ESMO-MCBS v2.0 and the ASCO Value Framework. Data were extracted from the latest available publications. Key endpoints included invasive disease-free survival (iDFS), overall survival (OS), pathological complete response (pCR), treatment-related adverse events (TRAEs), and quality of life (QoL). According to ESMO MBCS v2.0, standardized AT annotations for ≥30% G≥3 AEs, or ≥10% premature treatment discontinuation or hospitalization, and PT annotations for ≥20% persistent grade ≥3 toxicity, were retrieved . Concordance between ESMO MCBS and ASCO net health benefit (NHB) scores was assessed using Cohen’s Kappa coefficient. Results: The trials KATHERINE, NOAH, PEONY, KEYNOTE-522, CREATE-X, OlympiA, monarchE, and NATALEE achieved the highest ESMO-MCBS scores (Grade A) owing to improvements in iDFS and quality of life. In contrast, NeoSphere, APHINITY, and ExteNET were rated Grade C due to more modest absolute benefit. Notably, KATHERINE, KEYNOTE-522, OlympiA, CREATE-X, ExteNET, NATALEE, NOAH, and PEONY were annotated for acute toxicity (AT) under ESMO-MCBS v2.0, reflecting substantial rates of severe adverse events or treatment discontinuation. Among ASCO net health benefit (NHB) scores, NOAH (74), OlympiA (72), and CREATE-X (52) recorded the highest values, whereas NATALEE (12), ExteNET (5), and APHINITY (1) had the lowest scores. The APHINITY trial also showed the lowest toxicity score (-20), primarily due to the incidence of grade 3-4 diarrhoea (9.8% with pertuzumab vs. 3.7% with placebo). Overall, concordance between ESMO-MCBS v2.0 and ASCO VF was limited (Cohen’s Kappa -0.143; percentage agreement 33.3%). Conclusions: The revised ESMO-MCBS v2.0 provides a more rigorous assessment of clinical benefit by refining thresholds for absolute gains and systematically evaluating acute and persistent toxicities. NOAH and OlympiA achieved the most favourable benefit-risk balance across both tools. APHINITY and NATALEE recorded the lowest ASCO VF NHB scores, whereas APHINITY, NeoSphere and ExteNET were graded C under the ESMO-MCBS. These results emphasise differences in methodology and support the combined application of both frameworks to guide evidence-based choices in early breast cancer care.
Presentation numberPS4-11-13
Psychosocial determinants in patients with early breast cancer undergoing neoadjuvant therapy: initial results from a prospective study
Yael Bar, Tel Aviv Medical Center and The Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
Y. Bar1, N. Keren2, D. Eisenstadt2, Y. Hamama-Raz3, S. Lerner2, I. Zada2, I. Shiran1, Y. Leshem1, S. Strulov Shachar1, A. Sonnenblick1; 1Oncology Division, Tel Aviv Medical Center and The Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, ISRAEL, 2Oncology Division, Tel Aviv Medical Center, Tel Aviv-Yafo, ISRAEL, 3The Department of Social Work, Ariel University, Ariel, ISRAEL.
Background: The use of neoadjuvant therapy (NAT) for early-stage breast cancer has increased significantly in recent years. Emerging evidence suggests a link between psychological factors, such as anxiety and depression, and tumor behavior, potentially mediated by the immune microenvironment. While previous studies have demonstrated an association between emotional distress and treatment response in patients with metastatic cancer, data in the early breast cancer setting remain limited. We are conducting a prospective study to examine the relationships between psychosocial factors, immunological biomarkers, and response to NAT in patients with early breast cancer. Here, we present our preliminary findings on the psychosocial characteristics of this patient population. Methods: Patients with early-stage breast cancer who were referred for neoadjuvant chemotherapy with or without biological therapy at a single large academic institution were eligible for the study. Eligible NAT regimens included ddACT, TCHP, ddACTHP, and the KEYNOTE-522 protocol (TCAC + pembrolizumab). After signing informed consent, and prior to administration of the first treatment, blood samples were collected for immuno-proteomic analysis. On the same day, patients completed psychological questionnaires assessing anxiety (GAD-7), self-efficacy in coping with cancer (Cancer Behavior Inventory), social support (MSPSS), and socio-demographic characteristics. Clinical characteristics of the patients and their tumors were extracted from the electronic medical records. Results: At data cutoff, 40 patients had enrolled and completed pre-treatment questionnaires. Among these patients, 6 (15%), 9 (22.5%), 9 (22.5%), and 16 (40%) had estrogen receptor-positive/HER2-negative (ER+/HER2−), ER+/HER2+, ER−/HER2+, and triple-negative breast cancer, respectively. Median age was 46 (range: 25-77). Prior to treatment initiation, 11 patients (27.5%) reported moderate to high anxiety levels, while 29 (72.5%) reported low or no anxiety. No significant correlations were found between breast cancer subtype, tumor size, or nodal involvement and levels of anxiety, self-efficacy, or perceived social support. Women in a relationship reported significantly lower anxiety levels, higher self-efficacy and greater perceived social support compared to single women (p = .033, p = 0.018 and p = 0.017, respectively). Women with lower income levels or those who were unemployed reported significantly lower self-efficacy (p = .022 and p = .007, respectively). Among the 29 patients who completed neoadjuvant treatment and underwent surgery, no significant correlations were found between psychosocial factors and pathological complete response (pCR). However, both chemotherapy dose reduction (DR) and lower relative dose intensity (RDI), each significantly associated with lower pCR rates (p = .044 and p = .039, respectively), were also significantly correlated with lower perceived social support (p = .040 for DR and p = .021 for RDI) and lower self-efficacy (p = .025 for RDI). As expected, pCR rates were significantly associated with breast cancer subtype (p = .019). Conclusions: Moderate to high anxiety was reported by approximately one-quarter of patients with early breast cancer prior to starting NAT. Being in a relationship was associated with lower anxiety, higher self-efficacy, and greater perceived social support. Lower self-efficacy and perceived social support were significantly associated with reduced chemotherapy RDI, which in turn was associated to lower pCR rates. The study is ongoing, and additional data are needed to further explore the associations between psychosocial factors, immunological biomarkers, and response to NAT.
Presentation numberPS4-09-01
Listening to the Community: Barriers and Beliefs About Clinical Trials Among Black Women with Breast Cancer
Sabrina Mayhew, QLHC, San Francisco, MI
S. Mayhew1, S. Colen2, L. Patel2, S. Horton3, K. E. Jackson4, C. Allen5, O. Akanni6, J. LaCoursiere7, T. Heather8, E. Laura9; 1Access and Diversity, QLHC, San Francisco, MI, 2BreastCancerTrials.org, QLHC, San Francisco, MI, 3Clinical Operations, QLHC, San Francisco, MI, 4Founder and CEO, Sisters Network Inc, Houston, TX, 5Vice President, Sisters Network Inc, Houston, TX, 6Community Outreach, Sisters Network Inc, Houston, TX, 7Clinical Data, QLHC, San Francisco, MI, 8Patient Engagement, QLHC, San Francisco, MI, 9Breast Care Center, UCSF, San Francisco, CA.
Purpose African American breast cancer patients remain significantly underrepresented in clinical trials, despite experiencing more aggressive disease and disproportionately worse outcomes. While awareness of clinical trials has increased, there is still a gap between awareness and participation, especially due to limited access to personalized support and actionable next steps. BreastCancerTrials.org and Sisters Network® Inc. partnered to assess attitudes, knowledge, beliefs, and experiences related to clinical trials as part of a larger initiative to improve trial awareness and enrollment in African American women. Methods Sisters Network® Inc distributed a survey to over 100 African American breast cancer survivors affiliated with the organization to gather insights on their knowledge, experiences, and beliefs about clinical trials. As the largest national African American breast cancer survivorship organization with more than 30 chapters across the U.S., Sisters Network® Inc utilized its extensive network to reach a broad and diverse community of survivors. The survey asked about participants demographics, breast cancer details, and familiarity with clinical trials. It covered communication with healthcare providers, trial eligibility and experience, barriers to participation, challenges finding trial information, and interest in using clinical trial navigation for support. Results Preliminary findings show that more than 65% of respondents were familiar or somewhat familiar with clinical trials. When asked about participation, 44% said they would join a trial if their healthcare provider offered it as an option. Most participants lived in urban (51%) or suburban (46%) areas and were early-stage breast cancer survivors. Specifically, 14% were diagnosed at stage 0, 34% at stage 1, 46% at stages 2 or 3, and 3% had metastatic cancer. Regarding biomarker status, 34% reported being ER/PR positive, 21% had Triple Negative breast cancer, and 22% were unsure of their status. Thirty percent of respondents had discussed clinical trial participation with their healthcare team. Of those, 67% said the healthcare team initiated the conversation, while 16% reported bringing it up themselves. Key barriers to clinical trial participation included difficulty determining eligibility, challenges finding a trial, and limited understanding of how trials work. Despite these challenges, most respondents expressed interest in learning more. Over half indicated they would be interested in clinical trial navigation services, with 52% identified as having interest in receiving education and support from a clinical trial navigator. Conclusion This study further demonstrates the barriers that African American breast cancer patients experience, and the fact that they could benefit from culturally competent, personalized navigation. This survey was the first step in a broader effort to build trust and increase clinical trial knowledge, awareness, and participation among African American breast cancer patients through the development of customized, culturally responsive education programs and navigation support. BreastCancerTrials.org and Sisters Network® Inc. efforts to improve access and diversity in clinical trials will support broader initiatives within both organizations. Insights from this study will also help refine our enrollment strategies for the I-SPY clinical trials.
Presentation numberPS4-09-02
Mortality trends in Mexico: Epidemiological Analysis from 83 444 cases
Claudia H Arce-Salinas, Instituto Nacional de Cancerologia, CDMX, Mexico
J. L. Aguilar-Ponce1, M. Patiño-Gonzalez2, A. Nares-Ovando3, I. Molina-Mandujano4, P. S. Del Moral Villavicencio5, J. E. Perez Olguin6, M. R. Vargas Garcia7, P. A. Cabrera Galeana8, C. H. Arce-Salinas9; 1Comité de Moleculas Nuevas y Subcomite de Productos Biotecnologicos, IMSS Bienestar, CDMX, MEXICO, 2Direccion de Desarrollo e Integracion de Medicina Basada en la Evidencia, Secretaría de Salud, CDMX, MEXICO, 3Gestion de Indicadores de Servicios, Secretaría de Salud, CDMX, MEXICO, 4Evaluacion de Programas, Secretaría de Salud, CDMX, MEXICO, 5Desarrollo e Integracion de Medicina Basada en la Evidencia, Secretaria de Salud, CDMX, MEXICO, 6Modernizacion del Sector Salud, Secretaria de Salud, CDMX, MEXICO, 7Direccion General de Evaluaciòn del Desempeño, Secretaria de Salud, CDMX, MEXICO, 8Subdireccion de Oncologia y Hematologia, Instituto Nacional de Cancerologia, CDMX, MEXICO, 9Medical Oncology, Instituto Nacional de Cancerologia, CDMX, MEXICO.
Background: Breast cancer (BC) is the leading cause of death in women. Despite declining BC mortality rates worldwide, rates in Mexico continue to rise. The absence of nationwide epidemiological data hinders the implementation of effective mortality reduction measures. This analysis aims to characterize BC mortality patterns across Mexico.Patients and methods: This descriptive and retrospective analysis included mortality records (DATA_CAMAMA_2012-2023.csv) from all 32 Mexican states between 2012 and 2023. Analyzed variables included gender, age at death, state of residence, and cause of death. Cases with inconsistent or duplicate birth dates, or causes of death other than BC, were excluded. Statistical analysis included the Kolmogorov-Smirnov test for normality (p<0.05). Data are presented as median and interquartile range. Gender comparisons were performed using the Mann-Whitney U test (p<0.001). State-level analyses were conducted using the Kruskal-Wallis test with post-hoc Dunn and Bonferroni correction for multiple comparisons. A predictive model was constructed using Ln (Yt) = α + βt + εt, with validation R² = 0.9989, p < 0.005. Analysis was done with Artificial Inteligence Claude Sonnet 4.Results: A total of 83,444 cases were registered; the median age was 59 (49-70), and 99.32% were women. From 2012 to 2023, the mortality rate increased annually, from 16.2 to 20.6 per 100,000. Colima and Sonora had the highest mortality rates, 1.54 and 1.43 times the national rate, respectively. The north, northeast, northwest, and middle-west regions exhibited the highest mortality rates, with significant impact. The projected trend indicates a 22.7% increase in the estimation rate by 2030, corresponding to 10,875 new deaths.Conclusions: This is the first nationwide analysis of BC mortality in Mexico. The mortality rate is steadily increasing, the opposity to mortality trends worldwide. Measures focused on early diagnosis and prompt treatment are imperative to reduce mortality. The impact that this results had on the Public Health Policies should be taken into account, like the National Cancer Plan and the National Health Plan.
Presentation numberPS4-09-03
Impact of comorbidities and race in breast cancer survival across cancer stages
Matthew Sanborn, Lankenau Medical Center, Wynnewood, PA
M. Sanborn1, M. Koury1, A. Mittapalli1, M. Qazi1, A. Maity2, S. Kjelstrom3, A. Ghaneie2; 1Internal Medicine, Lankenau Medical Center, Wynnewood, PA, 2Hematology and Oncology, Lankenau Medical Center, Wynnewood, PA, 3Lankenau Institute for Medical Research, Lankenau Medical Center, Wynnewood, PA.
Title: Impact of comorbidities and race in breast cancer survival across cancer stages Background: Breast cancer survival varies by race, influenced by factors like socioeconomic status, access to care, and comorbid conditions. While comorbidities independently worsen survival, their role in shaping racial differences across cancer stages is less clear. Emerging data suggest that racial disparities in survival may be masked when comorbidities are present and only become apparent among patients without other medical illnesses. Understanding how comorbidities and race interact, and whether removing the comorbidity burden reveals racial disparities, is crucial for addressing inequities and informing next steps, including evaluating the impact of socioeconomic factors on these disparities. Methods: We conducted a retrospective cohort study of breast cancer patients diagnosed between 2018 and 2023 using the N-Power Medicine database of cancer patients in the United States. We grouped patients as White/Asian (W/A) or Black/Other (B/O), combining Black, Native American, and Pacific Islander patients into the latter category, and compared their baseline demographics and comorbidity profiles using standard statistical methods. We used Cox regression and random forest models to analyze factors associated with all-cause mortality (ACM). Comorbidities analyzed included cardiovascular disease (CVD), diabetes (DM), chronic obstructive pulmonary disease (COPD), congestive heart failure (CHF), peripheral vascular disease (PVD), and chronic kidney disease (CKD). Additional analyses were performed, stratifying patients by cancer stage and the presence or absence of comorbidities, to evaluate racial differences in survival outcomes. Results: Among 31,150 patients (W/A = 26,136; B/O = 5,014), the median age was 61 and 62 years, respectively. B/O patients demonstrated higher ACM when controlling for comorbidities (HR 1.3 (1.2-1.4)). All comorbidities increased ACM in both racial groups as follows: CVD (HR 2.5 (1.6, 3.9)), DM (HR 1.6 (1.2, 2.0)), COPD (HR 1.5 (1.0, 2.1)), CHF (HR 2.5 (1.8, 3.7)), PVD (HR 2.3 (1.6, 2.4)), CKD (HR 2.6 (2.0, 3.3)). The most significant comorbidities for predicting ACM were CKD and CHF. We did not identify a significant interaction between comorbidities and race, specifically: CVD (p = 0.665), DM (p = 0.665), COPD (p = 0.06), CHF (p = 0.06), PVD (p = 0.373), and CKD (p = 0.844). Stage-stratified analysis revealed significant racial disparities in survival only among patients without comorbidities, with B/O patients exhibiting higher mortality compared to W/A patients across stages I-IV (HR range 1.2-1.6). This racial difference was notably absent among patients with comorbidities. Conclusion: Among breast cancer patients, comorbidities and race each independently increased ACM. However, racial differences in survival emerged only among patients without comorbidities, suggesting that the comorbidity burden may mask underlying racial disparities across cancer stages. These findings suggest that racial differences in breast cancer outcomes remain relevant and warrant further investigation, even when comorbidities are present and can obscure these disparities. Future work will explore socioeconomic factors, treatment patterns, and the cumulative impact of multiple comorbidities to clarify the drivers of these disparities and inform targeted interventions.
Presentation numberPS4-09-04
Global disparities in breast cancer clinical trials and their impact on clinical evidence, driven by underrepresentation of low- and middle-income countries.
Varsha Gupta, Roswell Park Comprehensive Cancer Center, Buffalo, NY
V. Gupta1, V. Singh1, M. Alharbi1, N. Sharma2, L. Alessi3, A. Roy4, S. Gandhi5, S. Kabraji1; 1Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Medical Oncology, VA Southern Nevada Healthcare System, Las Vegas, NV, 3Medical Library, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 4Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 5Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA.
Background: Breast Cancer (BC) remains a major health challenge globally and with higher mortality in low/medium- income countries (LMIC). Generalizability of evidence from clinical trial (CT) depends on many factors, one of which is selection and recruitment of representative population. Most CT sites are in high-income countries (HICs) which creates unfair generalization as there are unique challenges for patients in LMIC including disease presentation, genetics, comorbidity, lifestyle, resources etc. We evaluated disparity in global distribution of BC CTs and contribution to clinical guidelines based on country’s economic status and geography. Methods: We searched ClinicalTrials.gov (NCT) to identify BC CTs between 2000-23. Search strategy was developed by a medical librarian. World Bank country’s income level classification 2022-23 was used to classify the countries into HIC, LMIC and 7 geographic regions. CTs with at least 1 site in LMIC were qualified as LMIC CT. Data regarding CT phase, funding, design, sites were extracted. Contribution to medical evidence was quantified based on reference of CT in BC-NCCN guideline. Pearson chi-square tests used for comparison of categorical data. Results: We identified 5518 eligible BC CTs in the database. 1,630 (29.5%) trials had at least one site in LMICs compared to 4505 (81.6%) trials in HICs. Only 19.7% (86.0% in HIC) of phase 1, 27.2% (82.3% in HIC) of phase 2, 50.7% (73.4% in HIC) phase 3 had a site in LMIC countries (p < 0.01). Industrial funding was the most common source of funding for LMIC (55.4% vs 35.2% for HIC) compared to academic funding for HIC (56.6 % vs 19.8 % for LMIC, p < 0.01). 906 (55.6%) of LMICs CTs were randomized allocation compared to 1664 (36.9%, p < 0.01) for HIC. Proportion of CTs in LMIC increased from 14.1% in 2000 to 43.2% in 2023 with most significant increase in East Asia Pacific (EAP) region from 2.4% in 2000 to 39.1% in 2023, whereas it remains almost constant for South Asia (SA) (7.1% to 6.4%), Sub Saharan Africa (AFR) (2.4% to 3.2%), Latin America (LA) (8.2% to 8.9%). Proportion of CTs in North American (NA) sites decreased from 70.6% to 49.6% and were driven by increasing number of EAP CTs (4% vs 60.3% of non-NA CTs).173 CTs were referenced in BC-NCCN guidelines, 148 were registered in NCT. 92/148 were done after 2000. 64 (69.5%) included LMIC site compared 86 (93.5%, p < 0.01) for HIC. Only 43 (46.7%) in SA, 46 (50%) EAP, 15 (16.3%) in AFR, 57 (62%) LA, compared to 77 (83.7%) in Europe/Central Asian and 71 (77%) in NA. 78 (84.8%) were phase 3, 83 (90.2%) were randomized, 54 (58.7%) were industry sponsored. Conclusions: LMIC continues to be markedly underrepresented in BC CTs leading to inequity/disparity in medical evidence. Studies in the future should aim to ensure fair representation of the LMIC, so that we have improved/reliable evidence for the use of guideline-based treatment in these populations.
Presentation numberPS4-09-06
Retrospective evaluation of neutropenia and Duffy-Null Associated Neutrophil Counts (DANC) among breast cancer patients receiving chemotherapy
Bhawneet Chadha, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
B. Chadha1, Y. Xu2, R. Hadidchi3, T. Duong3, X. Xue2, D. Levitz1, D. Makower1; 1Hematology/Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, 2Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, 3Radiology, Albert Einstein College of Medicine, Bronx, NY.
Background: Duffy-Null Associated Neutrophil Count (DANC) is a non-pathologic state characterized by a low absolute neutrophil count (ANC) without increased risk of infections. Approximately 60% of people identifying as Black are estimated to have the Duffy-null phenotype, and Duffy-null phenotype is also seen in people of Middle Eastern descent and Hispanic ethnicity. Cancer patients (pts) with DANC have often been excluded from clinical trials due to low ANC and may receive inappropriate dose delays or reductions of standard anticancer therapy. Testing for Duffy-null phenotype is readily available, but no standard of care exists for management of chemotherapy dosing in breast cancer (BC) pts with DANC. There is little data assessing the impact of low baseline ANC and Duffy-null phenotype on chemotherapy dose delays and dose reductions in triple-negative breast cancer (TNBC) pts. We conducted a retrospective study evaluating baseline ANC, chemotherapy dose delays and reductions, and use of Duffy antigen testing, in pts with TNBC treated at our institution, an urban academic medical center. Methods: Pts with Stage I to III TNBC treated from 2016 to 2021 were identified from the Montefiore Einstein Comprehensive Cancer Center (MECCC) tumor registry. Baseline ANC levels were correlated with dose reduction, delay or discontinuation of chemotherapy using logistic regression models, while adjusting for stage, chemotherapy regimen, baseline white blood count cell and absolute neutrophil count, as well as race and ethnicity. Results: 159 pts [77 (48%) Black, 69 (43%) Hispanic] met inclusion criteria. Median age was 59 yrs. 50 pts (31.4%) received chemotherapy in the neoadjuvant setting, 80 pts (50.3%) in the adjuvant setting, and 29 pts (18.2%) in both. 94 pts (59.1%) received anthracycline and taxane based chemotherapy; 41 pts (25.8%) received taxane without anthracycline; 24 pts (15.1%) received neither agent. Median baseline ANC was lower in Black pts than in Hispanic or Non-Hispanic white (NHW) pts (p=0.034), but no racial or ethnic differences in dose delays, dose reductions, early chemotherapy discontinuation, or neutropenic fever were seen. Low baseline ANC (defined as baseline ANC below the median level for each race/ethnicity group) was associated with higher likelihood of chemotherapy dose delay for neutropenia compared to high baseline ANC (38% vs 16%, p=0.002), and with trend to higher likelihood of dose reduction for neutropenia (18% vs 7.4%, p = 0.056). Within the low ANC group, those with dose delays had a lower baseline. ANC value as compared with those that did not have dose delays (1.7 vs 2.7, p<.001), but no correlations between baseline ANC and neutropenic fever was seen (ANC 2.8 vs 2.6, p=0.9). Three pts in our cohort had been told of low ANC prior to BC diagnosis: one underwent early discontinuation of chemotherapy due to neutropenia, and two experienced dose delays and dose reductions. No pt underwent formal testing for Duffy-null phenotype. Conclusion: In our diverse cohort of TNBC BC pts, Black pts had lower baseline ANC than Hispanic or NHW pts, but did not have more chemotherapy dose delays or reductions for neutropenia, and did not have higher risk of neutropenic fever. Lower baseline ANC, regardless of race or ethnicity, was associated with greater likelihood of chemotherapy dose delays and reductions for neutropenia, but not with higher risk of neutropenic fever. Testing for Duffy-null phenotype in pts with low ANC was not utilized. Duffy antigen testing of TNBC pts with low ANC may reduce unwarranted chemotherapy dose reductions or delays. Prospective evaluation of the utility of Duffy antigen testing in BC pts is warranted, and testing should not be limited to Black patients. Guidelines for management of chemotherapy dosing in BC pts with DANC should be developed.
Presentation numberPS4-09-07
Insurance Status as a significant contributor of disparities in outcomes among young breast cancer patients- A retrospective analysis from National Inpatient Sample
Vaishali Deenadayalan, Roswell Park Comprehensive Cancer Center, Buffalo, NY
V. Deenadayalan1, V. Muthusamy Kumarasamy1, L. Calvin Yee Fen2, S. Gandhi3; 1Department of Hematology and Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 2Department of Internal Medicine, Catholic Health System/ University at Buffalo, Buffalo, NY, 3Department of Hematology and Medical Oncology/ Breast Oncology, Emory Winship Cancer Institute, Atlanta, GA.
Background:Young women with breast cancer often experience more aggressive disease and worse outcomes. Beyond biological factors, we aimed to evaluate whether insurance status also contributes to disparities in clinical outcomes. This study compares clinical characteristics and outcomes of young breast cancer patients with private insurance versus Medicaid. Methods:We conducted a retrospective cohort analysis using National Inpatient Sample 2016-2020. Young female patients (age 18 – 50) with breast cancer were stratified by insurance status: Private insurance and Medicaid. Baseline characteristics, comorbidities, hospital features, and primary and secondary outcomes were compared. Multivariate logistic regression was used to derive adjusted odds ratios (aOR), while adjusting for age, race, Charlson Comorbidity Index, hospital region, teaching status of hospital, hospital bedsize, urban versus rural location, and ZIP code-based income quartiles. Results:A total of 144,080 young breast cancer patients were included in the study, of whom 81,025 (66.4%) had private insurance and 40,930 (33.6%) were insured through Medicaid.Patients with Medicaid were younger (mean age 41 vs 42.17 years, P<0.001), more likely to be African American (30.1% vs 16.1%) or Hispanic (22.9% vs 11.3%), and from lower-income ZIP codes (P<0.001). They had higher rates of comorbidities including anemia (37.2% vs 30.6%), diabetes (11.6% vs 6.8%), and smoking (31.7% vs 20.2%) (P<0.001 for all) Despite these disparities, there was no significant difference in in-hospital mortality between Medicaid and privately insured patients (4.33% [n = 1,770] vs 3.31% [n = 2,680]; aOR 1.004, P = 0.955). However, Medicaid patients had longer hospital stays (4.92 vs 3.74 days; aOR 1.21, P<0.001) and lower total hospitalization charges ($64,150 vs $71,780; aOR 0.87, P<0.001). Medicaid coverage was independently associated with higher risks of sepsis (aOR 1.15, P=0.041), acute kidney injury (AKI) (aOR 1.26, P<0.001), acute respiratory failure (ARF) (aOR 1.17, P=0.026) and pulmonary embolism (aOR 1.33, P=0.024) (Table 1). Conclusions:Although in-hospital mortality was similar between Medicaid and privately insured young breast cancer patients, those with Medicaid experienced significantly more complications, longer hospital stays, and came from more socioeconomically disadvantaged backgrounds. The paradox of lower hospitalization charges despite longer hospital stay and increased complications may reflect systemic inequities in care delivery and resource allocation. These findings underscore the need for targeted interventions and equitable access to mitigate disparities.
| Primary Outcomes | Private insurance | Medicaid | Adjusted Odds Ratio | P Value |
| Mortality | 3.31 | 4.33 | 1.004 | 0.955 |
| Length of Stay | 3.74 | 4.92 | 1.21 | <0.001 |
| Total Hospitalization Charges in $ | 71,780 | 64,150 | 0.87 | <0.001 |
| Secondary Outcomes | ||||
| Sepsis | 4.65 | 6.36 | 1.15 | 0.041 |
| Intubation | 1.61 | 2.52 | 1.23 | 0.073 |
| Pressor | 0.83 | 0.87 | 0.92 | 0.645 |
| Acute Kidney Injury | 4.81 | 7.27 | 1.26 | <0.001 |
| Acute Respiratory Failure | 4.20 | 5.96 | 1.17 | 0.026 |
| Pulmonary Embolism | 1.19 | 1.87 | 1.33 | 0.024 |
| Blood Transfusion | 6.47 | 7.96 | 1.09 | 0.137 |
Presentation numberPS4-09-08
Expanding Diversity through Innovation: Development of the I-SPY ACCESS Initiative for the I-SPY2.2 and DCIS RECAST Clinical Trials
Dame A. Idossa, Mayo Clinic (Rochester), Rochester, TX
S. L. Horton1, D. A. Idossa2, K. C. Hewitt3, J. Perlmutter4, L. Van’t Veer5, N. Hylton6, A. Borowsky7, F. Symmans8, D. Yee9, C. Yau10, P. Pohlmann11, J. Boughey12, L. Esserman10; 1Access and Diversity, Quantum Leap Healthcare Collaborative, San Francisco, CA, 2Oncology, Mayo Clinic (Rochester), Rochester, TX, 3Surgery, Huntsman Cancer Institute at University of Utah, Salt Lake City, UT, 4Cancer Advocacy, Gemini Group, Ann Arbor, MI, 5Applied Genomics, University of California San Francisco, San Francisco, CA, 6Radiology, University of California San Francisco, San Francisco, CA, 7Pathology, University of California, Davis, Sacramento, CA, 8Pathology, University of Texas MD Anderson, Houston, TX, 9Dept. of Medicine, University of Minnesota, Minneapolis, MN, 10Dept. of Surgery, University of California San Francisco, San Francisco, CA, 11Div. of Cancer Medicine, University of Texas MD Anderson, Houston, TX, 12Dept. of Surgery, Mayo Clinic (Rochester), Rochester, MN.
IntroductionCancer clinical trials have historically faced challenges in achieving fully representative trial population, particularly in terms of including patients from diverse ethnic and racial groups, those with low socioeconomic status, and those living in rural regions. This has contributed to inequities in access to novel therapeutic options. BackgroundThe I-SPY adaptive platform trials have prioritized diversity since inception, consistently enrolling in >10% of trial patients being Black and/or Hispanic individuals. The majority of participating institutions in the I-SPY 2.2 study are academic or large well-resourced health systems. In 2023, the I-SPY ACCESS (Achieving Cancer Clinical Trial Equity in Socioeconomically Diverse Sites) Initiative was launched to enhance representation by engaging and involving nontraditional trial sites- such as safety-net hospitals, community cancer centers, and rural cancer centers- that serve historically excluded patient populations. Methods The I-SPY ACCESS Initiative offers customized and personalized tailored support to under-resourced sites. This includes comprehensive feasibility and readiness assessments, onboarding guidance, enhanced site-specific training, centralized institutional review board (IRB) navigation, site peer mentorship, and educational materials for both patient and provider. Additionally, patients gain access to I-SPY advocate peer trial navigators. Sites are provided culturally responsive patient education tools- including multilingual brochures, a dedicated patient website, and content co-developed with community partners. A Clinical Trial Navigator supports both patients and sites, helping to locate logistical, social, and trial specific resources.Tools such as virtual Clinical Research Coordinators (vCRC’s) and OneSource technology are deployed strategically based on assessed site need and resource availability. OneSource is an innovative platform that enables direct extraction of clinical data from electronic health records into trial databases, reducing site workload and improving data quality. Funding models are currently being explored to extend OneSource and vCRC services across more ACCESS sites. In the future, there are plans to pilot financial navigation services and integrative care resources for all patients enrolled in I-SPY with particular focus on those enrolled at ACCESS sites. Results and ProgressAs of June 2025, seven sites have been assessed as ACCESS sites for I-SPY 2.2, with two sites having successfully completed activation and currently enrolling participants. Beginning in mid-2024, one DCIS RECAST ACCESS site has been activated and 11 additional sites are in the onboarding process. The modest pace of I-SPY 2.2 activation reflects the complexity and operational demands associated with resource-intensive platform trial. In contrast, the DCIS RECAST protocol was designed with a lower burden, facilitating easier implementation by under-resourced sites, reflected by increased interest in the study among ACCESS sites. The ACCESS framework is being explored for potential future application in the Pre I-SPY Phase 1 platform trial, particularly considering the historical underrepresentation of such sites in the early-phase clinical research. Conclusion and Future GoalsThe I-SPY ACCESS initiative demonstrates that investing in thoughtful, innovative, and effective means for trial delivery, as well as incorporating community-based support, can strengthen representation in complex, adaptive breast cancer trials. The initiative offers a flexible, equity-driven framework that is impactful now and scalable for future innovation in oncology research.
Presentation numberPS4-09-09
Distinct Immune and Metabolic profiles in Latin American breast cancer patients with obesity enrolled in FLEX
Marcela Mazo Canola, Mays Cancer Center, UT Health San Antonio, MD Anderson Cancer Center, San Antonio, TX
M. Mazo Canola1, A. Santillan2, J. Alberty-Oller3, E. Dias4, V. Kaklamani5, A. Elkhanany6, K. Hoskins7, M. Habibi8, R. Hampton9, J. L. Barone10, N. M. Johnson11, N. Gordon12, N. Sookhan13, T. Bah14, P. John15, E. Aponte16, S. Diab17, V. Klimberg18, N. Stivers19, C. Page19, S. Uygun20, W. Audeh19, J. O’Shaughnessy21; 1Breast Medical Oncology, Mays Cancer Center, UT Health San Antonio, MD Anderson Cancer Center, San Antonio, TX, 2Medical Oncology, Texas Breast Specialists-San Antonio Medical Center, San Antonio, TX, 3Breast Medical Oncology, Kings County Hospital Center, Brooklyn, NY, 4Surgical Oncology, Tennessee Oncology, Nashville, TN, 5Breast Medical Oncology, UT Health San Antonio, Mays MD Anderson Cancer Center, San Antonio, TX, 6Medical Oncology, Lester & Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 7Medical Oncology, University of Illinois College of Medicine Chicago, Chicago, IL, 8Medical Oncology, Northwell Health Florina Rusi Marke Comprehensive Breast Center, Staten Island, NY, 9Medical Oncology, Luminis Health Breast Center, Lanham, MD, 10Medical Oncology, Mission Hope Cancer Center, Santa Maria, CA, 11Surgical Oncology, Legacy Health, Portland, OR, 12Surgical Oncology, AIS Cancer Center, Adventist Health Bakersfield, Bakersfield, CA, 13Breast Medical Oncology, Trinity Health, Waterbury, CT, 14Medical Oncology, MetroHealth Medical Center, Cleveland, OH, 15Medical Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX, 16Breast Medical Oncology, Texas Oncology – San Antonio Medical Center, San Antonio, TX, 17Medical Oncology, Intermountain Health Good Samaritan Hospital, Aurora, CO, 18Surgical Oncology, UTMB Health, University fo Texas Medical Branch, League City, TX, 19Medical Affairs, Agendia, Inc., Irvine, CA, 20Bioinformatics, Agendia, Inc., Irvine, CA, 21Medical Oncology, Baylor University Medical Center, Texas Oncology, Dallas, TX and Sarah Cannon Research Institute, San Antonio, TX.
Background: Latin American (LA) women are more likely to be diagnosed with aggressive early-stage breast cancer (EBC) compared to Non-Hispanic White (NHW) women, yet population-specific tumor biology remains underexplored. Previously, we reported elevated immune gene expression in BluePrint® (BP) Luminal B tumors in LA patients (pts) with EBC and obesity compared to Black and NHW cohorts. Here, we present updated clinical comparisons between LA, NHW and Black pts with EBC and whole transcriptome analysis (WTA) between BP Luminal B and Basal BC in pts with obesity. Methods: Clinical and WT data were analyzed from 15,577 EBC pts self-identified as LA, Black, or NHW enrolled in FLEX (NCT03053193), a prospective observational study with MammaPrint® risk of recurrence and BP molecular subtyping assays, and WT profiles. ImPrint, 53-gene immune signature, results (+/-) were generated for pts with hormone receptor positive (HR+) EBC. Chi-square and t-tests were conducted on clinical groups using arsenal R package. For WT comparisons, 215 obese LA pts with BP Luminal B and 77 with BP Basal EBC were matched to corresponding NHW and Black pts by age, T and N status. Differentially expressed genes (DEGs) were evaluated using limma, and pathway enrichment was performed using gene set enrichment analysis with Hallmark gene sets. Significant results were reported with adjusted p < 0.05. Results: Compared to Black and NHW pts, LA pts were younger and more often premenopausal (Table). Both LA and Black pts had statistically significantly higher rates of obesity compared to NHW pts. Significantly higher rates of MP High Risk 2, BP Basal and ImPrint+ tumors were observed in LA and Black pts vs NHW. WT comparisons among the obese subpopulations revealed that metabolic pathways, including adipogenesis, angiogenesis, epithelial-mesenchymal transition, and oxidative phosphorylation were significantly downregulated in EBC in LA pts vs NHW and Black cohorts. Conversely, immune-related pathways such as allograft rejection and interferon gamma response were enriched among LA pts with Basal cancers compared to NHW and Black groups. These coordinated pathway alterations suggest unique metabolic and immune profiles in LA EBC subtypes with a median absolute fold change of 1.3 among DEGs. Conclusion: The data suggest that signals from metabolic pathway alterations from modest immune-related gene upregulation may contribute to more aggressive tumor behavior in obese LA pts with EBC. Clinical and transcriptomic analyses demonstrated differences in Luminal B and Basal EBC biology among self-identified LA pts compared to NHW and Black cohorts, consistent with prior research. With further research, these findings may offer treatment targets which may enhance outcomes and reflect the importance of including racially and ethnically diverse pts in clinical trials to characterize population-specific differences in EBC outcomes.
| – | LA (n=1,446) | Black (n=1,656) | NHW (n=12,475) | p value Black vs LA | p value NHW vs LA |
| Age | |||||
| Median | 57 | 59 | 63 | < 0.001 | < 0.001 |
| Mean (Std Dev) | 56.6 (12.3) | 58.4 (12.6) | 61.5 (12.16) | ||
| Menopausal status | < 0.001 | < 0.001 | |||
| Pre-/Peri- | 434 (31.8%) | 390 (25.6%) | 2338 (20.0%) | ||
| Post- | 931 (68.2%) | 1133 (74.4%) | 9375 (80.0%) | ||
| BMI | < 0.001 | < 0.001 | |||
| Underweight (<18.5) | 17 (1.2%) | 13 (0.8%) | 199 (1.6%) | ||
| Normal (18.5-24.9) | 229 (16.2%) | 184 (11.6%) | 3141 (26.0%) | ||
| Overweight (25-29.9) | 451 (31.9%) | 406 (25.6%) | 3665 (30.4%) | ||
| Obese (≥30) | 718 (50.7%) | 984 (62.0%) | 5058 (41.9%) | ||
| MammaPrint | < 0.001 | < 0.001 | |||
| UltraLow Risk | 225 (15.6%) | 151 (9.1%) | 1962 (15.7%) | ||
| Low Risk | 465 (32.2%) | 410 (24.8%) | 4618 (37.0%) | ||
| High Risk 1 | 464 (32.1%) | 653 (39.4%) | 4351 (34.9%) | ||
| High Risk 2 | 292 (20.2%) | 442 (26.7%) | 1543 (12.4%) | ||
| BluePrint | < 0.001 | < 0.001 | |||
| Luminal A | 667 (47.3%) | 545 (33.9%) | 6307 (52.6%) | ||
| Luminal B | 506 (35.9%) | 694 (43.1%) | 4344 (36.2%) | ||
| HER2 | 65 (4.6%) | 74 (4.6%) | 411 (3.4%) | ||
| Basal | 173 (12.3%) | 297 (18.4%) | 925 (7.7%) | ||
| ImPrint (HR+ subgroup with full genome available; n=12,040) | 0.049 | < 0.001 | |||
| ImPrint- | 870 (91.0%) | 1066 (88.4%) | 9418 (95.3%) | ||
| Imprint+ | 86 (9.0%) | 140 (11.6%) | 460 (4.7%) |
Presentation numberPS4-09-10
Geographic Barriers are Key Predictors of Clinical Trial Enrollment in Breast Cancer: A National Cancer Database Analysis
Ana Sandoval-Leon, Miami Cancer Institute, Miami, FL
A. Sandoval-Leon1, Y. Chamorro1, M. Roy1, M. Rubens1, N. Dempsey1, L. Carcas1, M. Ahluwalia1, L. Dumeny2, R. Mahtani1; 1Medical Oncology, Miami Cancer Institute, Miami, FL, 2Family Medicine, Florida International University, Miami, FL.
Background: Clinical trials are essential to improving cancer outcomes, yet racial and ethnic minorities remain underrepresented. Structural and social barriers such as limited access to care, geographic distance, financial burden, lack of information, and cultural mistrust contribute to this persistent underrepresentation. Despite longstanding initiatives to improve inclusivity, recent data show most clinical trial participants continue to be White (W). An understanding of the factors that influence trial enrollment, including both sociodemographic and geographic determinants, is essential to designing more equitable and representative research. In this study, we used the National Cancer Database (NCDB) to evaluate disparities in clinical trial enrollment among patients (pts) with breast cancer (BC), with a particular focus on the impact of race, income, education, and geographic access. Method: We conducted a retrospective cohort study using data from the NCDB including pts with complete data on race/ethnicity, clinical trial enrollment status, clinical stage, sociodemographic variables, and survival time. Race/ethnicity in the NCDB may be based on self-report, administrative data, or observation. Education and income quartiles were assigned based on ZIP code-level U.S. Census data, per NCDB methodology. Descriptive statistics were used to compare enrollment rates across racial and ethnic groups. Multivariable logistic regression was performed to estimate adjusted odds ratios (AORs) for clinical trial enrollment, with race/ethnicity as the primary predictor and adjustment for age, stage, grade, Charlson-Deyo comorbidity index, income, education level, facility type, and geographic location. A p-value of < 0.05 was considered statistically significant. All analyses were conducted using SAS software version 9.4. Results: A total of 1,798,690 BC pts were included in the analysis. The median age was 62 years. The cohort was 79.7% W, 10.9% Black, 4.9% Hispanic, and 4.6% other races/ethnicities. The majority of pts resided in metro areas (84.7%), followed by urban (13.3%) and rural (2.0%). Only 1,645 (<1%) were documented as enrolled in clinical trials. In multivariable logistic regression, rural residence was independently associated with lower clinical trial enrollment (AOR= 0.381; 95% CI: 0.24-0.60; p 50 miles to the treating facility (AOR= 0.44-0.52; p< 0.0001). Treatment at non-academic center was associated with a higher enrollment compared to academic centers (AOR 1.438, 95% CI 1.15-1.80; p<0.0012). After adjustment for clinical and sociodemographic variables, race/ethnicity, income and education level were not significant predictors of clinical trial participation. Conclusion: In this large national cohort of BC pts, clinical trial enrollment was strikingly low, with fewer than 1% of pts documented as participants. Rural residence and long travel distances to treatment facilities were significantly associated with lower enrollment, underscoring the critical role of geographic and structural barriers in accessing research opportunities. Unexpectedly, patients treated at non-academic centers had higher odds of trial enrollment, a finding that may reflect variation in reporting practices rather than true differences in access. Race, income, and education were not independent predictors after adjustment, suggesting access may be the primary driver of disparities. A key limitation is the NCDB’s underreporting of trial participation. The very low number of patients identified as enrolled highlights the urgent need for improved and standardized reporting mechanisms. Enhanced data accuracy is essential to guide equity-focused interventions and ensure representative participation in cancer research.
Presentation numberPS4-09-11
Associations of patient-reported social determinants of health (SDOH) with treatment deferral and clinic no-show rates in a diverse population of patients with breast cancer
Emily L Podany, Washington University in St. Louis, St. Louis, MO
E. L. Podany1, B. Bowe2, J. Hesse1, D. Cicka1, S. Addison1, A. Golden1, N. Katakam1, F. Fa’ak1, K. Weilbaecher1, A. A. Davis1; 1Internal Medicine, Washington University in St. Louis, St. Louis, MO, 2Surgery, Washington University in St. Louis, St. Louis, MO.
Background: SDOH are known to affect treatment and survival in breast cancer. However, most studies rely on census- or neighborhood-level data to determine SDOH risk. A gap in the research is how patient-level SDOH impact treatment. Here, we used patient-reported SDOH to describe associations with treatment delays and no-show rates in breast cancer clinics with diverse populations. Methods: We implemented a self-administered, 19 question, validated, electronic medical record (EMR)-integrated SDOH screener in North County Siteman Cancer Center breast cancer clinics after IRB approval. Evaluated SDOH risks included financial resource strain, food insecurity, housing instability, transportation availability, and depression. We assessed SDOH risk associations with infusional treatment deferrals, number of no-shows and cancellations of clinic and treatment visits, and emergency room (ER) visits within 12 months. We performed a regression adjusting for demographics to determine risk factors for infusional delays. Area deprivation index (ADI), a validated measure based on 9-digit zip code, was used to define neighborhood deprivation quartiles to compare the lowest quartile (<64) and highest quartile (≥90), with higher numbers indicating increased poverty. Results: 547 patients with breast cancer had at least one response to an SDOH evaluation from July 2023 to June 2025. 267 (48.8%) self-identified as Black, 280 self-identified as White (51.2%), and 81 (14.8%) had stage IV disease. Black patients were significantly more likely than White patients to be at high risk of food insecurity (p<0.001), housing instability (p<0.001), transportation difficulty (p=0.008), a higher stage (p=0.001), and be insured on Medicaid (p<0.001). Patients at high risk for financial resource strain, housing instability, transportation difficulty, or food insecurity had significantly more no-shows and cancelled appointments than those at low risk (p<0.05). There was a significant increase in no-shows and ER visits for patients with Medicaid compared to Medicare and private insurance (p<0.001). Of the 356 patients who received infusional therapy, patients with Medicaid were significantly more likely than patients on Medicare or private insurance to have a treatment deferral (p=0.02) and a higher number of deferrals (p=0.01). Patients with a high ADI had an increased number of deferrals (p=0.003) and a larger total deferral duration (p=0.005). Patients with high risk of financial resource strain, housing instability, food insecurity, or depression were significantly more likely to have a deferral in a treatment cycle (p<0.05). Patients with more than one SDOH risk factor were significantly more likely to have a deferral (p=0.002). On regression analysis, patients had a higher risk of infusional therapy deferrals if they had financial resource strain (Odds ratio [OR] 5.51, 95% confidence interval [CI] 1.20-25.40, p=0.03) and housing instability (OR 4.00, 95% CI 1.09 – 14.70, p=0.04). Discussion: In this patient-level, prospective study using granular SDOH questions, we found significantly higher no-show and cancellation rates in patients with financial resource strain, housing instability, transportation difficulty, food insecurity, and Medicaid insurance. There were significantly higher infusional therapy deferral rates in patients with financial resource strain, housing instability, food insecurity, depression, Medicaid, and in higher ADI neighborhoods. Our findings show that individual screening of SDOH with real-time EMR integration in patients with breast cancer is feasible and represents an opportunity for targeted interventions to improve patient outcomes.
Presentation numberPS4-09-12
Adverse Social Determinants of Health Are Associated With Lower Rates of Cardiotoxicity Surveillance in Trastuzumab-Treated Breast Cancer Patients
Divya Samat, Tower Health-Reading Hospital, Reading, PA
D. Samat1, S. Singh1, V. Shah2, S. Iqbal3, A. Guha4; 1Internal Medicine, Tower Health-Reading Hospital, Reading, PA, 2Vascular Biology Center, Augusta University, Augusta, GA, 3Hematology/Oncology, Tower Health-Reading Hospital, Reading, PA, 4Cardiology, Augusta University, Augusta, GA.
Background: Trastuzumab poses a risk of cardiotoxicity, necessitating routine cardiac surveillance. Current guidelines recommend an echocardiogram at baseline, followed by repeat assessments every 3 months during therapy, to detect early decline in left ventricular function. Social determinants of health (SDOH) may influence patients’ ability to undergo recommended cardiac surveillance, potentially leading to delayed recognition of cardiotoxicity and worse clinical outcomes. This study investigates the impact of adverse SDOH on guideline-directed cardiac surveillance in breast cancer patients receiving trastuzumab. Methods: We conducted a retrospective cohort study using the TriNetX global federated health research network. Adult patients (≥18 years) with a diagnosis of breast cancer who received trastuzumab between January 1, 2000, and January 1, 2024, were identified. Patients were stratified into two cohorts based on the presence or absence of documented social determinants of health (SDOH) factors, identified by ICD codes Z55-65. Propensity score matching (1:1) was performed across 35 baseline characteristics to balance demographics, comorbidities and medications between cohorts. The primary outcome was the proportion of patients undergoing transthoracic echocardiogram (TTE) during 0-3 months, 3-6 months, and 6-12 months following trastuzumab initiation. Comparative analyses were conducted using risk differences with 95% confidence intervals, hazard ratios (HR) derived from Cox proportional hazards models, and Kaplan-Meier survival estimates for time-to-event analyses. Statistical significance was set at p < 0.05. Results: Before matching, patients with adverse SDOH were slightly younger at index (58.5 vs. 56.7 years), more likely to be Black (18.1% vs. 12.2%), and had higher rates of comorbidities including hypertension (56.5% vs. 33.5%) and heart failure (15.4% vs. 4.2%) compared to those without SDOH risk factors. After propensity score matching, each cohort consisted of 2,554 patients with baseline characteristics well balanced between the two cohorts, and no significant differences in age, sex, race, or major cardiovascular comorbidities. Medication use, including anthracyclines and cardioprotective therapies like beta blockers and ACE inhibitors, was also similar post-matching. At 0-3 months, 33.9% of patients with documented adverse SDOH (cohort 1) received a TTE compared to 45.0% of those without SDOH risk factors (cohort 2), representing a risk difference of 11.1% (HR 0.76, 95% CI 0.69-0.83; p<0.001). The median number of TTEs was 1 in both cohorts among the patients receiving at least 1. At 3-6 months; 27.7% of cohort 1 underwent TTE versus 48.9% of cohort 2, with a risk difference of 21.2% (HR 0.51, 95% CI 0.46-0.56; p<0.001). Median TTE instances remained 1 in both groups. In the 6-12 month interval, surveillance remained low for cohort 1 at 31.1%, compared to 54.5% for cohort 2 with a risk difference of 23.4% (HR 0.49, 95% CI 0.45-0.54; p<0.001). Among the patients receiving surveillance, the median number of TTEs was 1 in cohort 1 versus 2 in cohort 2. Conclusion: Our findings reveal significant disparities in adherence to guideline-recommended echocardiographic surveillance among breast cancer patients with adverse SDOH, persisting across all 3 intervals. There was a substantial decline in follow-up surveillance among the adverse SDOH group over time. These disparities highlight the need for targeted interventions to improve equitable cardiac monitoring and reduce long-term cardiotoxicity risk in vulnerable populations.
Presentation numberPS4-09-13
Elevated Genomic Risk and Treatment Gaps in Black Patients with HR+ Early Breast Cancer: A MammaPrint-Based Retrospective Analysis
Srishti Sareen, University of Tennesse, Memphis, TN
S. Sareen1, A. Naik1, C. Kent1, G. Vidal2, A. Hendrix1, P. Kheirkhah Rahimabad3, A. Bagchi1; 1Hematology Oncology, University of Tennesse, Memphis, TN, 2Hematology Oncology, West Cancer Center & Research Institute, Germantown, TN, 3Radiation Oncology, University of Tennesse, Memphis, TN.
Background: Black women with hormone receptor-positive (HR+) early-stage breast cancer (EBC) experience a 38% higher mortality rate than White women, even after accounting for socioeconomic factors and healthcare access. Biologic and genomic differences are increasingly recognized as contributors to this disparity. Gene expression (GE) assays help predict recurrence risk and guide treatment decisions. However, evidence suggests some of these tests may underestimate risk in Black patients. MammaPrint, however, among GE tools, has demonstrated enhanced sensitivity in identifying high-risk disease in Black women, suggesting improved risk stratification in this population.Objective: To analyze MammaPrint risk classifications and treatment patterns in a real-world, exclusively Black cohort with HR+ early breast cancer. We examined associations with tumor stage, molecular subtype, and chemotherapy use, and compared findings with historic, predominantly White populations such as the MINDACT trial.Methods: We conducted a retrospective chart review of 118 self-identified Black/African American women aged ≥18 years with Stage I-III HR+ breast cancer who had MammaPrint testing done and had clinic visits between October 2024 to June 2025 at West Cancer Center and University of Tennessee in Memphis, Tennessee. One patient with Stage IV disease was grouped with Stage III for analysis. Tumor characteristics, molecular subtype, and treatment data were extracted from electronic medical records. Statistical analyses included descriptive statistics, Chi-square, Fisher’s Exact, and Cochran-Armitage trend tests. This pilot phase of 118 patients will be expanded to approximately 1,000 patients from the same two clinics seen in the past year to further evaluate correlations and validate findings.Results: Among 118 patients, 65 (55.1%) were classified as High Risk and 53 (44.9%) as Low Risk by MammaPrint. The mean age was 62.5 years (SD = 12.9). MammaPrint risk was not significantly associated with tumor stage (P = 0.07), though a significant trend showed 50% of Stage I, 67% of Stage II, and 88% of Stage III/IV patients classified as High Risk (P trend = 0.02). MammaPrint risk correlated strongly with molecular subtype (p < 0.001), with 94.5% of Low Risk patients having Luminal A tumors versus 87.8% of High Risk patients having Luminal B tumors.Despite over half the cohort classified as High Risk, only 28% received neoadjuvant chemotherapy and 9.3% received adjuvant chemotherapy. These low treatment rates raise concerns about potential undertreatment, consistent with FLEX registry findings.In contrast, the MINDACT trial, with a largely White population, reported a High Risk proportion of 35.8% in HR+ early breast cancer patients. This suggests the genomic risk burden may be substantially higher among Black women, underscoring the need for more inclusive validation of genomic tools and equitable precision oncology application.Conclusions: In this exclusively Black cohort, MammaPrint identified a significantly higher proportion of High Risk patients than historic White-majority populations, such as in MINDACT. The majority of High Risk tumors were Luminal B; however, chemotherapy use was low, highlighting gaps in clinical application of genomic data. These low rates may reflect the predominant use of Oncotype to guide chemotherapy decisions, as it remains the standard of care. These results emphasize the importance of MammaPrint in risk stratification for Black patients and the need to address disparities in guideline-based treatment.
Presentation numberPS4-09-14
Timing delays from diagnosis to first treatment in black and hispanic versus white women with triple-negative breast cancer
Marcela Mazo Canola, UT Health San Antonio, San Antonio, TX
M. Mazo Canola1, C. Hogea2, K. Backus3, T. L. Buckingham2, E. Gillen4, N. Barrow4, A. Schroeder4, M. Khan4, N. Mercado4, M. Karhade4, L. T. Housman4; 1Medicine/ Division of Hematology and Oncology, UT Health San Antonio, San Antonio, TX, 2Patient-Focused Implementation Science, Gilead Sciences, Inc, Foster City, CA, 3Medical Affairs, Gilead Sciences, Inc, Foster City, CA, 4Healthcare Data Analytics, Avalere Health, Washington, DC.
Purpose: Disparities for Black and Hispanic versus White women with triple-negative breast cancer (TNBC) have been identified in literature. This study used a mixed-methods approach to capture the lived experiences of women treated for TNBC. Qualitative interviews contextualized the quantitative analysis. This approach prioritizes the patient voice and patient reported outcomes when translating claims-based data. Methods: The quantitative analysis used commercial (n=4494) and Medicaid managed care (MMC; n=1851) claims. Study inclusion required claims for breast cancer and for TNBC-related therapy ≤12 months postdiagnosis. Qualitative interviews (n=42) were conducted with women diagnosed with TNBC, though not the same women from the quantitative sample. Results: In the quantitative analysis, compared to White women in MMC, Black women had over 5 weeks longer between diagnosis and treatment (82.1 days [95% CI 75.3, 89.4] versus 44.2 days [40.7, 48.0]), and Hispanic women had almost 3 weeks longer (63.2 days [58.1, 68.8]). Compared to White women, the qualitative analysis found Black and Hispanic women more frequently reported discrimination and care team dismissiveness, exacerbating the impact of delays in care and contributing to worse outcomes. Conclusions: Black and Hispanic women experienced longer wait times between TNBC diagnosis and treatment, compared to White women. These delays in care must be considered within the context of other barriers to obtaining timely treatment. The qualitative study identified barriers, such as provider dismissiveness, which could have compounding effects on care. Further research is required to improve the standard of care for all women with TNBC, fostering better patient and provider relations, and reducing treatment delays.
Presentation numberPS4-09-15
More than a number: Does the NCCN Distress Score Capture Key Social Determinants of Health Risks in Patients with Breast Cancer?
Amanda Golden, Washington University in St. Louis, St. Louis, MO
A. Golden1, E. L. Podany2, F. Fa’ak2, B. Bowe3, A. A. Davis2; 1Internal Medicine and Geriatrics, Washington University in St. Louis, St. Louis, MO, 2Hematology/Oncology, Washington University in St. Louis, St. Louis, MO, 3Surgery, Washington University in St. Louis, St. Louis, MO.
Introduction: Research has demonstrated that social determinants of health (SDOH) can impact time to diagnosis and access to treatment, leading to poorer outcomes and overall survival for cancer patients (pts). The National Comprehensive Cancer Network (NCCN) Distress Thermometer (DT) is a tool used to identify, monitor, and document a pt’s overall distress score (DS). This study aims to identify if the DT captures key SDOH risks for pts with breast cancer. Methods: Pts seen at North St. Louis County Siteman Cancer Center breast cancer clinics from 07/2023 to 06/2025 completed an optional 19 question, validated, screening questionnaire with real-time electronic medical record integration that categorized pts as “high risk” or “low risk” for individual SDOH factors based on responses. Categories of questions included financial resource strain, transportation availability, depression, food insecurity, and housing instability. Pts also completed the NCCN DT, which provided a DS of 0-10. We evaluated patients as “low distress” if DS 4 based on established literature. We compared DS with risk category of several pertinent SDOH factors using the closest DT completed within 6 months of the SDOH evaluation. If a pt had unanswered questions on the questionnaire, those were counted as missing. Statistical testing was performed using ANOVA, t-test, and chi square analysis. Results: We evaluated a total of 268 pts who completed both the NCCN DT and the SDOH evaluation. 125 (47.5%) self-identified as Black and 131 (49.8%) as White. We found that pts who were high risk for depression on SDOH screening had significantly higher reported DS than those who were at low risk (p=0.0003). Pts at high risk for transportation difficulty were more likely to have a higher DS than those at low risk (p=0.03). Pts on Medicaid were more likely to have a higher DS than those on Medicare or commercial insurance (p=0.002). There was no significant association between risk of financial resource strain, food insecurity, or housing instability and DS (Table 1). We further investigated NCCN DS as a continuous variable and found consistent results, with no significant association of DS with high risk of food insecurity, financial resource strain, or housing instability. Conclusions: Our study evaluated the NCCN DT compared to a validated SDOH questionnaire in a diverse population. While the DT accurately identified pts as high risk for depression and transportation difficulty based on our SDOH screening tool, other key risk categories including financial resource strain, food insecurity, and housing instability were not associated with DS. Research has shown that these risk categories are linked to poorer clinical outcomes. Because certain high risk SDOH factors (or pts) were not identified based on the NCCN DT, this points for the need for more granular screening of SDOH in clinical practice.
| Distress Score <=4 | Distress Score >4 | P-Value | |
| Depression | 0.0003 | ||
| High Risk | 4 (2.9%) | 9 (18.4%) | |
| Low Risk | 133 (97.1%) | 40 (81.6%) | |
| Financial Resource Strain | 0.26 | ||
| High Risk | 7 (11.5%) | 5 (20.8%) | |
| Low Risk | 54 (88.5%) | 19 (79.2%) | |
| Food Insecurity | 0.09 | ||
| High Risk | 16 (19.3%) | 10 (34.5%) | |
| Low Risk | 67 (80.7%) | 19 (65.5%) | |
| Housing Instability | 0.09 | ||
| High Risk | 10 (14.5%) | 7 (30.4%) | |
| Low Risk | 59 (85.5%) | 16 (69.6%) | |
| Transportation Needs | 0.03 | ||
| High Risk | 4 (4.7%) | 5 (17.2%) | |
| Low Risk | 82 (95.3%) | 24 (82.8%) |
Presentation numberPS4-09-16
Role of surgeon discussions in post-mastectomy reconstruction racial disparities
Faith Dickerson, University of Wisconsin School of Medicine and Public Health, Madison, WI
F. Dickerson1, M. C. Saucke2, C. R. Breuer2, J. R. Schumacher3, H. B. Neuman1; 1Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, 2Wisconsin Surgical Outcomes Research Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 3Surgery, University of North Carolina School of Medicine, Chapel Hill, NC.
Introduction:Surgeons often serve as the “gatekeepers” of breast surgery, including decisions for post-mastectomy reconstruction. The options surgeons present and how recommendations are communicated can influence which treatments patients consider. This dynamic suggests that surgeon framing could play a key role in perpetuating or mitigating reconstruction disparities. We analyzed surgeon-patient consultations to describe surgeons’ discussion of reconstruction overall and by patient race. Methods: This study was a secondary analysis of Alliance clinical trial A231701CD. We included women aged ≤60 years who underwent breast cancer surgery without neoadjuvant therapy and had a transcript of their surgical consultation available (n=182). Transcripts were reviewed by 2 or more researchers to categorize discussions: preference for breast conserving surgery (BCS) without consideration of mastectomy or reconstruction, discussion of mastectomy but not reconstruction, or discussion of mastectomy with reconstruction. For consults with a reconstruction discussion, we coded who initiated the topic (surgeon vs patient/support person) and how reconstruction was presented (neutrally vs suggestion for or against). We generated summary statistics and used multivariable logistic regression models to identify factors associated with reconstruction discussions, with the first model including patient age alone and then adding race, insurance, and income. We used chi square test to assess differences in how reconstruction was presented by patient race. Results:Most patients were White (63%), with 23% Black and 10% Asian. Patient median age was 51 (range 28-60). A minority of consultations did not discuss reconstruction (preference for BCS [29%, n=53] or discussed mastectomy but not reconstruction [4%, n=8]). Older patient age was associated with lower odds of discussing reconstruction. Black patient race was significantly associated with a lower odds of discussing reconstruction when controlling for age (Table), but not after inclusion of private insurance or income. Surgeons initiated the discussion in most consults (86%). Reconstruction was presented neutrally in 67% of consults, with surgeons making a suggestion for or against reconstruction in 27% and 6% of consults, respectively. There was no statistically significant difference in how reconstruction was presented by patient race (p=0.30). Discussion:Our findings suggest that Black patient race is associated with a lower odds of discussing reconstruction. The strength of this association was tempered by insurance type and income, although the pattern of findings remained consistent. This suggests that reconstruction disparities may partially stem from a lack of conversation during the initial surgical consultation. Additional work should focus on developing interventions to increase consistency in discussions about surgical options.
| Model 1 | Model 2 | Model 3 | Model 4 | |
| (*Statistically significant at p<0.05) |
Odds Ratio (95% Confidence Interval) |
Odds Ratio (95% Confidence Interval) |
Odds Ratio (95% Confidence Interval) |
Odds Ratio (95% Confidence Interval) |
| Age | 0.93 (0.89-0.97)* | 0.92 (0.88-0.97)* | 0.92 (0.87-0.97)* | 0.92 (0.87-0.97)* |
| Race: White | Ref | Ref | Ref | |
| Race: Black | 0.39 (0.18-0.84)* | 0.47 (0.21-1.1) | 0.51 (0.22-1.2) | |
| Race: Other | 0.51 (0.20-1.3) | 0.50 (0.19-1.3) | 0.51 (0.19-1.3) | |
| Insurance: Other/Self-Pay | Ref | Ref | ||
| Insurance: Private | 1.8 (0.78-4.1) | 1.8 (0.65-5.3) | ||
| Household Income: <$25,000 | Ref | |||
| Household Income: $25-49,999 | 0.68 (0.17-2.8) | |||
| Household Income: $50-99,999 | 0.69 (0.17-2.8) | |||
| Household Income: >$100,000 | 1.2 (0.29-5.1) | |||
| Household Income: Missing | 0.58 (0.14-2.5) |
Presentation numberPS4-09-17
Breast Cancer in Transgender Patients Following Medical and Surgical Intervention
Ayush Thakur, Case Western Reserve University School of Medicine, Cleveland, OH
D. Gilbert1, A. Thakur1, T. Tabernacki1, M. Loria1, M. A. Mart2, S. Rhodes3, S. Gupta3, K. Mishra4, M. McNamara5; 1School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, 2Center for Clinical Research, University Hospitals Cleveland Medical Center, Cleveland, OH, 3Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 4Urology, MetroHealth Medical Center, Cleveland, OH, 5Internal Medicine, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH.
Background: Current breast cancer (BCa) screening guidelines do not adequately address transgender populations, despite known differences in hormone exposure and post-operative anatomy between transgender and cisgender patients of the same gender. The effects of gender-affirming hormone therapy (HT) and surgical intervention (SX) on BCa risk in transgender individuals remain poorly understood.Objective: To evaluate the incidence and predictors of BCa in transgender men and women, stratified by gender-affirming interventions (HT and SX).Methods: Using TriNetX (TriNetX, Inc., Cambridge, MA, United States), a de-identified health research network, we identified adults with a diagnosis of Gender Identity Disorder (GID) (ICD-10: F64.x) from 2003-2023. Patients were divided by sex assigned at birth and categorized into cohorts based on gender-affirming intervention: no intervention (NI), HT alone (≥1 year), and HT with SX. SX was limited to top surgeries, such as breast contouring and mastectomies. Patients with a past history of BCa, bottom surgery, or inconsistent medical timelines were excluded. The primary outcome was a diagnosis of malignant neoplasm of the breast (ICD-10: C50.x), recorded in at least two instances. Logistic regression was used to identify predictors of BCa risk.Results: Among 33,775 transgender women (assigned male at birth), 12 (0.036%) were diagnosed with BCa. Of these, 83.3% had no intervention, 16.7% received HT, and none received both HT and SX. HT alone was not associated with increased risk of developing BCa. Independent predictors of BCa included age at GID diagnosis (OR =d 1.07 per year, 95% CI: 1.05-1.09, p < 0.001) and Black race (OR = 2.71, 95% CI: 1.13-5.76, p = 0.015). Among 44,117 transgender men (assigned female at birth), 37 (0.084%) were diagnosed with BCa. Most cases occurred in those with no intervention (72.9%), followed by HT+SX (21.6%), and HT alone (5.4%). Hormone therapy alone was not associated with increased risk. Individuals who received both HT and SX had significantly higher BCa prevalence compared to those with HT alone (0.25% vs. 0.07%, p = 0.01). Multivariate analysis showed increased odds of BCa with combined HT and SX (OR = 3.62, 95% CI: 1.02-10.14, p = 0.024), Black race (OR = 3.35, 95% CI: 1.20-8.15, p = 0.012), and age at GID diagnosis (OR = 1.07 per year, 95% CI: 1.05-1.09, p < 0.001). Conclusion: This is the largest cohort study to date assessing BCa risk among transgender patients, and the first study to stratify risk by level of gender-affirming intervention. HT alone does not appear to increase BCa risk in transgender individuals. However, among transgender men, the combination of HT and SX was associated with significantly elevated risk. These findings may reflect differences in tissue preservation during mastectomy, lower screening rates, or increased pathological surveillance post-surgery. Black race and age at GID diagnosis were also associated with greater risk among both trans men and women. Our findings underscore the need for revised BCa screening guidelines that reflect the unique risk profiles of transgender populations and address intersectional disparities in care.
Presentation numberPS4-09-18
Factors with Immunotherapy Use Among Patients with Triple Negative Breast Cancer in California: A Population-Based Study
Jackelyn Moya, UCSF, San Francisco, CA
J. Moya1, A. Vu2, L. Huppert3, E. Ziv4, S. Lin Gomez2; 1Department of Surgery, Breast Care Center, UCSF, San Francisco, CA, 2Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, 3Breast Medical Oncology, UCSF, San Francisco, CA, 4Department of General and Internal Medicine, UCSF, San Francisco, CA.
TITLE: Factors with Immunotherapy Use Among Patients with Triple Negative Breast Cancer in California: A Population-Based Study BACKGROUND: Immunotherapy with immune checkpoint inhibitors has been approved for triple negative breast cancer (TNBC) since 2019. However, uptake of new therapies may be delayed by a variety of factors that affect access, such as facility level factors and individual demographic factors such as race/ethnicity and age. We investigated individual-, facility- and neighborhood-level factors associated with use of immunotherapy among females diagnosed with TNBC in California from 2019-2021 using statewide cancer registry data. METHODS: We used the California Cancer Registry to identify 7,495 women ages 18 and over diagnosed with first primary microscopically confirmed malignant TNBC diagnosed in 2019-2021. We used registry data about immunotherapy use to infer use of immune checkpoint inhibitor. Patient-level factors include age at diagnosis, AJCC stage, grade, other treatments received, marital status, and health insurance status. We used multivariable logistic regression to calculate odds ratios (OR) and 95% confidence intervals (CI) factors associated with immunotherapy use, including independent variables that were statistically significant at p<0.1 for inclusion in the final logistic model. RESULTS: Overall, 16% (1,219 of7,495) women with TNBC received immunotherapy with use increasing from 7.8% in 2019 to 28.3% in 2021. In multivariable models, patient-level factors associated with lower likelihood of immunotherapy use included Non-Hispanic Black females (Non-Hispanic Black vs. Non-Hispanic White OR 0.61; 95% CI:0.47-0.80), earlier stage disease (Stage I vs. Stage IV OR: 0.14; 95% CI: 0.10-0.19), older age (Age >70 vs. Age <40 OR: 0.48; 95% CI: 0.39-0.61). Facility level factors associated with lower likelihood of immunotherapy use included no National Cancer Institute (NCI) designation status (non-NCI-designated vs. NCI-designated OR: 0.54; 95% CI: 0.43-0.67), facility with predominantly low socioeconomic status (low SES) patients (predominantly low SES vs. high SES OR: 0.61; 95% CI: 0.46-0.82). CONCLUSION: Use of immunotherapy among patients in California lagged among women seen at low SES hospitals, non-NCI designated facilities, and among older and Non-Hispanic Black patients. These results provide direction towards future research to better understand the patient, institutional, and neighborhood level barriers.
Presentation numberPS4-09-19
Factors influencing time to treatment initiation for breast cancer in Ethiopia
Anteneh A Kibret, La Trobe University, Melbourne, Australia
A. A. Kibret1, H. Jiang1, E. Woldetsadik2, M. Tafese3, B. Deressa4, G. Liu1; 1Public Health, La Trobe University, Melbourne, AUSTRALIA, 2Public Health, Addis Ababa University, Addis Ababa, ETHIOPIA, 3Oncology, Jimma University, Jimma, ETHIOPIA, 4Public Health, Adama Hospital Medical College, Adama, ETHIOPIA.
AbstractBackground: Breast cancer is becoming a challenging health condition in Ethiopia with a high rate of morbidity and mortality. Timely care is crucial for improving breast cancer survival. However, women in Ethiopia continue to encounter multiple barriers at various stages of the care pathway from symptom recognition to treatment initiation. This study aimed to assess the factors influencing the interval from symptom detection to treatment initiation among breast cancer patients in Ethiopia.Methods: A cross-sectional study was conducted between July and September 2024 among 458 women with histologically confirmed breast cancer who initiated treatment at three tertiary hospitals in Ethiopia. Data were collected through structured interviews and medical record reviews. Five key intervals from symptom detection to treatment initiation were measured using the Model of Pathways to Treatment. Andersen’s Behavioural Model guided the categorization of explanatory variables into predisposing, enabling, need-related, and contextual factors. Accelerated Failure Time (AFT) models were used to identify predictors of delay, with model selection based on the best-fitting distribution (Weibull, lognormal, or log-logistic). Time ratios (TRs) with 95% confidence intervals (CIs) were reported, and a p-value of <0.05 was considered statistically significant.Result: The study identified multiple factors associated with delays in breast cancer care in Ethiopia. Rural residence (TR = 1.84; 95% CI: 1.20-2.80) and presentation with painless breast mass (TR = 1.94; 95% CI: 1.26-3.00) were linked to longer intervals between symptom detection and first contact with health care providers (HCP). Low breast cancer literacy and use of traditional healing prior to diagnosis were consistently associated with delays across all stages of care. Consulting three or more healthcare providers significantly prolonged both the interval from symptom detection to first healthcare provider contact (TR = 3.13; 95% CI: 1.96-4.98) and the interval from symptom detection to diagnosis (TR = 1.63; 95% CI: 1.02-2.62). Lack of cancer suspicion by the first HCP was linked to longer time from first contact with HCP to diagnosis (TR = 3.07; 95% CI: 2.15-4.39). Similarly, the absence of referral by the first healthcare provider was associated with prolonged intervals from symptom detection to diagnosis (TR = 1.47; 95% CI: 1.06-2.03) and from symptom detection to treatment (TR = 1.47; 95% CI: 1.11-1.95). Being unmarried (TR = 1.46; 95% CI: 1.14-1.87), low-income (TR = 1.48; 95% CI: 1.05-2.08) and using traditional healer after diagnosis (TR = 2.25; 95% CI: 1.33-3.80) were associated with longer diagnosis-to-treatment intervals, while women aged ≥60 years had shorter diagnosis-to-treatment intervals (TR = 0.62; 95% CI: 0.41-0.93).Conclusion: Delays in breast cancer care in Ethiopia are influenced by a combination of predisposing, enabling, need-related, and contextual factors. Key contributors include rural residence, low breast cancer literacy, low household income, use of traditional healing before and after diagnosis, and limited HCP provider capacity for early detection and referral. To improve timely access to breast cancer care, interventions should focus on enhancing community awareness, integrating traditional healers into formal referral pathways, strengthening primary care and referral systems, building the capacity of frontline health workers, and addressing key financial and geographic barriers.
Presentation numberPS4-09-20
Trends and Disparities in Mortality Among U.S. Females with Breast Cancer and Hypertensive Diseases, 1999-2023
Abat Khan, Memorial Healthcare System, Pembroke Pines, FL
A. Khan1, S. Ali1, H. Habib2, A. I. Butt2, Z. A. Siddiqi3, A. Saleem2, I. A. Magsi4, I. Khan5, M. Jan2, O. Mohamed6, M. Raif7, S. Musleh Ud Din1, A. Kiamos1, S. Streit1, A. Castrellon1; 1Hematology and Oncology, Memorial Healthcare System, Pembroke Pines, FL, 2Internal Medicine, Allama Iqbal Medical College, Lahore, PAKISTAN, 3Internal medicine, James Cook University Hospital, Middlesbrough, England, UNITED KINGDOM, 4Internal Medicine, Shaheed mohtarma benazir bhutto medical college, Karachi, PAKISTAN, 5Internal Medicine, FMH College of medicine and dentistry, Islamabad, PAKISTAN, 6Internal Medicine, Jubilee Mission Medical College and Research Institute, Kerala, INDIA, 7Internal Medicine, King Edward Medical University, Lahore, PAKISTAN.
Trends and Disparities in Mortality Among U.S. Females with Breast Cancer and Hypertensive Diseases, 1999–2023 Background:Breast cancer remains a leading cause of cancer-related mortality in women, while hypertension is a prevalent comorbidity that can worsen outcomes. Despite their combined clinical significance, national mortality trends in women affected by both conditions remain underexplored.Methods:We analyzed data from the CDC WONDER database for U.S. female decedents aged ≥25 years from 1999 to 2023, using ICD-10 codes C50 (breast cancer) and I10–I15 (hypertensive diseases). Crude death rates (CDRs) and age-adjusted mortality rates (AAMRs) per 100,000 population were calculated. Trends were assessed using Joinpoint regression to estimate annual percent change (APC) and average annual percent change (AAPC).Results:From 1999 to 2023, 136,837 deaths were attributed to concurrent breast cancer and hypertensive diseases. Most deaths occurred at home (n=47,612), followed by nursing homes (n=38,608), medical facilities (n=35,200), hospice (n=788), and other locations (n=7,214).The AAMR increased from 2.11 in 1999 to 5.80 in 2023 (AAPC: 4.17%; 95% CI: 3.52–4.94). A sharp rise occurred between 1999–2001 (APC: 27.73%; 95% CI: 13.38–39.55), followed by stability until 2017, then another increase from 2017–2023 (APC: 7.25%; 95% CI: 4.43–15.30).Non-Hispanic Black females had the highest AAMRs (mean: 6.36). Their rates rose from 3.87 in 1999 to 5.95 in 2001 (APC: 18.91%) and from 6.24 in 2018 to 8.36 in 2020 (APC: 17.21%). Non-Hispanic White females experienced an increase from 1.94 in 1999 to 3.18 in 2001 (APC: 28.33%), followed by a slower rise until 2018 (APC: 0.75%) and another jump to 5.12 in 2020 (APC: 12.74%). Among Hispanic women, AAMRs rose from 1.4 in 1999 to 2.85 in 2013 (APC: 3.36%) and again from 2.71 in 2018 to 3.77 in 2020 (APC: 17.99%).No statistically significant AAMR changes were observed in American Indian or Alaska Native women. Asian or Pacific Islander women showed a consistent upward trend (AAPC: 2.68%; 95% CI: 1.93–3.79).Regionally, the Midwest had the highest AAMR (4.24). Trends showed an initial rise (1999–2007; APC: 4.21%), a decline (2007–2016; APC: −2.96%), and a rise again (2016–2023; APC: 4.41%). Similar patterns of increase were observed in the South, West, and Northeast, particularly in the early 2000s and post-2016.Rural areas had higher AAMRs (4.25) than urban areas (3.80). In rural regions, AAMR rose sharply between 1999–2001 (APC: 27.91%) and again between 2018–2020 (APC: 15.31%). Urban areas showed significant increases in similar timeframes.By age group (1999–2023), the highest CDR was in women aged ≥85 years (58.05), followed by ages 75–84 (20.49), 65–74 (7.67), and 55–64 (2.62). Death rates were substantially lower in younger women.Conclusion:Mortality from coexisting breast cancer and hypertensive diseases has risen significantly over two decades, with substantial disparities by race, region, and age. These findings underscore the urgent need for integrated care approaches and equity-focused interventions in this vulnerable population.
Presentation numberPS4-09-21
Disparities in Breast Cancer Mortality Among Individuals with Mental and Behavioral Disorders: A 25-year population-Based Analysis
Anas Al-Zubaidi, The University of Texas Medical Branch, Galveston, TX
A. Al-Zubaidi1, E. Cohen2, G. Whitman2, B. Adrada2, T. Moseley3, A. Robinson4; 1Radiology, The University of Texas Medical Branch, Galveston, TX, 2Radiology – Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Radiology – Breast Imaging, The University of Mississippi Medical Center, Jackson, MS, 4Radiology – Breast Imaging, The University of Texas Medical Branch, Galveston, TX.
Background: Breast cancer (BC) mortality has declined in the general U.S. population over recent decades. However, individuals with mental and behavioral disorders remain an understudied and potentially high-risk subgroup. This study aims to assess national trends in BC mortality among women with mental and behavioral disorders, stratified by race and ethnicity. Method: We studied a cumulative population of 9.17 billion by using the CDC Multiple Cause of Death database (ICD, 10th revision) between 1999 and 2023. We identified all women who died of BC, code C50, as underlying cause of death (UCD), regardless of mental and behavioral disorders (MBD). Then we identified the same UCD among women with MBD, code F01-F99, as multiple cause of death. Age-adjusted mortality rates (AAMR) per million persons (PMP) were calculated, standardized to 2000 US census, and stratified by ethnicity and race [Hispanic (H), non-Hispanic (NH), Black (B), White (W), Asian or Pacific Islander (A/PI), and American Indian or Alaska Native (AI/AN)]. Annual percentage changes were calculated to assess the mortality trends. Result: In the general population (GP), a total of 2,059,043 deaths from BC were identified, with an overall AAMR of 188.3 PMP (B: 296.3, W: 219.9, A/PI: 117.1, AI/AN: 123.5, H: 143.9, NH: 229.3). AAMR declined from 1999 to 2023 by 28% among B from 349.8 to 252.2 PMP, 30% among W from 259.7 to 183 PMP, 12% among A/PI from 126.2 to 111.7 PMP, 34% among AI/AN from 154.9 to 102.7 PMP, 18% among H from 164.3 to 135.5 PMP, and 27% among NH from 272.1 to 194.4 PMP. In the population with MBD, a total of 175,198 BC deaths were identified, with an overall AAMR of 8.7 PMP (B:12.5, W: 11.2, A/PI: 3.4, H: 4.5, NH: 11.86). AAMR increased from 1999 to 2023 by 222% among B from 4.9 to 15.8 PMP, 230% among W from 4.4 to 14.5 PMP, and 243% among NH from 4.4 to 15.1 PMP. Excluding years with unreliable data, the AAMR doubled among H, rising from 2.5 to 5.0 PMP (a 100% increase) from 2002 to 2023, and remained relatively stable among A/PI, declining slightly by 12.1% from 3.3 to 2.9 PMP between 2010 and 2023. No reliable data were identified among the AI/AN. Conclusion: This is one of the largest population-based studies analyzing BC mortality in women with MBD. While breast cancer mortality declined across all racial and ethnic groups in the GP over the past two and a half decades, substantial increases were observed among those with MBD. In sharp contrast to declining rates in the GP, AAMR more than doubled among most groups with MBD—rising over 200% among B, W, and NH individuals, and doubling among H. These findings underscore a widening mortality gap and highlight the urgent need for targeted interventions addressing disparities in breast cancer outcomes among those with MBD.
Presentation numberPS4-09-22
Uneven Progress: A Two-Decade Analysis of Regional Variations in Breast Cancer Mortality in the United States.
Anas Al-Zubaidi, The University of Texas Medical Branch in Galveston, Galveston, TX
A. Al-Zubaidi1, A. Robinson1, G. Whitman2, T. Moseley3, B. Adrada2, E. Cohen2; 1Radiology – Breast Imaging, The University of Texas Medical Branch in Galveston, Galveston, TX, 2Radiology – Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Radiology – Breast Imaging, The University of Mississippi Medical Center, Jackson, MS.
Background: Breast cancer (BC) remains a leading cause of cancer death among U.S. women. Despite significant advances in BC screening and treatment leading to declining national mortality rates, regional disparities in healthcare access and socioeconomic factors may contribute to uneven progress across U.S. geographic regions. This study aims to quantify these disparities and identify areas where tailored interventions may be needed. Method: We conducted a retrospective cohort study using the Centers for Disease Control and Prevention (CDC) WONDER database (ICD-10). We identified all women who died of BC (C50), as the underlying cause of death (UCD) between 2000 and 2023. Age-adjusted mortality rates (AAMRs) were calculated per 100,000 persons, standardized to the 2000 U.S. Census, and stratified by the census region [Northeast (NE), Midwest (MW), South (S), and West (W)]. AAMRs were analyzed using linear regression models over a 24-year period to determine average annual reduction (AAR) and overall trends. 95% confidence intervals (CIs) were used to assess the reliability of mortality rate estimates and validate the linear regression analysis. Regional comparisons were conducted to assess geographic disparities in mortality outcomes. Results: At the national level, a total of 995,078 deaths from BC were identified, with an overall AAMR of 21.8 (95% CI: 20.7 – 22.9), and region-specific AAMRs of 21.8 (95% CI: 21.3 – 22.3) in the NE, 22.4 (95% CI: 21.9 – 22.8) in the MW, 22.3 (95% CI: 21.9 – 22.7) in the S, and 20.8 (95% CI 20.4 – 21.3) in the W.Between 2000 and 2023, the overall AAMR declined by 31.3%, with a consistent national AAR of 1.30% per year. Regional AAMRs declined by 39.4% from 27.9 (95% CI: 27.4 – 28.5) to 16.9 (95% CI: 16.5 – 17.3) with an AAR of 1.68% per year (95% CI: 1.64% to 1.72%) in the NE, 33.2% from 27.7 (95% CI: 27.2 – 28.3) to 18.5 (95% CI: 18.1 – 18.9) with an AAR of 1.44% per year (95% CI: 1.34% to 1.54%) in the MW, 24.4% from 26.2 (95% CI: 25.8 – 26.7) to 19.8 (95% CI: 19.5 – 20.1) with an AAR of 1.02% per year (95% CI: 0.88% to 1.15%) in the S, 27.7% from 25.3 (95% CI: 24.7 – 25.9) to 18.3 (95% CI: 18 – 18.7) with an AAR of 1.20% per year (95% CI: 1.05% to 1.35%) in the W. Conclusion: This is one of the largest population-based studies analyzing nearly one million deaths from breast cancer in women over the past two decades. While there has been significant national progress in breast cancer mortality reduction across all U.S. regions (31.3% decline, 1.30% annually), substantial regional disparities persist. The NE is leading the nation in mortality reduction (39.4% reduction, 1.68% annually) while the S showed the most modest gains (24.4% reduction, 1.02% annually). These findings underscore the need for region-tailored strategies—enhancing screening outreach, closing healthcare access gaps, and targeting socio-demographic risk factors—to ensure mortality reductions are equitably realized nationwide.
| Census Region |
AAMR Percent Decline (2000-2023) |
Average Annual Reduction | Notable Observation |
| National Average |
31.3% (26.8 → 18.4) |
1.30% | Sustained substantial improvement at the national level. |
|
Northeast |
39.4% (27.9 → 16.9) |
1.68% | Leading national improvement model. |
| South |
24.4% (26.2 → 19.8) |
1.02% | The South trails the national average, exhibiting the smallest decline in mortality among all regions. Urgent intervention target. |
Presentation numberPS4-09-23
Palliative Care Utilization and Associated Outcomes in Hospitalized Breast Cancer Patients: Using The National Inpatient Sample 2021
Srinishant Rajarajan, AGH, Pittsburgh, PA
S. Rajarajan1, K. Babu1, M. Jin1, G. Gorecki1, R. Dileo1, S. Arivazhagan1, Y. Khalid2, C. Hilton3; 1Internal Medicine, AGH, Pittsburgh, PA, 2Internal Medicine, UAB Montgomery, Montgomery, AL, 3Haematology Oncology, AGH Cancer Center, Pittsburgh, PA.
Title:Palliative Care Utilization and Associated Outcomes in Hospitalized Breast Cancer Patients: UsingThe National Inpatient Sample 2021Background:While palliative care (PC) is increasingly recognized as essential in oncologic care, its utilizationamong breast cancer patients remains underexplored at a national level. We examined predictorsand outcomes associated with inpatient PC use in breast cancer hospitalizations.Methods:We conducted a retrospective cross-sectional analysis using the National Inpatient Sample (NIS)2021. Adult hospitalizations with a diagnosis of breast cancer and Palliative encounter wereincluded using the appropriate ICD-10 codes. Survey-weighted logistic and linear regressionmodels were employed to assess factors associated with PC utilization and its impact on inpatientmortality, length of stay (LOS), and total hospitalization charges. Covariates includeddemographics, comorbidities (Charlson Index), income quartile, insurance type, hospital teachingstatus, region, and bed size.Results:Among 32,719 breast cancer hospitalizations, PC was utilized in 12.1% (95% CI: 11.6–12.6%).Patients receiving PC were older (mean age: 66.8 vs. 65.1 years, p < 0.001), had higher comorbidityburden, and were more frequently treated at teaching hospitals (p < 0.001). White patients wereslightly more likely to receive PC compared to Black and Hispanic patients. Insurance status was astrong predictor (p < 0.001), with higher PC utilization observed among Medicaid and Medicarerecipients compared to those with private insurance. Additionally, patients treated at teachinghospitals and larger hospitals were more likely to receive PC, with both hospital teaching status (p <0.001) and bed size (p = 0.016) showing significant associations.Adjusted analysis showed that PC was independently associated with higher odds of in-hospitalmortality (aOR: 13.36, 95% CI: 11.91–14.99, p < 0.001), longer LOS (adjusted mean difference:+1.83 days, 95% CI: 1.55–2.12, p < 0.001), and increased total charges (adjusted mean difference: +$9,614, 95% CI: $5,163–$14,066, p < 0.001). Disparities were observed across racial andsocioeconomic strata, with patients from higher income quartiles and teaching hospitals more likelyto receive PC.Conclusions:Palliative care was used in 12.1% of breast cancer hospitalizations, with notable disparities by race,insurance, and hospital characteristics. Utilization was higher among older patients, those withMedicaid/Medicare, and at teaching hospitals. PC was associated with higher in-hospital mortality,longer stays, and increased charges. These patterns suggest appropriate use in severe illness but alsoreflect access gaps. Efforts are needed to promote equitable palliative care delivery across allsettings.
Presentation numberPS4-09-24
Clinical and economic impact of the 21-gene assay for guiding treatment in patients with HR+/HER2- early breast cancer across racial and ethnic subgroups in the US.
Gebra Cuyun Carter, Exact Sciences, Madison, WI
G. Cuyun Carter1, C. Moyon2, V. Berdunov3, M. Gouldson4, J. Racz5, J. Bennett6, O. Kantor7, Y. Abdou8; 1Health Economics & Outcomes Research, Exact Sciences, Madison, WI, 2Health Economics & Outcomes Research, Putnam, Paris, FRANCE, 3Health Economics and Market Access, Evidera, London, UNITED KINGDOM, 4Health Economics & Outcomes Research, Putnam, London, UNITED KINGDOM, 5Medical Affairs, Exact Sciences, Madison, WI, 6Clinical Biostatistics, Exact Sciences, Madison, WI, 7Breast Surgical Oncology Fellow, Brigham and Women’s Hospital, Boston, MA, 8Division of Oncology, University of North Carolina, Chapel Hill, NC.
Background: Racial and ethnic disparities in treatment and outcomes persist among breast cancer patients in the United States (US), raising concerns about equitable access to precision oncology tools. The 21-gene assay, which estimates distant recurrence risk and predicts the clinical benefit of chemotherapy for hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) early-stage breast cancer, has become a key decision-making tool. This study assessed the clinical and economic value of the 21-gene assay compared to relying on clinical-pathologic risk factors alone, stratified by race and ethnicity. Methods: A validated cost-effectiveness model was adapted to reflect patients with node-negative (N0) or node-positive (N1) HR+/HER2− early breast cancer across four US racial and ethnic subgroups: non-Hispanic Black (NHB), non-Hispanic White (NHW), Asian, and Hispanic. Clinical inputs were derived from published data, including TAILORx, RxPONDER, and the SEER database. Subgroup-specific assumptions were reviewed and confirmed by US clinical experts. Outcomes were expressed as cost per quality-adjusted life year (QALY) over a lifetime horizon. Sensitivity analyses tested the robustness of results. Results: The 21-gene assay was cost-effective across all racial and ethnic subgroups, offering improved outcomes at lower costs compared to clinical-pathologic factors alone. The assay was dominant (lower cost, greater QALYs) in both N0 and N1 populations (Table 1). The greatest cost savings were seen in the N0 NHB subgroup, driven by more precise chemotherapy use and reduced risk of disease progression. Sensitivity analyses supported the robustness of these findings across a range of assumptions and willingness-to-pay thresholds. Conclusions: The 21-gene assay improves patient outcomes and reduces costs compared to using clinical-pathologic assessment alone across diverse racial and ethnic subgroups with HR+/HER2- N0 and N1 early-stage breast cancer. In addition to the clinical benefit observed across all subgroups, the greatest cost savings were observed in the N0 NHB subgroup, highlighting the assay’s potential to support more equitable care and reduce disparities in treatment decision-making.
| Population | N0 | N1 | ||||||||
| Outcomes | Overall | NHB | NHW | Asian | Hispanic | Overall | NHB | NHW | Asian | Hispanic |
| ∆ Costs | -$10,886 | -$18,018 | -$9,380 | -$12,917 | -$9,104 | -$4,921 | -$2,107 | -$4,771 | -$5,810 | -$4,454 |
| ∆ QALYs | 0.154 | 0.169 | 0.156 | 0.228 | 0.153 | 0.076 | 0.050 | 0.063 | 0.102 | 0.072 |
Presentation numberPS4-09-25
Variation in diagnostic imaging features of breast cancer by race and ethnicity
Ashley Odai-Afotey, Dana-Farber Cancer Institute, Boston, MA
A. Odai-Afotey1, Q. Jin2, N. Tayob2, B. N. Durieux3, C. Lindvall3, J. Vincuilla4, T. A. King4, E. A. Mittendorf4, R. A. Freedman1; 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 3Department of Supportive Oncology, Dana-Farber Cancer Institute, Boston, MA, 4Division of Breast Surgery, Department of Surgery, Dana-Farber Brigham Cancer Center, Boston, MA.
BACKGROUND: Though racial and ethnic differences in tumor characteristics are well described, less is known about the potential variability in diagnostic and imaging features at presentation. Here, we examined variations in imaging features of breast cancer (BC) by race and ethnicity and explored their associations with clinical outcomes. METHODS: Diagnostic imaging features, clinicopathological characteristics and race and ethnicity information for adult females with a first diagnosis of stage I-III BC during 2016-2023 were obtained from our NCI-designated center’s prospectively maintained database, which captures all patients having breast surgery at our high-volume center. Occult disease was defined as a lack of detection on diagnostic mammography. Multifocal or multicentric disease was defined as having this feature on any imaging modality — mammogram, MRI, or ultrasound. Natural language processing (NLP) extracted breast density information from mammograms in the electronic health record (EHR). Additional demographics were extracted from the EHR including education and residential zip code, with zip codes used to calculate social deprivation index (SDI). Multivariable logistic regression was used to examine racial and ethnic differences in the presence of mammographically occult, multifocal, and multicentric disease as well as breast density, adjusting for age and tumor features. Cox proportional hazards model evaluated overall survival (OS), recurrence free survival (RFS), and distant recurrence-free survival (DRFS) by race and ethnicity, accounting for clinicopathological, diagnostic, and demographic characteristics. RESULTS: Among 10038 patients, mean age was 58.5 [SD 13.3]); 86.5% were White, 4.7% Black, 4.4% Asian, and 3.9% Hispanic. Most patients had clinical T1 (51.6%), node-negative (85.0%) and HR+HER2- (75.2%) disease. 5.5% had occult disease; 33.9% multifocal and 12.7% multicentric disease on imaging. 6.6% were in the most deprived SDI quintile. Occult disease was most prevalent among Hispanic patients (9.4%), while Asian patients had the highest rates of multifocal (43.4%), multicentric (22.0%) disease and extremely dense breast (24.7%). Black patients most frequently fell into the most deprived SDI quintile (34.6%). In multivariable analyses, after adjusting for clinicopathological features, Asian (vs. White) race was significantly associated with higher odds of multifocal (OR 1.24, 95% CI 1.01-1.52) and multicentric (OR 1.59, 95% CI 1.23-2.03) disease and extreme breast density (OR 1.81, 95% CI 1.39-2.34) (all p< 0.001). Hispanic (vs. non-Hispanic) ethnicity was associated with higher odds of occult disease (OR 2.08, 95% CI 1.35-3.13, p=0.001). Black (vs. White) women had lower odds of having extremely dense breasts (OR 0.61, 95% CI 0.42-0.87, p=0.008). Occult and multicentric disease were associated with extremely dense breasts (all p < .05). After adjusting for all diagnostic features, lymphovascular invasion (LVI), triple negative BC (TNBC), older age, higher stage and grade were associated with worse RFS, DRFS and OS (p< 0.05), and residing in a more deprived area was associated with lower RFS and DRFS (HR 1.37, p< 0.05). Race, ethnicity, breast density, multifocal, and multicentric disease were not associated with RFS, DRFS or OS. CONCLUSIONS: In this unique, large sample of women with breast cancer, we identified racial and ethnic differences in imaging features of breast cancer, particularly among Asian and Hispanic patients. Although we observed differences in breast density, multifocality and multicentricity, after adjusting for more traditional features (e.g., subtype), these dissimilarities were not associated with RFS, DRFS or OS and may not be prognostic. Longer follow-up is needed to see if accounting for more enhanced diagnostic characteristics impacts outcomes.
Presentation numberPS4-09-26
Implementation of Integrated Mammography and Genetic Evaluation Services (IMAGES) in Underserved Communities
Jacynda Casey, University of Texas MD Anderson Cancer Center, Houston, TX
J. Casey1, D. Kizub2, E. Patino1, L. Balderas3, M. Valerio-Shewmaker4, A. Ruiz1, G. Whitman5, B. Arun2; 1Clinical Cancer Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 3Office of Health Policy, University of Texas MD Anderson Cancer Center, Houston, TX, 4School of Public Health, University of Texas Houston, Houston, TX, 5Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Annual screening mammography has played a significant role in breast cancer diagnosis, lowering mortality and morbidity. Uninsured women are 40% less likely to receive guideline-based mammogram screening resulting in more advanced stage breast cancer, requiring more intense treatment or having fewer treatment options. Additionally, 10-15% of all breast cancers are due to an underlying germline mutation, which increases an individual’s lifetime risk of developing breast cancer. Identified women with an underlying germline mutation may qualify for increased surveillance or risk reducing surgeries. Despite this, fewer than 15% of women from underserved areas eligible for genetic testing are referred and tested. To bridge this gap, the Integrated Mammography and Genetic Evaluation Services (IMAGES) program was created at MD Anderson Cancer Center (MDACC). IMAGES is a collaboration between Project Valet (PV), a MDACC mobile mammography service, and the MDACC’s Clinical Cancer Genetics (CCG) program to provide access to screening mammograms and hereditary breast cancer risk assessment to uninsured women throughout the surrounding Houston area. IMAGES aims to increase access to screening mammography, identify women at risk for a hereditary breast cancer syndrome, and assist in the navigation and coordination of diagnostic services or cancer treatment. Methods: PV has mobile mammography units established with 8 partnering sites. Uninsured, asymptomatic women ages 40-74 years located in one of the four rural counties southeast of Houston are eligible to participate in IMAGES. Eligible women are referred by a primary care provider or designated medical staff at a collaborating federally qualified health center. At the screening appointment, a community health worker completes patient registration, offers a 1-page genetic cancer risk assessment (GCRA), which collects personal and family history of cancer, and obtains all relevant consent forms. Once the patient completes the mammogram, PV staff will inform the patient and their health care provider of the results and coordinate additional follow-up, as needed. If the patient elected to complete the GCRA, a genetic coordinator scores it to determine if the patient meets NCCN criteria for genetic testing of hereditary breast cancer syndromes (ATM, BARD1, BRCA1, BRCA2, CDH1, CHEK2, NF1, PALB2, PTEN, RAD51C, RAD51D, STK11 and TP53). The genetic coordinator will contact patients eligible for genetic testing to offer and coordinate genetic testing via saliva samples. Negative genetic test results are disclosed to the patient by the genetic coordinator while variant of uncertain significance and positive results are returned by an MDACC genetic counselor. Descriptive demographic and behavioral statistics will be used to analyze data and summarize outcomes of the program between and within community health clinic sites. Results: This project started March 1, 2024, and is currently ongoing. To date, 1529 of women have received screening mammograms and 334 have completed diagnostic follow-up. Of those screened, 886 completed the GCRA, 117 were eligible for genetic testing, and 3 have completed genetic testing. Conclusion: IMAGES is an ongoing service project designed to increase access to screening mammography and genetic evaluation for women in underserved communities in the surrounding Houston area. So far, we have shown it is feasible to integrate the GCRA into mammography screening clinics. Efforts are being made to streamline the genetic testing process. Once the project is completed, further analysis will be done to assess total outcomes. Funding: This project is supported by the Cancer Prevention & Research Institute of Texas (CPRIT, #PP240012). Contact: Jacynda Casey, MS, CGC, jawoodman@mdanderson.org
Presentation numberPS4-09-27
Global inequalities in female breast cancer burden by decomposition and age-period-cohort analysis, 1990-2021
Ke Li, Peking University People’s Hospital, Beijing, China
K. Li1, Y. Zhang1, Z. Liu2, W. Shu1; 1Breast Center, Peking University People’s Hospital, Beijing, CHINA, 2Department of Cardiac Surgery, Peking University People’s Hospital, Beijing, CHINA.
Background: Breast cancer is the most common cancer among women globally, accounting for 25% of all female cancer cases and is the fourth leading cause of cancer-related deaths. It is influenced by a complex interplay of reproductive, lifestyle, genetic, and environmental factors, with early diagnosis significantly improving prognosis and survival rates. However, disparities in survival rates persist between high- and low-income regions, highlighting the need for tailored regional strategies to address these inequities and improve outcomes.Methods: Comprehensive data on female breast cancer were extracted from the Global Burden of Disease (GBD) 2021 study. All-age rate and age-standardized rate (ASR) of Disability-adjusted life years (DALYs) were quantified from 1990 to 2021, through socio-demographic index (SDI) stratification. The age-period-cohort (APC) model was used to elucidate the effects of age, period and cohort, and to estimate overall annual percentage changes in DALYs (net drifts). Decomposition analysis further attributed changes in disease burden to epidemiologic trends, population growth, and aging. The slope index of inequality (SII) and the concentration index (CI) were calculated to quantify the absolute and relative cross-country inequalities in the burden of female breast cancer.Results: From 1990 to 2021, APC models showed elevated risk in postmenopausal women and period-related declines in high SDI countries. In contrast, risks rose or plateaued in low SDI regions. Cohort analysis revealed decreasing risks in high SDI countries but rising risks in younger cohorts elsewhere. Age-specific net-drifts showed declining DALY rates overall, but varied by SDI level. High and high-middle SDI regions peaked in early adulthood and post-menopause, with lower risks at ages 40-60. Middle SDI areas had gradual changes, while low-middle and low SDI regions had the highest DALY rates after 60 and 80, with sharp rises in the oldest groups. Decomposition analyses showed population growth as the primary driver of increasing DALYs globally, with ageing overtaking epidemiologic change around 2005. Ageing was dominant in high and high-middle SDI countries by 2021, while population growth was more important in low SDI countries, where ageing had minimal impact. Inequality shifted, from 1990 to 2021, the SII shifted from 5 to −12 and CI from 0.09 to −0.02, indicating a reversal of the breast cancer burden toward disadvantaged populations. These findings highlight persistent inequities and resource constraints in low and middle SDI regions.Conclusion: The global burden of female breast cancer continues to rise with marked regional and age disparities, highlighting the urgent need for region-specific interventions. High SDI regions should enhance early detection and care, while middle and low SDI countries need to improve access to screening and health system capacity to mitigate disparities.
Presentation numberPS4-09-28
Air Pollution and Oncotype DX recurrence socre: implications for racial disparities in breast cancer
Lindsay J Collin, Emory University, Atlanta, GA
L. J. Collin1, T. Armide1, L. E. Barber1, K. Bishop2, C. M. Destin1, M. L. Maliniak1, J. M. Switchenko3, L. E. McCullough1; 1Epidemiology, Emory University, Atlanta, GA, 2Environmental Science and Policy, University of Miami, Miami, FL, 3Biostatistics, Emory University, Atlanta, GA.
Background Nearly 70% of breast cancer patients are diagnosed with estrogen receptor (ER) positive, human epidermal growth factor-2 (HER2) negative breast cancer—a subtype with generally more favorable survival. Precision medicine for this subtype has been informed by gene expression assays, such as the Oncotype DX (ODX) recurrence score, that assess the 10-year risk of distant recurrence (Low, Medium, or High) and guide treatment decisions. Air pollution is recognized has been linked to more aggressive breast cancer subtypes, suggesting that exposure to air pollution may contribute to more aggressive tumors. However, no study has examined air pollution in relation to ODX, which can be used as a marker of tumor aggressiveness. We evaluated the associations of two air pollutants (PM2.5 and NO2) with ODX recurrence score among Black and White breast cancer patients. Methods We identified all women aged ≥18 years, diagnosed with a stage I-III first primary breast cancer between 2010 and 2015 in the Georgia Cancer Registry. Breast cancer patients were included if they were non-Hispanic Black (NHB) or non-Hispanic White (NHW) and had an ODX recurrence score. Daily ambient PM2.5 and NO2 concentrations were predicted at a 1-km resolution for the years 2005-2010 using an ensemble of three machine learning models available from the Environmental Protection Agency. For each patient, we assigned PM2.5 and NO2 exposures based on the average value in the 5-years prior to their diagnosis. We used multinomial regression to estimate the associations between a 5-unit increase in PM2.5 and NO2 and higher ODX scores (Intermediate vs Low and High vs Low), overall and by race. Models were adjusted for age at diagnosis (years), race (overall model only), and rurality. Results In the Georgia cohort (77% NHW, 23% NHB), 6985 women received ODX testing—4090 (56%) were classified as Low, 2394 (34%) as Intermediate, and 501 (7.2%) as High risk. A larger proportion of those with a High ODX recurrence score were NHB (30%) compared with a Low recurrence score (21%). In the overall study population, a 5-unit increase in PM2.5 was associated with higher odds of both an Intermediate (odds ratio [OR]=1.20, 95%CI: 1.02, 1.41) and High (OR=1.31, 95%CI: 0.97, 1.76) recurrence score compared with those in the Low-risk group (Table 1). Among NHW patients, these associations were attenuated (Intermediate vs Low: OR=1.14, 95%CI: 0.95, 1.36; High vs Low: OR=1.23, 95%CI: 0.87, 1.75). However, among NHB women, the associations were more pronounced (Intermediate vs Low: OR=1.48, 95%CI: 1.05, 2.09; High vs Low: OR=1.56, 95%CI: 0.88, 2.76). We did not observe associations between NO2 and ODX groups. Conclusions Our preliminary results suggest that air pollution, particularly PM2.5, may influence tumor aggressiveness. Moreover, this association may differentially impact NHB breast cancer patients, who are also more likely to live in areas with higher air pollution.
| Overall | Non-Hispanic White | Non-Hispanic Black | ||||||||
| Oncotype DX Group | Pollutant | N | OR | 95%CI | N | OR | 95%CI | N | OR | 95%CI |
| Low | PM2.5 | 6,985 | 1.00 | Ref. | 5,363 | 1.00 | Ref. | 1,622 | 1.00 | Ref. |
| Intermediate | PM2.5 | 6,985 | 1.20 | 1.02,1.41 | 5,363 | 1.14 | 0.95,1.36 | 1,622 | 1.48 | 1.05,2.09 |
| High | PM2.5 | 6,985 | 1.31 | 0.97,1.76 | 5,363 | 1.23 | 0.87,1.75 | 1,622 | 1.56 | 0.88,2.76 |
| Low | NO2 | 6,985 | 1.00 | Ref. | 5,363 | 1.00 | Ref. | 1,622 | 1.00 | Ref. |
| Intermediate | NO2 | 6,985 | 1.02 | 0.99,1.06 | 5,363 | 1.02 | 0.98,1.07 | 1,622 | 1.03 | 0.96,1.11 |
| HIgh | NO2 | 6,985 | 0.97 | 0.91,1.04 | 5,363 | 0.94 | 0.86,1.03 | 1,622 | 1.03 | 0.91,1.16 |
Presentation numberPS4-09-29
Variation in weight loss and intervention engagement in the Breast Cancer Weight Loss (BWEL) trial by language and ethnicity (Alliance)
Ashley Odai-Afotey, Dana-Farber Cancer Institute, Boston, MA
A. Odai-Afotey1, K. V. Ballman2, L. M. McCall3, C. Cao1, C. Alfano4, V. Bernstein5, T. E. Crane6, L. M. Delahanty7, L. Frank1, P. J. Goodwin8, O. Hahn9, D. L. Hershman10, J. O. Hopkins11, M. Irwin12, E. L. Mayer1, L. Minasian13, L. Nebeling14, M. Neuhouser15, E. D. Paskett16, P. A. Spears17, V. Stearns18, C. A. Thomson19, T. A. Wadden20, A. Weiss21, J. White22, C. Hudis23, E. P. Winer24, L. Carey25, A. H. Partridge1, J. Ligibel1; 1Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, 3Alliance Statistics and Data Management Center, Duke University, Durham, NC, 4Cancer Institute, Northwell Health, New Hyde Park, NY, 5Department of Medical Oncology, BC Cancer/University of British Columbia, Victoria, BC, CANADA, 6Department of Medical Oncology, Miller School of Medicine, University of Miami, Miami, FL, 7Diabetes Research Center, Massachusetts General Hospital, Boston, MA, 8Department of Medical Oncology, Sinai Health/Lunenfeld-Tananbaum Research Institute, University of Toronto, Toronto, ON, CANADA, 9Department of Medical Oncology, UChicago Medicine Comprehensive Cancer Center, Chicago, IL, 10Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, NY, 11Department of Medical Oncology, SCOR NCORP/Novant Health Cancer Institute, Kernersville, NC, 12Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, 13Division of Cancer Prevention, National Cancer Institute, National Institute of Health, Bethesda, MD, 14Division of Cancer Control and Population Sciences, National Cancer Institute, National Institute of Health, Bethesda, MD, 15Cancer Prevention Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 16Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, 17Office of Community Outreach and Engagement, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 18Department of Medicine, Weill Cornell Medical Center, New York, NY, 19Health Promotion Sciences, College of Public Health, University of Arizona, Tucson, AZ, 20Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 21Department of Surgery, University of Rochester, Rochester, NY, 22Department of Radiation Oncology, Kansas University Medical Center, Kansas City, KS, 23Breast Medicine Service, Solid Tumor Division, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 24Department of Medical Oncology, Yale Cancer Center, New Haven, CT, 25Department of Medicine, Lineberger Comprehensive Cancer Center, Chapel Hill, NC.
BACKGROUND: Obesity is prevalent in Hispanic patients (pts) with breast cancer (BC), but this population is often underrepresented in studies of weight loss interventions (WLIs). The BWEL trial (Alliance A011401; NCT02750826) is a phase III randomized trial testing the impact of a 2-year (yr) telephone-based WLI on invasive disease-free survival in pts with stage II-III HER2-negative BC and a body mass index (BMI) > 27 kg/m2. At 12 months (mo), Hispanic pts lost less weight than White pts on the WLI. Here, we evaluate differences in intervention engagement over the first 12 mo by language preference and ethnicity among BWEL pts on the WLI. METHODS: BWEL randomized pts 1:1 to WLI + health education (HE) or HE alone. WLI +HE pts received semi-structured telephone-based health coaching in English or Spanish, in addition to access to an activity monitor, wireless scale and meal replacement shakes. Pts self-reported ethnicity and their preferred language for the WLI. Kruskal Wallis and chi-square tests were used to assess language and ethnicity differences in demographic and treatment factors for continuous and categorical variables, respectively. Percent weight change and mean values for intervention attrition, number of calls, call duration, number of shake shipments, and frequency of activity monitor use and weight tracking over 12 mo were compared between Spanish-preferred Hispanic pts (SH), English-preferred Hispanic (EH) pts and non-Hispanic (NH) pts using ANOVA. Pts with missing language, ethnicity or 12 mo weight data were excluded from analysis. RESULTS: 1591 pts were randomized to the WLI + HE from 08/2018 to 02/2021. 80.5% of pts were White, 12.8% Black and 7.1% Hispanic. 1190 pts had 12 mo weight and language available. Among the 87 WLI + HE pts who had available data and identified as Hispanic, 34% were SH and 66% EH. SH WLI pts were younger (in yrs: 49.5 SH, 51.1 EH, 53.9 NH, p=0.006) and had lower BMI (32.2 SH, 35.1 EH, 34.5 NH, p=0.007). At 12 mo, SH WLI pts lost 2.4% of baseline body weight vs 3.3% in EH and 5.1% in NH (p=0.051) (Table). Compared with EH and NH WLI pts, SH WLI pts participated in fewer calls (15.7 SH, 21.1 EH, 24.3 NH, p< 0.0001), had fewer days of activity monitor use (134.2 SH, 177.3 EH, 205.3 NH, p=0.015) and fewer days of weight tracking (65.3 SH, 96.8 EH, 148.2 NH, p<0.0001), but had more shake shipments (3.9 SH, 3.5 EH, 3.0 NH, p=0.015). There were no significant differences in attrition rate or call duration by language preference or ethnicity. CONCLUSIONS: In a large phase III WLI study in breast cancer patients, engagement with the WLI appeared lower in SH pts, despite provision of the intervention in Spanish. More work is needed to improve accessibility and efficacy of WLI studies among non-English speaking populations. Support: U10CA180821, U10CA180882, UG1CA189823; https://acknowledgments.alliancefound.org.
| Type of 12 month Outcome
Mean (SD)
|
Non-Hispanic (NH) pts
N=1103
|
English-preferred Hispanic (EH) pts
N=57
|
Spanish-preferred Hispanic (SH) pts
N=30
|
p-value
(ANOVA)
|
| Withdrew from intervention
n (%)
|
46 (3.3%) | 1 (1.5%) | 2 (5.1%) | 0.53 |
| Percent weight change | -5.1% (7.9) | -3.3% (6.7) | -2.4% (4.9) | 0.051 |
| No. of calls | 24.3 (6.3) | 21.1 (8.0) | 15.7 (9.9) | <0.0001 |
| Call duration (minutes) | 34.2 (6.8) | 32.9 (5.8) | 32.1 (9.3) | 0.09 |
| Days of Fitbit activity monitor use | 205.3 (116.2) | 177.3 (130.3) | 134.2 (109.4) | 0.015 |
| Days of weight tracking | 148.2 (107.8) | 96.8 (94.5) | 65.3 (82.8) | <0.0001 |
| No. of shake shipments | 3.0 (1.9) | 3.5 (2.4) | 3.9 (1.2) | 0.015 |
Presentation numberPS4-09-30
Mapping Inequity: Racial Variations in Breast Cancer Hospital Admissions and Clinical Outcomes in the U.S.
Tijin Ann Mathew, Southeast Health, Dothan, AL
T. A. Mathew1, A. Al Sharie2, M. Gevorgian3, B. Easow1, S. Valasareddi4; 1Internal Medicine, Hematology/Oncology, Southeast Health, Dothan, AL, 2Internal Medicine, Southeast Health, Dothan, AL, 3Internal Medicined, Alabama College Of Osteopathic Medicine, Dothan, AL, 4Hematology Oncology, Southeast Health Cancer Center, Dothan, AL.
Introduction: Race may affect the clinical outcomes in breast cancer. We aimed to identify how race impacts admission, clinical outcomes, and disposition of breast cancer patients. Methods: We performed a retrospective analysis of breast cancer-related hospital admissions using the 2022 National Inpatient Sample (NIS). Patients were stratified according to race into the following groups: White, African American, Hispanic, Asian or Pacific Islander, Native American, and Other. The data analysis was performed using STATA/BE version 18.5. ANOVA, Chi-square, and multivariate logistic regression analysis were performed to evaluate the impact of race on the clinical outcome of the study population. Results: In 2022, there were 33,597 hospitalizations for breast cancer in the United States; of these, 64.7% were White, 16.7% African-American, 11.1% Hispanic, 3.7% Asian/Pacific Islander, 0.4% Native American, and 2.7% Other. The average age at breast cancer hospitalization showed significant racial differences, with means of 67 years for Whites, 62 years for African-Americans, 60 years for Hispanics, 61 years for Asian/Pacific Islanders and Others, and 59 years for Native Americans (p < 0.001). In-hospital mortality rates for breast cancer differed significantly by race, ranging from 5.0% in Whites to 6.0% in African-Americans (p = 0.02). Regarding the in-hospital outcomes with comparsion to White patients, African-Americans had substantially higher odds of AKI (OR = 1.42, p < 0.001), while patients categorized as “Other” had significantly lower odds (OR = 0.82, p = 0.046); no significant differences were observed for Hispanics, Asian/Pacific Islanders, or Native Americans. Referencing Whites, African-Americans had significantly higher odds of pulmonary embolism (OR = 1.27, p < 0.001), whereas Hispanics (OR = 0.79, p = 0.004), Asian/Pacific Islanders (OR = 0.53, p < 0.001), Native Americans (OR = 0.29, p = 0.036), and Others (OR = 0.68, p = 0.018) had significantly lower odds. The distribution of hospital teaching status varied by race, with the majority of African-American, Hispanic, and Asian/Pacific Islander receiving care at teaching hospitals (over 83%), compared to 76.6% of White patients and 73.4% of Native Americans; non-teaching hospital admissions were highest among Native Americans (26.6%) and Whites (25.4%). Hospital region varied significantly by race (p < 0.001), with most African-American patients admitted in the South (54.2%), Whites primarily in the South (36.1%) and Midwest (24.8%), and Asians/Pacific Islanders predominantly in the West (50.5%). Median household income also differed by race (p < 0.001), with nearly half of African-American patients (46.7%) in the lowest income quartile, compared to 20.6% of Whites and 49.6% of Asians/Pacific Islanders in the third quartile. Discharge disposition showed racial variation (p < 0.001), with Hispanics and Asians/Pacific Islanders more often discharged home (60.2% and 58.0%, respectively) compared to Whites and African-Americans (~49.5%). Insurance status differed significantly (p < 0.001); Medicare coverage was highest among Whites (63.6%), while Medicaid was more common in African-Americans (19.7%) and Hispanics (27.2%), and private insurance predominated among Asians/Pacific Islanders (37.8%). Conclusion: Significant racial disparities exist in breast cancer hospitalizations across the United States, affecting patient age, outcomes, comorbidities, and healthcare access. African-American patients experience higher risks of complications such as AKI and pulmonary embolism and are more likely to be treated in teaching hospitals and lower-income regions. These findings highlight the need for targeted interventions to address racial inequities in breast cancer care and outcomes.
Presentation numberPS4-11-16
Treatment Delays and Testing Disparities May Contribute to Racial Differences in Node-Positive ER+/HER2- Breast Cancer Outcomes
Lesley Coe, Albert Einstein College of Medicine, Bronx, NY
L. Coe1, M. Wood1, S. Jao2, F. Bhimani2, M. Sheckley2, E. Ravetch2, A. Gupta2, S. Feldman2, M. McEvoy2; 1Medical Student, Albert Einstein College of Medicine, Bronx, NY, 2Division of Breast Surgical Oncology, Montefiore Medical Center, Bronx, NY.
Background: Despite significant advances in breast cancer treatment, racial disparities in outcomes persist. Recent data from the National Cancer Database (NCDB) show Black patients with node-positive (N+) ER+/HER2- breast cancer receiving neoadjuvant chemotherapy (NAC) had worse overall survival (OS) compared to White patients (HR 1.15, p = 0.001) (Moldoveanu 2024). To understand these disparities, we conducted a comparative analysis of N+ ER+/HER2- breast cancer patients treated at a single large academic institution in the Bronx and those represented in the NCDB. Methods: We conducted a retrospective chart review of patients with N+ ER+/HER2- breast cancer (2018-2021) treated with NAC at an institution in the Bronx. Separately, we identified patients meeting the same criteria in the NCDB Participant Use File. Cohorts were analyzed separately, then compared to assess differences. We examined potential drivers of disparities such as molecular assay use, treatment delays, and income.Results: Among the 38,134 patients identified in the NCDB, 65% were White, 16% Black, 11% Hispanic, 4% Asian, and 5% other. Black patients were less likely than White patients to receive Oncotype DX testing (6.9% vs. 8.9%, p < 0.001). Black patients had longer times from diagnosis to the start of chemotherapy (43 vs. 38) and surgery (163 vs. 154 days) (p < 0.001) when compared to White patients. Furthermore, they were 2.6 times more likely to fall into the lowest income quartile (<$46,277, p < 0.001), a status also associated with longer times to chemotherapy (42 vs. 36 days) and surgery (163 vs. 159 days) (p < 0.001) compared to those earning over $74,063 annually. Patients in the slowest quartile for time to chemotherapy had a lower rate of nodal response compared to those in the fastest quartile (27% vs. 34%, p < 0.001). Of the 99 patients treated at the Bronx institution, 48% were Hispanic, 31% Black, 16% White, 2% Asian, and 3% other. Bronx patients (83.8% non-White) experienced longer times from diagnosis to chemotherapy (48 vs. 39 days, p = 0.02) and to surgery (217 vs. 156 days, p < 0.001) compared to the NCDB. Conclusion: Our findings highlight that delays in treatment initiation and lower utilization of Oncotype DX among Black patients may contribute to racial disparities in breast cancer outcomes. As demonstrated by the NCDB and Bronx cohort, non-White and low-income patients experience delays in initiating treatment and undergoing surgery when compared to White patients and higher-income patients. In the Bronx cohort, these delays were more pronounced than those observed in the national dataset. Longer time to chemotherapy was associated with reduced nodal response rates, underscoring the potential clinical consequences of these delays. Though these delays may in part be due to structural determinants of health, further studies are needed to identify all contributing factors.
| NCDB (Race) | White (n=24,701) | Black (n=6,223) | p-value |
| Age | 57.6 ± 13.8 | 54.9 ± 13.6 | < 0.001 |
| Oncotype DX Testing Ordered (%) | 2201 (8.9%) | 432 (6.9%) | < 0.001 |
| Ki-67 Ordered (%) | 13060 (52.9%) | 3272 (52.6%) | 0.72 |
| Medicaid (%) | 1898 (7.7%) | 1197 (19.2%) | < 0.001 |
| Medicare (%) | 7886 (31.9%) | 1559 (25.1%) | < 0.001 |
| Private Insurance (%) | 14053 (56.9%) | 3080 (49.5%) | < 0.001 |
| < $46,277 Income (%) | 3153 (13.1%) | 1843 (34.9%) | < 0.001 |
| > 15.3% No HSD (%) | 3061 (12.4%) | 1816 (29.2%) | < 0.001 |
| Time to Chemotherapy (Median Days (IQR)) | 38 (28-55) | 43 (31-62) | < 0.001 |
| Time to Surgery (Median Days (IQR)) | 154 (6-208) | 163 (4-218) | < 0.001 |
| NCDB (Income) | < $46k (n=5,235) | > $74k (n=12,802) | p-value |
| Oncotype DX Testing Ordered (%) | 407 (7.8%) | 1097 (8.6%) | 0.08 |
| Ki-67 Ordered (%) | 2682 (51.2%) | 7242 (56.6%) | < 0.001 |
| Time to Chemotherapy (Median Days (IQR)) | 42 (30-62) | 36 (25-50) | < 0.001 |
| Time to Surgery (Median Days (IQR)) | 163 (9-220) | 159 (4-205) | < 0.001 |
| Bronx vs. NCDB | Bronx (n=99) | NCDB (n=38,134) | p-value |
| Age | 58.1 ± 12.5 | 56.3 ± 13.7 | |
| Asian (%) | 2 (2%) | 1788 (5%) | |
| Black (%) | 31 (31%) | 6223 (16%) | |
| White (not Hispanic) (%) | 16 (16%) | 24701 (65%) | |
| Hispanic (%) | 48 (47%) | 1421 (4%) | |
| Other (%) | 3 (3%) | 1788 (5%) | |
| Time to Chemotherapy (Median Days (IQR)) | 48 (33-68) | 39 (28-56) | 0.02 |
| Time to Surgery (Median Days (IQR)) | 217 (187-259) | 156 (6-210) | < 0.001 |
Presentation numberPS4-11-18
Financial Toxicity in Breast Cancer Patients Assessed by the COST Score: A Systematic Review and Meta-analysis
Natalia Cristina C. Nunes, Brazilian National Cancer Institute – INCA, Rio de Janeiro, Brazil
N. C. Nunes1, J. da Silva1, K. S. de Medeiros2, G. Hora1, M. Teixeira1, L. Leite1, L. Zanetti1, B. Linhares3, A. de Melo1; 1Division of Clinical Research, Brazilian National Cancer Institute – INCA, Rio de Janeiro, BRAZIL, 2Institute of Education, Resarch and Innovation,, Liga Contra o Câncer, Rio Grande do Norte, BRAZIL, 3Clinical Research, Instituto Americas, Rio de Janeiro, BRAZIL.
Financial Toxicity in Breast Cancer Patients Assessed by the COST Score: A Systematic Review and Meta-analysis Background:Financial toxicity (FT) refers to the negative impact on patients and their families caused by significant financial strain resulting from the diagnosis or treatment of illnesses such as cancer. This effect can significantly affect their quality of life, survival, or daily functioning. The Comprehensive Score for Financial Toxicity (COST) is a validated patient-reported outcome measure developed to assess FT in cancer patients. COST scores range from 0 (worst FT) to 44 (no FT). The tool has been translated into over 30 languages and is now used globally in diverse healthcare systems. Breast cancer is associated with a high risk of FT due to prolonged treatment duration, long-term toxicities, and its frequent occurrence in working-age women. Although many studies have evaluated FT in this population, reported prevalence and methodological approaches vary widely. This study aims to determine the FT rate among breast cancer patients using the COST tool and to identify particularly vulnerable patient populations. Methods: A systematic review and meta-analysis (registered in PROSPERO, ID 1084262) was performed following PRISMA guidelines. Searches were performed in PubMed, Cochrane, LILACS and Embase. Eligible studies included breast cancer patients assessed with the COST tool. Data on FT prevalence and mean or median COST scores were extracted. Random-effects models were used to calculate pooled estimates. Heterogeneity was quantified using the I² statistic. Meta-regression and a bubble plot were used to assess temporal trends. The Newcastle-Ottawa Scale was used to assess the risk of bias. Results: Thirteen studies published between 2016 and 2024 were included, representing over 8,400 patients across China (n=5), the United States (n=6), South Korea (n=1), and Indonesia (n=1). Sample sizes ranged from 99 to 4,297. Most participants were women of working age, with ages ranging from 43 to 58 years. Educational background and employment status varied widely, with employment rates ranging from 7.7% to 70%. Insurance types reflected local health system structures, including public, private, and uninsured populations. Systemic therapies commonly include chemotherapy, endocrine therapy, and targeted agents. The pooled prevalence of FT was 66.9% (95% CI: 45.1-88.7%; I2 99.9%). The pooled mean COST score was 23.87 (95% CI: 21.25-26.50; I2 99.8%), reflecting moderate financial distress. A slight but non-significant decrease in COST scores over time was observed (regression coefficient: -0.40; 95% CI: -1.46 to 0.66; p = 0.432), suggesting a possible worsening of FT in recent years. No study was classified as having a “high risk” of bias. Conclusion: FT is highly prevalent among breast cancer patients, with a moderate burden as observed in COST scores. These findings highlight the need for targeted financial support strategies and health policy interventions to mitigate FT, particularly in vulnerable populations. Keyword: Breast cancer; Financial toxicity; COST; Meta-analysis; Systematic reviews.
Presentation numberPS4-11-19
Investigating Genomic and Treatment Heterogeneity in HER2+ and Triple Negative Breast Cancers
Sarah C Van Alsten, University of North Carolina at Chapel Hill, Chapel Hill, NC
S. C. Van Alsten1, C. Clayhold2, K. E. Reeder-Hayes1, K. A. Hoadley3, M. A. Troester4; 1Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 2School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 3Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 4Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC.
Background: Black women have a higher incidence of aggressive breast cancer subtypes, including both HER2+ and triple negative breast cancers (TNBCs), and this increased incidence contributes to racial disparities in outcomes. Differences in tumor biology and treatment have been well-studied for estrogen receptor (ER) positive/ HER2- disease, but less attention has been given to the interplay of biologic and access differences in explaining racial differences in outcomes of HER2+ and TNBC. We hypothesized that differences in tumor biological features and treatment patterns among ER-negative cancers are important contributors to disparate outcomes. Methods: The Carolina Breast Cancer Study Phase 3 (diagnosed 2008-2013) included clinical and molecular data from 391 women with TNBC and 451 women with HER2+ breast cancer (305 HR+/HER2+, 144 HR-/HER2+; 2 missing HR status). For TNBC cases, days between diagnosis and first course therapy were abstracted from medical records. For HER2+ cases, receipt and time to receipt of trastuzumab were abstracted along with receipt of endocrine therapy (for HR+/HER2+ cases). RNA expression was assessed for a panel of 219 genes [including PAM50 subtype, immune response, p53 status and proliferation] on the NanoString platform. Within each clinical subtype (HER2+ or TNBC) we applied consensus clustering to identify de novo genomic subtypes. We estimated odds ratios (ORs) and 95% confidence intervals (CIs) for the associations between self-reported race, trastuzumab/endocrine therapy receipt [in HER2+ cases only], and molecular subtypes using logistic and multinomial logistic regression. Differences in time to therapy were assessed by Wilcoxon tests. Separately in HER2+ cases and TNBCs, we used Cox regression to estimate unadjusted race-, treatment- and subtype-specific hazard ratios (HRs) and 95% CIs of recurrence over 10 years of follow up. Results: Among TNBCs, we observed substantial gene expression heterogeneity, including three subtypes: 1) one ‘Immunogenic’ type with high expression of immune genes, 2) one ‘Luminal/Androgen Receptor (LAR)’ with higher proportions of Luminal A/B tumors and higher expression of estrogen signaling genes, and 3) one ‘Basal-like (BL)’ group with higher expression of proliferation genes. Black women with TNBC were significantly less likely than non-Black women to present in the Immunogenic (39% vs 43%; OR= 0.67, 95% CI = 0.45 – 0.98) and LAR classes (20% vs 27%; OR = 0.57, 95% CI = 0.38 – 0.86) relative to the BL class, and had longer median time to first treatment (26 vs 18 days, p < 0.001). However, there was no difference in risk of recurrence by race among TNBCs (HR = 1.06, 95% CI = 0.65, 1.73). In contrast, Black women with HER2+ disease had a significantly higher risk of recurrence than non-Black HER2+ women (HR = 1.79, 95% CI = 1.08, 2.98), with differences most pronounced among HR-/HER2+s (HR = 2.71, 95% CI = 1.08, 6.84). A similar proportion (89% Black vs. 88% non-Black) of HER2+ cases received guideline-concordant trastuzumab, with no association between race and trastuzumab receipt (OR = 1.16, 95% CI = 0.65 – 2.07) nor between race and time to trastuzumab receipt (Black women mean = 73 days vs non-Black mean= 61 days, p = 0.15). Likewise, 90% of both Black and non-Black HR+/HER2+ cases received endocrine therapy, with no association between race and endocrine receipt in this subgroup. Conclusions: Black women with HER2+ breast cancers experienced worse prognosis, despite similar guideline-concordant treatment receipt and treatment timeliness. Discontinuation of trastuzumab and other treatment differences and/or access issues merit further consideration to understand HER2+ disparities. We did not find evidence of racial disparities in outcomes among TNBCs, but TNBCs have meaningful biological heterogeneity in outcomes.
Presentation numberPS4-11-20
Comparative Adherence to Adjuvant Hormonal Therapy and Associated Mortality in Male and Female Veterans with Breast Cancer: A Nationwide Cohort Analysis
Sonika Prasad, Saint Louis Univeristy, Saint Louis, MO
S. Prasad1, J. Gruber2, R. Aft3, A. Naaseh4, M. Schoen5; 1Hematology Oncology, Saint Louis Univeristy, Saint Louis, MO, 2Statistics, The Veterans Research and Education Foundation of St. Louis, Saint Louis, MO, 3Surgical Oncology, Washington University, Saint Louis, MO, 4SURGERY, Washington University, Saint Louis, MO, 5Hematology Oncology, St. Louis Veterans Affairs Medical Center, Saint Louis, MO.
Background: Male breast cancer is rare and understudied,with most treatment paradigms based on evidence from females. Existing studies suggest that men may have lower adherence to hormonal therapy, but large-scale data are limited. Hormonal therapy adherence significantly impacts outcomes in breast cancer treatment. However, differences in adherence patterns between male and female veterans remain understudied. Understanding these patterns is crucial for developing targeted interventions to improve adherence and patient outcomes.Methods: We conducted a retrospective cohort study using national data from the Veterans Health Administration (VHA) Informatics and Computing Infrastructure database. Veterans diagnosed with breast cancer and prescribed adjuvant hormone therapy (tamoxifen,anastrozole, letrozole, or exemestane) between January 1, 2005, and December 31, 2019,were included. Demographic and clinical characteristics were collected, and adherence was assessed. Multivariable logistic regression was utilized to identify factors associated with adherence, comparing male and female veterans.Results: A total of 7,155 veterans (1,139 males and 6,016 females) were analyzed. Male veterans were older at initial prescription (69.8 ± 10.0 years vs. 58.2 ± 10.7 years for females, p<0.001) and had higher adherence rates (74% vs. 65%, p<0.001). Multivariable regression indicated that adherence significantly increased with older age (Adj. OR: 1.03, 95% CI: 1.02-1.03;p<0.001) and among non-Hispanic/Latino individuals (Adj. OR: 1.39, 95% CI: 1.10-1.75;p=0.005), but decreased among Black veterans compared to White veterans (Adj. OR: 0.60,95% CI: 0.53-0.67; p<0.001), divorced/separated individuals (Adj. OR: 0.86, 95% CI: 0.75-1.00; p=0.049), widowed individuals (Adj. OR: 0.76, 95% CI: 0.60-0.95; p=0.018), and those with unknown cancer stage (Adj. OR: 0.80, 95% CI: 0.65-0.98; p=0.030). After adjustment, gender alone was not predictive of adherence (Adj. OR: 1.02, 95% CI: 0.87-1.20; p=0.771). Importantly, adherence to hormone therapy was significantly associated with reduced mortality risk (Adj. OR: 0.76, 95% CI: 0.67-0.86; p<0.001).Conclusion: Adherence to hormone therapy significantly improves survival outcomes among veterans with breast cancer. While male veterans demonstrated higher adherence rates, gender was not independently associated with adherence after adjustment. Targeted interventions addressing adherence disparities—particularly among younger, Black, divorced/widowed individuals,and patients with uncertain staging—could improve outcomes and reduce health disparities.
Presentation numberPS4-11-21
Breast Cancer Trends and Outcomes Among Young Hispanic and Asian/Pacific Islander Women Age < 40 in the National Cancer Database (NCDB) adjusted for U.S. population data
Poornima Saha, Endeavor Health, Evanston, IL
P. Saha1, K. Kuchta2, D. Thompson3, K. A. Yao2; 1Medicine, Hematology Oncology, Endeavor Health, Evanston, IL, 2Surgery, Endeavor Health, Evanston, IL, 3Surgery, University of Chicago, Chicago, IL.
Title: Breast Cancer Trends and Outcomes Among Young Hispanic and Asian/Pacific Islander Women Age < 40 in the National Cancer Database (NCDB) adjusted for U.S. population data Authors: Poornima Saha, MD, Kristine Kuchta, MS, Danielle Thompson, MD, Katharine A. Yao, MD, MS Background Breast cancer incidence is rising nationally, particularly among young women. While many studies have documented the rising incidence in Black women, few studies have investigated these trends in Asian/Pacific Islanders (API) and Hispanic women. The primary objective of this study was to analyze trends in the incidence of breast cancer in API and Hispanic women under forty years old adjusted for U.S. population data. Methods This study employed a retrospective cohort design to analyze trends in breast cancer diagnoses among women under and over forty years of age from 2010 to 2022 using data from the National Cancer Database (NCDB). The NCDB was queried for women diagnosed with breast cancer from 2010-2022. Patients were stratified by age 40 at time of breast cancer diagnosis. Breast cancer incidence data from the NCDB were analyzed using U.S. Census data population estimates. Results: Of 4,195,384 women (mean age 61±13), 188,311 were under 40 and 4,007,073 were over 40. Among women with breast cancer under age forty, 63.5% were White, 16.4% Black, 11.2% Hispanic, 6.3% API, and 2.6% Non-Hispanic Other. In contrast, in women with breast cancer diagnosed at age ≥40, a larger majority (77.5%) were white with lower proportions of Black (11.3%) and Hispanic (5.6%) patients. Women age 40 to have human epidermal growth factor-2 negative (Her2-) breast cancers (16.2% vs 8.1%) and triple negative breast cancers (TNBC) (12.8% vs 6.6%). Women age 40 to be diagnosed with grade 3 tumors (50.8% vs 28.7%), Stage III-IV disease (20.0% vs 11.1%), and be treated with chemotherapy (70.5% vs 31.0%). From 2010 to 2022, incidence in young women rose 15.4% in all races/ethnicities. Broken down by subgroup, the incidence rose 19.9% in white, 5.3% in black, 27.7% in Hispanic and 18.9% in API women. These findings confirm an increase in incidence rates among young women across all racial/ethnic groups. Our study shows the greatest relative increase in breast cancer rates in young Hispanic women and Black women experiencing the lowest relative increase. Conclusions:This study examined trends and outcomes in young minority women diagnosed with breast cancer age < 40. Hispanic women < 40 experienced the highest rise in incidence at 27.7% over 14 years followed by API women < 40 with 18.9% rate of incidence. The dramatic increase in breast cancer incidence among young women of color, particularly Hispanic and API women, is notable and warrants further research into factors contributing to these disparities.
Presentation numberPS4-11-22
Molecular characteristics of TNBC and pathologic response to chemoimmunotherapy: a focus on racial disparities
Melanie Sheen, Ochsner MD Anderson Cancer Center, New Orleans, LA
M. Sheen1, E. Biggs2, C. Taylor1, M. Lakey3, R. DeArmitt4, R. Emnance5, M. Bratton4; 1Oncology, Ochsner MD Anderson Cancer Center, New Orleans, LA, 2Center for Outcomes Research, Ochsner Health Institute, New Orleans, LA, 3Pathology, Ochsner MD Anderson Cancer Center, New Orleans, LA, 4Biospecimen & Core Research Lab, Ochsner Health Institute, New Orleans, LA, 5Biospecimen & Core Research Lab, Ochsner MD Anderson Cancer Center, New Orleans, LA.
Background: Triple-negative breast cancer (TNBC) is a high-risk subtype characterized by aggressive clinical behavior with limited targeted therapies. The addition of pembrolizumab to neoadjuvant chemotherapy (NACT) has improved pathologic complete response (pCR) rates in early-stage TNBC. However, racial disparities in outcomes remain, and limited data exist on how these disparities may impact pathologic response to chemoimmunotherapy. Our prior retrospective analysis revealed statistically significant differences in pCR among Black or African-American (AA) women following NACT. This study evaluates genomic differences among TNBC patients treated with a pembrolizumab-based NACT regimen at a regional health system, aiming to identify genetic markers that may predict treatment response and provide insight into observed racial disparities.Methods: Patient records from Ochsner Health, a regional healthcare network, were reviewed to identify individuals with early-stage TNBC treated with pembrolizumab-based NACT from 2020-2024. Exclusion criteria included unknown receptor status and age <18 years. Eligible patients were required to have archived formalin-fixed paraffin-embedded (FFPE) biopsy samples available at the main campus. Consented participants underwent tumor DNA sequencing performed by Tempus using their archived diagnostic tissue.Results: Archived FFPE tumor samples were obtained from 50 TNBC patients treated with NACT and pembrolizumab. Of these, 43 samples passed Tempus quality control measures, and 40 were included in the final analysis (excluding 1 patient who identified as “Other” race and 2 without definitive surgery or pCR data). Among the 40 analyzed patients, 25 identified as AA and 15 as White. The average number of mutations detected per patient was 16.5 with an average of 18 mutations/patient in AA patients and 13 mutations/patient in White patients (Table).
| pCR | no pCR | p-value | Race | Mutation | No Mutation | p-value | ||||
| KMT2C/D | mutation | 16 | mutation | 1 | 0.005 | AA | 12 | 1 | 0.512 | |
| no mutation | 12 | no mutation | 11 | White | 5 | 10 | ||||
| LRP1B | mutation | 8 | mutation | 1 | 0.233 | AA | 4 | 21 | 0.255 | |
| no mutation | 20 | no mutation | 11 | White | 5 | 10 | ||||
| RANBP2 | mutation | 4 | mutation | 1 | 1.00 | AA | 1 | 24 | 0.056 | |
| no mutation | 24 | no mutation | 12 | White | 4 | 11 | ||||
| MCL1 | mutation | 8 | mutation | 3 | 1.00 | AA | 9 | 16 | 0.158 | |
| no mutation | 20 | no mutation | 9 | White | 2 | 13 | ||||
| CKS1B | mutation | 11 | mutation | 1 | 0.067 | AA | 7 | 18 | 0.736 | |
| no mutation | 17 | no mutation | 11 | White | 5 | 10 |
95% (38/40) of patients harbored mutations in the TP53 gene—substantially higher than reported in broader breast cancer cohorts. A correlation was observed between variant allele frequency (VAF) of TP53 mutations and pCR status (p=0.047). Conclusion: With small numbers, the prevalence of the most frequently mutated genes was not statistically significant. Mutations in KMT2C/D were statistically significant. These methyltransferase mutations may serve as biomarkers for immunotherapy response in TNBC given the high pCR rates seen in these, regardless of race. Response to immunotherapy among patients with these mutations is consistent with findings in other cancer types. Patients with a higher VAF of TP53 had worse pCR rates, regardless of race, suggesting that mutation burden may influence treatment response. As more data is collected, it may reveal insight into racially distinct genomic signatures that could serve as predictors of response to NACT with immunotherapy.
Presentation numberPS4-11-23
Correlates of Distance Traveled to Treatment Centers Among Breast Cancer Patients: The Role of Race/Ethnicity and Insurance Status
Diya Desai, Vanderbilt University, Nashville, TN
D. Desai; Medicine, Health, and Society, Vanderbilt University, Nashville, TN.
Background: Black women and uninsured patients have a higher breast cancer mortality rate and are often diagnosed at more advanced stages. Prior research suggests transportation is a barrier to completing recommended treatment. This study examines whether race/ethnicity and insurance status are associated with the distance traveled to breast cancer diagnosis and treatment facilities. Methods: The study employed data from the National Cancer Database (NCDB) from 2013-2022 and included patients who were diagnosed with any stage of breast cancer at Commission on Cancer-certified centers in the United States. Patients with missing information on their racial/ethnic group, distance traveled, or insurance status were not included. Six ordinal categories of distance traveled (100 miles) were compared across racial/ethnic groups and insurance coverage groups. Z-tests were run to compare each proportion against the white patient group and privately insured patient group, and a chi squared test was run to determine if there was a correlation between insurance status and distance traveled and race/ethnicity status and distance traveled. Results: There was a significantly higher proportion of Black, Hispanic, and Asian American and Pacific Islander (AAPI) patients who traveled under 5 miles to their treatment compared to White patients. 18.8% of Black patients traveled under 5 miles compared to 13% of White patients. However, American Indian and Alaskan Native (AI/AN) patients had a greater proportion of patients that traveled over 100 miles to receive a diagnosis or treatment compared to White patients. Uninsured people were most correlated with shorter distance traveled to facilities, followed by Medicaid then Medicare patients. People with other forms of government insurance had a higher proportion than privately insured patients that traveled 50-99 miles and over 100 miles to facilities. Conclusion: There is a correlation between race/ethnicity and distance traveled and between insurance status and distance traveled with racial and ethnic minority groups and uninsured patients being more correlated with traveling smaller distances. Racial and ethnic minority groups and uninsured patients may lack tools for extensive travel. Even though racial and ethnic minority populations and people without private insurance travel less far than white patients, disparities remain. Further research into how disparities stem from structural racism and shape access to health providers is necessary. Disclaimer: The NCDB is a joint project of the Commission on Cancer of the American College of Surgeons and the American Cancer Society. The data used in the study are derived from a de-identified NCDB file. The American College of Surgeons and the Commission on Cancer have not verified and are not responsible for the analytic or statistical methodology employed, or the conclusions drawn from these data by the investigator
Presentation numberPS4-11-24
Enhancing Quality of Care Using Project ECHO: Insights from Breast Density and Advanced Breast Cancer Treatment Programs
Allison Rosen, American Cancer Society, Atlanta, GA
A. Rosen1, M. Odom1, J. McBride2, C. Blackwater3, M. Black4, K. Marcelle1, B. Morris5, S. Sivendran6; 1Project ECHO, American Cancer Society, Atlanta, GA, 2Data and Impact Reporting Team, American Cancer Society, Atlanta, GA, 3Health-Related Social Needs Interventions, American Cancer Society, Atlanta, GA, 4Early Detection, American Cancer Society, Atlanta, GA, 5Navigation, American Cancer Society, Atlanta, GA, 6Cancer Treatment Support, American Cancer Society, Atlanta, GA.
As of 2020, more than 168,000 women in the United States were living with advanced breast cancer, which carries a 5-year relative survival rate of approximately 31%. Over the past decade, this survival rate has gradually improved, thanks to advances in treatment, earlier detection, and a deeper understanding of disease biology. With these developments—alongside regular updates to the National Comprehensive Cancer Network guidelines and growing awareness of risk factors such as breast density—care for patients with advanced breast cancer has become increasingly complex. To help ensure equitable access to emerging therapies and evolving standards of care, the American Cancer Society launched two ECHO (Extension for Community Healthcare Outcomes) programs: one focused on breast density and risk assessment, and the other aimed at enhancing treatment for advanced breast cancer. Project ECHO is an innovative telementoring model that moves knowledge instead of patients. Through an “all teach, all learn” approach, healthcare professionals enhance their skills and knowledge to improve patient outcomes across the cancer continuum through sharing evidence-based practices and resources. Through case-based learning and a series of interactive sessions, participants across the globe share, learn, and collaborate to ensure that health care professionals are equipped to provide the right care at the right time and place, giving everyone the opportunity to prevent, detect, treat, and survive cancer. The Addressing Breast Density & Risk in Primary Care ECHO, the first of the two breast-focused ECHO programs, addressed the unique needs of providing care to patients with dense breasts, family history, and genetic markers. The overall program goal was to advance the understanding of the role breast density plays in a patient’s overall cancer risk. By the end of the program, participants had the knowledge to communicate and integrate breast cancer risk assessment into primary care practice. This was achieved through six monthly sessions, held from November 2024 to April 2025 led by subject matter experts who delivered clinical presentations and facilitated interactive discussions. The ECHO reached a total of 121 unique participants across 31 states. Participant health systems were comprised of 50% urban, 32% suburban, and 18% rural communities. A participant echoed the program’s success with the comment on how they care for patients, “I counsel every patient now in a different manner. Discussing density is now routine, computing breast cancer (TC) risk scores as well.” The Improving Treatment of Advanced Breast Cancer ECHO program, launched in May and will conclude in November 2025, aims to address best practices in shared decision-making and high-quality treatment of advanced breast cancer. By the end of the program, multidisciplinary professionals will have the knowledge on strategies to address the optimal sequencing of systemic therapy, utilize novel radiotherapeutics for detecting and treating breast cancer, and leverage healthcare professionals to support the physical and psychosocial journey of patients undergoing treatment. In the first two sessions, a total of 100 unique attendees across 20 states participated. Upon completion, knowledge, confidence, and likelihood to recommend to colleagues will be reported. As advancements in the field of breast cancer accelerate, staying informed is no longer optional; it is essential. ECHO programs serve as a powerful catalyst for bridging knowledge gaps by connecting a global network of health care professionals. Through dynamic, peer-to-peer collaboration, these programs not only share best practices but they foster expertise, elevate clinical standards, and empower health care professionals to provide exceptional, evidence-based care to their patients.
Presentation numberPS4-11-25
Evaluating Underrepresentation in Breast Cancer Clinical Trial Enrollment at Yale Cancer Center: A Retrospective Demographic Study
Jessica Liu, Yale School of Medicine, New Haven, CT
G. Gong1, J. Liu1, M. Taylor2, S. Pandya3, C. Taborda2, J. Xie4, J. Parikh4, W. Wei2, M. Stefanou1, P. Kunz2, N. Fischbach2, T. Battaglia1, L. Mendez1, J. Gaddy2, I. Krop2, P. LoRusso1, M. Lustberg1, A. Silber1; 1Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Department of Internal Medicine, Division of Medical Oncology, Yale School of Medicine, New Haven, CT, 3Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, 4Yale College, Yale University, New Haven, CT.
Background: As therapeutic advances continue to reshape the standard of care, balanced representation in breast cancer clinical trials is essential to ensure that novel treatments benefit all populations. Yet, racial and ethnic minorities, older adults, non-English speakers (NES), and individuals with lower socioeconomic status remain underrepresented in trial cohorts. To address these imbalances, our team developed an automated Clinical Trial Patient Matching (CTPM) system powered by machine learning to identify eligible patients using real-time clinical data. To quantify existing gaps and assess outcomes of CTPM, we conducted a benchmarking analysis comparing the demographic characteristics of breast cancer clinical trial participants at Yale Cancer Center (YCC) over the past 10 years, before, and after CTPM implementation. Methods: Demographic data for breast cancer patients seen at YCC between 2013 and 2024 were obtained from three sources: the Epic electronic health record via the Yale New Haven Hospital (YNHH) Computational Health Platform, the Clinical Trials Management System (OnCore), and the CTPM system, which provides lists of trial-eligible patients identified using machine learning algorithms. We conducted a retrospective benchmarking analysis comparing the general breast cancer population, CTPM-identified eligible patients, and patients who consented to therapeutic clinical trials, as recorded in OnCore. Comparisons were stratified by time period (pre- and post-CTPM implementation: before July 2022 and before July 2024, respectively) and focused on age (18-39 and >70 years), race, ethnicity, primary language, geographic location, and insurance status. Results: Among 45,380 breast cancer patients treated between 2013 and 2024, baseline demographics included 1.89% young adults (18-39 years), 52.9% older adults (>70 years), 19.8% racial/ethnic minorities, 4.7% non-English speakers (NES), 3.2% Medicaid-insured, and 8.0% rural residents. Between 2022 and 2024, demographics shifted with a decrease in older adults (32.1%) and an increase among young adults (4.8%), racial/ethnic minorities (25.6%), NES patients (6.6%), Medicaid-insured (4.1%), and rural residents (8.5%). Among patients enrolled via manual chart review, 1.98% were aged 18-39, 36.1% were >70 years, 21.6% were racial/ethnic minorities, 4.6% Medicaid-insured, 5.8% NES, and 9.2% rural residents. Following implementation of the CTPM system across four breast cancer clinical trials, 662 patients were pre-screened and deemed eligible, and 99 patients were consented. Trial participation increased for young adults ages 18-39 (10.7%), racial/ethnic minorities (28.1%), and Medicaid-insured patients (7.2%). Conclusions: Manual chart review, while useful, does not provide a comprehensive analysis of breast cancer patients and obscures recent demographic changes. In contrast, the automated CTPM system identifies more patients eligible for clinical trials and reflects population trends. We successfully identified additional patients who met eligibility criteria from underrepresented groups such as young adults, racial/ethnic minorities, and Medicaid-insured patients. However, most patients who were identified and confirmed as eligible did not consent or enroll. These findings highlight the importance of combining automated patient identification with novel strategies to overcome barriers to consent and improve representative participation in breast cancer clinical trials.
Presentation numberPS4-11-26
First latin american consensus on the medical oncology management of early-stage hr+/her2− breast cancer: addressing regional disparities in spanish-speaking countries of central and south america
Denis Ulises Landaverde, Mexico Hospital, San Jose, Costa Rica
D. U. Landaverde1, F. Petracci2, A. Grano de Oro3, M. A. Bravo-Garzon4, M. I. Estevez5, I. González6, C. Arce-Salinas7, I. Veliz8, J. L. Amador9, O. Castillo10, V. Enciso11, L. Codas12, F. Valencia13, M. Galvez14, J. Valentin-Bejarano15, S. Sánchez16, I. V. Lemes-Pistón17, D. Hannois18, J. P. Miranda-Olivares19, G. Guillen20; 1Oncology, Mexico Hospital, San Jose, COSTA RICA, 2Oncology, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 3Oncology, Instituto de Oncología Dr. Heriberto Pieter, Santo Domingo, DOMINICAN REPUBLIC, 4Oncology, Centro de Tratamiento e Investigación sobre Cáncer Luis Carlos Sarmiento Angulo, Bogota, COLOMBIA, 5Oncology, Red Oncologica Dominicana Integral, Santo Domingo, DOMINICAN REPUBLIC, 6Oncology, Hospital San Juan de Dios, San Jose, COSTA RICA, 7Oncology, Instituto Nacional de Cancerología México, Ciudad de Mexico, MEXICO, 8Oncology, Hemato Oncologia de Panamá Especializada, Ciudad de Panama, PANAMA, 9Oncology, Centro Hemato Oncológico de Panamá, Ciudad de Panama, PANAMA, 10Oncology, Instituto Oncologíco Nacional, Ciudad de Panama, PANAMA, 11Oncology, Instituto de Prevision Social, Asuncion, PARAGUAY, 12Oncology, Instituto Nacional del Cancer, Asuncion, PARAGUAY, 13Oncology, Instituto Regional de Enfermedades Neoplasicas del Sur, Arequipa, PERU, 14Oncology, Instituto Regional de Enfermedades Neoplasicas del Norte, Libertad, PERU, 15Oncology, Instituto Redional de Enfermedades Neoplasicas del CENTRO, Concepcion, PERU, 16Oncology, Asociación Española y Hospital Policial, Montevideo, URUGUAY, 17Oncology, Cooperativa Médica de Rivera, Rivera, URUGUAY, 18Oncology, Clínica INDISA, Providencia, CHILE, 19Oncology, Hospital Clínico de la Fuerza Aérea de Chile, Santiago, CHILE, 20Oncology, Sociedad de Lucha Contra el Cáncer del Ecuador, Manabi, ECUADOR.
Background: Despite the global progress in early-stage hormone receptor-positive, HER2-negative (HR+/HER2−) breast cancer management, Latin America faces persistent and significant disparities in access to genomic testing, systemic therapies, fertility preservation, and multidisciplinary care. This work represents the first Latin American consensus developed exclusively by experts from Spanish-speaking countries in Central and South America, aiming to adapt international recommendations to the realities of the region and reduce inequities in care delivery.Methods: A systematic review of the literature was performed focusing on international guidelines (ESMO, ASCO, NCCN, SEOM). A multidisciplinary panel of 24 oncology experts from 11 Latin American countries participated in a Delphi-based consensus process. Statements were rated on a 9-point Likert scale; consensus was defined as a mean score ≥7.0 with ≤1 outlier. Data were collected using REDCap, and descriptive statistics were used for analysis.Results: Consensus was reached on 90% of 32 proposed statements. Key recommendations included the use of clinical-pathological and molecular factors (nodal status, tumor grade, ER, Ki-67, gene signatures) for risk stratification and adjuvant therapy decision-making. Genomic assays were endorsed for selected cases, though their availability remains limited or restricted to the private sector in most countries. The panel agreed that testing should be omitted in very low clinical-risk tumors and in patients with ≥4 positive nodes. The consensus supports ovarian suppression plus endocrine therapy for premenopausal women at high risk, and highlights the unmet need for routine fertility preservation counseling, sexual health management, and access to imaging and genetic counseling. The experts underscored the regional heterogeneity in resource availability, infrastructure, and bureaucratic barriers as major obstacles to equity in breast cancer care.Conclusions: This is the first regional consensus developed by Spanish-speaking oncology experts from both Central and South America to address the challenges of HR+/HER2− early breast cancer in Latin America. The recommendations reflect the urgent need to bridge the gaps in access, infrastructure, and implementationacross diverse healthcare systems. This consensus represents a critical step toward promoting contextualized, equitable, and evidence-based oncology care throughout the region.
Presentation numberPS4-11-27
Acceptability of a web-based narrative video intervention to promote clinical trial decision making among Black women with breast cancer
Ryan Nguyen, University of Illinois Chicago, Chicago, IL
N. Hippalgaonkar1, D. Patel1, C. Hollahan2, A. White3, M. Carrasquillo3, M. Karan3, R. Warren4, M. Kromm4, G. Vass4, P. Khosla5, V. Henderson6, K. Hoskins1, L. Carnahan2, R. Nguyen1; 1Medicine, University of Illinois Chicago, Chicago, IL, 2School of Public Health, University of Illinois Chicago, Chicago, IL, 3Cancer Center, University of Illinois Chicago, Chicago, IL, 4Filming, Solid Line Media, Chicago, IL, 5Medicine, Sinai Chicago, Chicago, IL, 6Cancer Prevention Program, Fred Hutch Cancer Center, Seattle, WA.
Background: Black women are more likely to present with advanced stage disease and to die following a breast cancer (BC) diagnosis compared with women from other racial and ethnic groups. This persistent health inequity is in part driven by underrepresentation in clinical trials (CTs), as well as structural and social drivers of health. Objective: The overall objective of this study is to develop, optimize, and pilot-test a narrative decision aid (web-based video) that is culturally sensitive, grounded in a socioecological framework, informed by in-depth, iterative, and community-engaged qualitative research, and designed to support clinical trial decision-making among Black breast cancer patients receiving care in safety-net oncology settings. Here, we report outcomes from an acceptability study conducted at a safety-net oncology practice among end-users. Methods: To assess acceptability of the web-based, narrative video intervention, we conducted a study in a clinic population at the University of Illinois Cancer Center oncology clinic. Black women with a diagnosis of BC (any stage, any point during treatment or follow-up) were identified by their oncologist and invited to participate,. Consenting participants completed a paper-based pre-survey, viewed the 12-minute video intervention on an electronic device, and completed a paper-based post-survey. The survey included items related to demographics, prior exposure to clinical trials, clinical trial knowledge, self-efficacy, and video acceptability. Participants received $50 for completing all study activities. Data were entered into a REDCap database and exported to SPSS for analysis. We calculated descriptive statistics (mean, standard deviation) for pre- and post-intervention scores and used paired t-tests to analyze the change in scores from pre- to post-intervention. Results: Twenty Black BC patients participated in the study. Prior to the intervention, 85% reported that a member of their medical team had discussed clinical trials, 75% had been invited to participate, and 60% had previously enrolled in a clinical trial. The correct response rate to 5 items on clinical trial knowledge increased from 85.8% at baseline to 92.0% post-viewing (p = 0.029). Confidence in making an informed decision about clinical trials also improved significantly. Self-efficacy was measured using an 11-item scale (0 = not at all confident; 4 = very confident); mean scores increased from 3.35 at baseline to 3.50 post-viewing (p < 0.001). At baseline, 75% of participants strongly agreed or agreed with the statement, “If I had the option, I would definitely consider joining a clinical trial;” this increased to 85% post-viewing. The video was highly acceptable: 100% of participants indicated they were extremely or somewhat likely to share it with family or friends; 95% would not change anything about the video; 80% reported it would motivate them or a loved one to speak with their doctor about clinical trials; and 80% found the content trustworthy. Conclusions: This study demonstrated that a culturally tailored, narrative-based video intervention is both acceptable to Black breast cancer patients and associated with improvements in clinical trial knowledge, decision-making self-efficacy, and intention to participate in trials. These findings support the feasibility of implementing such interventions in safety-net oncology settings. A pilot study of the intervention with a quasi-experimental design is ongoing and will evaluate whether integrating the intervention into standard education for newly diagnosed Black BC patients is associated with increase in clinical trial enrollment. Results of the pilot study will be presented.
Presentation numberPS4-11-28
Breast Clinical Reporting and Data System (BCRADS): A Pragmatic, Low-Cost Tool for Early Detection of Breast Cancer in LMICs
Soumen Das, Institute of Breast Disease, NCRI Hospital, Kolkata, Kolkata, India
S. Das1, R. Agarwal1, T. Mandal2, A. Ali Mallick3, R. Paul4, D. Sarakar5; 1Surgical Oncology, Institute of Breast Disease, NCRI Hospital, Kolkata, Kolkata, INDIA, 2Medical Oncology, Institute of Breast Disease, Kolkata, Kolkata, INDIA, 3Surgery, IPGME&R,SSKMH, Kolkata, INDIA, 4Microbiology, College of Medicine and JNM Medical College, Kalyani, INDIA, 5Surgery, IPGME&R, SSKMH, Kolkata, INDIA.
BackgroundLMICs face challenges in early breast cancer detection due to limited mammography availability, with average tumor sizes at diagnosis around 3.5 cm. The WHO Global Breast Cancer Initiative aims for >60% early-stage detection, necessitating effective triage tools. We developed and evaluated the Breast Clinical Reporting and Data System (BCRADS) to standardize clinical breast examination (CBE) in LMICs to improve early detection while reducing unnecessary referrals.MethodsA prospective observational study was conducted at a tertiary care breast clinic in India, enrolling 418 women with breast symptoms between [Insert Dates]. Cancer group: 240 biopsy-confirmed breast cancer patients. Benign group: 178 patients with benign breast conditions. Each patient underwent structured history and systematic CBE, with BCRADS scoring assigned as follows: BCRADS Scoring: History: – Age: 50 (2) – Family history: No (0), Yes (1) – Prior chest wall radiation: No (0), Yes (1) – BRCA carrier: No (0), Yes (2) Clinical Examination: – Asymmetry: Absent (0), Present (1) – Skin ulcer/PDO: Absent (0), Present (2) – Hard fixed lump: Mobile (0), Fixed (2) – Axillary lump: Absent (0), Present (2) Total score range: 0-15. Higher scores indicate greater suspicion for malignancy. Analysis: BCRADS scores were compared between groups. ROC curve analysis was performed to evaluate discriminative ability, with optimal cut-off identified via Youden’s index. Sensitivity, specificity, PPV, NPV, and accuracy at the cut-off were calculated.ResultsThe mean BCRADS score was significantly higher in the cancer group (8.2 ± 2.4) compared to the benign group (3.1 ± 1.9, p < 0.001). ROC analysis showed AUC = 0.89 (95% CI: 0.86-0.92), indicating excellent discrimination. At the optimal cut-off of 6: – Sensitivity: 88% – Specificity: 79% – PPV: 85% – NPV: 83% – Accuracy: 84%ConclusionsThe BCRADS tool demonstrates high accuracy in predicting malignancy using standardized clinical assessment, enabling effective early detection triage in LMIC settings where mammography is limited. Integration of BCRADS into primary care workflows can reduce unnecessary referrals, prioritize imaging, and align with WHO goals of increasing early breast cancer detection. Future multicentric validation and integration with digital platforms for scale-up in LMICs are planned.
Presentation numberPS4-11-29
Pathological Complete Response and Breast-Conserving Surgery in Patients Undergoing Neoadjuvant Chemotherapy for Breast Cancer in an Institution fully Funded by the Brazilian Public Health System in Bahia, Brazil
Daniela Cristina Camarotti, Hospital Santo Antônio/Obras Sociais Irmã Dulce, Salvador, Brazil
D. C. Camarotti1, E. D. Moreira2, M. F. Assunção1; 1Mastology, Hospital Santo Antônio/Obras Sociais Irmã Dulce, Salvador, BRAZIL, 2Laboratório de Epidemiologia Molecular e Bioestatística, Instituto Gonçalo Moniz/ Fiocruz, Salvador, BRAZIL.
Introduction: Breast cancer (BC) incidence has increased in developing countries, showing high mortality rates despite lower incidence compared to developed nations. In 2023, 73,610 new BC cases were reported in Brazil, with 80% of patients relying solely on the Brazilian public health system (SUS) to get treated. Limited early detection in primary care contribute to 40% of cases being diagnosed at advanced stages. The prevalence of aggressive subtypes such as triple-negative (TN) and HER2+ (HER2) differ between regions in the country. The state of Bahia has a predominantly Black/mixed-race population (81%), however, data on tumor subtype distribution and neoadjuvant treatment outcomes are scarce. Objective: To evaluate pathological complete response (pCR) rates in the breast and axilla, following neoadjuvant chemotherapy (NACT) in BC patients treated at an institution fully funded by SUS in Bahia. Secondary objectives included assessing pCR rates by tumor subtype and breast-conserving surgery (BCS) rates. Methods: This case series study included 286 women who underwent surgery after NACT between Jan/2018 and Dec/2021. Patients with multifocal/multicentric tumors, bilateral BC, metastasis, or who received NACT elsewhere were excluded. Tumors were categorized into molecular subtypes (luminal, TN, HER2/HR+, HER2/HR−). Use of trastuzumab with number doses were documented for HER2 cases. Surgeries were classified as conventional BCS, oncoplastic BCS, or mastectomy (with/without reconstruction). pCR was defined as the absence of invasive disease in the breast and axilla. Results: Mean age was 51 ± 12 years; 82% self-identified as Black or mixed-race. Tumor subtypes: 41% luminal, 31% TN, 13% HER2/HR+, 15% HER2/HR−. TN was more frequent in mixed-race (36%), and HER2 in Black women (33%). 66% had locally advanced disease; 93% received ACT-based chemo. Among HER2+ cases, 63% received trastuzumab, but only 32% got 4+ doses. Overall, pCR was 22% and by subtype: HER2/HR− (49%), TN (26%), HER2/HR+ (25%), luminal (8%), by race: Black (27%), mixed-race (20%), White (17%). Mastectomy was done in 64% (19% with reconstruction); 36% had BCS, with 22% using oncoplastic techniques. In non-inflammatory cases, 42% had conservative surgery. Among cT3 patients with BCS, 41% had reconstructive surgery. Considering Luminal subtype, oncoplastic techniques were used in 36% of BCS. Discussion: This is the first study to assess pCR in a predominantly Black/mixed-race population in Brazil. The high prevalence of aggressive tumor subtypes and advanced-stage diagnoses likely influenced the pCR outcomes. Despite the recommendation for trastuzumab use since 2013, access remains limited and inconsistent in the SUS. Compared to international studies, pCR rates by subtype were lower, probably reflecting disparities in access to modern targeted therapies and immunotherapy. Higher pCR among Black/mixed-race women is likely linked to the greater frequency of responsive subtypes. BCS occurred in 42% of non-inflammatory breast cancer cases, lower than international figures. Oncoplastic techniques helped expand BCS eligibility in cT3 tumors and also in Luminal tumors, which tend to shrink less, benefiting from these surgical techniques. Conclusion: This study provides important insight into breast cancer treatment outcomes in an institution served exclusively by SUS in Brazil in a region with a predominantly Black/mixed-race population. The findings emphasize the urgent need for public health strategies that ensure earlier diagnosis and improve equitable access to targeted therapies, aiming to reduce disparities and improve outcomes for underserved populations.
Presentation numberPS4-11-30
A Retrospective Review of Demographics and BMI on ER Positive and Negative Breast Cancer in Rural Southeast Alabama
Namitha Thotli, Southeast Health, Dothan, AL
N. Thotli1, R. Bandarnaike1, S. Valasareddi2; 1Internal Medicine Residency, Southeast Health, Dothan, AL, 2Hematology and Oncology, Southeast Health, Dothan, AL.
Background:Breast cancer remains a leading health concern among women, with Estrogen Receptor (ER) status influencing prognosis and treatment. Rural populations often face disparities in access and outcomes, yet data remains limited. This study evaluates the relationship between Body Mass Index (BMI), demographics, and ER status in breast cancer patients from Southeast Alabama. By analyzing a large rural cohort, we highlight how elevated BMI, type 2 diabetes, tobacco use, race, and family history may influence ER(+) disease, offering insights that may guide future preventive strategies and targeted screening efforts in underserved communities. Methods:We reviewed data from July 2018 to June 2025 using our institution’s Epic database, SLICER DICER. BMI was categorized according to the World Health Organization (WHO) adult classification. International Classification of Disease (ICD) codes were used to extract data from Slicer Dicer. The study population (n=1421) consisted of individuals from rural communities in Southeast Alabama. Results:Comparisons were made between ER(+) breast cancer and ER(-) breast cancer in adult BMI classifications per WHO. Average age for ER(+) breast cancer was 70.1+.7 years and average age for ER(-) breast cancer was 73.1+.7 years. The ER(+) population was 70.9% white, 26.6% black, and 2.5% other. The ER(-) population was 62% white, 37.1% black, and 0.9% other. Comparisons between BMI for ER(+) vs ER(-) breast cancer respectively; BMI less than 18.5 was 2.67% vs 2.83%; BMI 18.5-25, 18.08% vs 17.56%; BMI 25-30, 29.34% vs 26.91%; BMI 30-35, 24.34% vs 25.5%, BMI 35-40, 15.62% vs 17%. Comparison with respect to Type 2 Diabetes Mellitus diagnosis for ER(+) vs ER(-) breast cancer respectively was 31%+3.2% vs 30.4+6.7%. Comparison with respect to positive family history for ER(+) vs ER(-) breast cancer respectively was 30.9%+3.3% vs 36.5%+7%. The comparison of tobacco use with respect to ER (+) vs ER(-) breast cancer was 40.1%+3.4% vs 40.9%+7.2%. Analysis using χ² test (χ²=889.32) with p<.001 showed that there is an association between BMI and developing breast cancer. Conclusion:In this large rural cohort, elevated BMI, type 2 diabetes, and tobacco use were significantly associated with ER(+) breast cancer (p<.001). White patients were more likely to have ER(+) breast cancer (p<.001 OR=1.6) at our institution. Black women are more likely to have ER(-) breast cancer (p<.001 OR=1.5). The obesity prevalence in post- menopausal women in our population (54%) surpasses national prevalence of obesity in post-menopausal women (43%) per National Institute of Health (NIH), highlighting a critical regional disparity. Our findings suggest that modifiable metabolic and lifestyle factors may contribute to increased ER(+) breast cancer risk in underserved communities. Patients may benefit from further emphasis being placed on metabolic health and early intervention to address prevention. This also brings into light the lack of access to metabolic care in the area. These results underscore the need for future studies and public health initiatives focused on prevention, early detection, and risk stratification tailored to rural populations.
Presentation numberPS4-12-02
Challenges in Breast Cancer Screening and Awareness in Sierra Leone – Initiatives by Thinking Pink Breast Cancer Foundation and Breast Cancer Hub
Lopamudra Das Roy, Breast Cancer Hub, Concord, NC
C. Parkinson Pratt1, A. Barrie1, R. BREWAH1, J. Kanu1, A. Sandy1, E. Foday2, L. Das Roy3; 1Research, Thinking Pink Beast Cancer Foundation, Freetown, SIERRA LEONE, 2Youyi Building-Ethics Committee, Ministry of Health, Freetown, SIERRA LEONE, 3Cancer Screening, Treatment Support and Research, Breast Cancer Hub, Concord, NC.
Background: Breast cancer is a major public health concern in Sierra Leone, contributing significantly to female cancer mortality. Late-stage presentation, lack of screening infrastructure, pervasive stigma, and absence of a national cancer registry hinder effective intervention [1,2]. Limited healthcare access, insufficient trained professionals, and low public awareness further exacerbate the crisis, necessitating community-based solutions and public health education [3]. Patients typically presented only when symptoms were severe, reflecting widespread unawareness, myths, traditional/religious beliefs and financial/geographic barriers to care [4]. Methods:This study presents data from community outreach and screening initiatives conducted by the Thinking Pink Breast Cancer Foundation (TP), a Sierra Leone-based nonprofit, and its collaboration with Breast Cancer Hub (BCH), a U.S.-based nonprofit established in 2017, that joined forces with TP in 2021. BCH expanded the impact through treatment aid for underprivileged patients since 2021 and launched the “One Community Per Month Campaign” in 2024, promoting monthly free screenings and awareness campaigns in schools, markets, and underserved communities. Data was collected by field teams and analyzed using descriptive statistics. Results: From 2012 to 2024, 20,968 individuals (19,719 females and 1,249 males) were screened. Of these, 5,537 showed abnormal symptoms such as pain, itching, swelling, or nipple discharge, leading to ~26.41% abnormality. During the 2024 “One Community Per Month Campaign,” 772 individuals (683 females, 89 males) were screened, and 91 abnormal cases were identified for further evaluation. Overall, we executed 1,169 lumpectomies. A total of 164 breast cancer cases were confirmed—all at advanced stages. Among them, 100 patients underwent mastectomy, while 63 advanced cases were untraceable due to family-related challenges. Mortality was extremely high: 100 patients died (92 females, 8 males), with only one known survivor. Alarmingly, knowledge of breast cancer symptoms was negligible. None of the participants reported ever performing a Breast Self-Exam (0%) or undergoing a Clinical Breast Exam (0%). Between 2012 and 2024, none of the 4,419 individuals over 40 years (4,328 from general outreach and 168 from the 2024 One Community-Per Month campaign) had ever received a screening mammogram or breast ultrasound—reflecting a 0% prior screening rate in this high-risk group. Visits to healthcare facilities were typically initiated only after symptoms became severe. Geographic and financial barriers, lack of awareness, and limited access to diagnostic services were significant contributors to delayed care. Discussion: The findings highlight a critical public health gap in breast cancer awareness, screening, and early intervention in Sierra Leone. Late-stage diagnoses and near-total mortality among confirmed cases underscore the urgent need for national breast cancer programs. TP and BCH have pioneered community-centered approaches, providing free screenings, awareness education, and patient aid. Their collaborative model demonstrates the potential for grassroots action to drive systemic change. Continued expansion of such programs, combined with national policy support, is essential to reduce preventable deaths and improve survival outcomes in Sierra Leone. References:
- WHO. Cancer Country Profiles – Sierra Leone. WHO; 2020.
- Jalloh MB et al. Breast cancer in Sierra Leone: a retrospective review. BMC Cancer. 2024;24(1):112.
- Kamara S et al. Barriers to early breast cancer detection in Sierra Leone. Int J Cancer Epidemiol. 2022;11(3):204-212.
- Conteh F et al. Community perceptions and screening behaviors for breast cancer in Freetown. Afr Health Sci. 2023;23(1):55-63.
Presentation numberPS4-12-03
Access-her: assessing consequences of standard-of-care gaps in her2-positive breast cancer in the brazilian healthcare system
André Mattar, Hospital da Mulher, SÃO PAULO, Brazil
A. Mattar1, F. Cavalcante2, C. La-Scala3, E. Pavanel3, G. Tiguman4, I. Silveira3, N. Correa-Netto3, V. Cano3; 1Núcleo de Mastologia, Hospital da Mulher, SÃO PAULO, BRAZIL, 2Núcleo de Mastologia, Hospital Geral de Fortaleza, SÃO PAULO, BRAZIL, 3Medical, Produtos Roche Químicos e Farmacêuticos, SÃO PAULO, BRAZIL, 4HTA, Produtos Roche Químicos e Farmacêuticos, SÃO PAULO, BRAZIL.
Background: Dual HER2 blockade with pertuzumab and trastuzumab, followed by adjuvant trastuzumab-emtansine (T-DM1) for residual disease, has been the undisputed global standard of care for high-risk HER2-positive early breast cancer (EBC). These regimens have demonstrated significant improvements in pathological complete response and overall survival in pivotal trials. However, neither pertuzumab nor T-DM1 is currently available in the Brazilian Unified Health System (SUS), leading to a critical gap between internationally recommended treatment and what is offered to patients in the public sector. This disparity may lead to worse oncological outcomes, including increased mortality. The present study aimed to estimate the number of preventable deaths associated with the lack of access to these therapies in non-PCR patients in Brazilian non-PCR patients in Brazil. Methods: We conducted a retrospective, scenario-based modeling study using real-world data from DATASUS, the national public health database in Brazil. The study population included women aged 18 years or older who initiated treatment for stage II or III HER2-positive early breast cancer (EBC) within the SUS in 2023. Three treatment scenarios for patients with residual disease after neoadjuvant therapy were modeled. The first scenario, representing current practice in SUS, consisted of trastuzumab neoadjuvant followed by adjuvant trastuzumab alone (SUS Standard of Care). The second scenario (Scenario 2) incorporated adjuvant trastuzumab emtansine (T-DM1) for patients with residual disease. The third scenario (Scenario 3) simulated neoadjuvant dual HER2 blockade with pertuzumab and trastuzumab, followed by adjuvant T-DM1 in non-pCR cases. Model parameters, including total pathologic complete response (tpCR) rates and 7-year overall survival (OS), were derived from the pivotal NeoSphere and KATHERINE trials, respectively. Estimated 7-year mortality was calculated for each scenario, and preventable deaths were defined as the difference in projected mortality between the SUS Standard and the two alternative scenarios. Results: A total of 6,061 women with stage II-III HER2-positive early breast cancer initiated treatment within the SUS in 2023, including 1,381 patients with stage II disease and 4,680 with stage III. Based on the scenario model, an estimated 744 deaths over seven years would occur under the SUS Standard of Care Scenario. In contrast, Scenario 2 projected 520 deaths, and Scenario 3 projected 402 deaths. These represent relative reductions in 7-year mortality of approximately 30% and 46%, respectively, when compared to the SUS Standard of Care. When stratified by clinical stage, Scenario 2 was estimated to prevent 51 deaths in stage II and 173 deaths in stage III. The Scenario 3, incorporating neoadjuvant pertuzumab and adjuvant T-DM1, was estimated to prevent 78 deaths in stage II and 264 in stage III. In total, adoption of the Scenario 3 could result in more than 342 preventable deaths per annual cohort. Conclusion: This analysis demonstrates that the absence of pertuzumab and T-DM1 from the Brazilian public health system may lead to over 342 preventable deaths annually among patients with high-risk HER2-positive early breast cancer. The estimated reductions in long-term mortality with access to standard-of-care regimens underscore the urgent need to align national treatment protocols with internationally accepted guidelines. Incorporating these therapies into the SUS is essential to address systemic inequities in breast cancer care and to improve survival outcomes for patients treated in the public sector in Brazil.
Presentation numberPS4-12-04
Late-stage breast cancer in young appalachian women: missed hereditary risk and persistent disparities in a community health setting
Jasneet Gill, Tennova North Knoxville Medical Center, Powell, TN
J. Gill1, N. Lopetegui-Lia2; 1Internal Medicine, Tennova North Knoxville Medical Center, Powell, TN, 2Medical Oncology, The Ohio State University, Columbus, OH.
Background: The incidence of breast cancer among young women is on the rise. In rural regions, a combination of socioeconomic disadvantages, poor health literacy, and limited access to specialized care exacerbates diagnostic delays, even more so in patients with hereditary risk. This study investigates the stage of presentation, family history, genetic testing uptake, and social determinants of health among young women diagnosed with breast cancer in a community health setting in Appalachia. Methods: This retrospective cohort study included 1,359 patients diagnosed with breast cancer at Tennova North Knoxville Medical Center from 2020 to 2024. Patients were stratified by age, i.e. young patients (<45 years) versus (vs) average-onset patients (age ≥45), stage at diagnosis, insurance status, urban vs rural residence, educational attainment, reported family history of cancer, and whether they underwent genetic testing and/or counseling (if positive family history of any cancer). Descriptive statistics were calculated, and bivariate comparisons were performed using Chi-square and t-tests. Logistic regression models were constructed to identify predictors of advanced-stage disease (stages III and IV) at diagnosis and the likelihood of undergoing genetic testing. Results: Of the 1,359 patients included, 16.3% (n=222) patients were < 45 years old at diagnosis. Among these young patients, 80.2% (n=178) presented with stage III or IV disease, while 6.3% (n=14) and 13.5% (n=30) were diagnosed at stages I and II, respectively. 81.1% (n=180) reported a first-degree family history of cancer, yet only 16.2% (n=36) received genetic testing and/or counseling. In bivariate analysis, patients with Medicaid or no insurance were significantly more likely to present with advanced stage breast cancer compared to those with private insurance (OR 4.5; 95% CI: 2.6-7.8, p < 0.001). Rural residence was also significantly associated with late-stage diagnosis (OR 3.7; 95% CI: 2.0-6.9, p = 0.002). Lower educational attainment (less than high school diploma) was associated with decreased likelihood of receiving genetic testing (OR 0.34; 95% CI: 0.15-0.78, p = 0.01). In multivariate logistic regression, both uninsured status (p < 0.001) and absence of genetic counseling among patients with positive family history of cancer (p = 0.002) independently predicted advanced-stage presentation at diagnosis. Conclusions: This study highlights a troubling pattern of late-stage breast cancer diagnosis in young Appalachian women, despite the presence of family history of cancer in > 80% of cases. Profound disparities in genetic testing utilization, driven by socioeconomic status, rurality, and health literacy, likely contribute to missed opportunities for early detection. These findings underscore the urgent need for community-based hereditary risk screening programs, access to genetic counseling, and culturally competent education strategies within rural healthcare systems.
Presentation numberPS4-12-05
Real-world evidence of ethnic-racial disparities in tumor-infiltrating lymphocytes and survival outcomes in early-stage triple-negative breast cancer
Jessé Lopes da Silva, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
J. da Silva1, L. de Albuquerque1, F. Rodrigues2, G. de Mesquita1, P. Fernandes2, L. da Silva1, L. Thuler1, A. de Melo1; 1Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL, 2Division of Pathology, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL.
Background: Triple-negative breast cancer (TNBC), comprising 10-15% of all breast cancer cases, is characterized by an aggressive course and high early recurrence rates. Neoadjuvant chemotherapy (NACT), with the recent addition of pembrolizumab, stands as the standard treatment for early-stage TNBC, where achieving a pathologic complete response (pCR) is a strong predictor of favorable long-term survival. Ethnicity is considered a crucial factor in TNBC prognosis; notably, African American women experience higher breast cancer mortality compared to their White counterparts. This disparity may partially result from differences in the tumor microenvironment (TME). Increasing evidence points to the TME’s key role, mainly the composition and density of tumor-infiltrating lymphocytes (TILs), in determining TNBC outcomes. However, the impact of racial and ethnic disparities on the prevalence of TILs and their prognostic significance remains inadequately explored. Understanding these factors could be pivotal in understanding disparities and tailoring more effective, equitable treatment strategies. Methods: The internal database of the Brazilian National Cancer Institute was queried to identify women diagnosed with TNBC who underwent NACT followed by curative surgery between January 2010 and December 2014. Patients were assigned to White or Black/Mixed-race groups for comparison. Core biopsy specimens were analyzed by immunohistochemistry (IHC) for CD4, CD8, FOXP3, and PD-L1 CPS. Complete blood counts were assessed to evaluate the pre-NACT neutrophil-to-lymphocyte ratio in peripheral blood. The correlation between racial groups and the TME composition by TILs subpopulations was evaluated. Progression-free survival (PFS) and overall survival (OS) estimates for each racial group were determined using the Kaplan-Meier method. Results: A total of 169 patients were included: 78 (46.2%) White and 91 (53.8%) Black/Mixed-race women. Clinical characteristics showed no significant differences: mean age was 50.9 (SD 11.3) vs. 50.0 (SD 10.2) years (p=0.555); mean BMI was 28.1 (SD 5.2) vs. 29.0 (SD 6.4) (p=0.351). Tumor stage II was present in 12 (15.4%) White and 16 (17.6%) Black/Mixed patients (p=0.861); tumor grade 3 in 50 (64.1%) vs. 63 (69.2%) (p=0.653). High CD4 expression was observed in 66 (88.0%) White and 70 (83.3%) Black/Mixed patients (p=0.542); high CD8 in 40 (53.3%) vs. 45 (49.4%) (p=0.738). FOXP3 high expression occurred in 38 (50.7%) vs. 35 (41.7%) (p=0.328). PD-L1 CPS ≥1% was present in 32 (41.0%) White and 37 (41.1%) Black/mixed patients (p=1.000). The CD4/FOXP3 high ratio was observed in 37 (49.3%) vs. 47 (56.0%) (p=0.499); CD8/FOXP3 high in 64 (85.3%) vs. 79 (86.7%) (p=0.979). The CD4/CD8 high ratio was more frequent in Black/Mixed (13; 15.7%) vs. White (4; 5.3%) (p=0.066). Neutrophil-to-lymphocyte ratio was similar in both groups. With a median follow-up of 62.5 months, the 5-year event-free survival (EFS) was 53.9% vs. 47.3% (p=0.45), and the 5-year overall survival (OS) was 56.1% vs. 58.6% (p=0.78) in White and Black/Mixed groups, respectively. Conclusions: These findings indicate clinical and pathological similarities between White and Black/Mixed-race women with TNBC undergoing NACT. The prevalence of TILs subtypes and PD-L1 expression showed no significant racial differences, and survival outcomes were comparable between the groups. These results suggest that differences in the TME across races are unlikely to explain racial outcome disparities.
Presentation numberPS4-12-06
Breast Cancer in Appalachia: A Comprehensive Analysis of Behavioral and Socioeconomic Factors Influencing Stage at Diagnosis
Jasneet Gill, Tennova North Knoxville Medical Center, Powell, TN
J. Gill1, N. Lopetegui-Lia2; 1Internal Medicine, Tennova North Knoxville Medical Center, Powell, TN, 2Medical Oncology, The Ohio State University, Columbus, OH.
Background: Breast cancer remains a leading cause of morbidity and mortality, with disparities in outcomes influenced by behavioral, demographic, and socioeconomic factors. The Appalachian region faces unique challenges due to limited access to healthcare, economic disparities, and high rates of tobacco and alcohol use. This study aims to assess whether alcohol use, smoking history, insurance status, and family history of cancer influences breast cancer staging at diagnosis. Methods: A retrospective analysis was conducted using medical records from patients diagnosed with breast cancer at Tennova North Knoxville Medical Center between 2020 and 2024. A total of 1,359 patients with breast cancer were identified. The study examined the association between alcohol use, smoking history, family history of cancer, diabetes mellitus type 2, insurance status, and breast cancer staging (AJCC 8th edition). Data on age and socioeconomic status were also considered. Descriptive and biostatistical analyses, including chi-square tests, logistic regression, and multivariable models were performed. Results: Of the 1,359 patients with breast cancer included in this study, 49.3% were diagnosed with stage III and 3.0% with stage IV breast cancer. Current alcohol use (55.7%) and smoking (14.5% current; 24.7% former smokers) were significant predictors of advanced stages (stage III or IV) at diagnosis, with current alcohol use increasing the odds of advanced disease by 2.1 times (OR 2.1, 95% CI [1.5, 2.9], p < 0.01) and current smokers being 2.5 times more likely to present with stage III (OR 2.5, 95% CI [1.8, 3.6], p < 0.01). A family history of cancer was protective, with those reporting it being less likely to present with advanced disease (OR 0.6, 95% CI [0.4, 0.9], p < 0.05). Medicaid patients and patients without insurance had significantly higher odds of presenting at advanced stages, with Medicaid being 3 times more likely to present with stage III or IV (OR 3.0, 95% CI [2.1, 4.5], p < 0.01), and uninsured patients being 5 times more likely (OR 5.0, 95% CI [2.3, 10.5], p < 0.01). Diabetes mellitus type 2 was associated with an increased risk of advanced disease (OR 1.6, 95% CI [1.2, 2.1], p < 0.01). Younger patients (<45 years) had a 2.4 higher likelihood of presenting with advanced breast cancer compared to older patients (≥ 45 years) (OR 2.4, 95% CI [1.5, 3.7], p < 0.01), suggesting delays in diagnosis, likely due to lower screening rates. Conclusions: This study performed in the Appalachian region identifies critical factors, including behavioral factors and socioeconomic disparities, and its influence on breast cancer stage at diagnosis. Current alcohol use, smoking, and government insurance/lack of insurance were found to be significant predictors of advanced-stage diagnoses, while family history of cancer was associated with breast cancer being diagnosed at an earlier stage. Additionally, Medicaid and uninsured patients presented with more advanced diseases at diagnosis, indicating that they may face barriers to early detection. These findings underscore the importance of addressing behavioral risk factors and targeting socioeconomic disparities to improve timely access to care in rural populations. Public health initiatives in the region should focus on community awareness, screening and early cancer detection programs, reducing/limiting smoking and alcohol use, and expanding access to insurance to mitigate these disparities.
Presentation numberPS4-12-07
Correlations between Race and Area Deprivation Index (ADI) on Treatment and Outcomes in Metastatic TNBC
Anumita Chakraborty, University of Pittsburgh Medical Center, Pittsburgh, PA
A. Chakraborty1, B. Jambunathan1, A. C. Haley2, X. Pei3, M. Q. Rosenzweig3; 1Hematology/Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA, 2School of Medicine, West Virginia University, Morgantown, WV, 3School of Nursing, University of Pittsburgh, Pittsburgh, PA.
Introduction: Previous data has determined that social determinants of health (SDOH) contribute to disparities in breast cancer outcomes. Barriers to treatment and treatment disparities could contribute to this difference, especially as the treatment landscape changes with targeted therapies. This study aims to evaluate the relation between race and area deprivation index (ADI) on treatment disparities and outcomes in patients with metastatic triple negative breast cancer (TNBC). Methods: This is a retrospective review of Caucasian and Black patients treated at the University of Pittsburgh Magee-Womens Hospital diagnosed with metastatic TNBC between 1996-2022. ADI was calculated through the Neighborhood Atlas interactive website from the University of Wisconsin School of Medicine and Public Health, as a percentile ranking from 1 to 100 where higher numbers indicate greater “disadvantage”. Targeted therapies included PARPi, PD-1 and PD-L1 inhibitors, bevacizumab, sacituzumab-govetican. De novo metastatic disease was defined by having initial and metastatic TNBC diagnoses within 2 months. Significance of results was determined using chi-square tests. Results: Of 270 total patients, 237 (87.8%) were Caucasian and 33 (12.2%) Black. ADI quartiles were stratified as Q1 ≤47, Q2 48-69, Q3 70-80, and Q4 >80. 59 (21.9%) were diagnosed with primary breast cancer <40 years of age. Median overall survival (OS) of the cohort was 14.1 months after diagnosis of metastatic disease. 46 (17%) patients presented with de novo metastatic disease; 41 (17.3%) Caucasian and 5 (15.2%) Black (p = 0.76). Median time to initiate systemic therapy after diagnosis of metastasis was 35 days in Caucasian patients and 33 days in Black patients. Both Caucasian and Black patients received a median of 3 lines of chemotherapy. 74 (33.9%) of Caucasian patients and 9 (30.0%) of Black patients received targeted therapy (p=0.668). Median time to progression (PFS) was 5.0 months in Caucasian patients and 5.5 months in Black patients. Median OS after diagnosis of metastatic disease was 14.4 months in Caucasians and 13.5 months in Blacks. 110 (46.4%) of Caucasian patients and 18 (54.5%) of Black patients developed CNS metastases (p = 0.381). Stratified by ADI, there was no significant difference in the number of patients presenting with de novo metastatic disease, 12 (17.9%) patients in Q1, 8 (12.7%) in Q2, 9 (13.8%) in Q3, and 12 (19%) in Q4, p = 0.71. Patients in Q1, Q3, and Q4 all had a median of 3 lines of chemotherapy, and Q2 had a median of 2 lines of chemotherapy. No significant difference was seen in the proportion of patients who received targeted therapies between quartiles (Q1 21 (31%), Q2 18 (30%), Q3 18 (29.5%), Q4 26 (42.6%), p = 0.37). Median PFS was similar across Q1-3 at 4.8-5 months, and highest in Q4 at 6.3 months. Median OS after diagnosis of metastatic disease was similar across ADI groups (14.9 months Q1, 14.8 months Q2, 12.8 months Q3, and 14.2 months Q4). Conclusions: There was no significant difference in time to start systemic therapy, number of lines of chemotherapy and proportion receiving targeted therapy in patients with metastatic TNBC, stratified by race and ADI. Similarly, with no difference in treatment parameters, including proportion of patients who received targeted therapy, there was no significant difference seen in progression and OS when stratified by race and ADI. Limitations include that this was a single-center study, and a limited sample size. Further investigation in determining correlations between SDOH, treatment parameters, and outcomes is required in patients with TNBC.
Presentation numberPS4-12-08
Oncotype DX prediction of recurrence by race
Quinn S Solfisburg, Boston Medical Center, Boston, MA
Q. S. Solfisburg1, K. P. Verma1, R. Soumya1, N. Garg1, L. Hildebrand1, L. J. Oshry1, M. Cassidy2, R. Dries1, N. Y. Ko3; 1Hematology and Medical Oncology, Boston Medical Center, Boston, MA, 2Surgical Oncology, Boston Medical Center, Boston, MA, 3Hematology and Medical Oncology, NYU Langone, New York, NY.
Background: The 21-gene recurrence assay from Oncotype DX is widely used in clinical practice to help predict recurrence risk and the likelihood of benefit of chemotherapy in patients with hormone receptor positive breast cancer. Worse breast cancer outcomes among Black women have been a pernicious and well-documented problem. The role of precision medicine and technical advancements in mitigating or exacerbating these disparities is an important and quickly evolving area of investigation. The TailorRX and RxPonder trials showing the important benefit of Oncotype DX were comprised of 70-85% White patients, with 6-7% Black patients, making real-world outcomes in a diverse population of particular interest. Methods: We conducted a retrospective analysis of patients with hormone receptor positive early breast cancer, who had Oncotype DX testing performed between 2010 and 2021 at a single institution, urban safety-net hospital. Patients with intermediate risk scores, defined as 11-25, were further analyzed for recurrence. Chart review was performed April-July 2025 to assess for disease recurrence. Race was self-reported by patients and extracted from the medical record. Receipt of chemotherapy was defined as chemotherapy given to treat the breast cancer for which Oncotype testing was performed, excluding therapy for subsequent recurrences. Statistical significance was assessed using chi-square testing and logistic regression, with all analyses conducted in Python. Results: Our cohort included 245 patients with intermediate Oncotype scores of 11-25: 72 (29%) Black and 173 (71%) non-Black. Mean and median age at diagnosis were the same for Black and non-Black patients in our cohort, 54 and 55 respectively. Black patients were less likely to have intermediate Oncotype scores (48% vs 62%, p=0.03). In univariate logistic regression, odds ratio for recurrence among those with intermediate Oncotype scores was 2.19 for Black patients compared to non-Black, 95% CI 1.02-4.72, p=0.045. Comparing Black patients to White patients specifically (N=124 White patients), the odds ratio for recurrence was further increased to 2.46, 95% CI 1.05-5.75, p=0.038. In multivariate analysis (N=219 because of data availability), adjusting for age at diagnosis and receipt of chemotherapy, Black patients with intermediate Oncotype scores were still significantly more likely to have recurrence compared to non-Black patients, OR 2.34, 95% CI 1.01-5.42, p=0.047. 6% of Black patients with intermediate Oncotype scores received chemotherapy, while 12% of non-Black patients received chemotherapy, but this was not statistically significant, p=0.31. Conclusions: In our study, Black patients with intermediate Oncotype DX scores were more likely to have disease recurrence compared to their non-Black counterparts. Our findings among a diverse patient cohort at a single urban safety-net hospital suggest that 21-gene recurrence scores may have disparate predictive power for patients of different races. Further research is needed to understand the underlying factors that contribute to these disparities, particularly in settings with comparable access to care. Identifying strategies to enhance validity of predictive tools across diverse patient populations may improve risk stratification and help mitigate racial disparities in breast cancer outcomes.
Presentation numberPS4-12-09
Racial and Ethnic Disparities in Enrollment to Breast Cancer Clinical Trials from 2000 to 2025
Fatma Nihan Akkoc Mustafayev, Miami Cancer Institute, Baptist Health South Florida, Miami, FL
V. Andion Camargo, F. Akkoc Mustafayev, K. Qidwai, M. Jaramillo, S. Lin, Z. Sarfraz, L. S. Spiegelman, K. Mustafayev, M. A. Ganiyani, M. S. Ahluwalia, R. L. Mahtani; Medical Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL.
Background: Breast cancer (BC) remains the most common malignancy among women worldwide and is the leading cause of cancer-related mortality. Despite significant advances in treatment and an increase in clinical trial (CT) activity over the past two decades, equitable access to trials has not kept pace. Racial and ethnic disparities in trial enrollment persist, limiting the generalizability of findings and perpetuating outcome gaps. Non-Hispanic Black women, in particular, face higher BC mortality despite similar or lower incidence compared to non-Hispanic White women. Understanding trends in CT representation is essential to addressing these inequities. This study aimed to evaluate global enrollment patterns by race and ethnicity (R/E) in completed BC CTs. Methods: We conducted a retrospective analysis of completed BC CTs registered on clinicaltrials.gov between 1/1/2000 and 6/1/2025. A total of 283 phase III-IV interventional studies were included. Trials were reviewed for reporting of R/E. The key outcome was to evaluate enrollment by R/E, in 5-year intervals to assess temporal trends. For each trial, the proportion of participants (pts) by R/E was extracted, and aggregate enrollment ratios were calculated across all reporting trials. Descriptive statistics were used for data analysis. Results: Of the 283 eligible trials, 120 (42%) reported race and 63 (22%) reported ethnicity. Most studies were conducted in North America and Europe. Among trials reporting race, White pts were the most represented, while Native Hawaiian and Pacific Islander pts were the least represented. Temporal trends demonstrated a decrease in White race enrollment from 88.9% (2000-2005) to 66.7% (2015-2021), alongside an increase in Asian enrollment from 4.5% to 21.5% over the same period. Among the 63 trials reporting ethnicity, 62 included non-Hispanic and 56 included Hispanic pts. Non-Hispanic individuals accounted for 81.4% of total enrollment across these trials. Overall, 75% of all pts identified as White, while Black pts accounted for only 3.4. Notably, 15 trials reported only a single race, often reflecting geographic enrollment patterns. Conclusions: Over the past 25 years, Black and Hispanic patients remained underrepresented in BC CTs. While modest improvements have occurred over time, particularly in Hispanic and Asian enrollment, persistent disparities limit the generalizability of trial findings and hinder progress toward equitable cancer care. This analysis is limited by the lack of standardization in how R/E are defined and reported, absence of geographic or population-adjusted benchmarks, and reliance on registry-reported data may underestimate disparities or mask regional variation. Greater transparency and standardization in demographic reporting, along with proactive efforts to diversify CT participation, are essential to ensure all patient populations are equitably represented in BC research.
| Racial Groups Represented, n(%) |
Trials (n=120) |
Participants (n=85,756) |
| White | 106 (88.3) | 64,314 (75) |
| Asian | 99 (82.5) | 13,215 (15.4) |
| Black/African American | 97 (80.8) | 2,935 (3.4) |
| American Indian or Alaska Native | 53 (44.2) | 765 (0.9) |
| Native Hawaiian or Pacific Islander | 33 (27.5) | 91 (0.1) |
| Unknown | 69 (57.5) | 2,681 (3.1) |
| More Than One Race or Other(s) | 55 (45.8) | 1,755 (2.1) |
| Ethnicity Categories Represented, n(%) |
Trials (n=63) |
Participants (n=29,830) |
| Non-Hispanic | 62 (98.4) | 24,290 (81.4) |
| Hispanic | 56 (88.9) | 3,290 (11.1) |
| Ethnicity Unknown | 41 (65.1) | 2,250 (7.5) |
Presentation numberPS4-12-10
A novel therapeutic targeting triple negative breast cancer brain metastases
Hanna Y. Irie, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
S. J. Baker1, A. Boire2, E. Reddy1, H. Y. Irie3; 1Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 2Neuro-Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 3Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
A novel therapeutic targeting triple negative breast cancer brain metastasesStacey J. Baker, Adrienne Boire, E.Premkumar Reddy, Hanna Y. IrieBackground/Rationale: Patients diagnosed with triple negative breast cancer (TNBC) have the lowest five-year survival rates compared to those diagnosed with other subtypes. Many patients develop metastatic recurrences soon after their initial diagnosis. Particularly problematic are central nervous system (CNS) metastases including brain and leptomeningeal metastases, for which patients with TNBC are at higher risk. Even after initial multimodality treatment, recurrent disease is common and contributes to the poor prognosis of patients with CNS disease. Cancer stem cells (CSC) defined as CD44 high/CD24 low-EpCAM+, and/or positive for high levels of aldehyde dehydrogenase-1 (ALDH1hi), play a critical role in treatment resistance of many cancers. CSCs are resistant to chemo/immunotherapy, as well as radiotherapy, and evade immune responses. Many TNBC’s are enriched for CSCs, and CSCs have been identified or isolated from brain metastases of multiple tumor types. Therefore, to achieve more durable remission and improve survival, CNS-targeted treatments need to eradicate CSCs in addition to bulk tumor cells. Although several CSC-inhibiting therapeutics have been proposed, the efficacy of most have been limited by single pathway-targeting. We recently discovered and reported on a novel compound, 108600, that potently and simultaneously targets multiple pathways activated in TNBC CSCs.108600, an inhibitor of CK2/Dyrk1/TNIK kinases, induces apoptosis of CSCs, as well as bulk tumor cells CK2/Dyrk1/TNIK expression is significantly higher in brain metastases vs. the paired primary breast tumors (GSE184869). Our studies investigated 108600 as a potential novel therapeutic for TNBC brain metastases.Methods: TNBC cell lines that spontaneously metastasize to the brain (BrM) were treated in vitro with 108600. Cell viability and apoptosis of bulk culture, as well as the CSC subpopulation, were assessed using Cell TiterGlo assays and flow cytometry. Pharmacokinetic studies were performed to assess bioavailability of 108600 in the brain. In vivo efficacy studies were initiated with human TNBC tumors in the brain generated by intracranial injection. Progression of tumor growth in the brains of mice treated with vehicle or 108600 were monitored by serial IVIS Imaging. Results: MDA-MB231, E0771 or 4T-1 cell line variants that spontaneously metastasize to the brain (BrM) were treated with vehicle or 108600 in vitro. 108600 potently inhibited viability of bulk culture BrM cells. There is significant enrichment of CD44hi/CD24low CSCs in MDA-MB231 BrM cells (~60%).108600 treatment decreased viability of the CSC subpopulation and induced their apoptosis, as assessed by flow cytometry (Annexin V staining). In pharmacokinetic studies, levels of 108600 in the brain after a single intraperitoneal (IP) injection (50mg/kg) were significantly higher than the in vitro IC50 values for growth inhibition using BrM cells. In vivo efficacy studies were initiated using tumor models generated by direct intracranial injection of MDA-MB231/Luciferase cells for maximal consistency of intracranial tumor formation. Treatment with 108600 (IP) significantly inhibited growth of TNBC tumors in the brain. Ongoing and future studies aim to assess the efficacy of 108600 treatment against tumors formed by spontaneous metastases and effects on survival.
Presentation numberPS4-12-11
Covid-19 and breast cancer disparities: were our fears realized?
Ruvarashe Rumano, The Ohio State University, Columbus, OH
R. Rumano1, R. Andrea1, V. Heh2, A. Clark2, E. Paskett1, B. Oppong1; 1Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 2Department of Surgery, The Ohio State University, Columbus, OH.
Background: Black women historically present with more advanced stages of breast cancer (BC) and experience 40% higher mortality than White women. The COVID-19 pandemic strained healthcare systems and reduced access to routine care, including screening. These disruptions raised concerns about widening racial disparities in BC diagnosis and treatment. We previously found no significant worsening in stage at diagnosis during the pandemic. Here, we evaluate racial differences in time to first definitive treatment and treatment outcomes before and during the pandemic. Methods: We conducted a retrospective cohort study of self-identified Black and White women aged 18+ diagnosed with invasive BC at a large comprehensive breast center between January 2018 and December 2021. We defined “pre-COVID” as 2018-2019 and “mid-COVID” as 2020-2021. Demographic, clinical, and tumor characteristics, including receptor status (ER, PR, HER2) and clinical stage (AJCC 8), were abstracted from the electronic medical record. Clinical stage was grouped into early (Stage I/II) vs. late (Stage III/IV). Time to first definitive treatment (surgery or chemotherapy) was calculated from diagnosis, with delay defined as >60 days. Neighborhood-level social vulnerability was derived from census tract data. Chi-square, Wilcoxon, and logistic regression analyses were used to assess associations by race and time period. Results: Of 3,399 patients, 10.4% were Black and 89.6% White. Black patients were more likely to live in urban areas (61.3% vs. 13.7%, p<0.0001), have public insurance (56.9% vs. 43.0%, p<0.0001), and reside in distressed communities (28.2% vs. 9.9%, p<0.0001). Compared to White women, Black women were more often diagnosed with Stage II disease (25.1% vs. 15.2%, p<0.0001) and had a higher prevalence of hormone receptor-negative tumors (ER-negative: 29.5% vs. 17.3%, PR-negative: 42.9% vs. 26.8%; both p<0.0001).Black women had a longer median time to first treatment compared to White women (48 vs. 43 days, p=0.05) and were more likely to experience treatment delays of 60 days or more (34.9% vs. 26.8%, p=0.01). The distribution of diagnoses across the pre- and mid-COVID periods did not differ significantly by race. In the pre-COVID period (2018-2019), 56.4% of Black patients and 56.3% of White patients were diagnosed, compared to 43.6% and 43.7%, respectively, in the mid-COVID period (2020-2021; p=0.97). Conclusion: Black patients experienced longer delays to first BC treatment and lower treatment rates compared to White patients in both the pre-COVID and mid-COVID periods. Our findings suggest that racial disparities in timely treatment persisted but were not amplified by the COVID-19 pandemic. Ongoing analyses will assess associations between these delays and outcomes such as recurrence and mortality.
Presentation numberPS4-12-12
Racial Disparities in Pathologic Complete Response in Patients with HER2 Positive Breast Cancer Treated with Neoadjuvant Chemotherapy
Regina Matar-Ujvary, Cleveland Clinic Foundation, Cleveland, OH
R. Matar-Ujvary1, R. Rangan1, W. Wei2, Z. Al-Hilli1; 1Department of Breast Service, Cleveland Clinic Foundation, Cleveland, OH, 2Department of Quantitative Health Sciences, Cleveland Clinic Foundation, Cleveland, OH.
Background: Black patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancers have lower rates of pathologic complete response (pCR) following neoadjuvant chemotherapy (NAC). This study aims to identify factors that contribute to disparities in achieving a pCR in HER2-positive breast cancers. Method: A retrospective review of an institutional tumor registry identified women with HER2-positive breast cancers who underwent NAC followed by surgery from 2016 to 2022. Sociodemographic factors, clinicopathologic features, and treatment variables were compared between patients self-identified as Black, White, and Other. Results: The study included 445 women with a median age of 54 (range 22-86) and a median follow-up of 48 months (range 7-110). A total of 355 (79.8%) self-identified as White, 58 (13.0%) Black, and 32 (7.2%) Other. There were no differences in Charlson Comorbidity Index, type of insurance, hormone receptor status, HER2 immunohistochemical score, clinical stage, overall grade, chemotherapy regimen, and completion of chemotherapy between racial groups. There was no difference in median number of days from diagnosis to initiation of chemotherapy or median number of days from completion of chemotherapy to surgery between racial groups (Black = 30.5, White = 30, Other = 31.5 [p=0.85] and Black = 22, White, 19, Other = 21 [p=0.26]). Following NAC, Black patients achieved a pCR in both the breast and axilla less frequently than White patients or Other (24 [41%] Black patients, 191 [54%] White patients, 20 [63%] Other), although this difference was not statistically significant (p=0.11). Factors associated with achieving pCR included age of diagnosis <50 years (p=0.02), private insurance (p=0.02), hormone receptor-negative tumor subtype (p<0.001), HER2 immunohistochemical score of 3+ (p=0.001), and grade 3 disease (p=0.01). Race, Charlson Comorbidity Index, tumor histology, clinical stage, and chemotherapy regimen were not associated with achieving pCR. Conclusion: Black patients had the lowest rate of pCR compared to patients self-identified as White or Other, although this difference was not statistically significant. The higher likelihood of Black patients being diagnosed with HER2-positive breast cancer at ≥50 years may account for the differences in achieving a pCR, however more research to understand disparities in outcomes are needed.
Presentation numberPS4-12-13
Real-world comparison of tumor-infiltrating lymphocytes and survival in advanced triple-negative breast cancer across racial groups in brazil
Jessé Lopes da Silva, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
J. da Silva1, A. dos Santos1, L. de Albuquerque1, A. Neto1, C. da Silva1, L. Cerva1, I. Small1, F. Rodrigues2, F. de Macedo2, P. Fernandes2, L. da Silva1, A. de Melo1; 1Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL, 2Division of Pathology, Brazilian National Cancer Institute, Rio de Janeiro, BRAZIL.
Title: Real-World Comparison of Tumor-Infiltrating Lymphocytes and Survival in Advanced Triple-Negative Breast Cancer Across Racial Groups in BrazilJessé Lopes Da Silva1, Alexssandra Lima Siqueira Dos Santos1, Lucas Zanetti De Albuquerque1, Antônio Lucas Araújo Neto1, Cecília Ferreira Da Silva1, Luana Aguiar Mesquita Cerva1, Isabele Avila Small1, Fabiana Resende Rodrigues2, Fabiane Carvalho De Macedo2, Priscila Valverde Fernandes2, Luís Felipe Leite da Silva1 and Andréia Cristina de Melo1 1Division of Clinical Research and Technological Development, Brazilian National Cancer Institute, Rio de Janeiro, Brazil2Division of Pathology, Brazilian National Cancer Institute, Rio de Janeiro, BrazilBackground:TNBC is an aggressive breast cancer subtype affecting younger, non-white women in Brazil, associated with higher recurrence and lower survival. Although chemotherapy remains the primary treatment for advanced TNBC, recent advances in immunotherapy highlight the importance of immune biomarkers such as tumor-infiltrating lymphocytes (TILs) and programmed death-ligand 1 (PD-L1) expression in predicting treatment response. TIL subtypes and PD-L1 are important in tumor immunity and treatment response. Their prognostic value and racial differences in Brazil require further investigation. Methods:A retrospective database query was performed within the Brazilian National Cancer Institute to identify women with recurrent or metastatic TNBC treated between 2018 and 2022. Patients were categorized as White or Black/Mixed-race. Biopsy specimens obtained before chemotherapy underwent immunohistochemistry (IHC) assessment using the PD-L1 CPS by 22C3 pharmDx assay, along with markers including CD3, CD4, CD8, CD68, FOXP3, and PD-1. Associations between the prevalence of TILs subtypes and ethnicity were evaluated. Progression-free survival (PFS) and overall survival (OS) were estimated with the Kaplan–Meier method.Results:A total of 148 patients with advanced TNBC were included: 44 (29.7%) White and 104 (70.3%) Black/Mixed-race. Median age was similar—White 52.0 years (SD 12.6) and Black/Mixed 51.5 years (SD 13.4). De novo disease was present in 31 (70.5%) White and 81 (77.9%) Black/Mixed patients. Immune marker expressions showed modest differences: median CD3 was 5.0 (IQR 1.0–16.2) in White and 5.0 (1.0–30.0) in Black/Mixed; CD4 median was 10.0 (1.0–16.2) vs. 10.0 (1.0–30.0); CD8 median was 5.0 (1.0–20.0) in both groups. CD68 median was higher in White patients—12.5 (1.0–75.0) vs. 10.0 (1.0–40.0). FOXP3 median was 0.0 (0.0–1.0) in White and 0.0 (0.0–3.0) in Black/Mixed; PD-1 median was 0.0 (0.0–1.0) in White and 1.0 (0.0–1.0) in Black/Mixed. PD-L1 CPS distribution was similar: CPS <1 in 25 (56.8%) White and 58 (55.8%) Black/Mixed; CPS 1–9 in 10 (22.7%) White and 24 (23.1%) Black/Mixed; CPS ≥10 in 9 (20.5%) White and 22 (21.2%) Black/Mixed. Median PFS was 4.6 months (95% CI 3.8–7.7) in White and 5.1 months (4.7–6.4) in Black/Mixed; 2-year PFS rates were 2.8% (0.4–19.4%) and 1.7% (0.3–10.8%), respectively. Median OS was 9.9 months (6.5–13.9) in White and 8.7 months (7.1–11.7) in Black/Mixed; 2-year OS rates were 17.9% (8.8–36.5%) and 6.9% (3.1–15.3%), respectively.Conclusions:In this Brazilian real-world cohort with advanced TNBC, immune marker expression and PD-L1 CPS were similar between White and Black/Mixed-race groups. Minor TIL differences did not affect PFS or OS, indicating comparable immunologic profiles across racial groups and suggesting comparable benefit from immunotherapy.Keywords: Triple-negative breast cancer; tumor-infiltrating lymphocytes; PD-L1; race disparities.
Presentation numberPS4-12-14
Clinicopathological Features of Gestational Breast Cancer in Syria: A Comparative Study
Ayla Kouli, Damascus University Faculty of Medicine, Damascus, Syrian Arab Republic
M. Al ali, A. Kouli, A. Al-Bitar, M. Saifo; Hematology/Oncology, Damascus University Faculty of Medicine, Damascus, SYRIAN ARAB REPUBLIC.
Title: Clinicopathological Features of Gestational Breast Cancer in Syria: A Comparative StudyFull author namesMousa Alali1, 2, Ayla Kouli2, Ahmad Al-Bitar2, and Maher Saifo1, 2*1Department of Oncology, Albairouni University Hospital, Faculty of Medicine, Damascus University, Damascus, Syrian Arab Republic2Faculty of Medicine, Damascus University, Damascus, Syrian Arab RepublicFull institutional mailing addressesMousa Alali, and Maher Saifo: Department of Oncology, Albairouni University Hospital, Harasta M5, Damascus University, Damascus, Syrian Arab RepublicAyla Kouli, Ahmad Al-Bitar, Mousa Alali, and Maher Saifo: Faculty of Medicine, Damascus University, Fayez Mansour Street, P. O. Box: 222, Damascus, Syrian Arab RepublicE-mail addressesMousa Alali: mousa.alali@damascusuniversity.edu.syAyla Kouli : aylakouly01@gmail.comAhmad Al-Bitar: dr.ahmad.al.bitar@gmail.comMaher Saifo: maher.saifo@damascusuniversity.edu.sy; maher.saifo@yahoo.com ORCID IDsMousa Alali: https://orcid.org/0000-0001-9132-1384Ayla Kouli: https://orcid.org/0009-0002-9337-250X Ahmad Al-Bitar: https://orcid.org/0009-0009-3173-1942Maher Saifo: https://orcid.org/0000-0002-5418-3186 *Corresponding authorMaher SaifoFaculty of Medicine, Damascus University, Fayez Mansour Street, P. O. Box: 222, Damascus, Syrian Arab Republic Telephone number: +963 944405424E-mail: maher.saifo@damascusuniversity.edu.sy; maher.saifo@yahoo.comList of presentations of the same material NoneCompeting interests The authors declare that they have no conflicts of interest.DisclosureThis is an original manuscript that has not been published previously and is not being submitted elsewhere.Word count283AbstractBackground: Syria faces a particularly high burden of breast cancer. Gestational breast cancer represents a distinct and complex subset of breast malignancies. It is defined as breast cancer diagnosed during pregnancy or within one year postpartum. It affects approximately one in every 3000 pregnancies. There is a notable gap in research comparing the demographic, clinical, and pathological characteristics of gestational breast cancer patients and other breast cancer patients within the Syrian context. This study aims to address this gap by providing a comprehensive comparison between these two distinct groups in Syria. Methods: This retrospective cohort study included patients with a histologically confirmed diagnosis of breast cancer between January 1, 2005, and June 1, 2025. Data were retrieved from Al-Bairouni University Hospital’s breast registry.Results: Our study included 164 female patients diagnosed with Breast Cancer. Patients were distributed among two groups: “Gestational breast cancer” group and “Other breast cancer patients” group. The gestational group consisted of 80 patients, while the other group consisted of 84 patients. Invasive ductal carcinoma was the most common histological subtype among both groups (90.2%). Tumors in the right breast were seen more in gestational breast cancer patients (60%) compared to the other group (38.8%). HER2-positive status was higher in the gestational group (59.7%). PR positivity was also higher in gestational breast cancer patients (56.3%). Conclusion: Our findings suggest that patients with gestational breast cancer in Syria may present with more aggressive biological features, particularly regarding HER2 expression. This could affect treatment strategies and disease prognosis. However, the endpoints of our study are identifying disease free survival and overall survival in both groups, which hasn’t been reached yet. Keywords: breast cancer, human epidermal growth factor receptor 2, pregnancy, Syria
Presentation numberPS4-12-15
Racial and Ethnic Disparities in Mental Health Care Among Breast Cancer Survivors with Serious Psychological Distress
Ahmed Allibhai, Southlake Hospital, Newmarket, ON, Canada
A. Allibhai1, A. Allibhai2; 1Student, Southlake Hospital, Newmarket, ON, CANADA, 2Student, Holy Trinity School, Richmond Hill, ON, CANADA.
Background: Breast cancer survivors with serious psychological distress (SPD) often require mental health support, yet access and utilization may differ by race/ethnicity. This study examined disparities in mental health service use and psychotropic medication utilization across racial and ethnic groups. Methods: We conducted a cross-sectional analysis using pooled 2018-2022 data from the Medical Expenditure Panel Survey. Adult female breast cancer survivors with SPD were identified and grouped as Non-Hispanic White (NHW), Non-Hispanic Black (NHB), Hispanic, and Non-Hispanic Other (NHO). We present sociodemographic characteristics (Table 1) and adjusted comparisons of counseling visits, provider visits, mental health costs, and psychotropic medication use (Table 2). Multivariable models adjusted for education, marital status, poverty level, and insurance type. Results: Compared to NHW patients, NHB patients had significantly fewer counseling visits (OR: -1.64, 95% CI: -2.59 to -0.72) and lower antidepressant (-14.6%, 95% CI: -24.9 to -5.7) and anxiolytic use (-12.8%, 95% CI: -19.6 to -4.7). Hispanic patients also had reduced antidepressant use (-9.9%, 95% CI: -18.9 to -0.6), and NHO patients had fewer psychiatrist visits (-1.19, 95% CI: -1.92 to -0.53) and lower anxiolytic use (-15.1%, 95% CI: -24.9 to -4.8). No significant differences in mental health visit costs or out-of-pocket spending were found across groups. Conclusion: Despite similar cost burdens, NHB and Hispanic breast cancer survivors with SPD are less likely to receive counseling and psychotropic medications than NHW peers. Targeted interventions are needed to reduce racial disparities in mental health care among cancer survivors.
| NHB vs NHW | H vs NHW | NHO vs NHW | |
| Office Based Mental Health Professional Visits | |||
| Psychiatrist Visits | 0.39 (-0.7, 1.41) | 1.68 (-0.13, 3.36) | -1.19 (-1.92, -0.53) |
| Psychologist Visits | -0.79 (-1.82, 0.21) | 0.82 (-1.39, 2.57) | 0.49 (-2.19, 3.03) |
| Counseling Visits | -1.64 (-2.59, -0.72) | -0.83 (-1.82, 0.35) | -0.83 (3.21, 1.45) |
| Expenditures (USD) of mental health professional visits | |||
| Total cost (USD) | -180 (-388, 9) | -2 (-214, 203) | -257 (-562, 39) |
| Out of pocket cost (USD) | -63 (-130, 19) | -49 (-93, 13) | -39 (-107, 36) |
| Doctor’s Prescription Patterns | |||
| Was medication prescribed during this visit | 22.59 (9.27, 34.36) | 8.04 (-1.4, 18.56) | 2.75 (-11.32, 17.32) |
| Psychotropic Medication Utilization (Purchased) | |||
| Any psychotropic medication | -15.6 (-24.6, -4.2) | 99.3 (-20.3, 0.6) | -17.7 (-33.6, -2.5) |
| Antidepressant | -14.6 (-24.9, -5.7) | -9.9 (-18.9, -0.6) | 15.2 (-31.2, 0.8) |
| Antipsychotic | 2.7 (5.1, 11.2) | 3.4 (4.6, 10.8) | 0.2 (-6.2, 6.8) |
| Anxioltyic | -12.8 (-19.6, -4.7) | 1.5 (-8.4, 12.5) | -15.1 (-24.9, -4.8) |
Presentation numberPS4-12-16
A Transitional Era of Breast Cancer Care after Sanctions Cessation in Syria: Current Challenges and Future Aspirations
Ayla Kouli, Damascus University Faculty of Medicine, Damascus, Syrian Arab Republic
A. Kouli, F. Al Jojo, M. Saifo; Hematology/oncology, Damascus University Faculty of Medicine, Damascus, SYRIAN ARAB REPUBLIC.
Ayla Kouli1, Fatima Al-Jojo1,2, and Maher Saifo1, 21Faculty of Medicine, Damascus University, Damascus, Syrian Arab Republic2Department of Oncology, Albairouni University Hospital, Faculty of Medicine, Damascus University, Damascus, Syrian Arab RepublicFull institutional mailing addressesAyla Kouli, and Maher Saifo: Faculty of Medicine, Damascus University, Fayez Mansour Street, P. O. Box: 222, Damascus, Syrian Arab RepublicMaher Saifo: Department of Oncology, Albairouni University Hospital, Harasta M5, Damascus University, Damascus, Syrian Arab RepublicE-mail addressesAyla Kouli: aylakouly01@gmail.comFatima Al-Jojo: Fatimaaljojo17@gmail.com Maher Saifo: maher.saifo@damascusuniversity.edu.sy; maher.saifo@yahoo.com ORCID IDsAyla Kouli: https://orcid.org/0009-0002-9337-250X Maher Saifo: https://orcid.org/0000-0002-5418-3186 *Corresponding authorMaher SaifoFaculty of Medicine, Damascus University, Fayez Mansour Street, P. O. Box: 222, Damascus, Syrian Arab Republic Telephone number: +963 944405424E-mail: maher.saifo@damascusuniversity.edu.sy; maher.saifo@yahoo.comList of presentations of the same material NoneCompeting interests The authors declare that they have no conflicts of interest.DisclosureThis is an original manuscript that has not been published previously and is not being submitted elsewhere.Background:Syria has been through years of armed conflict and prolonged international sanctions. Syria’s healthcare system has been severely damaged by this conflict, specifically cancer care. Breast cancer patients in Syria are usually unable to receive the adequate care. Methods:This is a cross-sectional observational study that is conducted at Al-Bairuni University Hospital in Syria. It aims evaluate the state of breast cancer care in Syria during the past five years. It focuses on diagnostic methods, treatment access, and healthcare delivery to Breast Cancer patients in Syria. Data is collected by reviewing medical records of all patients diagnosed with breast cancer between 2019 and 2024 at Al-Bairuni University Hospital.Results:Al-Bairuni University Hospital is the country’s primary cancer center. It receives around 50% of all cancer patients in Syria. 12330 patients were diagnosed with breast cancer between 2019 and 2024. This is approximately 25% of all cancer patients at Al Bairuni University Hospital between 2019 and 2024. A majority of these patients had advanced diseases. This is likely due to lack of knowledge, early screening methods, and diagnostic techniques. Access to immunotherapy and targeted therapy is limited, and chemotherapy agents are usually intermittently available. Radiation therapy is also affected by the shortage of functioning machines and electronic power supply. Breast oncologists encounter high volume of patients daily (~ 200), which compromises care quality. Furthermore, the need for a national cancer registry is limiting epidemiological studies and research on surveillance. Clinical trials are still absent in Syria likely due to healthcare deterioration. Conclusion:Syria’s transition into an era without conflict and international sanctions, raises hopes for a better future for breast cancer patients. Reconstructing the oncology infrastructure in Syria must be a global concern. International collaborations must occur to increase drug availability for patients, advance diagnostic tools, and initiatiate clinical trials in Syria. Keywords: breast cancer, Syria
Presentation numberPS4-12-17
Analyzing Mortality Trends and Disparities in Pulmonary Embolism and Breast Cancer among U.S. Patients: A Decade Long Population-Based Retrospective Study, 2010-2020.
Alina Abbas, Mayo Hospital, Lahore, Pakistan
A. Abbas1, H. Habib1, A. Tayyab2, S. A. Khan3; 1Internal Medicine, Mayo Hospital, Lahore, PAKISTAN, 2Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 3Internal Medicine, Ghurki Trust Teaching Hospital, Lahore, PAKISTAN.
Background Patients with breast cancer are particularly vulnerable to developing Pulmonary Embolism (PE) due to factors like cancer-induced hypercoagulability, treatment with chemotherapy, and frequent hospital stays. Despite this known association, there is a lack of comprehensive, population level data tracking PE-related mortality trends in this population over time. Objectives To evaluate national trends and disparities in PE-related mortality among U.S. breast cancer patients from 2010 to 2020. Methods We conducted a retrospective analysis of adults aged 25 years of age and older from 2010-2020 using CDC WONDER Multiple Cause of Death Data. Relevant ICD Codes for Pulmonary Embolism (I26) and Malignant Neoplasm of Breast (C50) were utilized. Data for total number of deaths and Age Adjusted Mortality Rate (AAMR) per 100,000 population was extracted. Joinpoint Regression Program was used to analyze mortality trends and calculate Annual Percentage Changes (APC with relevant 95% Confidence Interval), with further analyses stratified by race and census regions. Results Between 2010 and 2020, a total of 488 deaths were seen in Hispanic or Latino Population (AAMR 0.19), and a total of 8020 deaths were noted among the Non Hispanic Population in the U.S. (AAMR 0.34) attributed to PE and Breast Cancers. APCs among the Non Hispanic Black or African American Population (2010-2012: 10.68, then 2012-2015: -10.91, and 2015-2020: 8.87) and White Population (2010-2012: 10.13, then 2012-2016: -2.18, and 2016-2020: 7.02) appeared to be uptrending initially; downtrending observed later on, and a significant rise being observed in the recent years. Among the Hispanic or Latino Population, APCs were downtrending initially with an uptrending noticed in the recent years (2010-2016: -3.46, then 2016-2020: 17.80). We were not able to calculate AAMR and APCs for American Indian/ Alaska Natives and Asian/ Pacific Islanders due to limited data reliability. Regionally, the South reported the highest number of deaths (3020), followed by the Midwest (1996), West (1923), and Northeast (1591). AAMRs were highest in the Midwest (0.36), followed by the West (0.34), Northeast (0.32), and South (0.31). For Northeast, mortality trends appeared to be increasing (APC: 0.21), while for the Midwest (2010-2017: -0.93, then 2017-2020: 15.82), South (2010-2018: -0.57, then 2018-2020: 18.86) and West (2010-2012: 17.24, then 2012-2017: -3.26, then 2017-2020: 12.08), a similar pattern of initial decline and a recent incline, particularly significant in the Midwest, was observed in the recent years. Conclusion Recent years have seen a concerning rise in mortality related to Pulmonary Embolism and Breast Cancer across the U.S., with notable disparities based on geography and demographics. These patterns highlight an urgent need for targeted research and tailored preventive efforts to close the gaps, and improve survival and patient outcomes.
Presentation numberPS4-12-18
Socioeconomic Status and Delayed Breast Cancer Care in Southeast Texas:ARetrospective Cohort Study
Abdullah Jamal, Baptist Hospitals of Southeast Texas, Beaumont, TX
A. Jamal1, J. Thompson2, S. Devi1, P. Jain1, N. Ganatra1, J. Anwar3, M. Khalil4, T. Terro5; 1Internal Medicine, Baptist Hospitals of Southeast Texas, Beaumont, TX, 2Department of Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Science Center,, El Paso, TX, 3Department of Medicine, Division of Hematology and Oncology, McGovern Medical School, UT Health, Beaumont, TX, 4Hematology Oncology, Baptist Hospitals of Southeast Texas, Beaumont, TX, 5Department of Special Populations Research and Novel Therapeutics, Baptist Hospitals of Southeast Texas, Beaumont, TX.
BACKGROUND: Breast cancer is the most common cancer diagnosis and the second leading cause of female cancer-related death in the US. Time-to-treatment-initiation (TTI) has been shown to be critical for improving disease and survival outcomes across cancer types, including breast cancer. Socioeconomic status (SES) and associated disparities are increasingly recognized as influential factors in access to and timing of cancer care. As such, this retrospective cohort study investigated associations between SES and delays in breast cancer TTI across various treatment modalities in Southeast Texas. METHODS: Logistic regression models assessed the associations between treatment delays and socioeconomic or clinical characteristics among 609 patients. RESULTS: Regarding overall treatment, males, unmarried patients, younger patients, and Medicaid users experienced increased delay odds, while females, widows, and Medicare users experienced decreased odds. For chemotherapy, Tricare coverage was associated with excessive delay odds. Regarding surgical intervention, African Americans, unmarried patients, younger patients, and Medicaid users experienced increased delay odds, while Caucasians, older patients, and Medicare users experienced decreased odds. For radiation therapy, divorcees and those with Medicare and Medicaid eligibility experienced increased delay odds.Regarding hormone therapy, younger patients and Medicaid users experienced increased delayodds, while older patients and Medicare users experienced decreased odds. For immunotherapy, unmarried patients experienced increased delay odds, while being married conferred lower odds. CONCLUSION: These results indicate that certain SES factors may significantly predict treatment delays in breast cancer care. As such, targeted strategies addressing disparities, social support, and care coordination are needed to reduce inequities and improve appropriate TTI.
Presentation numberPS4-12-19
Ethnoregional Disparities in Breast Cancer Screening: A National Database Analysis
Shubhank Goyal, University of Texas Rio Grande Valley, McAllen, TX
S. Goyal1, A. Calderon1, D. Hernandez1, S. Khan2, S. Chauhan2, D. Nguyen3; 1Internal Medicine, University of Texas Rio Grande Valley, McAllen, TX, 2Cancer Immunology and Microbiology, University of Texas Rio Grande Valley, McAllen, TX, 3Medical Oncology, University of Texas Rio Grande Valley, McAllen, TX.
Background:Despite breast cancer being the most diagnosed cancer among women in the United States, persistent disparities in screening mammography remain. Non-Hispanic Black, Hispanic, and American Indian/Alaska Native (AIAN) women are disproportionately affected by lower screening rates, contributing to delayed diagnoses and worse outcomes. This study aims to examine these disparities through a comprehensive analysis of national prevalence data. Methods: We used 2022 model-based, age-adjusted mammography prevalence data from the Behavioral Risk Factor Surveillance System (BRFSS), based on responses to the core question, “Have you had a mammogram in the past two years?” for women aged 50-74 years. Ethnic groups analyzed included Non-Hispanic White, Hispanic, Non-Hispanic Black, American Indian/Alaska Native (AIAN), and Asian/Pacific Islander (API). U.S. regions were defined using Census Bureau classifications: Northeast, Midwest, South, and West. All ethnic groups were included in descriptive analysis; however, stratified comparisons focused on historically underserved populations (Hispanic, Non-Hispanic Black, and AIAN) due to reliability and consistency of state-level estimates. The dataset comprised 95,728 respondents from all 50 states and D.C.(District of Columbia), with prevalence estimates age-standardized to the 2000 U.S. standard population. States or subgroups with fewer than 50 respondents were excluded. Differences across groups were assessed using Kruskal-Wallis tests and Dunn’s post-hoc comparisons with Bonferroni correction. Results: Stratified inferential analyses emphasized historically underserved populations due to consistency and completeness of data reporting. Statistically significant differences in screening prevalence were found by both ethnicity and region (Kruskal-Wallis p < 0.001). Non-Hispanic Black women had the highest screening rates nationally, while Hispanic and AIAN women had significantly lower rates. Regional analysis showed that Hispanic women in the West had markedly lower screening rates than those in the Northeast (adjusted p = 0.03, Dunn’s test). The most pronounced disparities were seen in the West, where both Hispanic and AIAN women were under-screened. In contrast, Non-Hispanic Black women maintained high screening coverage across all regions. The interaction effect between ethnicity and region was evident, with screening gaps varying by both demographic and geographic factors. These findings underscore the need for region- and ethnicity-specific screening interventions, particularly in areas like the Western U.S. where historically marginalized groups remain at elevated risk for under-screening. Conclusion: Improving screening coverage among Hispanic and AIAN populations—especially in underserved regions—will be critical to reducing late-stage diagnoses and improving outcomes. These findings support the urgent need for public health efforts that are community-driven, culturally tailored, and data-informed. As Medicare and Medicaid continue to evolve, proactive investment in regionally customized screening infrastructure, mobile outreach, and language-accessible education can help close persistent gaps. Continued national surveillance and improved data reporting for API and other racial groups are also essential to ensure equitable breast cancer prevention efforts. Further research is needed to understand how regional context influences screening and treatment disparities within the same ethnic populations.
Presentation numberPS4-12-20
Regional Variation in Guideline Concordance for Women with Triple-Negative and HER2+ Breast Cancer in Nova Scotia
Rachel Hemsworth, Izaak Walton Killam Health, Halifax, NS, Canada
H. Stewart, A. Mayo, T. Li, R. Hemsworth, G. Knapp, A. Drohan; Surgery, Izaak Walton Killam Health, Halifax, NS, CANADA.
Background: Triple-negative (TNBC) and human epidermal growth factor receptor 2 positive (HER2+) breast cancers are associated with an elevated risk of metastatic tropism and poor outcomes. Since 2020, systemic neoadjuvant therapy (NAT) has been recommended for both forms of the disease when tumor size at diagnosis is equal to or over 2 cm. The objective of this study was to quantify the proportion of patients in Nova Scotia (NS) with resectable TNBC and HER2+ breast cancer that received guideline concordant neoadjuvant therapy. Methods: We conducted a retrospective analysis using 2021-2023 data from the NS Breast Screening Program. Adult patients (18-80 years) with an incident diagnosis of non-metastatic, ≥T2, triple negative or HER2+ breast cancer were considered theoretically eligible for NAT and included in the analysis. The neoadjuvant intervention was considered given if the patient had a wait-time of ≥4 months between tissue diagnosis and surgery. Our primary outcome was the proportion of theoretically eligible patients who received guideline-concordant NAT. Secondary outcomes included the proportion of patients who experienced a pathologic complete response (pCR). Receipt of guideline concordant care was compared over time (2021-2023) and between health system administrative zones (Central, Western, Northern, Eastern). Results: A total of 291 women were included, with a median age of 59 (IQR: 48-67). Forty-five percent were treated in the Central Zone. Of the cohort, 49.5% were TNBC and 50.52% were HER2+. Overall, 64.6% of eligible patients appear to have received NAT. There were differences in receipt of NAT between administrative zones (Central 70.8%, Eastern 69.5%, Northern 55.6%, Western 52.1%, p=0.046). The overall pCR for the cohort was 37.8% and the response was consistent across the zones (% range: 34.2%-40.0%, p=0.951).
Conclusion: This study highlights regional disparities in the adherence to NAT guidelines for TNBC and HER2+ breast cancers. Future efforts should focus on strategies to improve NAT guideline adherence, which will improve outcomes for NS breast cancer patients.
Presentation numberPS4-11-14
Anthracycline-based Systemic Chemotherapy on Long-term Prognosis of Metaplastic Breast Cancer: A Real World Study with 637 Cases in 14 multi-center
Can Zhou, First Affiliated Hospital of Xi’ an Jiaotong University, Xi’ an, China
C. Zhou1, P. Ni2, Z. Dai3, G. Qiao4, Y. Ren1; 1First Affiliated Hospital of Xi’ an Jiaotong University, Xi’ an, CHINA, 2ShaanXi Provincipal People’s Hospital, Xi’ an, CHINA, 3First Affiliated Hospital, Zhe Jiang University, School of Medicine, Hang Zhou, CHINA, 4Yantai Yuhuangding Hospital, Yan Tai, CHINA.
Anthracycline-based Systemic Chemotherapy on Long-Term Prognosis of Metaplastic Breast Cancer: A Real World Study with 637 cases in 14 multi-center Abstract:Background: The standard chemotherapy treatment regimen for metaplastic breast cancer (MpBC) remained uncertain. This study aimed to assess the survival disparities between anthracycline-based and non-anthracycline-based regimens and investigate appropriate chemo-regimen for MpBC patients in real world.Patients and Methods: A total of 637 cases with MpBC in admitting diagnosis between 2002 and 2025 were collected from 14 large hospitals in China. Kaplan- Meier analysis, univariate and multivariate Cox proportion hazard models, and cumulative incidence flow were performed to estimate survival effect and identify prognostic factors comprehensively during the median follow-up time of 30 months. Results: Among the 373 patients enrolled in final study, totally 243 (65.1%) patients received anthracycline-based regimen (anthracycline cohort) whereas 130 (34.9%) patients with non-anthracycline-based regimen (non-anthracycline cohort). Subjects in anthracycline cohort had an absolute improvement of 13.6% (89.4% vs. 75.8%) and 7.5% (82.9% vs. 75.9%) in 10-year overall survival (OS) and 10-year local-regional recurrence free survival (LRFS), respectively, in comparison with those in non-anthracycline cohort. And anthracycline-based regimen was found to be an independently favorable factor of OS through multivariate Cox analysis. The local-regional recurrence occurred in 6.2% (23/373) of patients, while 13.1% (49/373) of MpBC patients developed distant metastases.Conclusion: This multi-center study identified the association between administration of anthracycline and improved OS, anthracycline-based regimen had higher likelihood to be the preferred protocol for MpBC.
Presentation numberPS4-10-26
Pretreatment neutrophil-to-lymphocyte ratio is associated with pathologic complete response following neoadjuvant chemo-immunotherapy in early triple-negative breast cancer
Alexis LeVee, UCLA David Geffen School of Medicine, Los Angeles, CA
A. LeVee1, E. Yang1, K. Tsai2, N. Baclig1, A. Soliman1, S. Zhang1, A. Kordic2, J. Mortimer2, M. Lechner1, S. Alkassis1, N. McAndrew1, M. Lipsyc-Sharf1, M. Sedrak1, R. Callahan1, A. Master1, D. Prager1, K. McCann1, A. Bardia1; 1UCLA David Geffen School of Medicine, Los Angeles, CA, 2City of Hope Comprehensive Cancer Center, Duarte, CA
Background: Neoadjuvant immunotherapy with chemotherapy is widely used for management of localized triple negative breast cancer (TNBC). While tumor infiltrating lymphocyte (TILs) have been associated with therapy response, the impact of absolute lymphocyte count (ALC) and neutrophil-to-lymphocyte ratios (NLR) in modulating response is not known. In this study, we evaluated the association between ALC, NLR, and therapeutic response to chemo-immunotherapy in patients with TNBC. Methods: In this multi-institutional retrospective study, patients with stage I-III TNBC diagnosed between 2019 to 2023 were included who were treated with at least one cycle of neoadjuvant pembrolizumab and chemotherapy at two comprehensive cancer centers. Pre-treatment laboratory values were collected for all patients. An NLR cut-off value of 5 was used based on previous studies. Univariate and multivariate logistic regression and Cox proportional hazards models were used to assess the impact of pretreatment NLR on pCR. Results: A total of 372 patients with early-stage TNBC were included with an overall pCR rate of 61.3%. The median baseline NLR was 2.21 (95% CI, 2.10-2.30), and 9.4% of patients had a baseline NLR >5. Patients with a baseline NLR >5 had significantly lower pCR rates compared to those with NLR <5 (45.7% vs. 62.9%; p=0.047). Similarly, baseline lymphopenia was also associated with lower pCR rates (45.9% vs. 63.0%; p=0.043). Multivariate logistic regression demonstrated that baseline NLR >5 was significantly associated with inferior pCR rates (odds ratio [OR], 0.49; 95% CI 0.24-0.997; p=0.049), adjusting for age, race and stage. Conclusions: This is the first study to demonstrate that absolute (and relative) lymphopenia is associated with lower pCR rates in patients with early-stage TNBC treated with neoadjuvant chemo-immunotherapy. Further research is needed to further evaluate lymphocyte count as a biomarker of neoadjuvant chemo-immunotherapy response and develop potential therapeutic strategies to improve lymphopenia in patients with early-stage TNBC.
Presentation numberPS4-11-15
Effect of exercise training during neoadjuvant chemotherapy on breast cancer pathological complete response
Ruffo Freitas-Junior, Federal University of Goias, Goiânia, Brazil
V. D. L. Rosa1, R. R. Alves1, C. S. Anjos2, R. M. S. Rahal1, D. V. G. Santana1, L. R. Soares1, C. A. Vieira1, R. Freitas-Junior1; 1Federal University of Goias, Goiânia, BRAZIL, 2Hospital Memorial Arthur Ramos, Maceio, BRAZIL.
Background: Physical activity is related to reduced mortality and recurrence in breast cancer patients, as well as fewer severe adverse effects during and following its treatment. The potential of exercise as a concurrent therapy for inhibiting primary tumor growth has been suggested by preclinical and observational studies. However, the impact of exercise on efficacy of neoadjuvant chemotherapy (NAC) for early breast cancer is unclear and prospective controlled studies are lacking. Therefore, we aimed to perform a meta-analysis exploring the effect of exercise training during NAC on the pathologic complete response (pCR) in breast cancer. Methods: We systematically searched PubMed, EMBASE and Cochrane databases for randomized controlled trials (RCTs) that compared exercise training or usual care during NAC in breast cancer patients. The primary endpoint of interest was pCR. Subanalyses were performed for pCR by breast cancer subtype. Heterogeneity was examined with I2 statistics. A random-effects model was used for outcomes with high heterogeneity. The protocol of this systematic review is registered with PROSPERO under the identifier CRD420251142764. Results: We included 5 RCTs with 465 patients undergoing NAC, met the inclusion criteria for this meta-analysis, of whom 258 (55.4%) received training. The mean age ranges from 48.4 to 52.3 years on the exercise training group and 45.0 to 53.3 years on control group. The pooled pCR rate was 51.5% in the exercise group compared with 46.3% in the usual care group. Overall, there was no significant difference between groups (odds ratio [OR] 1.28; 95% confidence interval [CI] 0.83-1.98; Figure 01). Similarly, the subgroup analysis revealed that pCR in breast cancer subtype hormone receptor-positive HER2-negative (OR 2.50; 95% CI 0.74-8.42), HER2-positive (OR 1.17; 95% CI 0.54-2.53) and triple negative (OR 1.19; 95% CI 0.40-3.54) were not significantly different between groups. Conclusion: Our meta-analysis suggests that the exercise training during neoadjuvant chemotherapy is not significantly different from usual care on breast cancer pathological complete response.
Presentation numberPS4-02-05
Patient-derived organoids (PDO) as functional models to capture interindividual heterogeneity in breast cancer chemotherapy response.
Erico Tosoni Costa, Institute of Education and Research of the Hospital Sírio-Libanês (IEP-HSL), Sao Paulo, Brazil
E. T. Costa1, L. D. Simões1, D. G. Giannotti2, E. X. dos Santos3, A. R. A. Canteli1, C. H. dos Anjos1, J. L. B. Bevilacqua1, R. M. Junior1, P. C. M. da Silva1, P. A. d. Teixeira4, L. A. J. Moyses2, L. A. Yamashita4, C. R. Saccarelli4, A. C. S. D. Barros1; 1Institute of Education and Research of the Hospital Sírio-Libanês (IEP-HSL), Sao Paulo, BRAZIL, 2Imaging Diagnostics Center of the Hospital Sírio-Libanês (IEP-HSL), Sao Paulo, BRAZIL, 3Institute of Education and Research of the Hospital Sírio-Libanês, Sao Paulo, BRAZIL, 4Medical Imaging Center of the Hospital Sírio-Libanês (IEP-HSL), Sao Paulo, BRAZIL.
Patient-Derived Organoids (PDO) as Functional Models to Capture Interindividual Heterogeneity in Breast Cancer Chemotherapy Response Erico T. Costa¹, Lucas D. Simôes¹, Larissa AJ Moyses², Carolina R. Saccarelli², Ernande X. dos Santos¹,Amanda R.A. Canteli¹, Patrícia AC Teixeira², Carlos H. dos Anjos¹, José L.B. Bevilacqua¹, Daniela G.Giannotti¹, Ronaldo Morales Jr.¹, Priscila C. Silva¹, Alfredo C.S.D. Barros¹ ¹Instituto Sírio-Libanês de Ensino e Pesquisa, Centro de Oncologia Molecular, Hospital Sírio-Libanês,São Paulo, Brazil. ²Núcleo de Diagnóstico por Imagem da Mama, Hospital Sírio-Libanês, São Paulo,Brazil. Background: Breast cancer is the leading cause of cancer-related mortality in women worldwide. Chemotherapy resistance remains a critical barrier to durable outcomes, and no robust predictive biomarkers are available for standard regimens.Consequently, many patients are exposed to empirically applied polychemotherapy with suboptimal efficacy and unnecessary toxicity. Recent evidence indicates that the clinical benefit of many combinations reflects independent drug action (IDA),whereby distinct patients benefit from distinct agents rather than from true pharmacologic synergy. This model highlights an opportunity to replace empiric combinations with personalized monotherapies. Methods: We established a functional precision oncology platform using PDOs to evaluate drug responses in breast cancer. Thirty biopsies from 29 patients with invasive carcinomas treated with the AC-T regimen (doxorubicin/cyclophosphamide and paclitaxel) were collected.PDOs were successfully established in 22 cases (76%) and preserved the histopathological and immunohistochemical profiles of parental tumors (ER/PR/HER2, Ki-67). Drug responses were quantified using Growth Rate (GR)metrics and integrated into a Sequential Efficacy of Chemotherapy (SEC) score aligned with clinically relevant free (unbound) plasma concentrations (Cmax, corrected for protein binding). Results: When challenged with the complete AC-T regimen, PDOs revealed that 20% of tumors were resistant and 80% were sensitive.Within the sensitive group, however, only 32% demonstrated evidence of synergistic interaction between doxorubicin and cyclophosphamide, whereas in 68% the effect could be attributed to a single agent, consistent with the Independent Drug Action (IDA) paradigm. Dissection of individual drugs confirmed pronounced heterogeneity:43% of PDOs were resistant to doxorubicin, 25% to 4-hydroxy-cyclophosphamide, and 71% to paclitaxel, with 6% showing cross-resistance to both anthracycline and taxane. Importantly, PDO profiling uncovered alternative vulnerabilities, such as sensitivity to epirubicin in some doxorubicin-resistant cases and to docetaxel in a subset resistant to paclitaxel. These results demonstrate that PDO assays can disentangle the relative contribution of each drug within combination regimens, providing a functional basis to understand clinical benefit as arising from patient-to-patient variability rather than from true pharmacologic synergy. Conclusions: PDO-based assays capture interindividual heterogeneity in drug response and align with the IDA paradigm, explaining why most patients benefit from only a subset of drugs in standard combinations. By prospectively identifying the most effective single agent per patient, PDO-guided therapy could shift breast cancer management from empiric polychemotherapy toward individualized monotherapies. This approach offers the dual advantage of maximizing efficacy and minimizing toxicity, with the potential to fundamentally redefine precision chemotherapy in breast cancer.
Presentation numberPS4-12-21
Breast Cancer Profile and Healthcare Practices Among Transgender Patients within the Brazilian Unified Health System: a mixed cross-sectional and survey study (TRANSformation)
Jesse Lopes da Silva, Sociedade Brasileira de Oncologia Clínica (SBOC), São Paulo, Brazil
I. C. Schultz1, J. L. da Silva2, L. Landeiro3, N. C. Nunes4, R. Sant’Ana5, G. Onzi1, N. Viana1; 1AstraZeneca, São Paulo, BRAZIL, 2Sociedade Brasileira de Oncologia Clínica (SBOC), São Paulo, BRAZIL, 3Grupo Brasileiro de Estudos em Câncer de Mama (GBECAM), São Paulo, BRAZIL, 4Rede Americas, Rio de Janeiro, BRAZIL, 5Escola de Enfermagem de Ribeirão Preto da Universidade de São Paulo, Ribeirão Preto, BRAZIL.
Introduction: Breast cancer (BC) is the most diagnosed cancer worldwide, predominantly affecting cisgender women, while in cisgender men it represents only 1-2% of cases. For the transgender population, estimated at 0.5-1% of the general population, there is a significant information gap. BC risk is more complex in this population due to gender-affirming hormone therapies and surgeries. In Brazil, data is even more scarce. Within the Brazilian Unified Health System (SUS) database, the absence of structured mechanisms for recording gender identity since records rely solely on sex assigned at birth renders transgender people largely invisible, hindering equitable care and compromising early detection and management. This is the first nationwide study assessing BC care for transgender patients in Brazil using two complementary approaches: a retrospective analysis of 18 years of SUS database data to identify barriers in care and registration, and a nationwide physician survey to gather insights on institutional preparedness, challenges, and perspectives for screening and treating BC in transgender individuals. Methods: Outpatient records from the SUS database (2008-2025) were analyzed, encompassing a cohort of patients identified through cross-referencing gender-affirming procedure registries as a proxy for transgender identity. BC cases were ascertained via diagnostic codes and oncological treatment records. Complementarily, a nationwide anonymous online survey was conducted with medical oncologists, breast surgeons, gynecologists and other physicians involved in the transgender patients journey across diverse Brazilian regions. The survey explored perceptions regarding screening practices, adherence to clinical guidelines, and institutional readiness to deliver equitable oncological care for this population. Results: Analysis of SUS database records (2008-2025) identified 12,648 patients who underwent gender-affirming procedures. Within this cohort, only six cases of breast cancer were recorded (five in trans men and one in trans woman) representing <0.0005% of the cohort. This figure likely reflects underdiagnosis and underreporting rather than a truly low incidence, although it aligns with trends observed internationally. Additionally, the survey with over 240 physicians, spanning public, private, and mixed healthcare services across all five Brazilian regions, exposed critical gaps in transgender oncology care. Over 65% reported not following specific protocols for transgender patients, nearly 50% acknowledged insufficient knowledge, and almost 80% had not received institutional training or guidance. Collectively, these results underscore the pressing need for inclusive clinical guidelines, structured professional training, and robust health information systems to ensure equitable cancer care for transgender populations. Conclusion: This first nationwide analysis of over a decade of SUS records exposes critical gaps in registration and care. The transgender population remains largely invisible in SUS’s information systems, compromising early detection and effective management of BC. The lack of documented cases, combined with systemic barriers such as inconsistent sex/gender registration and insufficient professional training, underscores the need for urgent action. These findings reveal a convergence between database analysis and physician perceptions, pointing to the same structural barriers. To address them, it is essential to improve health information systems, implement comprehensive professional training, and establish inclusive public health policies.
Presentation numberPS4-03-02
P-climb: pet-ct and liquid biopsy for monitoring breast cancer bone metastasis
Deloris Veney, The Ohio State University, Columbus, OH
D. Veney1, H. LeFebvre1, L. Wei1, G. Bader1, M. Cherian1, A. Davenport1, M. Gatti-Mays1, K. Johnson1, S. Sardesai1, R. Wesolowski1, N. Williams1, K. Shanahan2, V. Prasath1, B. To1, K. Hoskova3, M. Childress3, J. C. F. Quintanilha3, R. P. Graf3, M. Levy3, L. W. Pasquina3, E. Kubi-Appiah1, D. G. Stover1; 1The Ohio State University, Columbus, OH, 2METAvivor Research and Support, Elmwood, WI, 3Foundation Medicine Incorporated, Boston, MA
P-CLIMB: PET-CT and Liquid Biopsy for Monitoring Breast Cancer Bone Metastasis Background: Bone metastases (mets) develop in 50-70% of patients with metastatic breast cancer (MBC) and are associated with significant morbidity due to skeletal-related events (SREs), associated with significant decline in quality of life. Of the 180,000 patients living with MBC in the United States, 30-45% have bone mets as their only site of disease (‘bone-only’/BO-MBC). Patients with BO-MBC are frequently excluded from clinical trials because bone mets are not ‘measurable.’ In a large nationwide cohort, using the FoundationOne®Liquid CDx circulating tumor DNA (ctDNA) assay we previously demonstrated that ctDNA was just as likely to be detected and ctDNA tumor fraction (TF) remained prognostic in patients with BO-MBC. We hypothesize that ctDNA TF in combination with PET-CT will provide superior monitoring of bone mets. Methods: In this contemporaneously enrolled, retrospectively analyzed study, patients with any hormone receptor subtype receiving any line of systemic therapy undergoing standard-of-care PET-CT were identified for blood collection within 37 days of each PET-CT for ctDNA analyses. Patients were categorized as {BO or bone-lymph node (BO/L-MBC)} versus {bone mets + visceral mets (BV-MBC)} based on disease status. Plasma was banked from 20mL of blood collected at the time of clinical blood draws and underwent analysis by the laboratory developed test FoundationOneMonitor ctDNA assay for research assessment of ctDNA TF (accounting for aneuploidy, variant allele frequency, fragment length, clonal hematopoiesis [CH] and genomic alterations for 324 genes at each timepoint). PET-CT scans were deidentified and reviewed by a study radiologist on VISAGE PACS to determine SUVmax (PET Maximum Standardized Uptake Value) plus number of skeletal/non-skeletal lesions. Outcome measures included progression-free survival (PFS) and time to next therapy (TTNT). Results: A total of 42 patients (21 BO/L-MBC, 21 BV-MBC) completed a total 160 PET-CT & ctDNA pairs(2-7 per patient). ctDNA TF was detected at first available blood draw for 38% (8/21) with BO/L- and 57% (12/21) BV-MBC respectively (Fisher exact p=0.27). Overall, ctDNA TF value demonstrated higher correlation to SUVmax in both BO/L-MBC and BV-MBC cohorts at T2 (Spearman rho range 0.55-0.60, both p≤0.01) than T1 (range 0.43-0.47, p=0.03 and 0.05), with no significant correlation between TF and total number of lesions by PET-CT. Primary analyses focused on pairs of baseline (T1) and next restaging (T2) scan. T1:T2 TF change was not significantly different between the two cohorts (Wilcoxon rank sum p=0.90). For the total study population, TF change and SUVmax change from T1:T2 were each significantly associated with clinician-interpreted imaging response (TF change: Kruskal-Wallis p=0.006; SUVmax change: ANOVA p=0.002). When evaluating TF change from T1:T2, patients whose TF remained increased (relative to stable or declined) had significantly shorter PFS (hazard ratio [HR] 3.7, 95% confidence interval [CI] 1.7-7.8, log-rank p<0.001) and TTNT (log-rank p<0.001). When evaluating BO/L-MBC cohort whose first PET-CT imaging was ‘stable’, patients whose TF increased (relative to stable or declined) had significantly shorter PFS (HR 8.6, 95% CI 1.6-47, log-rank p=0.008). Detailed evaluation of specific genomic alterations revealed expansion of known resistance mutations (including ESR1, ERBB2, and PTEN) in advance of imaging and clinical progression. Conclusion: In patients with BO/L-MBC, ctDNA TF change was significantly associated with PFS and TTNT, even in the setting of PET-CT imaging interpreted as ‘stable.’ ctDNA sequencing offers a clinically useful adjunct to clinical imaging for bone-metastatic MBC, with the potential to expand the definition of measurability to include those with BO-/L-MBC.
Presentation numberPS4-05-02
Rapid and ultrasensitive detection of surgical margin and metastatic lymph node via EpCAM-targeted surface-enhanced Raman scattering biosensor in breast cancer
Kangliang Lou, School of Medicine, Xiamen University, Xiamen, China
K. Lou1, J. Bai2, S. Fu3, X. Shen1, Y. Gao1, S. Lin1, C. Li2, Y. Chen1, G. Zhang2; 1School of Medicine, Xiamen University, Xiamen, CHINA, 2The Breast Center and the Cancer Institute, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University & Peking University Cancer Hospital Yunnan, Kunming, CHINA, 3College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, CHINA.
Purpose: Breast-conserving surgery (BCS) and sentinel lymph node biopsy (SLNB) are cornerstones of precision treatment for early breast cancer, aiming to balance oncological efficacy with cosmetic and functional preservation. However, current intraoperative margin evaluation and SLNB rely on frozen section pathology, which faces inherent technical limitations, including prolonged procedural time and considerable false-negative rates in detecting micro-metastases. To address these challenges, we developed a novel surface-enhanced Raman scattering (SERS) biosensor, targeting breast cancer-specific biomarker epithelial cell adhesion molecule (EpCAM), for rapid evaluation of surgical margins and lymph node metastasis status. Methods: The SERS biosensor was consisted of Au-IR808@EP and Beats@EP. Gold nanoparticles (AuNPs) were synthesized by sodium citrate reduction, which was then functionalized with EpCAM antibody and the Raman reporter IR808 to construct Au-IR808@EP. After physicochemical characterization, EpCAM-targeting ability of the biosensor was evaluated using EpCAM antigen standards and breast cancer cell lines. Tissue homogenates prepared from mouse models bearing high EpCAM-expressing MDA-MB-231-Epcam tumors, MCF-7 tumors, and the metastatic lymph nodes (MLNs) were then analyzed under the SERS biosensor to evaluate the pathology of tissues. Finally, clinical application was validated using surgical specimens from 20 breast cancer patients across two centers to distinguish malignant from normal tissues and identify MLNs. Results: The EpCAM-targeted SERS biosensor was successfully fabricated and formed a stable double-antibody sandwich complex upon antigen binding. It exhibited a detection limit (LOD) of 6.96 pg/mL for EpCAM standard solution. Subsequently, the method demonstrated excellent targeting capability in the detection of various breast cancer cells, with an LOD of 341 cells for MCF-7. In mouse models, the biosensor enabled rapid EpCAM detection within 11 minutes, effectively differentiating between high-EpCAM (MDA-MB-231-EpCAM) and low-EpCAM (MDA-MB-231-NC) tumors and MLNs. Meanwhile, it precisely differentiated tumor from normal tissues in 30 MCF-7 tumor-normal pairs (AUC = 1), and achieved an AUC of 0.995 in detecting 28 MLNs. Clinically, ex vivo rapid evaluation via the SERS biosensor effectively discriminated malignant breast tissues from normal tissues lesions (AUC = 0.91) and identified metastatic lymph nodes (AUC = 0.889) in surgical specimens from 20 BC patients. Conclusion: We successfully constructed an ultrasensitive SERS biosensor for efficient EpCAM targeting and rapid dual-functional assessment of surgical margins and nodal metastasis. As BCS and SNLB procedures play an increasingly important role towards precision surgery, this innovative technology holds strong potential to achieve optimized cosmetic appearances and better clinical outcomes.
Presentation numberPS4-06-30
Increased Survival in Nude Mice Inoculated with MCF7 Breast Cancer (BC) in the Mammary Fat-Pad Achieved via a Single Injection of a Lipid-like Hydrogel Emulsion Containing a New Molecular Entity (NME) and N-allyl Noroxymorphone (NaN) Compared to Tamoxifen (TAM) and Abemaciclib (ABE)
Bryan S Margulies, Zetagen Therapeutics, Syracuse, NY
B. S. Margulies, J. C. Loy; Zetagen Therapeutics, Syracuse, NY
Background: First-line therapies after the diagnosis of a BC malignancy depends on the phenotype (e.g., HR+, HER2+, or triple negative BC) includes neo-adjuvant therapy, surgical intervention (e.g., mastectomy or lumpectomy), and sometimes post-surgical radiation. Both targeted therapies (e.g., TAM and ABE) and chemotherapies are systemically administered, which results in negative off-target effects. Initial results for Zeta-BC-007, which is a fixed-dose drug that comprises an NME and NaN in a lipid-like hydrogel emulsion, shows superior tumoricidal effects that increased survival after a single intratumoral injection relative to groups treated with systemically administered TAM or ABE. Importantly, other hydrophobic drugs, such as ABE, can be loaded into the lipid nanoparticles (LNP) for delivery via Zeta-BC-007. Methods: The NME (lanthanum (III) tri 9,10-diacetoxy octadecanoate) and the lipid-like hydrogel emulsion contains alginate and linoleic acid. All other drugs were obtained commercially. The NME is an intrinsic component of the LPNs in Zeta-BC-007 while other drugs were loaded onto LNPs (size = 200-nm) prior to incorporation into the lipid-like hydrogel emulsion. MCF7 cells treated with the NME, ABE, and NaN in vitro were assessed for cell proliferation and death. Nude mice were inoculated with the MCF7 HR+ BC cells (N=142 total; N=8/group) and treated when tumors were palpable (150-mm3). Mice were sorted into groups: 1) Control; 2) TAM (1-mg/mL) and 3) ABE (125-mg/mL) given via oral gavage every 72-hours; 4) Zeta-BC-007 with the NME only; 5) Zeta-BC-007 with the NME + NaN; 6) Zeta-BC-007 with the NME + ABE; and 7) Zeta-BC-007 with the NME + NaN + ABE. Tumor volumes were measured every 72-hours for 60-days. Excised tumors processed for H&E histology were evaluated with histomorphology (e.g., tumor volume and necrosis) and histopathology. Results: In vitro, a significant dose-dependent decrease in cell number was seen for the MCF7 cells through 72-hours for the NME, NaN, and ABE (p<0.05). The combination of 5-fold less NaN (1-mM) + 10-nM of ABE resulted in a 60% decrease in cell number (p<0.01), suggesting a synergistic effect from pairing the 2 drugs. In the mouse study, we found that TAM and ABE treatment decreased tumor volume 34% and 15%, relative to Controls. Treatment with Zeta-BC-007 with the NME only decreased tumor volume 27% while the tumor volume was decreased 53% for Zeta-BC-007 with the NME + NaN, 57% for Zeta-BC-007 with the NME + ABE, and 66% for Zeta-BC-007 with the NME + NaN + ABE. Zeta-BC-007 loaded with NaN or ABE alone or in combination resulted in a 30% – 60% decrease in tumor volume relative to TAM and ABE administered alone. By day 60, all mice in the Control, TAM, and ABE treated groups were dead. Histology showed a decrease in tumor volume and increased adipose tissue and necrosis for the mice treated with any of the Zeta-BC-007 drug combinations. Mice in the Zeta-BC-007 groups also had no ki67 staining (i.e., no tumor activity) and increased numbers of macrophages, the latter of which was confirmed by increased numbers of circulating monocytes. Conclusion: Treatment of BC via intratumoral injection is a novel therapeutic approach that promises to increase drug bioavailability while eliminating negative off-target effects. ABE and NaN appear to have synergistic treatment effects while the NME also induced significant cytotoxicity. Zeta-BC-007 showed significant reductions in tumor volume via increased necrosis that relates to increased cytotoxicity in the tumor. This increase tumoricidal effect from Zeta-BC-007 led to increased survival. Further work in needed; however, these initial results point to a new treatment paradigm.
Presentation numberPS4-12-22
Uncovering Barriers in Clinical Trials for Cancer Care at a tertiary oncology center – survey feasibility study
Anne-Laure Strong, Penn State College of Medicine, HERSHEY, PA
A.-L. Strong1, E. Wilson1, M. Vasekar2; 1Penn State College of Medicine, HERSHEY, PA, 2Penn State Cancer Institute, HERSHEY, PA
Purpose: Financial Strain and limited geographic access to tertiary treatment centers are known barriers to cancer clinical trial participation. A study conducted by Sydney Kimmel Cancer Center (SKCC) surveyed 2,744 adult residents in the Mid-Atlantic region, assessing their motivations and barriers to enter clinical trials and highlighting unique characteristics of their catchment area. Furthermore, this study proved useful to inform the development of targeted interventions and programs aimed at lowering barriers to participation. Therefore, a pilot study to explore the feasibility of conducting such a survey at Penn State Cancer Institute (PSCI), within its unique population that includes rural population as well as suburban ones was performed. This survey examined the motivation and specific barriers that PSCI’s patients face in relation to taking part in clinical trials. Experimental Design: We conducted a pilot study of 100 patients(pts) at PSCI using a 14-questions paper survey exploring patients’ demographics, attitudes toward clinical trials, and their motivations and perceived barriers to future participation. Patients were also asked to rank their most significant concerns. 100 consecutive pts were surveyed, significant proportion of patients had breast cancer diagnosis. Association between demographic data and willingness to participate in trials were evaluated with Fisher’s exact test and corrected for false discovery rate with Benjamini-Hochberg procedure. Results: The survey was completed by 100 pts within 3 business days, achieving an 82.2% participation rate. Data collection was managed by only 2 medical students, highlighting the high feasibility of this questionnaire to capture patients’ insights into trial participation. Median age was within 65-74 yrs and were 56% women, 88% white. Motivational findings were consistent with those reported by SKCC. (See Table 1) When asked about concerns preventing participation, the most commonly cited and ranked concerns was “having several health issues” (n=45), followed by “transportation cost or difficulties” (n=40). 100% pts reporting an annual household income (ahi) higher than $150k (n=10) expressed willingness to participate in trials, compared to 56% of ahi less than $150k (n=73, statistically significant (p=0.017)). Previous participation in trial was also associated with willingness to participate (p=0.018). Conclusions: We showed that this 14-questions survey exhibits high feasibility for capturing patient-reported barriers and motivations for clinical trial participation. Results were reproducible between 2 neighboring cancer treatment centers. Expanded studies are required to capture larger patient populations and demonstrate the survey potential at rural/suburban tertiary centers to support the design of further targeted interventions.
| Response | PSCI | SKCC | |
| Most common reason for participation | “I might learn more about my own health or medical condition” | 68.0% | 67.4% |
| Least common reason for participation | “My family would want me to participate” | 20.0% | 12.4% |
| Most common reason not to participate | “I am afraid of the side effects” | 52.0% | 52.6% |
| Willing to participate | 58.0% | 50.8% |
Presentation numberPS4-05-04
Evaluation of a 113-gene predictor of benefit from neoadjuvant T-AC treatment in an I-SPY2 cohort
Jacob H Niklassen, Aida Oncology, Copenhagen, Denmark
J. H. Niklassen1, T. Berg2, B. Ejlertsen3, B. Hahn1, J. Nart1, P. Buhl Jensen1, U. Hald Buhl1, I. Kappel Buhl1; 1Aida Oncology, Copenhagen, Denmark, 2Department of Oncology, Centre for Cancer and Organ Disease, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark, 3Danish Breast Cancer Group, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
Background: Neoadjuvant chemotherapy is standard in early-stage breast cancer (BC), enabling tumor downstaging and treatment tailoring. Taxane-based regimens are widely used in both hormone receptor (HR)-positive/HER2-negative and triple-negative BC, yet pathological complete response (pCR) rates remain modest. In the I-SPY2 trial, high-risk early-stage BC patients were randomized to standard taxane-anthracycline (T-AC) vs investigational regimens. We demonstrate that within the shared HER2-negative control arm, a 113-gene expression-based machine learning model can stratify patients by likelihood of benefit from T-AC, supporting biomarker-driven strategies to improve pCR. The 113-gene model, which was developed using data from T-FEC treated patients analyzed on the Affymetrix HG U133A and HG U133 Plus 2.0 arrays, has previously been tested in five independent cohorts (GEO IDs: GSE20194, GSE20271, GSE23988, GSE32646, GSE230881) and shown to discriminate patients achieving pCR to taxane-based therapy from those who do not (published at ESMO 2025). Here, we present further validation of this 113-gene signature in data from the I-SPY2-990 mRNA Data Resource and demonstrate its applicability to Agilent microarrays in addition to Affymetrix. Methods: The previously developed model was adapted to the Agilent platform, where 108 of the 113 genes were available, and then applied to gene expression data from 179 high-risk, early-stage BC pre-treatment tumor samples, from patients who were treated with T-AC and evaluated for pCR at time of surgery. Patients were stratified based on HR status and scored on a scale from 0-100. The association of patient score (per 50-point increase) with pCR was assessed using logistic regression models. Results: For each 50-point increase in patient score, the odds of achieving pCR were significantly higher with an Odds-ratio (OR) of 3.37 (95% CI 1.3-10.3, p = 0.02) in HR-negative tumors and an OR of 5.70 (95% CI 1.8-22.6, p = 0.006) in HR-positive tumors. The association with score remained significant in a multivariable model adjusting for age, HR status, and MammaPrint risk group (OR 4.41, 95% CI 1.8-11.5, p = 0.0015). Conclusions: In this analysis of 179 high-risk, early-stage, HER2-negative patients from a phase II prospective study, with gene expression data from the Agilent 4x44k microarray, we evaluated a 113-gene machine learning-based predictor of response to taxane-based regimens. The model, originally built on data from Affymetrix microarrays and previously evaluated on five external cohorts, was able to successfully distinguish patients with pCR from those without pCR after neoadjuvant T-AC treatment. The association between model score and treatment outcome was significant in both univariate and multivariable settings. These findings provide further validation of the predictor and support its robustness across different microarray platforms.
| Dataset / subtype | N | Obtained pCR | Response Rate | OR | p value | ||||||
| GSE20194 | 230 | 48 | 20.9% | 5.07 | <0.001 | ||||||
| GSE20271 | 178 | 26 | 14.6% | 11.06 | <0.001 | ||||||
| GSE23988 | 61 | 20 | 32.8% | 8.48 | <0.001 | ||||||
| GSE32646 | 115 | 27 | 23.5% | 7.64 | <0.001 | ||||||
| GSE230881 | 263 | 64 | 24.3% | 4.08 | <0.001 | ||||||
| I-SPY2 (GSE194040) | 179 | 31 | 17.3% | 4.50 | <0.001 | ||||||
| HR-negative | 85 | 17 | 20% | 3.37 | 0.02 | ||||||
| HR-positive | 94 | 14 | 14.9% | 5.70 | 0.006 |
Presentation numberPS4-05-09
Molecular Profiling of Malignant Effusions in Advanced Breast Cancer via cfDNA and Tumor Cells Analysis: A Complementary Approach to Guide Targeted Therapy
Ana Velasco, Hospital Universitari Arnau de Vilanova, Lleida, Spain
E. Marín1, S. García-Roman1, S. Morales2, J. Porcel2, J. Soberino3, A. Martínez-Bueno4, C. Mayo de las Casas C1, B. García-Peláez1, R. Roman1, M. Molina-Vila1, C. Aguado1, A. Velasco2; 1Pangaea Oncology, Barcelona, SPAIN, 2Hospital Universitari Arnau de Vilanova, Lleida, SPAIN, 3IOB Institute of Oncology Hospital Quirónsalud Barcelona, Barcelona, SPAIN, 4Instituto Oncologico Rosell, Barcelona, SPAIN.
Background:Malignant effusions (MEs) are a frequent complication of advanced breast cancer (BC), reflecting tumor dissemination and associated with poor prognosis. Liquid biopsy (LB), particularly via next-generation sequencing (NGS) of circulating cell-free DNA (cfDNA), has emerged as a valuable tool for real-time monitoring and molecular characterization. While blood is the most common fluid used, MEs offer a rich source of tumor-derived DNA and cells. In addition to cfDNA profiling, tumor cells (TCs) can potentially be isolated and cultured from MEs, harnessing these fluids as a valuable source to develop patient-derived models for research and drug testing. We hypothesized that MEs are a viable source for both cfDNA mutation analysis and TC culture, and that identified molecular alterations may correlate with drug sensitivity. Methods:MEs were prospectively collected from patients with advanced BC. cfDNA was extracted from fluid samples and subjected to targeted NGS for somatic mutation profiling. Cytology was used to confirm malignancy. Parallel attempts were made to isolate and culture TCs from effusion samples. In three successfully established 3D cultures, cell viability assays were performed with targeted therapies based on detected mutations. Clinical follow-up data were collected to evaluate prognostic associations. Results:A total of 28 MEs were collected, including 21 pleural and 7 ascitic effusions. cfDNA was successfully extracted from all MEs, enabling comprehensive molecular testing with an 11-gene NGS panel. Molecular alterations were detected in 54% of patients. The mutational landscape revealed PIK3CA as the most mutated gene (25% of patients), followed by TP53 (21%). ESR1 D538G was detected in two patients following palbociclib/letrozole and capecitabine treatments, respectively. Additionally, alterations in ERBB2, KRAS, FGFR1/2, PTEN, ARID1A and a germline BRCA1 mutation were also identified. Paired cytological analysis was available in 15 samples, showing a concordance rate of 80% with molecular testing results. A PIK3CA mutation was detected in a cytology-negative case, highlighting the usefulness of effusions’ analysis in identifying and characterizing tumoral cells. In two cases with positive cytology, no pathogenic alterations were detected by NGS. However, TCs could be isolated from the MEs, further confirming their malignancy. Drug sensitivity assays were performed in three patient-derived 3D TCs culture. In a sample harboring a PTEN deletion, sensitivity to everolimus was demonstrated, with a 60% reduction of cell viability at <0.001 µM dose. Additionally, in a sample with FGFR1 amplification, paclitaxel showed a 70% reduction of cell viability at <0.1µM. Finally, in a ERBB2 and KRAS G13D mutated case, TCs showed resistance to alpelisib, palbociclib, ribociclib, elacestrant and letrozole. Conclusion:Malignant effusions represent a valuable and overlooked source of tumor material for molecular diagnostics in advanced breast cancer. Although cfDNA NGS and cytology show high correlation, NGS can detect tumoral content in samples that tested negative for cytology, and TCs’ isolation is able to confirm malignancy even in cases with no molecular alterations detected. The feasibility of culturing TCs from MEs and testing drug sensitivity further underscores the potential of this approach for personalized treatment. Integrating cfDNA genotyping, cytology, and TC functional assays from MEs could enhance clinical decision-making in late-stage breast cancer.
Presentation numberPS4-12-23
Regional variability and healthcare disparities in the management of early-onset breast cancer in Peru
Iris Otoya, Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
I. Otoya1, C. Calle1, N. Valdiviezo2, Z. Morante1, J. Moreno3, S. Lozano4, M. Valdivia5, G. Valencia6, L. Sanchez7, T. Vidaurre1, V. Acuña8, S. Neciosup1; 1Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 2Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas,, Lima, Peru, 3Oncología Médica, Instituto Regional de Enfermedades Neoplásicas, Trujillo, Peru, 4Oncología Médica, Hospital Regional Lambayeque, Lambayeque, Peru, 5Oncología Médica, Hospital Goyeneche, Arequipa, Peru, 6Oncología Médica, Hospital Antonio Lorena, Cusco, Peru, 7Oncología Médica, Hospital de Apoyo II Santa Rosa, Piura, Peru, 8Universidad Nacional Mayor de San Marcos, Lima, Peru
Background: Breast cancer is the most common malignancy among women in Peru, with around 7,800 new cases and nearly 2,000 deaths each year. The National Cancer Institute (INEN) in Lima receives approximately 1,200 new cases annually, reflecting the high burden concentrated in the capital. Breast cancer in young women (≤45 years) represents a particular clinical challenge due to its association with more aggressive behavior and poorer prognosis. Although international evidence suggests that clinical presentation may vary by population and geography, data on regional differences within Peru remain scarce. Objective: To analyze regional differences in the distribution in young women breast cancer subtypes in Peru, including the incidence of Luminal A, Luminal B, HER2+, and triple-negative tumors, along with associated clinicopathological features and treatment response. Methods: This observational, cross-sectional, multicenter study included women aged ≤45 years with stage II-III breast cancer diagnosed between 2021 and 2023, all of whom received neoadjuvant therapy, across three Peruvian macro-regions: Lima, the Northern Coast, and the Southern Highlands. The sampling design was based on the regional epidemiological burden and operational capacity of participating centers, allocating 92 patients to Lima (≈60%), 34 to the Northern Coast (22.7%), and 23 to the Southern Highlands (15.3%). The sample size ensured 80% statistical power (α=0.05) to detect ≥20% differences in the most frequent subtypes, with Fisher’s exact test applied to comparisons with small frequencies to ensure robust interregional analyses. Results: A total of 149 young women with breast cancer were analyzed across three Peruvian macro-regions: Lima (93; 62.4%), Northern Coast (33; 22.1%), and Southern Highlands (23; 15.4%). Median age at diagnosis was 39, 41, and 38 years respectively, showing a homogeneous pattern with slight regional variation. Stages II-III predominated, with a higher proportion of advanced cases in the Southern Highlands (~65%). Invasive ductal carcinoma was the main histology (>85%), while high-grade tumors were more frequent in the Southern Highlands (~47%). Molecular subtype distribution varied: Luminal B was predominant in Lima (~42%), HER2+ in the Northern Coast (~36%), and triple-negative in the Southern Highlands (~26%). Pathological complete response (RCB 0) was highest in Lima (~28%), intermediate in the Northern Coast (~21%), and lowest in the Southern Highlands (~13%). Breast-conserving surgery was more common in Lima (~35%), while mastectomy predominated in the Northern Coast and Southern Highlands (>70%). Radiotherapy coverage was uniform across regions (>85%). Conclusions: These findings reveal regional differences in young women breast cancer across Peru, likely influenced by inequities in access to specialized care and treatment standardization. This highlights the need for strategies to improve early diagnosis and harmonize management to achieve better outcomes. Larger studies with more representative samples and reduced bias are required to better reflect the national reality
Presentation numberPS4-06-28
Initial clinical and pharmacology results from START-002: 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 mTNBC or HR+/HER2- mBC
Steven J Isakoff, Mass General Cancer Center, Boston, MA
S. J. Isakoff1, A. Martynova2, P. L. Bedard3, A. Bardia4, M. E. Gatti-Mays5, N. LeVasseur6, A. Varkaris1, A. Bayliffe7, W. Randolph7, K. Srinivasan7, S. McCue7, K. Liu7, Z. Su7, K. Chin7, V. Kaklamani8, K. McCann4, E. Hamilton9; 1Mass General Cancer Center, Boston, MA, 2USC Norris Comprehensive Cancer Center, Los Angeles, CA, 3Princess Margaret Cancer Centre, Toronto, ON, Canada, 4UCLA Health Jonsson Cancer Center, Los Angeles, CA, 5The Ohio State University Comprehensive Cancer Center, Columbus, OH, 6BC Cancer Vancouver Centre, Vancouver, BC, Canada, 7Marengo Therapeutics, Cambridge, MA, 8UT Health San Antonio, San Antonio, TX, 9Sarah Cannon Research Institute, Nashville, TN
Background:Invikafusp alfa (“invika”) 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 TILs. Clinically, invika monotherapy generates a potent and selective expansion of mainly CD8+ Vβ6/ Vβ10 T cells. 0.08mg/kg was selected as RP2D and has demonstrated clinically meaningful single-agent anti-tumor activity in anti-PD(L)-1 resistant tumors, including objective responses in patients with TMB-H TNBC, CRC, NSCLC, and other tumor types resistant to anti-PD(L)-1. Based on these initial results, US FDA granted Fast Track Designation for invika in TMB-H CRC. Sacituzumab govitecan (SG), an antibody drug conjugate (ADC), is FDA approved for patients with metastatic TNBC and HR+/HER2- mBC. Preclinical studies indicate ADCs enhance tumor immunogenicity by promoting immunogenic cell death, increased antigen presentation, and tumor immune infiltration. Combination of ADCs with immunotherapy has the synergistic potential to enhance tumor eradication, help overcome resistance mechanisms, and improve overall treatment outcomes in breast cancer patients. Methods: START-002 is a Ph 1b/2 study to evaluate invika in combination with SG in patients with mTNBC or HR+/HER2- MBC. In Ph1b, patients were enrolled to 2 safety run-in cohorts at 0.04 mg/kg or 0.08mg/kg of invika with standard dose of SG (10mg/kg). Ph2 includes expansion cohorts for patients with mTNBC or HR+/HER2- mBC. Primary objective is to characterize the safety of invika + SG and to evaluate preliminary anti-tumor activity of the combination. Results: As of 15 Sep 2025, 11 patients with mTNBC or HR+/HER2- mBC were enrolled in 2 safety run-in cohorts. No DLTs were observed. Most common AEs (occurring in ≥2 patients) included neutrophil count decreased, diarrhea, alopecia, and stomatitis considered related to SG; cytokine release syndrome related to invika; fatigue and platelet count decreased considered related to either or both drugs. No ICANS or irAEs were reported. Treatment-related AEs were predominantly low grade 1 or 2, with exception of neutrophil count decreases, which were mainly grade 3. In the 0.04 mg/kg safety run-in cohort, 5 patients had at least 1 tumor assessment. Four experienced disease control (1 confirmed partial response (cPR) +3 SD) and 1 disease progression. The cPR was observed in a patient with HR+/HER2- mBC with liver and bone metastases, who progressed after 4 lines of endocrine and chemo-based regimens. The 3 patients with SD all experienced some degree of tumor reduction at the first scan but did not meet criteria for PR. Enrollment to the 0.08 mg/kg safety cohort has been completed with the first 3 patients reporting SD after completing at least one tumor assessment. Nanostring gene expression analyses in Ph1b demonstrated unequivocal expansion of Vβ6/10 T cells after the 1st dose of invika in mBC patients treated with this combination, similar to the selective expansion of Vβ6/10 T cells observed with invika monotherapy. Conclusions: Invika in combination with SG is feasible and safe at the doses tested. Combination safety profile is consistent with the known safety profile of each individual agent. Consistent with its mechanism of action, invika demonstrated unequivocal expansion of Vβ6/10 T cells when given with SG in mBC patients. Initial clinical anti-tumor activity including a cPR has been observed in patients with previously treated mBC. A RP2D has been selected and enrollment to Ph2 expansion cohorts is ongoing to further investigate the clinical activity of this promising novel combination therapy.
Presentation numberPS4-06-22
Niche-specific clustering of immune-suppressive cell populations and signals in breast cancer bone metastases
Déja Grant, Meharry Medical College, Nashville, TN
D. Grant1, G. Joseph2, M. Searcy3, R. Johnson3; 1Meharry Medical College, Nashville, TN, 2Vanderbilt University, Nashville, TN, 3Vanderbilt University Medical Center, Nashville, TN
Breast cancer cells frequently metastasize to bone and home to the osteogenic niche (bone surface), where they are thought to lay dormant until reactivated. Our knowledge of how tumor cells are impacted by the bone microenvironment is limited, as the spatial integrity of bone is often compromised. We hypothesized that myeloid populations infiltrate the osteogenic niche and sustain tumor burden by creating an immunosuppressive environment. We performed Digital Spatial Profiling (DSP) of bone metastases from 6-week-old female C57Bl/6 mice challenged with E0771 mouse mammary carcinoma cells by intracardiac injection (n=3/group) and evaluated tumor and immune cells residing in the marrow or endosteum. Mice were treated with α-PD-1 every 3-4 days to activate T cells and create a pro-inflammatory microenvironment. Upon sacrifice, formalin-fixed bone tissue sections were decalcified, paraffin-embedded, sectioned, and stained with syto13, pan-cytokeratin (PanCK), and CD45 as morphological markers to identify nuclei, tumor, and immune cells respectively. Antibodies for 70+ proteins (e.g., immune activation, cell death, proliferation, and cell stress), were then hybridized to the bone tissue slides, and regions of interest (ROIs) were selected along the osteogenic niche (within 100µm of the bone surface) and the marrow region (>100µm from the bone surface). Each ROI (n=5-6/bone) was segmented into two areas of interest (AOIs), PanCK or CD45 positive staining, and antibody reads were collected in each segment using Nanostring GeoMx/nCounter instruments and normalized to GAPDH and Histone H3 housekeeping proteins. Within the CD45+ segments (n=9 AOIs), we observed increased CD163 (p<0.0043) and FOXP3 (p<0.0385) expression in the osteogenic niche compared to the marrow niche, suggesting immune-suppressive M2-like macrophages and T regulatory cells (Tregs) may cluster in the osteogenic niche. Moreover, immune checkpoint proteins PD-1 (p<0.0202) and PD-L1 (p<0.0253), which are also immune-suppressive, were enriched in the osteogenic niche compared to the marrow in the CD45+ segments. In PanCK+ segments, we observed elevated p38 (p<0.0012), MEK1 (p<0.0280), pan-RAS (p<0.0411), and phospho-ERK1/2 (p<0.0177) proteins in the marrow niche compared to the osteogenic niche, suggesting that oncogenic pathways are enriched in breast cancer cells in the marrow. These data suggest that while the osteogenic niche is immune suppressive and may therefore provide an environment conducive to breast cancer immune evasion, breast cancer cells may preferentially proliferate within the marrow niche, away from the osteogenic niche/bone surface.
Presentation numberPS4-12-24
Risk and Pattern of Relapse in Triple-Negative Breast Cancer with Pathological Complete Response after Neoadjuvant Treatment: Updated Results from the European GAMBIT Real-world Study
Maria Vittoria Dieci, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova; Oncology 2, Veneto Institute of Oncology IOV IRCCS, Padova, Italy
M. Dieci1, T. Foukakis2, S. Giacchetti3, C. Desmedt4, J. Frenel5, E. Gasparini6, M. Kok7, P. Vigneri8, W. Jacot9, C. Vernieri10, M. Lambertini11, F. Patanè12, F. Piacentini13, E. Bria14, A. Zambelli15, D. Trapani16, A. Botticelli17, O. Garrone18, D. Massa1, S. Lando19, A. Matikas20, L. Someil3, G. Floris21, M. Campone22, M. Ragazzi23, E. Lips24, F. Martorana25, G. Bonomi26, A. Papakostantinou27, L. Nicolé28, I. Zerdes29, P. Neven30, M. Rotolo31, H. Horlings32, S. Nucera8, A. Gudin de Vallerin33, A. Vingiani34, A. Dei Tos35, V. Guarneri1; 1Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova; Oncology 2, Veneto Institute of Oncology IOV IRCCS, Padova, ITALY, 2Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden ; Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Stockholm, Sweden, Stockholm, SWEDEN, 3APHP, Senology Unit, Saint Louis Hospital, Université Paris Cité Paris, Paris, FRANCE, 4Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, BELGIUM, 5Department of Medical Oncology, Institut de Cancerologie de l’Ouest, Saint Herblain, France ; Nantes Université, INSERM, Centre de Recherche en Cancérologie et Immunologie Nantes Angers, Nantes, FRANCE, 6Pathology Unit, Azienda USL – IRCCS di Reggio Emilia, Reggio Emilia, ITALY, 7Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, NETHERLANDS, 8Department of Clinical and Experimental Medicine, University of Catania; Medical Oncology Unit, Istituto Clinico Humanitas, Misterbianco, Catania, ITALY, 9Department of Medical Oncology, Institut du Cancer de Montpellier (ICM), Montpellier, FRANCE, 10Fondazione IRCCS Istituto Nazionale dei Tumori; IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, ITALY, 11Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy ; Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 12IRCCS Ospedale San Raffaele, Milano, ITALY, 13Division of Medical Oncology, Dept. of Medical and Surgical Sciences for Children and Adults, University Hospital of Modena and Reggio Emilia, Italy, Moden, ITALY, 14Department of Medical Oncology, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy ; Università Cattolica del Sacro Cuore, Rome, Italy ; Ospedale Isola Tiberina – Gemelli Isola,, Rome, ITALY, 15ASST Papa Giovanni XXIII, Bergamo, Italy ; School of Medicine and Surgery, Bicocca University, Milan, ITALY, 16Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan,, Milan, ITALY, 17Department of Experimental Medicine, Sapienza University of Rome,, Rome, ITALY, 18Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, ITALY, 19Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy; Unit of Biostatistics, Epidemiology and Public Health, University of Padova, Padova, ITALY, 20Department of Oncology/Pathology, Karolinska Institutet, Stockholm, Sweden ; Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Cente, Stockholm, SWEDEN, 21Department of Pathology, UZ Leuven, Leuven, BELGIUM, 22Institut de Cancérologie de l’Ouest Angers-Nantes, Saint-Herblain, FRANCE, 23Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Italy ; Department of Medical and Surgical Sciences for Children and Adults University of Modena and Reggio Emilia, Modena, ITALY, 24Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, NETHERLANDS, 25Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy ; Medical Oncology Unit, Istituto Clinico Humanitas, Misterbianco, Catania, Italy, Catania, ITALY, 26Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, ITALY, 27Department of Oncology/Pathology, Karolinska Institutet; Department of Breast Cancer, Endocrine Tumours, and Sarcoma, Karolinska University Hospital, Stockholm, SWEDEN, 28Pathology Unit, Angelo Hospital, Mestre, ITALY, 29Department of Oncology-Pathology, Karolinska Institutet; Breast Center, Theme Cancer, Karolinska University Hospital and Karolinska Comprehensive Cancer Center, Padova, ITALY, 30Department of Oncology, Gynecologic Oncology and Multidisciplinary Breast Center, University Hospitals Leuven/KU Leuven, Leuven, BELGIUM, 31Oncology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia; University of Modena and Reggio Emilia, Modena, Reggio Emilia, ITALY, 32Department of Pathology, The Netherlands Cancer Institute, Amsterdam, NETHERLANDS, 33Department of Pathology, Translationnal Research Unit, Institut du Cancer de Montpellier, Montpellier, FRANCE, 34Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, ITALY, 35Department of Integrated Diagnostics, Azienda Ospedale-Università Padova, Department of Medicine, University of Padua, Padova, ITALY.
Background: Pathological complete response (pCR) after neoadjuvant treatment (NAT) associates with improved outcomes in triple-negative breast cancer (TNBC), yet a subset of patients still experience relapse. We previously reported on risk and pattern of relapse in up to 733 patients with pCR after NAT (Massa D, SABCS 2024 and ESMO Breast 2025). Here, we present updated results on 1190 patients, with detailed analysis of site-specific relapse risk.Patients and Methods: 2458 patients with stage I-III TNBC (ER<10%, PgR <10%, HER2-negative) treated with NAT were included in the multicentric real-world GAMBIT study across 18 European centers. Of these, 1193 obtained a pCR (48.5%, pCR cohort) and 1265 had residual disease (RD cohort). TILs were assessed on treatment-naïve biopsies for 691 patients with pCR (pCR-TILs cohort). We collected the first site of distant relapse and analyzed site relapse patterns by competing risk analysis. Primary survival endpoint was distant relapse-free survival (DRFS).Results: In the pCR cohort, clinical nodal involvement (cN+) was independently associated with worse outcomes (DRFS HR 2.38 [95%CI 1.14-4.97]; overall survival [OS] HR 3.19 [95%CI 1.53-6.66]). In the pCR-TILs cohort, cN and TILs were independently prognostic in multivariable models. The co-occurrence of cN+ and TILs<30% (cN+/low-TILs) identified a subgroup of patients with distinctively high risk of relapse and death: 5-year DRFS 83.4% and OS 85.8% versus ≥95% and ≥97.3% in other strata (cN+/high-TILs; cN0/low-TILs; cN0/high-TILs). 5-year cumulative incidence of first distant relapse in pCR cohort, pCR groups defined by cN and TILs, and RD cohort are shown in the Table.While 5-year cumulative incidence of any CNS metastases as first site of relapse was lower in the overall pCR cohort vs the RD cohort (3.6% vs 7.0%, p=0.001), there was no difference in the incidence of CNS-only relapse without extracranial disease: 2.6% vs 2.1%; p=0.445. Moreover, within the pCR-TILs cohort, patients with cN+/low-TILs had 5-year cumulative incidence of CNS metastases that were comparable (and even worse for CNS-only) to patients with RD. Conclusions: Patients with TNBC and pCR after NAT are heterogeneous with regards to outcome, and the co-occurrence of low TILs and cN+ identify a subgroup of patients at clinically meaningful risk of relapse, and enriched in CNS metastases. These results challenge the assumption that pCR confers uniformly excellent prognosis and support risk-adapted allocation in post-pCR clinical trials, including escalation strategies and studies of CNS-active agents.
| Study cohorts | CNS any, % | CNS-only (no extracranial disease), % | Visceral, % | Distant any, % |
| pCR cohort (n=1190) | 3.6% | 2.6% | 5.6% | 7.2% |
| pCR-TILs cohort: cN0/high-TILs (n=127) | 0.0% | 0.0% | 1.3% | 1.3% |
| pCR-TILs cohort: cN0/low-TILs (n=255) | 2.8% | 0.9% | 4.4% | 5.0% |
| pCR-TILs cohort: cN+/high-TILs (n=105) | 0.0% | 0.0% | 2.3% | 4.6% |
| pCR-TILs cohort: cN+/low-TILs (n=202) | 7.5% | 6.9% | 10.3% | 13.3% |
| RD cohort (n=1259) | 7.0% | 2.1% | 20.3% | 29.9% |
Presentation numberPS4-12-26
Neosaci-io: neoadjuvant sacituzumab govitecan (sg) + pembrolizumab (pb) in patients (pts) with early-stage tnbc experiencing suboptimal response to keynote-522
Clinton Yam, The University of Texas MD Anderson Cancer Center, Houston, TX
C. Yam, T. Iwase, B. Nelson, R. L. Bassett Jr, A. Singareeka, G. Whitman, M. Karuturi, B. Adrada, D. Kizub, M. Guirguis, A. Buzdar, T. Moseley, A. Nwosu-Iheme, M. Kapoor, J. Sukumar, P. Thomas, C. H. Barcenas, N. Ibrahim, G. Moscol, A. Nasrazadani, D. Ramirez, M. Wright, J. Lee, W. A. Woodward, J. K. Litton, K. K. Hunt, S. Giordano, D. Tripathy, V. Valero, L. Huo, G. M. Rauch, N. T. Ueno; The University of Texas MD Anderson Cancer Center, Houston, TX
Background: Pts with residual disease (non-pCR) after the KEYNOTE-522 regimen face a 30-40% risk of recurrence within 3 years, underscoring the urgent need for more effective therapies. In preliminary analyses, we identified that ≤80% tumor reduction after 4 cycles of paclitaxel+carboplatin (PC) + pembrolizumab (Pb) predicted a pCR rate of only ~20%, defining a biologically aggressive subgroup at risk of early recurrence and reduced survival. SG has demonstrated superior clinical efficacy over conventional chemotherapy in metastatic TNBC and emerging data suggest that SG enhances the efficacy of anti-PD-(L)1 therapy. Given the paucity of available evidence regarding the effectiveness of SG with Pb in the neoadjuvant setting, particularly in pts with suboptimal response to PC+Pb, we conducted the first trial of SG+Pb as sequential neoadjuvant therapy (NAT) in pts with high-risk, early-stage TNBC (NCT05675579). Methods: Pts with stage II-III TNBC and suboptimal response to PC+Pb, defined as disease progression or a ≤80% reduction in tumor volume by breast imaging, were eligible. Pts received SG (10mg/kg IV, D1 & 8) + Pb (200mg IV, D1) every 21 days x 4 cycles as the second phase of NAT before undergoing surgery. Blood and tumor biospecimens were collected prior to initiating SG+Pb and at surgery for correlative studies. We employed the Bayesian Optimal Phase II design to detect an improvement in pCR rate from 20% to 45% in order to deem the regimen worthy of further study in a randomized, phase III trial. Success was defined as pCR in 9 out of 25 response-evaluable pts (alpha=4.6%, power=85.8%). Pts not evaluable for response were replaced to ensure N=25 response-evaluable pts. Results: 27 pts were enrolled from 6/22/2023-5/23/2025. All 27 pts were evaluable for safety, and 25 pts were evaluable for response (1 pt completed <2 cycles of study treatment due to an unrelated adverse event; 1 pt was found to have an ineligible histology at surgery). Among the 25 response-evaluable pts, the pCR rate was 48% (95% CI: 28-69%). Clinicopathological characteristics are described in Table 1. Treatment-related adverse events (TRAE, all grades) occurring in ≥20% of safety-evaluable pts included neutropenia (78%), fatigue (37%), leukopenia (30%), and anemia (26%). 16 pts (59%) experienced grade ≥3 neutropenia. Of the 27 safety-evaluable pts, 24 (89%) completed all 4 planned cycles of SG+Pb. The remaining 3 pts discontinued study treatment early due to TRAEs (n=1), AEs unrelated to study treatment (n=1), and disease progression (n=1). Conclusion: These data provide the first evidence of SG+Pb as an effective treatment option in pts with high-risk, early-stage TNBC experiencing poor response to PC+Pb (pCR=48% vs 20% in historical controls), with no new safety signals. Correlative proteomic (including TROP2 IHC), genomic, and immunological studies are ongoing.
| pCR (n=12) | Non-pCR (n=13) | p value | |
| Age at diagnosis – Median (range) | 49.8 (28.6-71.1) | 63.2 (38.8-77.1) | 0.19 |
| T stage – n (%) | |||
| T1 | 1 (8) | 0 | 0.65 |
| T2 | 9 (75) | 9 (69) | |
| T3 | 2 (17) | 4 (31) | |
| T4 | 0 | 0 | |
| Nodal involvement – n (%) | |||
| Positive | 5 (42) | 4 (31) | 0.69 |
| Negative | 7 (58) | 9 (69) | |
| Germline BRCA mutation status – n (%) | |||
| Mutant | 2 (17) | 1 (8) | 0.59 |
| Wild Type | 9 (75) | 11 (85) | |
| Not tested | 1 (8) | 1 (8) | |
| Histology – n (%) | |||
| Ductal | 9 (75) | 9 (69) | 1.00 |
| Metaplastic | 3 (25) | 3 (23) | |
| Lobular | 0 | 1 (8) | |
| Histologic Grade – n (%) | |||
| 2 | 3 (25) | 6 (46) | 0.41 |
| 3 | 9 (75) | 7 (54) | |
| Ki67 – n (%) | |||
| ≤35% | 2 (17) | 4 (31) | 0.65 |
| >35% | 9 (75) | 9 (69) | |
| Not Available | 1 (8) | 0 |
Presentation numberPS4-12-28
A novel approach for phenotyping triple negative breast cancer using an Artificial Intelligence digital pathology-based prognostic test to assess recurrence risk and response to therapy
Michael Donovan, Icahn School of Medicine at Mount Sinai; PreciseDx, Miami, FL
N. Stanzione1, G. Fernandez2, S. Vaisman3, A. Sainath Madduri3, R. Scott3, M. Prastawa3, X. Zhang3, M. Donovan4; 1David Geffen School of Medicine at UCLA, Los Angeles, CA, 2Icahn School of Medicine at Mount Sinai; PreciseDx, New York, NY, 3PreciseDx, New York, NY, 4Icahn School of Medicine at Mount Sinai; PreciseDx, Miami, FL
Background Triple negative breast cancer (TNBC) accounts for approximately 15-20% of incident breast cancers and until recently, was the only BC subtype without targeted therapies. Recent therapeutic advances, including the use of immunotherapy (IO), have highlighted the importance of the spatial tumor microenvironment which includes both the number and location of infiltrating cancer immune cells. We previously reported on a redistribution of recurrence risk in TNBC patients from Mount Sinai using the PreciseBreast (PDxBR) test that includes AI-derived morphologic features of tumor architecture, nuclear size, stroma, mitotic figures and lymphocyte content combined with age, tumor size, anatomic stage and lymph node status. We conducted this study to gain a better understanding of the prognostic performance of PreciseBreast (PDxBR) for TNBC in an independent patient cohort. MethodsPatients with H&E stained slides from surgical excisions of TNBC stored in the UCLA biorepository (2008-2017) were sent along with clinical outcomes data to perform the PDxBR test. H&E slides were digitized and imaged on a Philips Ultra-Fast whole slide scanning system. Images were deconstructed to morphologic analytes using an AI-enabled platform to quantify tumor cell and tissue architectural features. Eight quantitative morphologic features (AI-grade model) were combined with 4 clinical factors to determine risk of recurrence. The AI-grade model was also evaluated. We applied an optimized PDxBR test for TNBC with risk scores 0-100 using an adjusted threshold 50yrs, 86% non-black; 62% tumor size ≤2.5cm and 30% >2.5cm-5cm; 87% Stage 1/2, 68% pN0, 32% pN1-3+, 88% Grade 3; 82% received chemotherapy. There were 14 events (18%) including 6 D, 4 DM, 2 LR, 2 SP with a median follow-up of 7.9 yrs. PDxBR identified 30 (39%) as high risk, 47 (61%) as low risk compared to the AI-grade model with 64 (83%) high risk, 13 (17%) low risk. PDxBR, which includes the AI-grade model + clinical features, was the only model with a significant HR of 3.72 (95% CI, 1.26-10.97, p = 0.0174), with Se 0.57 (95% CI 0.32-0.79), Sp 0.65 (95% CI, 0.53-0.76), PPV 0.27 (95% CI 0.14-0.45) and NPV 0.87 (95% CI, 0.75-0.94). PDxBR classified 8/14 events as high risk including 2 DM, 2LR, 1SP, and 3D. 4 of 14 events (3 D, 1 DM) included tumors that were ≥ 3cm of which 2 had 1-2pN. 6/14 patients with events did not have chemotherapy and 5 of 6 were high risk by PDxBR. The most significant PDxBR morphologic features for event identification included mitotic figure ‘hot spot’ quantification and tumor architecture (i.e. degree of differentiation).ConclusionsPDxBR introduces a novel approach towards risk stratification and outcome prediction in a clinically high risk TNBC population. Standardized AI digital pathology-based platforms have the potential to accurately re-assess risk and response to chemotherapy and IO, especially in randomized clinical trials.
Presentation numberPS4-12-29
Backbone optimization with dose-dense anthracyclines and prophylactic G-CSF improves pCR without added febrile neutropenia in pembrolizumab-treated TNBC: real-world evidence from Latin America
Diego Bustos, FUNDACION ARTURO LOPEZ PEREZ, Santiago, Chile
D. Bustos, M. Callahan, M. Vera, I. Saffie; FUNDACION ARTURO LOPEZ PEREZ, Santiago, Chile
Background Pembrolizumab combined with neoadjuvant chemotherapy (NACT) is the standard of care for early triple-negative breast cancer (TNBC) based on KEYNOTE-522. Real-world data from Latin America are scarce, and the contribution of dose-dense anthracyclines (ACdd) to efficacy and safety remains underreported. Methods We conducted a retrospective, single-institution study including patients with stage II-III TNBC treated with pembrolizumab-NACT. The primary endpoint was pathologic complete response (pCR, ypT0/is ypN0). Secondary endpoints included Residual Cancer Burden (RCB), immune-related and hematologic adverse events, and febrile neutropenia (FN). Adverse events were graded per CTCAE v5.0. Results A total of 40 patients initiated pembrolizumab-NACT. ACdd was used in 32/40 (80%), with routine primary G-CSF prophylaxis.Among the 32 patients who underwent definitive surgery with evaluable pathology, pCR (RCB-0) was achieved in 26/32 (81.3%). Among non-pCR cases, 2 were RCB-1 (6.3%), 3 were RCB-2 (9.4%), and 1 was RCB-3 (3.1%).In the entire cohort (n=40), febrile neutropenia (FN) occurred in 5/40 (12.5%), while grade ≥3 neutropenia was observed in 25% (9 grade 3, 1 grade 4). The FN rate was comparable to KEYNOTE-522 (15.1%), despite the high prevalence of ACdd in this cohort. The most frequent immune-related adverse events were hypothyroidism (n=4) and adrenal insufficiency (n=4). Grade 3 toxicities included adrenal insufficiency (n=1), hepatitis (n=1), and rash (n=1). No grade 4-5 events were observed. Conclusions In this Latin American real-world cohort, pembrolizumab-NACT achieved a high pCR rate (81.3%) with an acceptable hematologic and immune-related safety profile. The widespread use of ACdd (80%) with prophylactic G-CSF may have contributed to enhanced efficacy without new safety concerns. These findings highlight the relevance of backbone optimization when integrating immunotherapy into TNBC treatment and provide valuable evidence from underrepresented regions. Disclosure of prior presentation: Portions of these data were previously presented at SCOM (Chile); this submission includes updated cohort size, ACdd utilization, and expanded efficacy and safety analyses.
Presentation numberPS4-12-30
Overall survival and prognostic factors in patients with triple-negative breast cancer treated with adjuvant capecitabine: real-world experience in a peruvian cohort
Miguel Chirito, Instituto Nacional de Enfermedades Neoplasicas, Lima, Peru
M. Chirito, Guillermo Valencia1, Zaida Morante1, Patricia Rioja1, Sandra Adrianzén2, Karen Cruz2, Antonio Nuñez1, Claudia Castillo1, Jorge Sánchez1, Astrid Cuyutupa1, Daniel Garcia1, Andrea Recines1, Raúl Mantilla3, Karina Aliaga1, Connie Rabanal1, Jorge Dunstan2, Silvia Neciosup1, Tatiana Vidaurre1; Instituto Nacional de Enfermedades Neoplasicas, Lima, PERU.
OVERALL SURVIVAL AND PROGNOSTIC FACTORS IN PATIENTS WITH TRIPLE-NEGATIVE BREAST CANCER TREATED WITH ADJUVANT CAPECITABINE: REAL-WORLD EXPERIENCE IN A PERUVIAN COHORT Miguel Chirito1, Guillermo Valencia1, Zaida Morante1, Patricia Rioja1, Sandra Adrianzén2, Karen Cruz2, Antonio Nuñez1, Claudia Castillo1, Jorge Sánchez1, Astrid Cuyutupa1, Daniel Garcia1, Andrea Recines1, Raúl Mantilla3, Karina Aliaga1, Connie Rabanal1, Jorge Dunstan2, Silvia Neciosup1, Tatiana Vidaurre1. 1. Department of Medical Oncology. National Institute of Neoplastic Diseases (INEN). Lima, Peru2. Department of Breast and Soft Tissue Surgery. National Institute of Neoplastic Diseases (INEN). Lima, Peru3. Research Department. National Institute of Neoplastic Diseases (INEN). Lima, Peru Corresponding author: Miguel Chirito Carbajal miguelchirito10@gmail.com INTRODUCTION Triple-negative breast cancer is a subtype with a poor prognosis, characterized by high rates of relapse and early mortality. Adjuvant therapy with capecitabine in patients with residual disease after neoadjuvant therapy has shown a benefit in overall survival (OS), as reported in the CREATE-X study with a 5-year OS of 79%; however, there is limited evidence in Latin America (LATAM) from real-world clinical practice. OBJECTIVE To analyze the results in patients with non-metastatic triple-negative breast cancer treated with adjuvant capecitabine at a Peruvian cancer center, and to identify prognostic factors associated with overall survival. METHODS A retrospective cohort study was conducted of patients diagnosed with non-metastatic triple-negative breast cancer treated between 2020 and 2023 at a Peruvian cancer center. Clinical, histopathological, and therapeutic variables were collected. OS and its association with clinical and pathological characteristics were estimated using Kaplan-Meier curves. RESULTS A total of 199 patients were evaluated, 67% of whom received adjuvant capecitabine. The median age was 47 years (range 42-56), with most (98.5%) having an ECOG performance status of 0-1. Regarding anatomical variables, the most frequent were T3-T4 (65%) and N1 (45%); the most common clinical stage was IIIB (31%). Among the pathological characteristics, the most frequent were ductal histology (73%), histological grade 3 (68%), with an average ki67 of 60%. The median time between the end of neoadjuvant chemotherapy and surgery was 51 days (30.5-79.5). During neoadjuvant therapy, 25% of patients used carboplatin. Regarding surgery, most patients underwent mastectomy (81%), with operative findings of ypT2 (37%), ypN0 (43.3%), and pathological stage III. During adjuvant therapy, 96% of patients received radiotherapy. With a median follow-up of 31 months (9-103), OS rates at 12, 36, and 60 months were estimated at 99%, 87%, and 58%, respectively, with no median OS reached. Conservative surgery (vs. mastectomy, NR vs. 15 months, p=0.043), absence of lymphovascular invasion (vs. presence, p=0.022), and involvement of ypT1-ypT2 lymph nodes (vs. ypT3, 80% vs. 48%, p=0.002) were associated with higher OS. Regarding recurrence patterns, recurrence was observed in 39% of patients, with distant recurrence being the most frequent in 74%. The most frequent site of metastasis was the lung. CONCLUSION In this Peruvian cohort of patients with triple-negative breast cancer, adjuvant capecitabine demonstrated an increase in OS at 5 years, showing aggressive disease behavior and a high tumor burden, predominantly locally advanced. Conservative surgery, absence of lymphovascular invasion, and low lymph node involvement were associated with higher OS. These findings consolidate capecitabine as part of the standard treatment for post-neoadjuvant residual disease in a Latin American population.
Presentation numberPS4-05-17
Identifying hub genes among different breast cancer subtypes and racial diversity through integrated bioinformatics analysis (WGCNA)
Efrain Daniel Lee-Kay-Pen, Clinica Padre Luis Tezza, Lima, Peru
Presentation numberPS4-06-01
Targeting eIF4A unleashes type I IFN immunity in triple-negative breast cancer
Na Zhao, Baylor College of Medicine, Houston, TX
N. Zhao1, E. Kabotyanski1, J. Lei2, A. Saltzman3, M. Lewis2, J. Yang4, J. Rosen1; 1Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 2Breast Center, Baylor College of Medicine, Houston, TX, 3Proteomics Core, Baylor College of Medicine, Houston, TX, 4Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, CHINA.
Background: Triple-negative breast cancer (TNBC) arises from aberrant control of gene expression at multiple levels. Because mRNA abundance explains less than half of the variance in protein levels, it is essential to consider post-transcriptional mechanisms, particularly translational regulation. Our previous publication (PMID: 37874652) suggested that the translation initiation factor eIF4A is a therapeutic vulnerability in TNBC. Targeting eIF4A with the first-in-class inhibitor zotatifin remodels tumor translatome, suppresses TNBC growth, and synergizes with carboplatin in syngeneic mouse models. Methods and Results: In the current study, we further explored the mechanism of zotatifin efficacy in TNBC mouse models and observed that zotatifin treatment induced a marked increase in IFN-β secretion and robust upregulation of interferon-stimulated genes in vitro and in vivo. Immunofluorescence in tumor cells demonstrated cytosolic dsRNA accumulation following eIF4A inhibition, which activates an MDA5-dependent type I IFN pathway in a tumor cell intrinsic manner. Functional blockade of IFNAR1 partially abrogated the efficacy of the combination treatment in syngeneic mouse models, confirming that type I IFN signaling is required for the therapeutic effect. To determine whether these mechanistic insights translate to human TNBC, we next evaluated efficacy across patient-derived xenograft (PDX) models. Across a panel of 8 TNBC PDX models, the zotatifin-carboplatin combination achieved complete responses in 1 model (1/8), partial responses in 3 models (3/8), and stable disease in 2 models (2/8). Zotatifin-containing arms triggered type I IFN transcriptional programs in both tumor and stromal compartments. Tumors with higher basal IFN-related gene expression exhibited greater treatment sensitivity, suggesting type I IFN activity as a potential predictive biomarker of response. Conclusions: These data define the mechanistic basis of eIF4A-targeted therapy, linking translational inhibition to innate immune activation, and provide a biomarker framework to guide future clinical trials combining zotatifin with chemotherapy in TNBC.
Presentation numberPS4-10-05
Dsc3 suppresses the ccl21-pdc-treg axis to reprogram immunosuppressive microenvironment and potentiate immunotherapy in triple-negative breast cancer
Yusong Wang, the first hospital of china medical university, Shenyang, China
Y. Wang, Y. Xu; Breast surgery, the first hospital of china medical university, Shenyang, CHINA.
The emergence of immunotherapy has revolutionized cancer treatment, particularly with the exploration of PD-1/PD-L1 checkpoints. However, its efficacy in triple-negative breast cancer remains limited. In this study, RNA-Seq analysis was performed on baseline samples from patients in the phase II single-arm clinical trial, TREND. By integrating data from public immunotherapy cohorts, we aimed to identify potential biomarkers. Desmocollin 3 (DSC3) was identified due to its strong association with pathological complete response. In animal models, DSC3-overexpressing tumors showed increased sensitivity to anti-PD-1 therapy, which was dependent on host-specific immunity. Tumor-infiltrating lymphocytes and draining lymph nodes exhibited an increased proportion of T cells with enhanced cytotoxic functions, including IFN-gamma and TNF-alpha production, which are critical for anti-PD-1 therapy efficacy. To explore how DSC3 enhances T cell cytotoxicity, we investigated dendritic cell subsets and other myleoids cell subsets. We found reduced plasmacytoid dendritic cells (pDCs) in DSC3-overexpressing tumors. Since pDCs recruitment is mediated by CCL21.e.g., we further examined its secretion to understand why pDCs were reduced. DSC3 overexpression led to decreased CCL21 secretion, impairing pDC recruitment and mitigating their interaction with regulatory T cells, which alleviated the local immune-suppressive tumor microenvironment and promoted T cell infiltration and cytotoxic function. To investigate the mechanism of reduced CCL21 secretion, we explored the relationship between DSC3 and the transcription factor SOX2. Molecular experiments, including ChIP, suggested that DSC3 mRNA competes with transcription factor SOX2, inhibiting its transcriptional activity on CCL21, in nucleus. Overall, our study highlights DSC3 as a potential biomarker for patient stratification in anti-PD-1 therapy and identifies novel immune system targets aimed at improving immunotherapy efficacy.
Presentation numberPS4-10-15
Accelerating an Anthracycline-Free Future: A New Drug in Clinical Testing Offers Patients Hope for Safer, More Effective Breast Cancer Therapy Combinations
Christine W Handy, Hamzeh Mystique Films, Swampscott, MA
C. W. Handy1, J. R. Rodrigues2, K. A. Guedes3, I. B. Walters4, L. H. Bender5; 1Patient Advocacy, Hamzeh Mystique Films, Swampscott, MA, 2Drug Development, Intensity Therapeutics, Inc., Shelton, CT, 3Clinical Development, Intensity Therapeutics, Inc., Shelton, CT, 4Medical, Intensity Therapeutics, Inc., Shelton, CT, 5Executive, Intensity Therapeutics, Inc., Shelton, CT.
Title: Accelerating an Anthracycline-Free Future: A New Drug in Clinical Testing Offers Patients Hope for Safer, More Effective Breast Cancer Therapy Combinations Christine Handy1, Joshua Rodrigues2, Kim Guedes RN2, Ian B. Walters M.D.2, Lewis H. Bender2 1Hamza Mystique Films, Swampscott, MA 2Intensity Therapeutics, Inc., Shelton, CT Note: Christine Handy is a breast cancer survivor, ASCO-designated patient advocate, author, and executive producer of the award-winning film, Hello Beautiful. Background: Doxorubicin is used in several chemotherapy regimens for many breast cancer subtypes, including triple-negative breast cancer (TNBC) and HER2-positive breast cancer before surgery (neoadjuvant). Doxorubicin can also be used in early-stage breast cancer in adjuvant therapy, after surgery, or in metastatic breast cancer. Patients refer to this drug as the “red devil.” It earns this moniker not just for its harsh red color, but for its side effects: fatigue, nausea, hair loss, and, far worse, permanent heart damage. Some patients die from neoadjuvant treatment itself. TNBC is an aggressive breast cancer subtype that affects 420,000 women worldwide and 56,000 in the US. Patients agree to intense neoadjuvant immunochemotherapy (Keynote-522), hoping to achieve a pathologic complete response (pCR), which means no cancer left at the time of surgery. Achieving pCR is strongly linked to an increase in event-free survival (EFS) and can serve as an endpoint to expedite FDA drug approvals. The SCARLET phase III trial, led by the National Cancer Institute, is studying whether an anthracycline-free regimen (NeoPACT) can remove doxorubicin in early-stage TNBC. While this trial gives patients hope, the EFS results are not expected until 2033 or 2034. Many patients are unable to wait so long, especially in the adjuvant or metastatic settings. Current Research Objectives and Rationale: INT230–6 is a new drug consisting of two potent cytotoxic chemotherapy agents (cisplatin and vinblastine sulfate) formulated with a diffusion and cell penetration enhancer (SHAO) in a single vial. The product candidate is designed for intratumoral injection. INT230-6 kills cancer cells locally and stimulates the immune system to recognize and attack cancer systemically. The drug, after being injected into the tumor, attacks the entire tumor. In early studies, the drug has been shown to cause up to 95% tumor necrosis in tumors as large as 4.4 cm. The ongoing INVINCIBLE-4 trial (NCT06358573) is a two-cohort randomized controlled study testing INT230-6 dosed before the standard Keynote-522 regimen, which includes doxorubicin (cohort A), and the KN-522 regimen alone (cohort B). The goal of INVINCIBLE-4 is to enhance initial tumor destruction and immune priming, thereby improving pathological complete response (pCR) rates over KN-522. With higher pCR rates and strong immune activation, a regimen without anthracyclines may become possible. Phase 3 Study Design and Rationale: If the results from the Phase 2 INVINCIBLE-4 study show a clinically meaningful increase in pCR using INT230-6 before KN-522, the next step would be a pivotal phase III trial design with three treatment arms in neoadjuvant TNBC: 1. INT230-6 followed by full KEYNOTE-522 regimen 2. INT230-6 followed by KEYNOTE-522 without anthracycline. 3. Standard KEYNOTE-522 regimen alone (with anthracycline) With pCR as the first primary endpoint for accelerated approval and enrolling sufficient TNBC patients for the endpoint of EFS, the three-arm 1:1:1 randomized study design comparing cohorts 1 or 2 separately to cohort 3 could enable a significantly faster approval pathway (for cohort 2). pCR results could be available years before the SCARLET study data. However, EFS will be required for full approval of INT230-6 with an anthracycline-free regimen.