Poster Session 3
Session Details
Presentation numberPS3-01-01
Impact of medically assisted procreation and pregnancy on breast and gynecologic cancer risk in women under 45: a real-world, propensity-matched analysis using the trinetx network
Giulia Miserocchi, IRST IRCCS Dino Amadori, Meldola, Italy
G. Miserocchi1, A. Farolfi1, F. Merloni1, C. Gianni1, N. Gentili1, M. Mariotti1, G. Di Menna1, O. Serra1, C. Casadei1, F. Rusconi2, A. Andalò1, S. Sabbioni1, A. Carlini1, D. Montanari1, M. Sirico1, R. Maltoni1, L. Cecconetto1, S. Sarti1, M. Palleschi1, A. Musolino1; 1Medical Oncology, Breast & GYN Unit, IRST IRCCS Dino Amadori, Meldola, ITALY, 2TrineTX, TrineTX, Cambridge, MA, USA, MA.
Impact of Medically Assisted Procreation and Pregnancy on Breast and Gynecologic Cancer Risk in Women Under 45: A Real-World, Propensity-Matched Analysis Using the TriNetX NetworkBackgroundThe potential oncologic risks associated with medically assisted procreation (MAP) remain a concern, particularly in young women undergoing hormonal stimulation. Although previous studies have suggested no significant increase in breast cancer (BC) risk after MAP, data remain limited for ovarian (OC) and endometrial (EC) cancers. Moreover, the protective role of pregnancy against BC is well established but less explored for other gynecologic malignancies. This study aimed to evaluate the risk of BC, OC, and EC in women under 45 undergoing MAP compared with women who had a delivery, and to assess the impact of pregnancy compared with nulliparity.MethodsThis was a retrospective analysis using real-world data from the TriNetX Global Collaborative Network. Two propensity-matched comparisons were performed: (1) MAP vs. a cohort of parous women undergoing mammographic screening (n=878 per group); (2) parous vs. nulliparous women undergoing mammographic screening (n=1,322 per group). Matching was performed on age, race, body mass index (BMI), genetic predisposition to malignancies, and previous oral contraceptive use. Outcomes included the incidence of BC, OC, and EC. Risk ratios (RR), odds ratios (OR), hazard ratios (HR), and cumulative incidence curves were calculated. The observation period extended up to 20 years.ResultsIn the first analysis, no significant difference was found in BC risk (3.6% vs. 4.3%; RR=0.84; HR=1.005; p=0.985). OC cases were absent in both cohorts. EC incidence was significantly higher in the MAP group, although the number of cases was low (1.1% vs. none; p=0.002). In the second analysis, parous women had a significantly lower BC incidence (3.3% vs. 7.3%; RR=0.45; HR=0.42; p=0.010). OC occurred only in nulliparous women (0.8% vs. none; p=0.002). Conversely, EC was higher in parous women (0.8% vs. none; p=0.002).ConclusionsThis real-world study suggests that MAP in women under 45 does not increase the risk of BC or OC compared with parous women. However, a possible increased risk of EC, although based on few cases, warrants attention. Moreover, pregnancy appears to confer a significant protective effect against BC and OC compared with nulliparity. Pending further long-term studies, these data support the oncologic safety of MAP for BC and OC, while highlighting the importance of individualized risk counseling regarding EC. Keywords breast cancer, ovarian cancer, endometrial cancer, medically assisted procreation, fertility treatments, pregnancy, nulliparity, TriNetX, real-world data, cancer risk.
Presentation numberPS3-01-03
Interactions between polygenic variants and clinical factors as predictors of breast cancer risk in women of self-reported Black/African ancestry
Timothy Simmons, Myriad Genetics, Inc., Salt Lake City, UT
T. Simmons, E. Hughes, M. Kucera, A. Gutin; Biostatistics, Myriad Genetics, Inc., Salt Lake City, UT.
Background Polygenic risk scores (PRSs) combine information from single-nucleotide polymorphisms (SNPs) across the genome to explain a substantial portion of genetic breast cancer (BC) susceptibility. In previous studies, a multiple-ancestry PRS (MA-385) based on 56 ancestry-informative and 329 BC-associated SNPs was accurate for diverse populations and ranked among the most significant factors affecting the risk of BC development. However, medical guidelines discourage the routine clinical use of PRS, citing a need for studies to evaluate potential interactions between SNPs and environmental and hormonal risk factors in diverse populations. Here, we investigate interactions of MA-385 and individual BC-associated SNPs with clinical risk factors in the widely used Tyrer-Cuzick (TC) model in a large cohort of self-reported Black/African women. Methods We examined clinical and genetic records from self-reported Black/African women who were referred for hereditary cancer testing with a multigene panel and tested negative for pathogenic variants in BC-associated genes. Multivariable logistic regression models adjusted for age, personal/family cancer history, and genetic ancestry were used to test individual TC risk factors, MA-385, and 5 individual BC SNPs with the greatest impact on BC risk for women of Black/African ancestry. Effect modification of MA-385 and individual SNPs by TC risk factors was evaluated by including interaction terms in the models. Odds ratios (ORs) and 95% confidence intervals (CIs) are reported per standard deviation of MA-385. Significance tests were performed using two-sided p-values based on likelihood-ratio test statistics. Results We identified 39,817 women who met study eligibility criteria. 8,802 (22.1%) women had been diagnosed with invasive BC prior to hereditary cancer testing. 17,529 women, independent of previous MA-385 development studies, were included in analyses of MA-385. Individual SNPs and clinical TC risk factors were examined in women with complete risk factor information. All 39,817 women had complete information for age and BC family history; sample sizes for other clinical risk factors ranged from 1,795 for breast density to 29,486 for height. The MA-385 was a highly significant predictor of invasive BC after accounting for clinical factors (OR=1.38; 95% CI 1.32-1.45, p=4.2 x 10-45). We found no evidence of interaction of MA-385 with any individual TC factor (all Bonferroni-adjusted p>0.05). The strongest evidence of interaction was for hormone replacement therapy status (unadjusted p=0.04; Bonferroni-adjusted p=0.66). The 5 SNPs with the greatest impact on BC risk for women of Black/African ancestry were distinct from those previously identified as having the greatest impact on BC risk for women of European ancestry. We found no evidence of interaction between these individual SNPs and TC risk factors (all Bonferroni-adjusted p>0.05). The strongest evidence for interaction was between a SNP on chromosome 19 (rs2363956) and BMI category (unadjusted p=0.04; Bonferroni-adjusted p=1). Conclusions In a large cohort of Black/African women, MA-385 was a highly significant predictor of BC and substantially improved risk prediction over clinical risk factors. In contrast, interactions of MA-385 and individual BC SNPs with clinical risk factors were not statistically significant. These findings suggest that TC factors have a minimal, if any, impact on the strength of the association between the MA-385 PRS and invasive BC in women of Black/African ancestry.
Presentation numberPS3-01-04
Single-cell transcriptomic and phenotypic profiling reveals T cell dysfunction in cancer-free BRCA1 germline mutation carriers
Huai-Chin Chiang, George Washington University, Washington, DC
H. Chiang1, R. Araya1, L. Qi2, H. Arestake1, S. Martinez1, A. Sanchez1, I. Kalia3, Y. Hu2, A. Horvath1, R. Li1; 1Biochemistry and Molecular Medicine, George Washington University, Washington, DC, 2Anatomy & Cell Biology, George Washington University, Washington, DC, 3Medicine, George Washington University-MFA, Washington, DC.
Women harboring germline mutations in BRCA1 have an approximately 80% lifetime risk of developing breast cancer. Extensive research on the breast epithelium, the cell of origin for BRCA1-related tumors, has shed light on the molecular functions of BRCA1 in tumor suppression. However, all somatic cells in a germline BRCA1 mutation carrier contain the same mutant allele, yet it remains unclear whether BRCA1 heterozygosity in stromal compartments could also contributes to the increased risk of BRCA1-associated cancer. To determine the impact of BRCA1 heterozygosity on T cells at the transcriptional and functional levels, we first investigated a cohort of T cells isolated from healthy individuals who are BRCA1 mutation carriers (BRCA1mut/+) and non-carriers, using single cell RNA sequencing (scRNA-seq) and high-parametric spectral flow cytometry. Single-cell transcriptomic analysis revealed a significant enrichment of a gamma-delta (γδ) T cell subpopulation in BRCA1mut/+ samples. A similar trend, albeit not statistically significant, was observed for a SOX4high T cell subpopulation. Functionally, BRCA1mut/+ T cells exhibited a delayed response to CD3/CD28 costimulation and showed an enrichment of naïve cells in the CD4 compartment. In addition, the production of cytokine IL-17 is significantly reduced in BRCA1mut/+ T cells after 24 hours of activation. To determine a causal effect of T-cell BRCA1 heterozygosity on tumorigenesis, we next deleted one allele Brca1 in mouse T lymphocytes. Implanted mammary tumors grew more robustly in T cell-specific Brca1-/+ mice than their Brca+/+ counterparts, supporting the notion that Brca1 heterozygosity in mouse T lymphocytes compromises the antitumor immunity. Furthermore, mechanistic studies indicate that BRCA1 regulates T cell-related gene expression in conjunction with other transcriptional coregulators. In summary, our data from clinical samples and preclinical models suggest that the heterozygous BRCA1 mutation status leads to T cell-intrinsic functional alterations and compromised antitumor immunity. We propose that germline BRCA1 mutations exert a “double whammy” effect—contributing to elevated cancer risk through their presence in both the cell of origin and the surrounding stromal compartments.
Presentation numberPS3-01-05
Prevalence and Characterization of Germline CDH1 Mutations in a Large Real-World Breast Cancer Cohort
Banu Arun, The University of Texas MD Anderson Cancer Center, Houston, TX
B. Arun1, S. Rivero-Hinojosa2, R. Green2, V. N. Aushev2, A. Antonopolous2, A. Rodriguez2, M. C. Liu2, J. E. Garber3; 1Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Oncology, Natera, Inc., Austin, TX, 3Division for Cancer Genetics and Prevention, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.
Background: Germline mutations in CDH1 are well-established as risk factors for hereditary lobular breast cancer (BC). However, the frequency and phenotypic landscape of CDH1 germline mutations in unselected BC populations remain incompletely defined. Here, we investigated the prevalence of CDH1 pathogenic/likely pathogenic (P/LP) germline variants in a large real-world BC cohort. Methods: We analyzed germline whole-exome sequencing (WES) data from 53,607 BC patients in Natera’s Real-World Database. CDH1 variants were annotated using Ensembl Variant Effect Predictor (VEP). Variants classified as “LOW” impact (synonymous or non-functional splice region variants) were excluded. Germline P/LP classification was sourced from ClinVar. In the absence of ClinVar annotation, truncating variants were designated as P/LP. Ancestry inference was determined using the EthSeq algorithm with four groups: African (AFR), East Asian (EAS), European (EUR), and South Asian (SAS). Global variant frequencies were also compared using 1000 Genomes Project data. Results: Among the 53,607 BC patients analyzed, the majority had ductal histology (68.55%), followed by lobular (10.36%), mixed (2.78%), and not available (18.31%). The median age in the cohort was 57 [range: 20-102]. Prevalence of germline CDH1 variants meeting the inclusion criteria was 11.0% (5892/53607) globally, with 10.9%, 11.9%, 7.7%, and 8.8% prevalence in EUR, AFR, EAS, and SAS populations, respectively. By histology, 11.1% of lobular and 11.7% of ductal cases presented a CDH1 germline variant. We observed 367 unique CDH1 allele variants, with 19 classified as P/LP. These included three splice acceptor site variants, twelve variants within the extracellular protein domain, one in the transmembrane domain, and three within the cytoplasmic domain. P/LP CDH1 variants were found in 23 individuals, representing 0.39% of patients with CDH1 germline variants and 0.25% of all patients with lobular breast cancer (0.042% of all BC patients analyzed). Among the patients with P/LP variants, 14 had lobular, 7 ductal, and 2 unknown histologies, with a median age of 54 [range: 38-71]. The majority of pathogenic variants were observed in patients of European ancestry. The variant spectrum included known loss-of-function alleles, such as truncating and splice-site alterations, as well as selected missense changes with predicted deleterious impact, indicating a range of functional consequences relevant to tumor biology. Conclusions: In this large real-world BC cohort, CDH1 germline variants were observed in 11% of patients with variable prevalence by genetic ancestry. We identified 19 P/LP CDH1 variants in 23 BC patients. These germline variants remain strongly enriched in lobular BC histology, consistent with prior literature, with 0.25% of patients with lobular histology carrying variants classified as P/LP. The majority of P/LP variants localized to functionally important regions of the E-cadherin protein, including the extracellular and cytoplasmic domains, underscoring their likely disruption of cell-cell adhesion. These findings support broader consideration of CDH1 in germline testing panels for BC risk assessment. Overall, this study reinforces the clinical value of CDH1 germline analysis, particularly in lobular BC.
Presentation numberPS3-01-06
Evaluating Liquid Biopsy as a Surrogate for Germline Testing in Metastatic Breast Cancer: Concordance and Discordance Analysis in the STING Program
Alessandra Spata, “Gustave Roussy” Institute, Villejuif, France
A. Spata1, V. Goldbarg1, M. Brichard2, M. Ibanez-Alda3, I. Valle4, F. Giugliano5, T. Grinda1, A. Viansone1, C. Poisson1, C. Bousrih1, S. Akla1, C. Roussel-Simonin1, E. Rassy1, A. Sabau1, L. Antoun1, C. Nicotra6, M. NgoCamus6, B. Verret7, A. Bayle6, E. Rouleau8, B. Pistilli1, A. Italiano9, O. Caron1, J. M. Ribeiro1; 1Department of Medical Oncology, “Gustave Roussy” Institute, Villejuif, FRANCE, 2Department of Medical Oncology, Cliniques universitaires Saint-Luc, Bruxelles, BELGIUM, 3Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, SPAIN, 4Clinical Oncology Unit, Careggi University Hospital, Firenze, ITALY, 5Unit 981, Molecular Predictors and Novel Targets in Oncology, “Gustave Roussy” Institute, Villejuif, FRANCE, 6Drug Development Department (DITEP), “Gustave Roussy” Institute, Villejuif, FRANCE, 7INSERM Unit U981, “Gustave Roussy” Institute, Villejuif, FRANCE, 8Medical Biology and Pathology Department, “Gustave Roussy” Institute, Villejuif, FRANCE, 9Department of Early Phase Trial Unit, Institut Bergonié Comprehensive Cancer Centre, Bordeaux, FRANCE.
Background: In France, access to PARP-inhibitors for metastatic breast cancer (mBC) is restricted to BRCA1, BRCA2 and PALB2 germline variant carriers. Genomic tumor sequencing or circulating tumor DNA assays (ctDNA or liquid biopsy (LB)) can incidentally reveal potential germline alterations. Currently, LB assays are nor designed nor validated for germline variant detection or interpretation, and it is not known whether their performances are good enough to replace a regular germline test. Our aim is to quantify the concordance between germline and LB in BC, and to explore the potential of LB to serve as a proxy for germline testing.Methods: Among patients (pts) with mBC enrolled between February 2021 and March 2025 in the STING molecular screening program (NCT04932525), pts with a pathogenic/likely pathogenic variant (P/LPVs) in BRCA1, BRCA2, and PALB2 genes in ctDNA were retrieved through the study database. LB in STING was carried out by using the FoundationOne Liquid CDx assay covering the analysis of 324 genes. We checked all the selected pts medical records to look for germline test results. We first performed descriptive analyses of clinical and pathological variables of the selected pts. We then assessed the positive predictive value (PPV) and false discovery rate (FDR) of ctDNA. Variant allele frequency (VAF) values were explored to evaluate their potential as a surrogate marker of germline origin, with a threshold of 40% VAF prospectively examined based on prior biological rationale.Results: Among 1,325 pts enrolled in STING program, 119 (9.0%) LB showed P/LPVs in BRCA1 (n=33; 27.7%), BRCA2 (n=71; 59.7%), or PALB2 (n=15; 12.6%). Germline genetic testing was performed in 93 of these pts (79.8%: 27 BRCA1, 56 BRCA2, and 12 PALB2).Among the entire cohort of 93 pts with BRCA1, BRCA2, or PALB2 mutations detected by LB and who underwent germline testing, 67 pts had confirmed germline P/LPV, resulting in an overall PPV of 72 %. Conversely, 26 pts were germline negative. In this subcohort, median age at diagnosis was 43.5 years (SD ± 12.1) for germline P/LPV carriers versus 45.6 years (SD ± 13.1) for somatic pts.Among the 27 LB BRCA1 pts, 21 (77.78%) pts showed concordance, all with a VAF >40%. LB FDR for BRCA1 variants was 22.2%. Among the 6 discordant pts, 5 pts harboured mutations with VAF <40% (median 4.9%), consistent with somatic origin, while one case involved a rearrangement intron 4 with a VAF of 42%.Among 54 LB BRCA2 pts, 39 (57.4%) showed concordance. Of these, 37 (94.9%) had VAF >40%, while 2 pts exhibited VAF between 26-38%. LB FDR for BRCA2 variants was 27.7%. Among the 15 discordant pts, 14 exhibited low VAF consistent with somatic origin; for one pt with exon 2 loss alteration, VAF was unavailable.Among 12 LB PALB2, 7 (58.3%) pts showed concordance, all with VAF >40%. LB FDR for PALB2 variants was 41.7%. Among the 5 discordant pts, all presented mutations with VAF <40% (median 0.28%) on LB, suggesting somatic variants. Interestingly, in our population, the PALB2 mutation rate was higher than previously reported.Conclusion: Our findings highlight a substantial concordance (PPV 98.5%) between germline and LB results for BRCA1, BRCA2, and PALB2 P/LPVs in mBC pts enrolled in the STING program, when applying a 40% VAF threshold. Although the overall FDR appears elevated, 92% of discordant cases involved variants with VAFs below 40%, which would not typically raise suspicion for germline origin in clinical practice. These results suggest that 40% VAF threshold could be a practical and reliable surrogate marker for germline status provided it is confirmed in larger independent cohorts. Data on LB false negative rates will be presented at the conference, to measure the likelihood of a positive germline test when the LB shows no PV and thereby evaluate the safety of omitting germline testing in LB-negative cases.
Presentation numberPS3-01-07
Advancing Risk-Informed Breast Cancer Screening: Development of a Digital Toolkit for Primary Care and OB/GYN Integration
Amanda Woodworth, Henry Mayo Hospital Keck Medicine of USC, Santa Clarita, CA
M. Thomas1, A. Woodworth2, A. Hazra3, Y. Duron4, J. Miller5, A. Patel6, C. Allen7, L. Schlager8, L. Soltani9, L. Meier10; 1National Breast Cancer Roundtable, American Cancer Society, Cincinnati, OH, 2Division of Breast & Soft Tissue Surgery, Henry Mayo Hospital Keck Medicine of USC, Santa Clarita, CA, 3Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, 4n/a, The Latino Cancer Institute, San Jose, CA, 5National Breast and Cervical Cancer Early Detection Program, Centers for Disease Control and Prevention, Rockville, MD, 6The Breast Care Center at Liberty Hospital, The University of Missouri-Kansas City School of Medicine, Liberty, MO, 7n/a, The Cancer Prevention and Research Institute of Texas, Austin, TX, 8Public Policy, Facing Our Risk of Cancer Empowered, Tampa, FL, 9n/a, El Rio Health Congress Health Center, Tucson, AZ, 10National Breast Cancer Roundtable, American Cancer Society, Atlanta, GA.
Title:Advancing Risk-Informed Breast Cancer Screening: Development of a Digital Toolkit for Primary Care and OB/GYN IntegrationAbstract:Background:Despite strong evidence supporting risk-informed breast cancer screening, implementation in primary care and OB/GYN settings remains limited. Barriers include lack of awareness, clinical time constraints, and inconsistent workflows for identifying and managing at-risk individuals. To address these challenges, the American Cancer Society National Breast Cancer Roundtable (ACS NBCRT) developed a digital Breast Cancer Risk Assessment Toolkit to support frontline providers in initiating, conducting, and acting on breast cancer risk assessment during routine care.Methods:The toolkit was developed over a 12-month period through a collaborative process led by the ACS NBCRT Early Action Priority Team and a national Advisory Group of experts in primary care, breast oncology, health equity, genetics, and public health. The group identified key barriers to implementation and defined practical tools needed to support guideline-concordant care. The resulting toolkit includes:1.Provider communication scripts2.Clinical workflow diagrams3.A risk assessment checklist4.A health system readiness assessment5.A comparison matrix of risk models6.Case studies from academic and community-based settingsContent was reviewed by ACS subject matter experts and aligned with ACS screening guidelines, while acknowledging variation in NCCN and USPSTF recommendations. Tools were designed to support both standalone and team-based care models, with consideration for feasibility in underserved and FQHC settings.Results:Toolkit reviewers—including clinicians and implementation scientists—reported high usability and clinical relevance. The materials supported patient-centered communication, flexible integration into EHR workflows, and strategic decision-making about referrals for genetic counseling or supplemental screening. Case studies highlighted implementation successes from both academic centers and bilingual, community-engaged programs serving Latina women. The model comparison table further supports providers in choosing risk tools that align with their population and system capabilities.Conclusions:The ACS NBCRT Risk Assessment Digital Toolkit offers a novel, digital-first approach to operationalizing breast cancer risk assessment in primary care and women’s health. By providing a streamlined, equity-informed package of tools, the initiative aims to improve early identification of high-risk individuals and expand access to personalized screening and prevention. Future directions include expansion of the case study library to feature additional models from rural and safety-net settings, as well as pilot testing the toolkit through practice-based dissemination and implementation research with primary care and OB/GYN users to assess feasibility, acceptability, and impact on clinical workflows. This scalable resource addresses a long-standing implementation gap and supports broad uptake of risk-informed breast cancer care.
Presentation numberPS3-01-08
Impact of allostatic load on breast cancer risk prediction among racially / ethnically diverse women
J Alex B Gibbons, Columbia University, New York, NY
J. B. Gibbons, V. Ro, J. E. McGuinness, R. Carlos, K. D. Crew; Department of Medicine, Division of Hematology / Oncology, Columbia University, New York, NY.
BackgroundCurrent breast cancer risk assessment tools tend to underestimate risk of invasive breast cancer (IBC) among Black or Hispanic women and include clinical risk factors, such as family history of breast cancer, that are not readily available or under-reported in the electronic health record (EHR). Allostatic load (AL)—a measure of cumulative physiological stress which is comprised of standard laboratory results and vitals—has been associated with IBC incidence and mortality. Our objective was to determine whether components of AL derived from the EHR can improve IBC risk prediction. MethodsWe conducted a retrospective analysis of women undergoing screening mammography at Columbia University Irving Medical Center from 11/1/14 to 4/7/25. We extracted EHR data on the earliest index screening mammogram, age at index scan, race, ethnicity, body mass index (BMI), mammographic density (MD, 4-category BIRADS classification), breast biopsy results, and family history of breast cancer. Patients with a diagnosis of IBC or ductal carcinoma in situ prior to the index scan were excluded. The following laboratory data and vitals were collected for AL biometrics representing major regulatory systems: metabolic (albumin, alkaline phosphatase, glucose), renal (blood urea nitrogen, creatinine), immune (white blood cell count), and cardiovascular (pulse, systolic and diastolic blood pressure). Patients with at least five of these components available in the two years preceding the index scan were included, and multiple imputation by chained equations was used to impute missing AL data. For modeling, AL-derived data were used as individual values and as a composite score (0-9), with one point assigned for each biometric in the highest quartile except albumin which was the lowest quartile. The primary outcome measure was development of IBC, defined as histologically-confirmed IBC at least six months after the index scan. Multiple logistic regression models were constructed, and area under the receiver operating characteristic curve (AUC) values were compared using DeLong’s test to a baseline model constructed from established IBC risk factors readily extracted from the EHR. ResultsAmong 27,155 evaluable patients, mean age was 59 years (SD 12) and 40% were Hispanic, 29% non-Hispanic White, and 14% non-Hispanic Black. A total of 135 (0.5%) patients had a subsequent IBC diagnosis. The baseline IBC risk prediction model of age, race / ethnicity, BMI, and MD achieved an AUC of 0.642; adding AL composite score or including breast biopsy results or family history did not significantly improve risk prediction. However, adding all nine individual AL-derived variables improved the AUC to 0.663 (p=0.011). Among non-Hispanic White women, the baseline model with AUC of 0.643 improved with the addition of breast biopsy results and family history (AUC of 0.656, p<0.001) and the AL-derived variables (AUC of 0.691, p=0.037). Among non-Hispanic Black women, the baseline model had an AUC of 0.634, which improved with the addition of breast biopsy results and family history to an AUC of 0.649 (p<0.001). Among Hispanic women, including AL-derived variables significantly improved the baseline model with an AUC of 0.611 to an AUC of 0.714 (p=0.002). Including the composite AL score did not improve prediction for any racial / ethnic group. DiscussionWe demonstrated that addition of AL-derived laboratory data and vitals extracted from the EHR significantly improved IBC risk prediction, particularly among Hispanic women, achieving AUCs comparable to existing well-validated breast cancer risk assessment tools. With further validation, incorporation of AL variables may facilitate efficient IBC risk prediction in the clinical setting to inform breast cancer screening and risk-reducing strategies among diverse women.
Presentation numberPS3-01-09
Associations of genetically predicted metabolites with mammographic breast density and breast cancer risk in premenopausal women
Ghazaleh Pourali, Washington University School of Medicine, Saint Louis, MO
G. Pourali1, L. Lyu2, X. Guo2, A. Toriola1; 1Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, 2Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN.
Background: Genome-wide association studies (GWAS) have identified genetic loci associated with mammographic breast density (MBD) and breast cancer risk, but the mechanisms underlying these associations remain unclear. Metabolite quantitative trait loci (mQTL) analyses can map genetic loci associated with metabolites, potentially offering insight into mechanisms involved in risk, but there is limited data on how genetically predicted metabolites are associated with MBD and breast cancer risk in premenopausal women. To address this, we performed mQTL analyses in non-Hispanic White (NHW) and non-Hispanic Black (NHB) women and examined their overlap with established GWAS loci for MBD and breast cancer. Methods: Our analysis included 494 NHW and 163 NHB women who had screening mammograms at Washington University in St. Louis. Plasma metabolomic profiling (Metabolon®) identified 1,074 metabolites, and genotyping (Infinium microarray, Illumina) covered 2,028,571 loci. Metabolite data were batch-normalized, metabolites with >10% missingness were excluded, and remaining missing values were imputed. We conducted mQTL mapping using linear regression in PLINK 2.0, stratified by race and adjusted for age, body mass index, alcohol use, oral contraceptive use, and first five genotype principal components. Significant mQTLs (FDR-p<0.05) were compared to GWAS loci for breast cancer, two fine mapping studies, and GWAS of MBD. Results: In NHW women, 30 mQTLs were associated with 54 metabolites overlapping breast cancer GWAS loci. Key pathways included xenobiotics (anti-inflammatory/immunosuppressant drugs, chemicals, tobacco metabolites), lipid (fatty acid, corticosteroid, bile acid metabolism), and amino acid (tryptophan, methionine/cysteine, tyrosine metabolism). In NHB women, 2 mQTLs were associated with 3 metabolites overlapping breast cancer GWAS loci: rs16866849 (diltiazem, ß =0.2, FDR-p=1.6×10⁻⁶, cardiovascular drug), and rs9397068 with two metabolites (2-hydroxyibuprofen, ß=5.3, FDR-p=9.6×10⁻³, analgesics; and 2,2′-methylenebis(6-tert-butyl-p-cresol), ß=14.6, FDR-p=9.6×10⁻³, chemical). Additionally, in NHW women, 5 mQTLs were associated with 5 metabolites overlapping GWAS loci of MBD. For dense area, these included rs150249911 (levulinate (4-oxovalerate), ß=6.5, FDR-p=2.3×10-7, food component/plant), rs150249911 (N-acetyltyrosine, ß=5.0, FDR-p=3.3×10-4, tyrosine metabolism), rs2042239 (5-acetylamino-6-formylamino-3-methyluracil, ß=1.3, FDR-p=3.3×10-2, xanthine metabolism), and rs833472 (3-hydroxystachydrine, ß=11.0, FDR-p=2.5×10-3, food component/plant). For percent mammographic density, rs61941038 (3-hydroxystachydrine, ß=10.4, FDR-p=7.4×10-3, food component/plant) was associated. For non-dense area, rs78395856 (taurochenodeoxycholic acid 3-sulfate, ß=3.7, FDR-p=3.3×10-2, bile acid metabolism) was associated. Conclusions: Our novel study identified genetic loci associated with metabolites that overlap with established MBD and breast cancer risk loci with differences in NHW and NHB women. These findings suggest biological mechanisms of genetic risk and provide a basis to prioritize metabolites and pathways for targeted prevention.
Presentation numberPS3-01-10
Patterns of metastasis according to germline BRCA1/2 mutation status in patients with HER2-negative metastatic breast cancer
Michele Bottosso, University of Padova, Padova, Italy
M. Bottosso1, G. Griguolo1, L. Trovò1, S. Tognazzo2, G. Faggioni3, E. Mioranza3, G. Vernaci3, D. Massa1, V. Faso1, D. Napetti1, A. Menichetti3, S. Zovato2, M. Dieci1, V. Guarneri1; 1Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, ITALY, 2Hereditary Tumors Unit, Istituto Oncologico Veneto, Padova, ITALY, 3Medical Oncology 2, Istituto Oncologico Veneto, Padova, ITALY.
Background: Germline pathogenic variants (PVs) in BRCA1/2 are found in approximately 5% of patients with breast cancer (BC) and are associated with different clinical characteristics and subtype distributions. BC exhibits subtype-specific patterns of metastasis according to hormone receptor (HR) and HER2 status. Although limited, existing evidence suggests that germline BRCA1/2 PVs may be associated with a higher frequency of metastasis to specific sites, such as the central nervous system (CNS). However, these findings are often confounded by differences in subtype distribution among BRCA1/2 PV carriers. In this study, we investigated the association between germline BRCA1/2 mutation status and the patterns and frequency of metastasis across BC subtypes. Methods: Patients treated for HER2-metastatic BC at Istituto Oncologico Veneto between 2007 and 2025 and with known germline BRCA1/2 mutation status were retrospectively identified. Data on metastatic sites at initial diagnosis of metastatic BC and throughout the disease course (last follow-up) were collected. CNS involvement was defined as the presence of either brain metastases or leptomeningeal disease. Overall survival (OS) was defined as the time from the first diagnosis of metastatic BC to death from any cause or last follow-up. Results: A total of 184 patients were identified: 33 BRCA1 PV carriers, 28 BRCA2 PV carriers, and 123 noncarriers. Overall, 131 patients presented a HR+ BC and 53 a triple negative BC (TNBC). TNBC was more frequent among BRCA1 carriers (n=21, 63.3%), while HR+ BCs were more common among BRCA2 carriers (n=23, 82.1%) and noncarriers (n=96, 78.0 %; p<0.001). At initial diagnosis of metastatic BC, BRCA1/2 carriers with a HR+ BC presented a lower incidence of bone metastases compared to noncarriers (31.4% vs 51.0%, p=0.046). In contrast, no differences in metastatic site distribution at first metastatic BC diagnosis were observed among patients with TNBC. Median follow-up from stage IV disease diagnosis was 80.7 months (95%CI 53.5-108.0). At last follow-up, BRCA1/2 carriers with a HR+ BC presented a higher incidence of CNS involvement compared to noncarriers (31.4% vs 14.6%, p=0.030). Among BRCA1/2 carriers with HR+ BC, CNS involvement appeared relatively early, with 23.4% cumulative incidence at 24 months as compared to 5.2% in noncarriers (Gray’s p=0.041). At last follow-up, among patients with TNBC, BRCA1/2 carriers presented a lower incidence of liver metastases compared to noncarriers (34.6% vs 70.4%, p=0.009), while no significant difference in terms of CNS involvement was observed, with high rates in both subgroups (57.7% vs 37%, p=0.219). No significant difference in terms of OS was observed between BRCA1/2 carriers and noncarriers, both among patients with HR+ BC (39.2 vs 47.6 months, respectively; p=0.246) and among those with TNBC (25.8 vs 15.8 months, respectively; p=0.467). Conclusions: Germline BRCA1/2 PV are associated with distinct metastatic patterns among patients with HER2-negative BC. In the HR+ disease, carriers had relatively fewer bone metastases at diagnosis and presented a higher risk of CNS involvement over time. These findings may inform tailored radiological surveillance strategies.
Presentation numberPS3-01-11
Breast Cancer Risk Prediction for Racially Diverse Women With Benign Breast Disease
Alzina Koric, Washington University in Saint Louis, Saint Louis, MO
A. Koric, Y. Park, J. Jiang, F. Boul, G. Colditz; Surgery, Washington University in Saint Louis, Saint Louis, MO.
Purpose: Existing breast cancer risk prediction models are ineffective for women with benign breast disease (BBD), especially in racially and ethnically diverse populations. We aimed to identify risk predictors for accurately estimating breast cancer development in a contemporary cohort of women from diverse racial backgrounds diagnosed with BBD. Patients and Methods: We identified 9,202 women diagnosed with histologically-confirmed benign lesions at the Joanne Knight Breast Health Siteman Cancer Center in St. Louis from 2010-2023. Risk modeling was performed with Cox regression using 95% confidence intervals (95%CI) and internally validated through a bootstrap resampling method with 100 replacements (i.e., corrected for overfitting). Discriminatory model performance was estimated by Harrell’s C-index. Calibration was assessed by estimating observed and predicted ratio (O/E) at 5-year BC predicted risk. Results: Among the 9,202 women with BBD, 64.3% were White, 32.5% were Black, and 3.2% were Asian women, 3.3% had a subsequent breast cancer at least six months following BBD. The final model predictors included: age at BBD diagnosis, family history of breast cancer, atypical hyperplasia (AH), proliferative disease without atypia (PDWA), breast density, and race. The model discrimination in the original sample had a Harrell’s C-index of 0.68 (95%CI 0.65, 0.71) and in the bootstrap sample of 0.67 (95%CI 0.63, 0.70). At the 5-year, the overall model calibration was good (O/E=0.98, 95%CI 0.86, 1.12). The model calibration was satisfactory across most risk-factor-defined subgroups; however, it tended to underestimate risk in the low-risk group and overestimate risk in the high-risk group (as defined by 20% quantiles). Conclusion: The final model outperformed existing breast cancer risk models among BBD women and future research will externally validate the model evaluating the risk among a subset of women with atypia.
Presentation numberPS3-01-12
Wisdom and mypebs: personalized breast cancer screening trials operating in distinct international contexts
Katherine Leggat-Barr, Tufts University School of Medicine, Boston, MA
K. Leggat-Barr1, P. Giorgi Rossi2, M. Guindy3, F. Gilbert4, M. Roman5, J. Burrion6, H. De Koning7, S. de Montgolfier8, L. Giordano9, D. Keatley10, J. Deleuze11, E. Gauthier12, S. Michiels13, C. Vissac-Sabatier14, H. Anton-Culver15, S. Borowsky16, S. Brain17, J. Esserman18, E. Ziv19, A. Fiscalini20, D. Goodman-Gruen21, D. Heditsian17, R. Hiatt22, V. Lee17, D. Moorehead23, A. Naiem24, O. Olopade25, H. Park26, B. Parker27, A. Petruse28, M. Scheuner29, L. van ‘t Veer30, V. Arasu31, M. Eklund32, L. Madlensky33, Y. Shieh34, N. Wenger35, J. Tice36, C. Kaplan37, A. Kaster38, R. Lancaster39, A. LaCroix40, S. Delaloge41, L. Esserman20; 1Medical School, Tufts University School of Medicine, Boston, MA, 2Reggio Emilia, Azienda Unita Sanitaria Locale di Reggio Emilia, Reggio Emilia, ITALY, 3Medical School, HaBarzel, Tel Aviv, ISRAEL, 4Department of Radiology, University of Cambridge, Cambridge, UNITED KINGDOM, 5Hospital, Hospital Parc de Salud Del Mar, Barcelona, SPAIN, 6Medical School, Institut Jules Bordet, Bruxelles, BELGIUM, 7Department of Public Health, Erasmus, Rotterdam, NETHERLANDS, 8Aix Marseille Univ, INSERM, IRD, SESSTIM, Marseille, FRANCE, 9l’Epidemiologia e la Prevenzione Oncologica, Centro di Riferimento per l’Epidemiologia e la Prevenzione Oncologica, Torino, ITALY, 10Independent Cancer Patient Voice, Independent Cancer Patient Voice, London, UNITED KINGDOM, 11Génomique Humaine, Centre National de Recherche en Génomique Humaine, Évry-Courcouronnes, FRANCE, 12Predilife, Villejuif France, Villejuif Cedex, FRANCE, 13Department of Biostatitics, Gustave Roussy, Villejuif, FRANCE, 14Unicancer, Unicancer, Paris, FRANCE, 15Department of Medicine, Genetic Epidemiology Research Institute, University of California, Irvine, Irvine, CA, USA., Irvine, CA, 16Department of Pathology, School of Medicine, University of California, Davis, Sacramento, CA, 17Breast Science Advocacy Core, Breast Oncology Program, University of California, San Francisco, San Francisco, CA, 18OB-GYN Department, Diagnostic Center of Miami, Miami FL USA, Miami, FL, 19Department of General and Internal Medicine, University of California San Francisco, San Francisco, CA, 20Department of Surgery, University of California, San Francisco, San Francisco, CA, 21Department of Epidemiology and Biostatistics, University of California, Irvine, Irvine, CA, 22UCSF Helen Diller Family Comprehensive Cancer Center, Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, 23Womens Cancer Resource Center, Womens Cancer Resource Center, Berkeley, CA, 24Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 25Center for Clinical Cancer Genetics and Global Health, Department of Medicine, The University of Chicago, Chicago, IL, 26Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, 27Moores Cancer Center, University of California San Diego, San Diego, CA, 28Clinical and Translational Science Institute Office of Clinical Research, University of California Los Angeles, Los Angeles, CA, 29Department of Medicine, University of California, San Francisco and San Francisco VA Health Care System, San Francisco, CA, 30Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, 31Division of Research, Kaiser Permanente, Pleasanton, CA, 32Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SWEDEN, 33Department of Medicine, University of California, San Diego, San Diego, CA, 34Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, 35Division of General Internal Medicine and Health Services Research, University of California, Los Angeles, Los Angeles, CA, 36General Internal Medicine, School of Medicine, University of California San Francisco, San Francisco, CA, 37Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA., San Francisco, CA, 38Edit Sanford Breast Center, Sanford Health, Fargo, SD, 39Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 40Medical School, University of California San Diego, San Diego, CA, 41Department of Cancer Medicine, Gustave Roussy, Villejuif, FRANCE.
Background WISDOM (Women Informed to Screen Depending on Measures of risk) (NCT02620852) and MyPeBS (My Personal Breast Screening) (NCT03672331) are two prominent risk-based breast cancer screening (RBS) trials based in the US and Europe & Israel, respectively. Both trials aim to assess whether RBS is as safe as current screening (no increase incidence of stage> II/IIB breast cancers) and can aid to enhance resources on those at higher risk, while minimizing harm to those at lowest risk. WISDOM and MyPeBS were conducted independently but with a planned joint analysis. Guidelines in the US range from annual (ACR, NCCN) to biennial screening starting at age 40 (USPSTF). In contrast, screening in MyPeBS countries is population-based by invitation every 2 years in (France, Belgium, Italy, Spain, Israel) and every 3 years in the UK starting at age 50. We will compare the baseline characteristics of both cohorts. Methods Eligibility included ages 40-74 in WISDOM and 40-70 in MyPeBS and no prior breast cancer history. Both are randomized 1:1 to RBS vs. country-based standard of care screening, though in WISDOM, women could choose their arm if they declined randomization (pragmatic, preference tolerant approach). Methods of recruitment reflect systematic/screening invitation (EU) vs. opportunistic (US). In WISDOM, the 5-year risk of invasive BC is estimated using the Breast Cancer Screening Consortium (BCSC) score, which includes clinical data & breast density, combined with a Polygenic Risk Score (PRS) (up to 126 single nucleotide polymorphisms (SNPs)), & a panel to identify germline mutations in 9 breast cancer risk genes. Four categories were used to classify risk (lowest, average, elevated, & highest), with corresponding screening assignments: age-based (mammogram (Mg) starting at 50), two-year (average), one-year moderate risk (MR), or Q6 (alternating Mg and MRI every 6 months). MR/Q6 includes breast health specialist consultation to discuss risk reduction and the Breast Health Decisions tool (automated risk/prevention recommendations, which is available to all patients). In MyPeBS, the 5-year risk is estimated using the Mammorisk™/BCSC model + PRS 313, or Tyrer-Cuzick (TC) + PRS 313 among women who have >1 first-degree family history of breast cancer (FDFH) with BC. Absolute risk cutoffs are defined as ‘low’ (1.66–6%), and ‘very high’ (≥6%) with corresponding screening assignments: next Mg at 4 years, Mg every 2 years, annually, or annually + MRI. Results WISDOM and MyPeBS enrollment was 46,289 and 53,143 women, respectively. Using the MyPeBS absolute risk cutoffs, the risk distribution for WISDOM and MyPeBS respectively were similar: ‘very high,’ (1% vs. 2%); ‘high’ (31% vs 33%), ‘average’ (26% vs 29%) & ‘low’ (40% vs 37%). Between the two trials, the screening recommendations for the low-risk group were the most different: every 2 years starting at 50 in WISDOM vs. next Mg at 4 years in MyPeBS. Median age was similar (54 vs. 55), though the percentage of women in their 40s was higher in WISDOM (38% to 23%). Baseline BMI was higher in WISDOM (27.6 vs 25.3). Differences (WISDOM vs MyPeBS) include: Education levels, with college degrees in 76% vs 52%; rates of prior biopsies 24% vs 14%; presence of a first-degree family member 24% vs 17%; and the use of hormonal replacement in the postmenopausal women 45% vs 17%. Chemoprevention use at baseline in both trials was 1% or less. Conclusion Within these two major trials, thresholds for risk assignment and proportion of patients in the personalized risk groups are very similar as are screening schemes except for low-risk women (every 2 yrs (WISDOM) vs. every 4 yrs (MyPeBS)). Variation provides the opportunity to learn from the effect of different practice patterns. Data emerging from both trials should be generalizable and have the potential to be practice-changing.
Presentation numberPS3-01-13
Evaluating Cancer Rates, Cancer Types, and Variant Hotspots Across Races and Ethnicities in Individuals with Li-Fraumeni Syndrome
Hillary Esplen, Baylor College of Medicine, San Antonio, TX
H. Esplen1, B. Arun2, C. DiNardo3, H. Abdel-Salam4, K. Richardson5, C. Peterson6, J. Corredor4; 1Department of Pediatrics, Baylor College of Medicine, San Antonio, TX, 2Departments of Breast Medical Oncology and Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Department of Clinical Cancer Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 5Department of Pediatrics, Division of Medical Genetics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth Houston) and Children’s Memorial Hermann Hospital, Houston, TX, 6Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: Li-Fraumeni Syndrome (LFS), caused by pathogenic/likely pathogenic (P/LP) variants in TP53, is a cancer predisposition syndrome that increases the risk for numerous cancer types in both children and adults. Many studies have shown that cancer rates can vary between races and ethnicities. For example, the 2024 Cancer Statistics published by the American Cancer Society reported the incidence for breast cancer to be highest for White individuals and lowest for Hispanic/Latino individuals. One LFS study showed East Asian individuals were more frequently diagnosed with gastric cancer compared to North Americans and Europeans. Other studies identified P/LP TP53 variants associated with specific populations, such as the South and Southeast Brazil founder variant, p.Arg337His. However, there lacks extensive research on the variable expressivity of cancers within the LFS population, based on genotype, race, and/or ethnicity. This study aims to describe the specific TP53 germline variants, cancer rates, and cancer types among individuals of different racial and ethnic groups diagnosed with LFS. Methods: A retrospective chart review of individuals with germline P/LP TP53 variants at the University of Texas MD Anderson Cancer Center was completed. Data collected includes race, ethnicity, number of cancer diagnoses, cancer types, and location of the TP53 variant (exon and codon). Individuals with a suspected somatic TP53 variant were excluded from this study. Racial groupings included Asian, Black or African American, White, and Other/Unspecified. Ethnicity was categorized as Hispanic, Non-Hispanic, and Other/Unspecified. There were also six cancer type categories: adrenocortical carcinoma, brain cancer, breast cancer, osteosarcoma, soft tissue sarcoma, and non-core LFS cancers. Descriptive and statistical analyses, including chi-squared or Fisher’s exact tests and pairwise comparisons, were performed using R software (v4.4.1). Results: Two hundred and fifty individuals with a confirmed P/LP TP53 variant were included in the analysis. Breast cancer was the most common cancer type diagnosed for all ethnic and racial groups, except Asian individuals, who most frequently had non-core LFS cancer diagnoses. There was a significant difference in the rate of breast cancer across racial groups (p = 0.037), with Black or African American individuals with LFS having the highest rate of breast cancer at 76.5%. Additionally, individuals who identified as Black or African American were 1.87 times more likely to develop breast cancer in comparison to those who identified as White. For cancer rates, most individuals in each racial and ethnic group were diagnosed with one primary cancer. Statistical analysis did not identify a significant difference in cancer rates across groups defined by race or ethnicity. Regarding the location of variants within the TP53 gene, there was a significant difference in the rate of variants in exon four (p = 0.021) and exon six (p = 0.016) across racial groups, with variants in exon four being more common in Asian individuals (33.3%) and variants in exon six being more common in Black or African American individuals (35.3%). Conclusion: This study highlights similarities in the rates and types of cancers seen across racial and ethnic groups within a cohort of individuals with LFS. However, this study also identified potential differences in the rates of breast cancer and variant location across racial populations within the cohort. The results of this study provide information that can lead to more personalized counseling for patients and train risk models to accurately predict cancer risk for individuals with LFS of differing races and ethnicities.
Presentation numberPS3-01-14
Oncological and Surgical Outcomes of Risk Reducing Nipple Sparing Mastectomy in Germline Pathogenic Variant Carriers
Francisco Pimentel Cavalcante, Hospital Geral de Fortaleza, fortaleza, Brazil
F. P. Cavalcante1, M. Antonini2, A. L. Frasson3, I. de Oliveira-Junior4, F. P. Brenelli5, A. Berretini Junior6, G. N. Garcia7, F. Bagnoli8, F. P. Zerwes9, E. C. Millen10, A. Mattar11, J. P. Reis12, R. Fenile13, R. A. da Costa Vieira14, H. R. de Oliveira Filho15, M. Lichtenfels16, R. Sandoval17, P. L. Quidute4, R. Z. Torresan5, L. M. Zuliani18, M. M. Bortoluzzi9, A. D. Lima19, R. S. Guindalini20, N. Polidorio21; 1Breast Service, Hospital Geral de Fortaleza, fortaleza, BRAZIL, 2Breast Service, Hospital do Servidor Público Estadual Francisco Morato de Oliveira, Sao Paulo, BRAZIL, 3Breast Service, Hospital Israelita Albert Einstein, Sao Paulo, BRAZIL, 4Breast Service, Barretos Cancer Hospital, Barretos, BRAZIL, 5Breast Service, Universidade Estadual de Campinas, Campinas, BRAZIL, 6Breast Service, Hapvida NotreDame Intermédica, Sao Paulo, BRAZIL, 7Breast Service, Oncoclinicas, Sao Paulo, BRAZIL, 8Breast Service, Santa casa de misericórdia de São Paulo, Sao Paulo, BRAZIL, 9Breast Service, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, BRAZIL, 10Breast Service, Hospital Geral de Fortaleza, Rio de Janeiro, BRAZIL, 11Breast Service, Hospital da Mulher, Sao Paulo, BRAZIL, 12Breast Service, Grupo Orizonti, Belo Horizonte, BRAZIL, 13Breast Service, Hospital Ipiranga, São Paulo, BRAZIL, 14Breast Service, Hospital do Câncer de Muriaé, Muriae, BRAZIL, 15Breast Service, School of Medicine, Federal University of Paraná, Curitiba, BRAZIL, 16Breast Service, Centro de Mama Santa Casa e Oncologia Hospital Nora Teixeira/Rede Einstein, Porto Alegre, BRAZIL, 17Breast Service, Medical Oncology Center, Hospital Sírio-Libanês, Brasilia, BRAZIL, 18Breast Service, Grupo Américas, Hospital Paulistano, Sao Paulo, BRAZIL, 19Breast Service, Universidade Federal do Rio de Janeiro, Rio de Janeiro, BRAZIL, 20Breast Service, Instituto D’or de Ensino e Pesquisa, Salvador, BRAZIL, 21Breast Service, Rede Americas Oncology, Sao Paulo, BRAZIL.
Background: Nipple-sparing mastectomy (NSM) is increasingly adopted as a risk-reducing surgical approach in women with germline pathogenic variants (PVs). Although oncologic outcomes are well established for risk-reducing mastectomies in BRCA1/2 carriers, data are limited for NSM as well as for individuals with other high- and moderate-penetrance PVs. This study aimed to evaluate the incidence of new breast cancer, cosmetic outcomes and surgical complications following risk-reducing NSM in PV carriers. Methods: We conducted a multicenter retrospective cohort study of risk-reducing NSM performed between 2008 and 2025 across 12 institutions. Asymptomatic patients (bilateral risk-reducing mastectomy [BRRM]) or individuals with unilateral breast cancer (contralateral risk-reducing mastectomy [CRRM]) were included. Patients with occult malignancy detected at NSM were excluded. Crude incidence rates were used to report new breast cancer diagnoses and complications. Cosmetic outcomes were assessed using the Harvard four-point scale. Risk factors for complications were analyzed using univariable (UVA) and multivariable (MVA) analyses. Results: A total of 524 patients underwent risk-reducing NSM: 219 BRRM and 305 CRRM (743 mastectomies). Most patients carried BRCA1 (46.0%) or BRCA2 (25.4%) PVs, followed by TP53 (15.1%). Median age was 40 years (range 20-68). At a median follow-up of 41.2 months (IQR 13.1-60.3), three patients (0.6%) developed breast cancer: one nodal recurrence post-BRRM, and two flap recurrences post-CRRM (BRCA1 [n=2] and BRCA2 [n=1]). Breast cancer-related death occurred in three patients (0.6%), all following CRRM. Cosmetic outcomes were favorable, with 90% rated as excellent or good. Surgical complications occurred in 101 patients (19.3%), more frequently after CRRM (24.6%) than BRRM (11.9%) (p<.001). On UVA, complications in BRRM were associated with obesity (OR 9.15, 95% CI 2.08-40.2, p=0.003) and Wise-pattern incision (OR 35.0, 95% CI 2.57-47.3, p=0.008). In CRRM, comorbidities (OR 2.76, 95% CI 1.38-5.54, p=0.004) and Wise-pattern incision (OR 7.33, 95% CI 1.65-32.5, p=0.009) were significant. On MVA, comorbidities (OR 2.59, 95% CI 1.23-5.47, p=0.012) and Wise-pattern incision (OR 7.83, 95% CI 1.73-35.5, p=0.008) remained independently associated with complications in CRRM; no independent predictors were found in BRRM. Conclusion: In this multicenter cohort of women with high- and moderate-penetrance germline PVs, risk-reducing NSM was associated with a very low incidence of new breast cancer (0.6%), supporting its oncologic safety. Most patients achieved favorable cosmetic outcomes. Surgical complication rates were higher after CRRM and associated with comorbidities and Wise-pattern incisions. These findings may guide surgical planning and patient counseling for risk-reducing NSM in genetically predisposed populations.
| Clinical and surgical characteristics, n (%) |
Overall (n=524) |
Bilateral risk-reducing NSM (n=219) |
Contralateral risk-reducing NSM (n=305) |
| Pathogenic variant gene | |||
| BRCA1 | 241 (46.0) | 111 (50.7) | 130 (42.6) |
| BRCA2 | 133 (25.4) | 61 (27.9) | 72 (23.6) |
| TP53 | 79 (15.1) | 17 (7.8) | 62 (20.3) |
| PALB2 | 36 (6.9) | 18 (8.2) | 18 (5.9) |
| CHEK2 | 19 (3.6) | 9 (4.1) | 10 (3.3) |
| ATM | 10 (1.9) | 3 (1.4) | 7 (2.3) |
| Others (PTEN/BARD1/RAD51C/RAD51D | 6 (1.2) | 0 (0.0) | 6 (2.0) |
| Age at risk-reducing NSM, median (range) | 40 (20-68) | 41 (20-67) | 40 (22-68) |
| Comorbidities | 60 (11.5) | 20 (9.1) | 40 (13.3) |
| BMI, median (range) | 24.2 (17.2–39.0) | 23.8 (18.0–36.3) | 24.5 (17.2–39.0) |
| Breast weight in grams, median (IQR) | 250.0 (185.5–325) | 226.0 (46.0–670.0) | 270.0 (35.0–1200.0) |
| Surgical complications | 101 (19.3) | 26 (11.9) | 75 (24.6) |
| Baker 3 Capsular contracture | 45 (8.6) | 13 (6.0) | 32 (10.5) |
| Additional reconstructive surgery | 38 (7.3) | 11 (5.0) | 27 (8.9) |
| Cosmetic outcome by Harvard Scale | |||
| Excellent | 230 (59.9) | 111 (71.6) | 119 (52.0) |
| Good | 113 (29.4) | 40 (25.8) | 73 (31.9) |
| Breast cancer after risk-reducing NSM | 3 (0.6) | 2 (0.9) | 1 (0.3) |
| Breast cancer death | 3 (0.6) | 0 (0.0) | 3 (1.0) |
Presentation numberPS3-01-15
Making informed choices on incorporating chemoprevention into care (MiCHOICE): Cluster randomized controlled trial of decision support for breast cancer chemoprevention, SWOG 1904
Katherine D. Crew, Columbia University Irving Medical Center, New York, NY
K. D. Crew1, G. L. Anderson2, K. B. Arnold3, A. Michel4, M. T. DeLucie4, C. W. Law4, S. Pruthi5, A. C. Sandoval Leon6, R. Shirley7, M. T. Grosse Perdekamp8, S. V. Colonna9, S. L. Krisher10, T. King11, L. D. Yee12, T. J. Ballinger13, C. Braun-Inglis14, D. A. Mangino15, K. B. Wisinski16, C. A. DeYoung17, M. Ross18, J. D. Floyd19, A. Kaster20, L. VanderWalde21, T. Saphner22, C. Zarwan23, S. Lo24, C. Graham25, A. K. Conlin26, K. J. Yost27, D. M. Agnese28, C. Jernigan29, D. L. Hershman1, M. L. Neuhouser30, J. A. Zell31, B. Arun32, R. Kukafka33; 1Medicine and Epidemiology, Columbia University Irving Medical Center, New York, NY, 2Public Health Sciences, SWOG Statistics and Data Management Center, Seattle, WA, 3Statistics, SWOG Statistics and Data Management Center, Seattle, WA, 4Medicine, Columbia University Irving Medical Center, New York, NY, 5General Internal Medicine, Mayo Clinic, Rochester, MN, 6Medicine, Miami Cancer Institute at Baptist Health South Florida, Miami, FL, 7Surgery, Good Samaritan Hospital Corvallis, Corvallis, OR, 8Clinical Sciences, Carle Cancer Center, Urbana, IL, 9Medicine, Huntsman Cancer Institute / University of Utah Medical Center, Salt Lake City, UT, 10Surgery, Holy Redeemer Hospital and Medical Center, Meadowbrook, PA, 11Surgery, Dana-Farber Brigham Cancer Center, Brigham and Women’s Hospital, Boston, MA, 12Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA, 13Medicine, Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN, 14Cancer Biology, University of Hawaii Cancer Center, Honolulu, HI, 15Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 16Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI, 17Medicine, Kaiser Permanente NCORP, Vallejo, CA, 18Medicine, Virginia Commonwealth University, Richmond, VA, 19Medicine, Cancer Care Specialists of Illinois, Heartland NCORP, Decatur, IL, 20Family Medicine, Sanford Roger Maris Cancer Center, Fargo, ND, 21Surgery, Baptist Memorial Health Care, Memphis, TN, 22Medicine, Aurora NCORP, Milwaukee, WI, 23Medicine, Lahey Hospital & Medical Center, Burlington, MA, 24Medicine, Loyola University Stritch School of Medicine, Maywood, IL, 25Surgery, Emory University Hospital/Winship Cancer Institute, Atlanta, GA, 26Medicine, Providence Cancer Institute, Portland, OR, 27Medicine, Cancer Research Consortium of West Michigan NCORP, Kalamazoo, MI, 28Surgery, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 29Patient Research Advocate, SWOG Prevention, Screening, & Surveillance Committee, Kansas City, MO, 30Epidemiology, Fred Hutchinson Cancer Center, Seattle, WA, 31Medicine, University of California, Irvine, Irvine, CA, 32Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 33Biomedical Informatics, Columbia University Irving Medical Center, New York, NY.
Background: Breast cancer (BC) chemoprevention with selective estrogen receptor modulators (SERMs) and aromatase inhibitors (AIs) remains underutilized among high-risk women. Improving knowledge about BC risk and chemoprevention among high-risk patients and their healthcare providers may enhance informed decision-making about this BC risk reduction strategy. Methods: We conducted a cluster randomized controlled trial to evaluate the effectiveness of web-based patient and provider decision support, RealRisks and BNAV, to improve chemoprevention informed choice among women with atypical hyperplasia (AH) or lobular carcinoma in situ (LCIS). Thirty-one sites, including academic and community practices across the U.S., were randomized to standard patient educational materials alone or combined with RealRisks and BNAV. A total of 412 patients and 210 healthcare providers were enrolled. Patient surveys were administered at baseline, 6 and 12 months. The primary outcome at 6 months was chemoprevention informed choice, defined as adequate chemoprevention knowledge and attitudes congruent with decision. Secondary endpoints included BC risk perceptions, worry, chemoprevention knowledge, decision conflict and chemoprevention decision. Assuming 10% loss to follow up (LTFU), roughly equal accrual across sites, and an intraclass coefficient (ICC) of 0.02, we had 90% power to detect a 15% absolute difference in informed choice if the event rate in the control arm was 10%. Results: Among 24 evaluable sites, 14 were randomized to the intervention arm and 10 to the control arm. Among 210 enrolled providers, median age was 46 (range, 28-83) and 78% were female; 75% physicians and 25% advanced practice providers/nurses; 53% medical oncologists, 29% surgeons, and 10% gynecologists/family practitioners/internists; and 83% had previously prescribed chemoprevention. Among 412 enrolled patients, median age was 53 (range, 35-74), including 77% White, 8% Black, and 10% Asian race, and 14% Hispanic ethnicity. In terms of BC risk factors, 77% had AH and 23% LCIS, 34% had a first-degree family history of BC, and 65% had heterogeneously or extremely dense breasts on mammography. Thirty percent were unevaluable for the primary endpoint (1% ineligible, 26% LTFU, 3% missing data). Among 288 evaluable patients (148 at intervention sites, 140 at control sites), the proportion of patients exercising informed choice at 6 months in the intervention and control arms was 35% vs. 27%, respectively (odds ratio [OR]=1.44, 95% confidence interval [CI]=0.84-2.46, p=0.19; observed ICC=0.006). Compared to patients in the control arm at 6 months, those in the intervention arm were more likely to have accurate BC risk perceptions (OR=1.80, 95% CI=1.11-2.91) and adequate chemoprevention knowledge (OR=1.62, 95% CI=1.00-2.61). We observed no significant differences in BC worry, decision conflict, or chemoprevention decision between study arms. At 6 months, 49% of women in the control arm and 51% in the intervention arm decided to take a SERM or AI for BC chemoprevention (p=0.58), a rate higher than anticipated. Conclusions: We did not observe a significant increase in informed choice among patients and providers assigned to chemoprevention decision support compared to standard educational materials alone. Among women with AH or LCIS seen mainly by breast specialists who prescribed chemoprevention, about half decided to take a SERM or AI. Therefore, relatively high chemoprevention uptake may be achieved among women with high-risk breast lesions managed by breast specialists. Chemoprevention decision support may improve decision antecedents, such as accurate BC risk perceptions and chemoprevention knowledge, but may have a limited role in changing health behaviors.
Presentation numberPS3-01-16
Gut microbial dysbiosis associates with the early-onset breast cancer in BRCA mutation carriers
Po-Han Lin, National Taiwan University Hospital, Taipei City, Taiwan
P. Lin1, Y. Chen2, T. Yen1, H. Chang1, Y. Chu1, Z. Liu1, S. Kuo3, C. Huang4; 1Medical Genetics, National Taiwan University Hospital, Taipei City, TAIWAN, 2Pediatrics, National Taiwan University Hospital, Taipei City, TAIWAN, 3Medical Oncology, National Taiwan University Hospital, Taipei City, TAIWAN, 4Surgery, National Taiwan University Hospital, Taipei City, TAIWAN.
Germline BRCA mutation carriers face a high risk of developing breast cancer, yet individual risk and age of onset vary significantly, influenced by both genetic and environmental factors. The gut microbiota is crucial for host health, regulating immunity, metabolism, and endocrine functions. Dysbiosis—an imbalance of gut microbiota—has been linked to diseases including cancer. We aimed to investigate whether specific gut microbes influence breast cancer risk in BRCA mutation carriers and to explore the underlying mechanisms. First, we studied 27 BRCA mutation carriers with early-onset breast cancer (60 years) without cancer. Fecal microbiota were analyzed using full-length 16S rRNA sequencing. The linear discriminant analysis (LDA) effect size (LEfSe) analysis showed that the Eggerthella lenta (E. lenta) and Dorea longicatena (D. longicatena) were the most significantly enriched bacteria in feces of the mutation carriers with early-onset breast cancer, compared to those in the old carriers without breast cancer. As these two bacteria are more common in older individuals, their presence in younger cancer patients suggests an association with cancer, independent of age. Second, we expanded the study to 102 BRCA mutation carriers (54 BRCA1, 48 BRCA2), 64 of whom had breast cancer. Mutation carriers with fecal E. lenta had a significantly earlier age of breast cancer than those without it (median age: 42.2 vs. 52.8 years-old, p<0.001), while D. longicatena showed no significant association. Subgroup analyses confirmed the link between E. lenta and early-onset breast cancer in both BRCA1 and BRCA2 groups (p<0.05). We then generated antibiotic-induced microbiome depletion (AIMD) model in the brca1-null mouse. The AIMD mice were gavaged with phosphate-buffered saline or E. lenta; mice gavaged with E. lenta developed breast tumors significantly earlier than those without E. lenta (p=0.022). We conducted untargeted metabolomics on urine samples from mutation carriers with early-onset breast cancer and from old cancer-free carriers (age >60). Tryptophan and its downstream metabolites—kynurenine, 3-hydroxykynurenine, 3-hydroxyanthranilic acid, and quinolinic acid—were significantly elevated in early-onset patients and positively correlated with E. lenta levels (p<0.05), compared to metabolites in the old cancer-free carriers. In mice fed E. lenta, serum levels of tryptophan/kynurenine pathway metabolites were elevated, too. To assess causality, brca1-null mice were intraperitoneal injected with quinolinic acid, a downstream end-product of the tryptophan/kynurenine pathway. The treated mice developed breast tumors significantly earlier than controls (p<0.01). Then, we tested the impact of quinolinic acid in the cells with heterozygous BRCA1 mutation. DNA fiber assay revealed significantly increased replication speed and impaired replication fork protection in the BRCA1 (185delAG/+) MCF10A cell line with co-culture of 10uM quinolinic acid, compared to those without quinolinic acid co-culture. We long-term (6 weeks) treated BRCA1 (185delAG/+) MCF10A cells with 10uM quinolinic acid. Whole exome sequencing revealed the treated cells harboring a significantly elevated homologous recombination deficiency score (by scarHRD software), compared to cells without quinolinic acid co-culture (p<0.05). These data suggested quinolinic acid enhanced the replication stress and caused the genomic instability in the human mammary cells harboring BRCA1 mutation. Our study demonstrated that dysbiosis is associated with early-onset breast cancer in BRCA mutation carriers. The E. lenta-derived tryptophan/kynurenine/quinolinic acid pathway induces the replication stress and genomic instability that drive breast cancer.
Presentation numberPS3-01-18
Gene expression signatures of beneficial response to tamoxifen for breast cancer prevention
Stacey J Winham, Mayo Clinic, Rochester, MN
S. J. Winham1, C. Wang1, Y. Liu1, B. M. McCauley1, D. J. Tabrizi1, D. L. Gehling2, J. L. Fischer2, L. R. Seymour2, M. H. Solanki3, M. E. Sherman4, K. R. Lohani2, T. L. Hoskin1, D. C. Radisky5, A. C. Degnim2; 1Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 2Department of Surgery, Mayo Clinic, Rochester, MN, 3Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 4Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 5Department of Cancer Biology, Mayo Clinic, Jacksonville, FL.
Uptake of tamoxifen is limited to 5-15% of all eligible women due to concerns regarding medication side effects combined with the lack of ability to predict which women will benefit from the therapies. We used a unique retrospective sample of n=92 high-risk women with paired benign biopsies obtained before (PreTam) and during tamoxifen (OnTam) to discover tissue gene expression biomarkers of biologic response, defined as change (Δ) in lobular Ki67. Ki67 was digitally quantified in normal lobules from PreTam and OnTam benign tissue samples and samples with PreTam Ki67 = 0 were excluded. Data were analyzed from 51 women with both PreTam and OnTam Ki67 (median age = 49 years and median BMI = 25.5 kg/m2 at PreTam biopsy). OnTam biopsies were collected a median of 11.5 months after PreTam biopsy (median 6.5 months after starting tamoxifen; dose 20 mg/day in 86%). Median Ki67 significantly decreased overall (Δ Ki67 = -0.29%, p=0.003), but the effect was restricted to women with baseline Ki67 ≥ 0.5% (n=38, Δ Ki67 = -0.78%, p=0.0002); no significant change occurred with baseline Ki67 14,600 genes), PreTam vs OnTam comparison across 33 pairs with baseline Ki67 ≥ 0.5% yielded 66 differentially expressed genes (fold-change > 2 and p<0.01). At p 0.50) and 14 genes with significant negative correlation (rho < -0.50) with Δ Ki67 (p<0.01). At p<0.01, ranked gene set enrichment analysis yielded 67 gene sets enriched with negative correlation (i.e. increased expression associated with a reduction in Ki67), including pathways related to ribosome biogenesis, DNA recombination, and nucleic acid metabolic processes; there were 62 gene sets enriched for positive correlation (i.e. decreased expression associated with a reduction in Ki67), including pathways such as VEGF and ERBB signaling. Our data support (i) baseline Ki67 ≥ 0.5 % and (ii) an 87‑gene baseline signature as complementary biomarkers of tamoxifen response. Prospective validation of this combined assay could target tamoxifen to the women most likely to benefit, potentially overcoming a major barrier to chemoprevention uptake.
Presentation numberPS3-01-19
Supplemental Imaging Improves Breast Cancer Early Detection but Remains Underutilized among High-Risk Women
Maryam Imran, The University of Texas MD Anderson Cancer Center, Houston, TX
M. Imran1, N. M. Kettner1, S. Hanash1, O. Weaver2, J. Leung2, J. Dennison1; 1Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX.
Background: The MERIT (Mammography, Early Detection, Risk Assessment, and Imaging Technologies) cohort was established in 2017 at MD Anderson to integrate clinical, imaging, and blood biomarker data for personalized risk assessment of breast and other cancers. This analysis evaluates breast cancer detection rates and staging outcomes among women undergoing annual screening mammography with or without supplemental imaging. Methods: Between 2017 and the present, 8,099 women receiving annual screening mammograms were enrolled in the MERIT cohort. Participants were categorized based on imaging modality: mammography (MG) alone or MG with supplemental imaging, defined as receipt of breast MRI or CEM within two years of enrollment. Clinical data, imaging outcomes, and cancer diagnoses were collected. Follow-up time was calculated from enrollment to the date of the last breast imaging exam or breast cancer diagnosis. Cancers were classified as ductal carcinoma in situ (DCIS) or invasive, with staging and receptor subtype data collected when available. Results: Over 28,153 person-years of follow-up, 236 breast cancers were diagnosed. Women with cancer had a mean follow-up time of 2.30 years (SD = 1.86), compared to 3.51 years (SD = 2.19) among women without cancer. Among 1,505 women who underwent supplemental imaging, 61 breast cancers were identified, resulting in an incidence rate of 1,046 per 100,000 person-years. In this group, 38 percent of cancers were DCIS and 62 percent were invasive. No stage III or IV cancers were detected among women who received supplemental imaging. In contrast, among 6,594 women who received screening mammography alone, 175 breast cancers were diagnosed, yielding an incidence rate of 784 per 100,000 person-years. In this group, 28 percent of cancers were DCIS and 72 percent were invasive. Eleven late-stage cancers were identified in the mammography-alone group, including five stage IIB, three stage IIIB, one stage IIIC, and two stage IV cancers. Notably, eight of these were triple-negative, and two stage IV cases were hormone receptor-positive. MRI utilization varied by both breast density and Gail model risk classification. Among high-risk women with dense breasts, 37 percent received MRI, compared to 19 percent of high-risk women with non-dense breasts. Among low-risk women, MRI use was 16 percent for those with dense breasts and 9 percent for those with non-dense breasts. Conclusions: Supplemental imaging was associated with a higher breast cancer detection rate and a greater proportion of early-stage disease compared to mammography alone. The absence of late-stage cancers in the supplemental imaging group, in contrast to 11 advanced-stage cases in the MG-alone group, suggests a potential benefit for earlier detection through supplemental imaging. However, supplemental MRI remains underutilized among high-risk women. These findings support the need for personalized, risk-aligned screening strategies that integrate both breast density and formal risk assessment to improve early detection of clinically significant breast cancer.
Presentation numberPS3-01-20
Prospective evaluation of breast mammographic density and biomarker changes induced by 4-hydroxytamoxifen gel application versus placebo to the breast of women at increased risk for breast cancer
Banu K Arun, UT MD Anderson Cancer Center, Houston, TX
B. K. Arun1, G. Gierach2, M. Scoggins3, E. A. Bowles4, S. A. Khan5, T. B. Bevers6, S. S. Rao7, J. E. Garber8, S. Raza9, N. B. Kumar10, H. S. Han11, J. Heine12, B. Niell13, P. Chalasani14, K. Fitzpatrick15, L. G. Wilke16, A. Eowler17, H. C. Beckwith18, J. E. Kuehn-Haijder19, C. Mays6, L. A. Vornik6, O. Lee20, E. Diamond21, M. Perloff22, W. Dong23, D. Liu24, J. J. Lee24, F. W. Symmans25, E. Vilar-Sanchez6, B. M. Heckman-Stoddard26, P. H. Brown6; 1Breast Medical Oncology and Clinical Cancer Genetics, UT MD Anderson Cancer Center, Houston, TX, 2Integrative Tumor Epidemiology, NCI, Rockville, MD, 3Radiology, UT Southwestern Medical Ctr, Dallas, TX, 4Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington Health Research Institute, Seattle, WA, 5Breast Surgical Oncology, Northwestern University, Chicago, IL, 6Cancer Prevention, UT MD Anderson Cancer Center, Houston, TX, 7Genetics, Northwestern Medical Center, Chicago, IL, 8Breast Medical Oncology and Clinical Cancer Genetics, Dana Farber Cancer Institute, Boston, MA, 9Radiology, Brigham & Women’s Hospital, Boston, MA, 10Cancer Epidemiology, Mofitt Cancer Center, Tampa, FL, 11Breast Medical Oncology, Moffit Cancer Center, Tampa, FL, 12Breast Medical Oncology and Clinical Cancer Genetics, Moffitt Cancer Center, Tampa, FL, 13Radiology, Moffitt Cancer Center, Tmpa, FL, 14Breast Medical Oncology and Clinical Cancer Genetics, University of Arizona Cancer Center, Tucson, AZ, 15Radiology, University of Arizona, Tucson, AZ, 16Surgery, University of Wisconsin, Madison, WI, 17Radiologycs, University of Wisconsin, madison, WI, 18Medical Oncology, University of Minnesota, Minneapolis, MN, 19Radiology, University of Wisconsin, Minneapolis, MN, 20Breast Medical Oncology and Clinical Cancer Genetics, Northwestern University, Chicago, IL, 21Division of Cancer Prevention, NCI, Bethesda, MD, 22Division of Cancer Prevention, NCI, Rockville, MD, 23Bioistatistics, UT MD Anderson Cancer Center, Houston, TX, 24Biostatistics, UT MD Anderson Cancer Center, Houston, TX, 25Department of Pathology, UT MD Anderson Cancer Center, Houston, TX, 26Division of Cancer Prevention, NCI, Houston, TX.
Background: Tamoxifen uptake for risk reduction has remained low due to concerns about toxicity despite available effectiveness data. Studies of tamoxifen in the adjuvant and preventive setting have demonstrated that a 10% or greater decline in mammographic density (MD) is associated with better outcomes; however, tamoxifen metabolism and associated MD declines are variable across patients. An alternative application of a bioactive tamoxifen metabolite may reduce side effects while maintaining efficacy. 4-hydroxytamoxifen topical gel (4-OHT) is a transdermal agent, shown in preliminary studies to be well-tolerated with similar decreases in Ki-67 to oral tamoxifen in presurgical DCIS studies and significant drug concentration in breast parenchyma but very low levels in the systemic circulation. Therefore, we conducted a multicenter clinical trial to evaluate changes in MD in women with dense breasts induced by 4-OHT versus placebo gel when applied to the breast for 12 months. Methods: Pre- and postmenopausal women between the ages 40-69 yrs, or less than 40 yrs with 5-year Gail risk greater than 1.66%, with heterogeneously or extremely dense breast tissue, were enrolled in this study. Participants were randomized (1:1): 2 mg 4-OHT gel or Placebo gel for 12 months (mo), applied daily to each breast. Participants underwent baseline mammograms, blood draws for toxicity and research biomarkers, and optional breast biopsy, all of which were repeated at the end of 12 mo. The primary endpoint was to estimate and compare the percent change in MD (measured on cranio-caudal mammograms using Cumulus) from baseline to mo 12 between 2 groups. With 64 women in each group, there is 80% power to detect a decrease in density of 6% in the 4-OHT gel vs 2% in the placebo group with a common SD of 8% using a two-sided t-test with a significance level of 0.05. Secondary endpoints included tolerability, measurement of drug metabolites in breast tissue and plasma, and research biomarkers. Results: This prospective, randomized, double-blind, placebo-controlled phase II study randomized 158 participants from Sept 2017 to October 2020 across 6 centers. 123 participants had both baseline and month 12 mammograms, 61 in 4-OHT and 62 in Placebo. The mean (standard deviation) breast density decrease over 12 month was 6.30% (9.11%) in the 4-OHT gel arm and 5.77% (11.24%) in the Placebo arm. The changes in breast density from baseline to month 12 were not significantly different between the two treatment arms (Hotelling’s T2 test p-value = 0.09). For those with available research biopsies, in the placebo arm (N=18), all 4-OHT and metabolite levels were below the quantifiable level (BQL). In the treatment arm (N=12), at months 6 and 12, 10 patients had Z-4-OHT tissue levels above BQL with an average of 10.09 ng/g. No grade 3/4 toxicity was observed in either arm. Twenty-nine participants (thirteen 4-OHT and sixteen Placebo) had 41 gr 1 or 2 adverse events related to the application of gel. Drug exposure serum toxicity, including FVIII and vWB, was not different between treatment arms. IGF-1 levels decreased from baseline to month 12 in both arms; the difference was not statistically significant, whereas IGFBP-3 increased significantly at month 12 in the 4-OHT arm (p=0.04). Conclusion: This prospective double-blind randomized study did not demonstrate a significant difference in the change of MD from baseline to the 12-month treatment between the 4-OHT arm and the placebo arm. The 4-OHT gel was well tolerated, and drug and metabolites were measurable in the breast tissue. Serum IGFBP-3 was found to be increased in the 4-OHT treatment group. The effect of baseline MD characteristics by automated software measurements and other tissue biomarkers will be analyzed and reported separately.
Presentation numberPS3-01-21
Prediction of Breast Cancer Risk and Tumor Aggressiveness in the Athena Breast Health Network
Katherine Leggat-Barr, Tufts University School of Medicine, Boston, MA
K. Leggat-Barr1, J. Tice2, K. Badal3, R. Sayaman4, B. Parker5, S. Hartman6, H. Anton-Culver7, H. Park8, A. Ziogas9, A. Petruse10, A. Naiem11, K. Malvin12, L. Sabacan13, T. Lewis14, T. Glatt1, S. Kapoor3, A. Verma15, P. Warner16, E. Tsopurashvili3, A. Griffin17, V. Lee18, J. McGuire19, S. Borowsky20, WISDOM Study and Athena Breast Health NetworkInvestigators and Advocate Partners21, C. Kaplan22, R. Hiatt23, A. Stover-Fiscalini3, K. Kerlikowske12, Y. Shieh24, L. Esserman3, L. van ‘t Veer4; 1School of Medicine, Tufts University School of Medicine, Boston, MA, 2Department of General and Internal Medicine, University of California, San Francisco, San Francisco, CA, 3Department of Surgery, University of California, San Francisco, San Francisco, CA, 4Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, 5Moores Cancer Center, University of California San Diego, San Diego, CA, 6Department of Family and Preventive Medicine, University of California, San Diego, San Diego, CA, 7Department of Medicine, Genetic Epidemiology Research Institute, University of California, Irvine, Irvine, CA, 8UC Irvine School of Medicine, Department of Pathology and Laboratory Medicine, Irvine, CA, 9Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, CA, 10Clinical and Translational Science Institute Office of Clinical Research, University of California Los Angeles, Los Angeles, CA, 11Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 12Departments of Medicine and Epidemiology/Biostatistics, University of California, San Francisco, San Francisco, CA, 13Department of Surgery, University of California San Francisco, San Francisco, CA, 14School of Medicine, University of California, San Francisco, San Francisco, CA, 15School of Medicine, Pritzker School of Medicine, Uchicago, Chicago, IL, 16School of Medicine, Columbia University, New York, NY, 17Cancer Registry, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 18Breast Science Advocacy Core, Breast Oncology Program, University of California, San Francisco, San Francisco, CA, 19Cancer Registry, University of California, San Francisco, San Francisco, CA, 20Department of Pathology, School of Medicine, University of California Davis, Saramento, CA, 2122Division of General Internal Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA., San Francisco, CA, 23UCSF Helen Diller Family Comprehensive Cancer Center, Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA USA, San Francisco, CA, 24Department of Population Health Sciences, Weill Cornell Medicine, New York, NY.
Background The Athena Breast Health Network (Athena) is a University of California (UC) and Sanford Health initiative integrating clinical care and research to drive improvements in breast screening. Here, we evaluated cancer occurrence and tumor type in our UC San Francisco (UCSF) Athena cohort. We assessed the association with 5-year invasive cancer risk level by the Breast Cancer Surveillance Consortium (BCSC) version 3 (BCSCv3) model and the development of aggressive or non-aggressive cancer. Methods BCSCv3 risk scores were calculated for 10,031 consented UCSF Athena participants enrolled 2011-2022 with a median follow-up of 6.9 years. The BCSCv3 model includes age, race/ethnicity, prior breast biopsy results, breast density, first and second-degree family history, body mass index, menopausal status, and age at first live birth. Risk information was gathered prior to breast cancer diagnosis. The ‘high’ risk threshold was previously set at >1.67% 5-year risk by the BCSCv3 model (PMID37976443). All others were classified as ‘low’ risk. To identify those who developed invasive breast cancer after intake, we performed a cancer linkage with the San Francisco Mammography Registry and the UCSF cancer registry (complete through Dec. 2018 and Jan. 2022 respectively). Tumor aggressiveness was defined in two ways. Advanced or non-advanced cancer was classified by the AJCC prognostic pathologic stage (>stage II is advanced, <stage II is non-advanced). Faster and slower growing cancer considered tumor biology only, where one or more of the following criteria defined faster: HR-negative, HER2-positive, and/or grade 3, all other were slower growing cancers. We used logistic regressions to examine associations between BCSCv3 5-year risk score and breast cancer incidence and tumor aggressiveness. Results Of the 10,031 participants, 2838 (28%) participants were designated as ‘high’ risk, and 7193 (72%) participants were ‘low’ risk. Overall, 213 (2%) developed breast cancer. 34 (16%) developed advanced cancer and 179 (84%) developed non-advanced cancer. Among women with breast cancer, 60 (28%) developed faster-growing cancers and 153 (72%) developed slower-growing cancers. The median 5-year BCSCv3 risk score for women with and without breast cancer was 1.73% and 1.38% respectively (p<0.001). The average 5-year BCSCv3 risk score for women with advanced and non-advanced cancers was 1.67% and 1.74%, respectively. The average 5-year BCSCv3 risk score for women with faster and slower growing cancers was 1.65% and 1.76%, respectively. In logistic regression analysis adjusted for age, women in the high-risk group had significantly higher odds of developing non-advanced cancer compared to healthy controls (OR = 3.08, 95% CI 1.87-5.08, p<0.001), whereas the association for advanced cancer was not statistically significant (OR = 1.24, 95% CI 0.56-2.77, p=0.59). Similarly, when analyzing slow vs. fast growing cancer, in logistic regression analysis adjusted for age, women in the high-risk group had significantly higher odds of developing slower-growing cancer compared to healthy controls (OR = 2.02, 95% CI 1.39-2.92, p<0.0001), whereas the association for faster-growing cancer was not statistically significant (OR = 1.62, 95% CI 0.88-2.99, p=0.12). Conclusions This study confirms that higher 5-year BCSCv3 risk scores are associated with higher overall breast cancer development. Participants with the highest 5-year BCSCv3 risk scores are more likely to develop cancers with less aggressive features, suggesting the BCSCv3 model may preferentially predict less aggressive tumors. Those with the highest 5-year BCSCv3 risk may be more likely to benefit from endocrine risk reduction therapy. The Analysis using the recently developed BCSC advanced cancer model is ongoing and may offer better discrimination for those at risk for advanced cancers.
Presentation numberPS3-01-22
Targeting Disparities: Impact of Mammographic Screening and Social Drivers on Advanced-Stage Triple-Negative Breast Cancer in Vulnerable Populations Across US Counties
Reine Abou Zeidane, University Hospitals Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH
R. Abou Zeidane1, C. Pisano1, F. Hussain2, W. Dong2, T. Lal3, L. Vu2, N. Mehta4, S. Koroukian2, J. Rose2, C. Speers1; 1Radiation Oncology, University Hospitals Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH, 2Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 3Surgery, division of Surgical Oncology, University Hospitals Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH, 4Family Medicine, University Hospitals Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, OH.
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with disproportionate mortality among underserved populations. Mammographic screening reduces breast cancer mortality, but disparities in access and utilization persist, particularly in African American (AA) communities and socioeconomically vulnerable areas. This study aims to quantify the relationship between mammography uptake, demographic factors, and social determinants of health (SDOH) with advanced-stage TNBC (AS-TNBC) across diverse U.S. populations. Methods: We conducted a cross-sectional analysis of 593 counties across multiple states using data from the CDC PLACES database (2020 release, representing 2018 data), the American Community Survey (ACS 2014-2018), and county-level SDOH indicators. Communities were aggregated using the Max-P Regions method to balance TNBC case counts and socioeconomic indices (adjusted for area deprivation index, ADI). Descriptive statistics were used to summarize key characteristics across regions. Then, multivariable Poisson and negative binomial regression models were used to evaluate associations between AS-TNBC diagnoses mammographic screening uptake, demographic (% AA, % Hispanic, % aged <45 years), and social vulnerabilities, including limited English proficiency, rurality, and insurance status. Results: The median percentage of AS-TNBC diagnoses across counties was 38.8% (IQR: 34.9-43.6%). Counties with higher AS-TNBC incidence exhibited significantly lower mammography use (71.5% vs. 72.6%, p 0.001), higher uninsured rates (18.7% vs. 15.7%, p <0.001), and increased proportions of AA residents (14.0% vs. 7.9%, p <0.001), limited English proficiency (1.7% vs. 1.5%, p 0.005), higher socioeconomic deprivation (ADI top quartile: 33.7% vs. 16.4%, p <0.001), and greater rurality (41.5% vs. 30.2%, p 0.006). Multivariable Poisson regression modeling demonstrated a protective effect of mammographic screening, where a 10% increase in screening uptake correlated with a 5.2% decrease in AS-TNBC diagnoses (RR 0.95, 95% CI 0.91-0.99, p 0.007). Conversely, higher proportions of AA residents (RR 1.04, p <0.001), rural populations (RR 1.05, p 0.02), and Hispanic residents (RR 1.002, p <0.001) significantly correlated with increased AS-TNBC rates. Negative binomial regression (adjusting for total breast cancer cases) reaffirmed the robust protective role of screening (RR 0.91, 95% CI 0.89-0.93, p <0.001). In the negative binomial model (adjusted for total breast cancer cases), mammographic screening remained a strong protective factor (RR 0.91, 95% CI: 0.89-0.93, p <0.001), indicating a 9.1% reduction in AS diagnosis per 10% increase in screening. Higher proportions of AA residents (RR 1.04, p <0.001), uninsured individuals (RR 1.04, p <0.001), and those in the most deprived areas (ADI top quartile, RR 1.04, p <0.001) were significantly associated with higher AS diagnosis rates, as well as younger population composition (age 18-44; RR 1.03, p 0.004). Conclusions: Mammographic screening disparities and adverse social determinants significantly influence the prevalence of advanced-stage TNBC. High-risk populations, particularly AA communities, rural areas, and regions with elevated socioeconomic vulnerabilities, face disproportionate impacts. These findings highlight the urgent need for targeted interventions aimed at enhancing access to culturally sensitive and linguistically appropriate screening and healthcare services to mitigate disparities and improve breast cancer outcomes.
Presentation numberPS3-01-23
National cost of the WISDOM risk-based breast cancer screening algorithm with comparison to advocated guidelines
Kimberly Badal, University of California San Francisco, San Francisco, CA
K. Badal1, J. Staib2, J. A. Tice3, M. Kim4, M. Eklund5, L. Wilson6, S. Dacosta Byfield2, K. Catlett7, L. Maffey2, R. Soonavala1, A. Stover Fiscalini1, L. Esserman1; 1Department of Surgery, University of California San Francisco, San Francisco, CA, 2Optum Labs, Optum Labs, Eden Prairie, MN, 3Department of Medicine, University of California San Francisco, San Francisco, CA, 4Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, 5Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Solna, SWEDEN, 6Department of Medicine and Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, 7Optum Center for Research and Innovation, UnitedHealth Group, Minnetonka, MN.
Background: The WISDOM (Women Informed to Screen Depending on Measures of risk) Study is testing the safety and effectiveness of risk-based breast cancer screening. We aimed to estimate the total, national, yearly cost of screening in the USA if WISDOM’s screening algorithm were used, with comparison to current screening guidelines. Methods: We used Optum Labs Data Warehouse (OLDW) to estimate 2D and 3D- mammograms, and MRI screening and recall costs and rates for commercial and Medicare Advantage insurance. OLDW contains longitudinal de-identified administrative, medical, and pharmacy claims for over 200 million commercial and Medicare Advantage enrollees and patients. Screening and recall costs were normalized to calendar year 2020. We used real-world participation rates by age of 54-78%. The risk-based strategy included a cost of USD$270 for genetic testing and risk assessment. We modeled three guidelines: 2021/2023 American College of Radiology (ACR), 2023 American Cancer Society (ACS), and 2024 United States Preventative Services Task Force (USPSTF). The percentage of high-risk/average-risk women per guideline were estimated using the literature. Total cost of screening was calculated as the sum of screening and recall costs. The average lifetime cost of screening per woman until 74 years was also calculated. One-way deterministic sensitivity analysis was used to determine the impact of inputs on the aggregate cost. Results: Median mammogram costs ranged from $139-$360, MRI costs ranged from $545-$2,439, while recall rates ranged from 9.2%-20.9%. The projected national cost of screening was $30B for ACR, $18B for ACS, $8B for USPSTF, and $7B for WISDOM. The average lifetime cost through age 74 years to screen a high-risk/average-risk woman was: $89,725/$13,416 for ACR, $89,725/$7,946 for ACS, $6,931/$6,931 for USPSTF, and $66,451/$6,618 for WISDOM. For USPSTF, the cost of screening high-risk/average-risk women were the same because these women were screened the same. Participation rates, proportion of women with a lifetime risk >20%, and commercial MRI and 3D costs had the largest impact on total costs. Conclusion: WISDOM’s risk-based screening algorithm can reduce national cost substantially, even with the added cost of genetic testing, while maintaining intensive screening for the highest-risk women. The resources saved can be used to improve screening for women at high risk for fast-growing cancers, which are often identified between screens, and to encourage improved adherence.
Presentation numberPS3-01-24
Results of a training workshop for genetic counseling students on improving patient research literacy and engagement in clinical research
Susan Joy Friedman, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL
S. J. Friedman1, R. H. Pugh Yi2, M. Dean3, P. L. Welcsh4, K. N. Owens4, J. D. Rogers1, D. B. Rose5, E. Kuhn6; 1Research and Education, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 2Program Evaluation, Akeso Consulting, LLC, Vienna, VA, 3Department of Communication, University of South Florida, Tampa, FL, 4Education, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 5Research, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 6Education, Susan G. Komen, Dallas, TX.
Background New cancer treatment and prevention strategies have led to an increase in clinical trials enrolling people with germline mutations linked to cancer risk. Yet, referral of patients remains low. In a FORCE survey, 14% of respondents at high risk for breast cancer were told by their healthcare providers about clinical research opportunities, while 75% expressed interest1. In a survey of genetic counselors, most had never referred breast cancer survivors (63%) or metastatic patients (69%) to clinical trials, and 55% listed research jargon/health literacy as barriers to referral2. We developed a training for genetic counseling students consisting of a presentation, activities and group discussions on factors that impede patients’ ability to find, understand, and enroll in hereditary cancer research, and ways that genetic counselors can address these barriers. We introduced tools to help people find research studies, including FORCE’s “Search and Enroll Tool” that provides plain-language descriptions of studies enrolling people with inherited mutations. Methods We conducted 17 workshops for genetic counselor training programs, and trained 256 students. Almost half completed feedback surveys. Three training program directors participated in post-training interviews. Results Students indicated that they plan to apply what they had learned in practice. They appreciated the plain-language features of FORCE’s tool and how it prioritizes studies enrolling people with germline mutations, and they plan to use the tool to connect patients to research opportunities. Course directors agreed that the training was appropriate to integrate into their curricula. They valued the fact that the training helps students recognize the importance of sharing clinical research opportunities with patients, teaches students how to find this information and present it to patients in easy-to-understand terms, and emphasizes the importance of representativeness in research. Students and directors recommended retaining the content, flow and presentation approach, activities and discussions, and emphasis on the practical application of material presented. Suggestions included limiting the training to 90 minutes, providing more hands-on experience with finding and discussing studies with patients, and developing a version that students can complete on their own. Discussion Students and faculty found the training to be informative, relevant, and engaging. Genetic counseling training programs could benefit from training on this critical topic that may not be offered elsewhere in their curricula. In addition to learning information and skills related to finding and communicating with patients about hereditary cancer research, students and faculty agreed that FORCE’s Search and Enroll Tool is useful for matching patients to relevant research opportunities.
| Question | Number of respondents who strongly agreed or agreed (%) |
| After the presentation I had a stronger understanding of the challenges of enrolling patients in cancer research. | 109 (96.4%) |
| After the presentation, I had a stronger understanding of how social determinants of health affect research participation. | 106 (93.8%) |
| After the presentation, I had a stronger understanding of how to find research studies for specific patient populations. | 109 (97.3%) |
|
The presentation will help me better communicate directly with people about research opportunities. |
109 (96.4%) |
|
The presentation provided me with useful tools to help me find and refer patients to clinical research studies. |
110 (98.2%) |
| The presentation made me more likely to refer patients to clinical research studies. | 102 (90.3%) |
| I plan to use FORCE’s Search and Enroll Tool to find studies enrolling patients with inherited mutations. | 106 (94.6%) |
| The presentation met the stated objectives. | 109 (98.2%) |
Presentation numberPS3-01-25
Replication stress as a driver of breast tumorigenesis in BRCA1 heterozygous<sup> </sup>carrier derived breast organoids
Ozge Somuncu, Dana Farber Cancer Institute, Boston, MA
O. Somuncu1, B. Lamarre1, S. Mukkavalli1, T. B. Branigan2, R. Ravindranathan1, N. V. Wietmarschen3, H. Nguyen1, N. Ashton4, K. Parmar1, G. Shapiro1, D. A. Dillon5, D. Chowdhury3, P. A. Konstantinopoulos3, J. E. Garber3, A. D. D’Andrea1; 1Center for DNA Damage and Repair, Dana Farber Cancer Institute, Boston, MA, 2Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 3Center for BRCA and Related Genes, Dana Farber Cancer Institute, Boston, MA, 4Department of Radiation Oncology, Dana Farber Cancer Institute, Boston, MA, 5Department of Pathology, Dana Farber Cancer Institute, Boston, MA.
Replication stress as a driver of breast tumorigenesis in BRCA1 heterozygous carrier-derived breast organoids Ozge S. Somuncu, Benjamin Lamarre, Sirisha Mukkavalli, Timothy B. Branigan, Ramya Ravindranathan, Niek Van Wietmarschen, Huy Nguyen, Nicholas Ashton, Kalindi Parmar, Geoffrey Shapiro, Deborah A. Dillon, Dipanjan Chowdhury, Panagiotis A. Konstantinopoulos, Judy E. Garber, Alan D. D’Andrea Germline mutations in BRCA1 significantly increase the risk of BRCA1(+/-) heterozygous carriers in developing early and rapid breast and ovarian cancers. However, the physiological alterations in heterozygous BRCA1(+/-) breast tissue necessary for malignant transformation, and the specific features predisposing this tissue to loss of heterozygosity (LOH) are poorly understood. BRCA1(+/-) carrier breast tissue is reported to contain a small population of cells with copy number alterations (CNAs) (1q gain and 16q loss), p53 mutations, and LOH of BRCA1. A previous study showed that BRCA1(+/-) carrier breast tissue fibroblasts and mammary epithelial cell lines have several features of replication stress (R/S), which may contribute to genomic instability and progression to cancer. Luminal progenitor cells (LPs) appear to be the primary cells leading to these cancers but the contribution of other subpopulations in breast tissue remains to be elucidated. Patient-derived BRCA1(+/-) carrier organoids are a living, physiological system that allows for this study of R/S, BRCA1 LOH, and acquired CNAs in all breast subpopulations in parallel. To further examine the phenotype of BRCA1(+/-) breast tissue, we generated viably-growing carrier organoids derived from breast tissues of seventeen BRCA1(+/-) women, who underwent prophylactic mastectomy, and from fifteen BRCA1(+/+) women who underwent reduction surgery as controls. Hydroxyurea induced R/S, as indicated by increased levels of pKAP1, yH2AX, and ELF3, in all subpopulations of the BRCA1(+/-) carrier organoids in comparison to the BRCA1(+/+) organoids. Consequently, BRCA1(+/-) carrier breast tissues and organoids exhibited shortened telomere length and high levels of ELF5, a premature aging marker, suggesting that BRCA1(+/-) carriers are predisposed to altered telomere replication leading to R/S. BRCA1(+/-) carrier organoids and tissues are enriched for G2/M markers where homologous recombination (HR) is a critical DNA damage pathway that resolves R/S. A BRCA1(+/-) carrier with PARPi sensitivity, indicative of HR deficiency, exhibited some of the common CNAs previously detected in BRCA1(-/-) breast tumors (1q gain and 16q loss) suggesting LOH in small fraction of cells. Overall, patient-derived organoid models allow for the evaluation of the BRCA1(+/-) pathologic phenotype and testing drugs for preventing the outgrowth of BRCA1(-/-) malignant clones. Acknowledgments We’d like to thank to patients for donating tissue, as well as Alicia Pollaci (CTO Sr Research Project Mgr) and Claire Capaldi (Research Lab Tech) for helping on the tissue collections. In addition, we would like to thank MacLean C. Sellars MD, PhD for the flow cytometry analysis.
Presentation numberPS3-01-26
Estrogen hormone replacement therapy (E-HRT) and Tamoxifen: Prevention versus Adjuvant setting
Joseph Ragaz, University of British Columbia, Vancouver, BC, Canada
J. Ragaz1, K. S. Wilson2, H. Wong1, H. Qian1, S. Shakeraneh3, J. J. Spinelli4; 1School of Population and Public Health, University of British Columbia, Vancouver, BC, CANADA, 2Division of Medical Oncology, BC Cancer Agency, Victoria, BC, CANADA, 3Infection Prevention and Control, Providence Health Care, Vancouver, BC, CANADA, 4Department of Statistics, BC Cancer Agency, Vancouver, BC, CANADA.
Objective: To determine the value of estrogen in overall management of breast cancer. Background: Despite the prior consensus of Estrogen [E] Breast Cancer [BrCa] harm, recent 20-year follow-up data from the double-blind randomized Women’s Health Initiative Trial 2 [WHI-2], show E-alone based Hormone Replacement Therapy [E-HRT] reducing BrCa incidence by 22%, and, unexpectedly, BrCa mortality by 40%.1 For comparison, the Tamoxifen randomized breast cancer prevention trial [IBIS-1] also reported a lower BrCa incidence [RR=0.71], but no mortality reduction [RR = 1.19, 95% CI].2,3 (Table 1). Comments: These prevention outcomes show both E-HRT and Tamoxifen equally effective in reducing incidence rates. The striking difference, however, is the significant mortality reduction with E-HRT, but not with Tamoxifen. Furthermore, E-HRT offers major benefits for quality-of-life [QOL] and for other rate reductions: Alzheimer dementia mortality: -26%; women age <60 at E-HRT start, both myocardial infarctions [-41%] and the all-cause mortality [-26%].3,4 In addition to these prevention benefit outcomes, estrogen emerged much more effective than Tamoxifen in stage IV BrCa [5 year overall survival: 35% vs 16%, adjusted p = 0.039].5 Conclusion: We believe the observed benefits of E-HRT* are sufficiently compelling to seek its guideline approval both in breast cancer prevention and in adjuvant settings, with or without anti-estrogens, with expectations of tens of thousands more avoided deaths per year, just in USA alone,4 while QOL is dramatically improved. *E-HRT: Estrogen alone for women with hysterectomy; Estrogen + progesterone for women with retained uterus References: 1. Chlebowski RT, Anderson GL, Aragaki AK, et al. Association of Menopausal Hormone Therapy With Breast Cancer Incidence and Mortality During Long-term Follow-up of the Women’s Health Initiative Randomized Clinical Trials. Jama. Jul 28 2020;324(4):369-380. 2. Cuzick J, Sestak I, Cawthorn S, et al. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. The Lancet Oncology. Jan 2015;16(1):67-75. 3. Manson JE, Aragaki AK, Rossouw JE, et al. Menopausal Hormone Therapy and Long-term All-Cause and Cause-Specific Mortality: The Women’s Health Initiative Randomized Trials. Jama. Sep 12 2017;318(10):927-938, Supplement. 4. Ragaz J, Shakeraneh S, Qian H, Wong H, Spinelli JJ, Wilson KS. Abstract PS7-05: Estrogen-based hormone replacement therapy [E-HRT] reduces all-cause, breast cancer, and Alzheimer’s dementia mortality. Cancer Research. 2021;81(4_Supplement):PS7-05-PS7-05. 5. Peethambaram PP, Ingle JN, Suman VJ, Hartmann LC, Loprinzi CL. Randomized trial of diethylstilbestrol vs. tamoxifen in postmenopausal women with metastatic breast cancer. An updated analysis. Breast cancer research and treatment. Mar 1999;54(2):117-22.
| BrCa outcomes |
E-HRT vs Placebo1,2 Hazards, 95% C.I. |
Tamoxifen vs Placebo3 Hazards, 95% C.I. |
|
Incidence |
0.78 [0.65-0.93] | 0.71 [0.60-0.83] |
|
Mortality |
0.60 [0.37-0.97] | 1.19 [0.68-2.10] |
Presentation numberPS3-01-27
Association between adherence to the mediterranean dietary pattern and PAM50-derived breast cancer proliferation and recurrence risk scores. Exploratory analysis from the Epigeicam study
Begoña Bermejo, Medicine Department, Hospital Clinico Universitario de Valencia, Instituto de Investigacion Sanitaria (INCLIVA), Universidad de Valencia; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Valencia, Spain
V. Lope1, P. Fernández1, Á. Guerrero-Zotano2, P. Sánchez-Rovira3, A. Antón-Torres4, M. Benavent-Viñuales5, J. Baena-Cañada6, S. Antolín7, M. Muñoz8, L. Paris9, J. Chacón10, C. Olier11, S. González12, J. García-Sáenz13, Á. Jimenez-Arranz14, A. Oltra15, J. Brunet16, M. Marin-Alcala17, A. De Juan18, B. Pérez-Gómez1, R. Rincón19, R. Caballero19, B. Bermejo20, M. Martín21, M. Pollan1; 1Environmental Epidemiology and Cancer Area, Unidad de Epidemiologia del Cancer y Ambiental, Departamento de Epidemiologia de Enfermedades Cronicas, Centro Nacional de Epidemiologia (CNE), Instituto de Salud Carlos III (ISCIII); GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 2Medical Oncology, Instituto Valenciano de Oncologia (IVO); GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 3Medical Oncology, Hospital Universitario de Jaén; GEICAM Spanish Breast Cancer Group, Jaén, SPAIN, 4Medical Oncology, Hospital Universitario Miguel Servet, Instituto Investigación Sanitaria Aragon, Departamento Medicina Universidad de Zaragoza; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 5Medical Oncology, Hospital Universitario Virgen del Rocio, Instituto de Biomedicina de Sevilla; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 6Medical Oncology, Instituto de Investigación e Innovación Biomédica de Cádiz; GEICAM Spanish Breast Cancer Group, Cádiz, SPAIN, 7Medical Oncology, Complejo Hospitalario de La Coruña (CHUAC); GEICAM Spanish Breast Cancer Group, A Coruña, SPAIN, 8Medical Oncology, Hospital Clinic i Provincial de Barcelona, FRCB-Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Department of Medicine, University of Barcelona; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 9Medical Oncology, Centro Oncológico de Galicia; GEICAM Spanish Breast Cancer Group, A Coruña, SPAIN, 10Medical Oncology, Hospital Univresitario de Toledo; GEICAM Spanish Breast Cancer Group, Toledo, SPAIN, 11Medical Oncology, Hospital Universitario Fundación Alcorcón, Universidad Rey Juan Carlos; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 12Medical Oncology, Research Foundation Mútua Terrassa/University of Barcelona, Hospital of Mútua Terrassa; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 13Medical Oncology, Department of Oncology and Instituto de Investigación Sanitaria Hospital Clinico San Carlos (IdISSC); GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 14Medical Oncology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC)-Hospital Universitario Reina Sofía, Universidad de Córdoba, Córdoba, SPAIN, 15Medical Oncology, Hospital Virgen de los Lirios; GEICAM Spanish Breast Cancer Group, Alcoy, SPAIN, 16Medical Oncology, Institut Catala d’Oncologia, Hospital Josep Trueta, Oncogir, IDIBGI; GEICAM Spanish Breast Cancer Group, Girona, SPAIN, 17Medical Oncology, Consorci Sanitari de Terrassa; GEICAM Spanish Breast Cancer Group, Terrassa, SPAIN, 18Medical Oncology, Hospital Universitario Marques de Valdecilla; GEICAM Spanish Breast Cancer Group, Santander, SPAIN, 19Translational, GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 20Medical Oncology, Medicine Department, Hospital Clinico Universitario de Valencia, Instituto de Investigacion Sanitaria (INCLIVA), Universidad de Valencia; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 21Medical Oncology, Department of Medical Oncology, Hospital General Universitario Gregorio Marañon Instituto de Investigacion Sanitaria Gregorio Marañon, Universidad Complutense; CIBERONC-ISCIII; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: There is growing evidence supporting the benefits of the Mediterranean diet in breast cancer (BC) prevention, but their contribution to prognosis remains unclear. Our objective is to investigate whether adherence to the Mediterranean dietary patterns before diagnosis is associated with the risk of BC recurrence and tumor proliferation in an exploratory cohort of the multicenter case-control EpiGEICAM study.Methods: Transcriptome-wide gene expression was assessed using the nCounter Breast Cancer 360 panel (NanoString). Formalin-fixed paraffin-embedded tumor tissues from an exploratory cohort of 89 pre- or perimenopausal BC patients from the EpiGEICAM study were used. The PAM50 gene expression signature was employed to classify tumors into the gold-standard BC intrinsic molecular subtypes, and to compute both the PAM50 risk of recurrence (ROR) and PAM50 proliferation scores. Dietary patterns over the five years prior to diagnosis were identified in the control population of the EpiGEICAM study, applying principal components analysis without rotation of the variance-covariance matrix over 26 inter-correlated food groups. The association between adherence to Mediterranean dietary patterns and both ROR and proliferation scores was analyzed using linear regression models with continuous outcomes. All models were adjusted for age and adherence to a Western dietary pattern, with additional adjustment for PAM50 molecular subtype in the proliferation score analysis. Results: The mean age was 44 years and 28% had a family history of BC. Regarding molecular subtypes, 49.4% corresponded to Luminal A, 22.5% to Luminal B, 13.5% to HER2-enriched and 14.6% to Basal-like subtype. High adherence to the Mediterranean dietary pattern was observed in 56% of participants, with higher prevalence among women with Luminal A and Luminal B subtypes. The mean ROR score was 51 (95%CI=46-57) in women with low adherence to the Mediterranean diet, and 46 (95%CI=40-51) in those with high adherence. Mean proliferation scores were 4.8 (95%CI=4.6-5.1) and 4.5 (95%CI=4.3-4.8), respectively. Although no statistically significant associations were observed, women with high adherence to the Mediterranean dietary pattern showed lower mean ROR score (β=-5.8; 95%CI=-14.0-2.5) and a reduced proliferation score (β=-0.14; 95%CI=-0.40-0.12) compared to those with low adherence. Conclusion: The present EpiGEICAM exploratory analysis showed that adherence to a Mediterranean dietary pattern prior to diagnosis may be associated with lower breast cancer PAM50 proliferation and risk of recurrence scores. Studies with larger sample sizes are needed to confirm these results and to clarify the effect of the Mediterranean diet on the biology and prognosis of this tumor.
Presentation numberPS3-01-28
Effects of 6 Months of Tirzepatide on Risk Biomarkers for Development of Breast Cancer
Carol J Fabian, University of Kansas Medical Center, Kansas City, KS
C. J. Fabian1, C. R. Beaver1, K. R. Powers1, L. Ranallo1, A. L. Kreutzjans1, K. Pittman1, C. Altman1, T. Metheny1, R. Biswell1, A. Zelenchuk1, H. Pathak2, A. Mitra2, A. K. Godwin2, D. P. Mudaranthakam3, E. D. Giles4, S. D. Hursting5, K. L. Cook6, B. F. Kimler7; 1Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 2Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 3Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 4Movement Science, University of Michigan, Ann Arbor, MI, 5Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 6Cancer Biology and Surgery, Wake Forest University School of Medicine, Winston-Salem, NC, 7Radiation Oncology, University of Kansas Medical Center, Kansas City, KS.
Background: Achieving and sustaining the ≥10% weight loss sufficient to reduce risk for breast cancer has been difficult with diet and exercise alone. Tirzepatide is a dual agonist of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptors. It is FDA-approved for obesity treatment with average weight loss of 14% at 24 weeks. Genetic variants of the GIP receptor (GIPR) that reduce GIPR signaling have been associated with increased breast cancer risk; however, little is known about the presence or localization of GIPR within breast tissue. High mammographic density, breast adipocyte size, breast epithelial Ki-67 ≥2%, and visceral fat of >1kg (iDXA) are all associated with increased risk of breast cancer. With the exception of BMI and insulin resistance, there is little information on the effect of tirzepatide on risk biomarkers for breast cancer. We sought to assess feasibility of a convenient clinical cohort trial model and gain preliminary information on biomarker modulation in a pilot study. Methods: Women with obesity and additional risk factors for breast cancer seen in the KUMC Weight Management Clinic (WMC) and Breast Cancer Prevention Clinic (BCPC), considering clinical use of tirzepatide, were approached for study participation. Procedures for assessment of risk biomarkers were performed by BCPC personnel before and after 6 months of tirzepatide. Inclusion criteria were age 35-64, BMI ≥30 kg/m2, ≥2x population risk for breast cancer, and self-pay or insurance coverage for tirzepatide. Exclusion criteria included prior invasive cancer or current insulin or antiestrogens. Prior to tirzepatide initiation, women underwent a fasting blood draw, 3D mammography, iDXA, random periareolar fine needle aspiration, and optional stool for microbiome. Tirzepatide was started at a dose of 2.5 mg/week, escalated every 4 weeks to a maximum of 15 mg under the guidance of the WMC. Key imaging risk biomarkers are absolute fibroglandular volume (FGV) via Volpara™ and visceral fat via iDXA. Tissue biomarkers performed in the BCPC or Biomarker Validation Core laboratories include Ki-67, GIPR, and estrogen response gene expression. Systemic markers include estradiol, estrone, SHBG, progesterone, insulin, adipocytokines, circulating microRNA, and Klotho. Breast adipocyte size is measured at the University of Michigan and microbiome analyzed at Wake Forest University. Objectives: 1) Feasibility as defined by ≥1 enrollee/month and >70% completion of study including biomarker procurement; 2) Proportion of women with detected GIPR in breast epithelium; and 3) Identification of reliably modulated biomarkers of breast cancer risk. Results: 23 women have been enrolled in 9 months with median age 49, BMI 35 kg/m2, IBIS 10-year breast cancer risk 6.3%, and visceral fat mass 1.7 kg. 13 are pre or perimenopausal and 10 postmenopausal. Of the 12 women completing study to date, median relative change in BMI is -15%, visceral fat -19%, lean mass -8%, FGV -22%, and HOMA-IR -31%. Median relative change in Ki-67 for the seven women with baseline Ki-67 ≥ 2% is -50%. Five of seven initial entrants have shown evidence at baseline of low, but detectable, levels of GIPR in breast epithelium by spatial genomics. Conclusions: Rapidity of recruitment, high retention, and completion of off-study biomarker assessments support expansion to a larger phase II trial where the directionality and magnitude of biomarker change can be evaluated with greater precision and confidence.
Presentation numberPS3-01-30
Change in mammographic density (MD) with metformin use: Alliance A211201, A companion study to NCIC study MA.32
Marie Wood, University of Colorado Anschutz Medical Campus, Aurora, CO
M. Wood1, M. I. Elsaid2, L. J. Grimm3, H. Liu4, C. Oswold5, C. S. Kuzma6, I. Bedrosian7, J. Ligibel8, P. J. Goodwin9; 1Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 2Biomedical Informatics and Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, 3Radiology, Duke University, Durham, NC, 4Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 5Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, 6Hematology/Oncology, First Health of the Carolinas, Pinehurst, NC, 7Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 8Medical Oncology, Dana-Farber Cancer Institute, Aurora, MA, 9Medical Oncology, University of Toronto, toronto, ON, CANADA.
Background: Metformin may impact tumorigenesis directly, through effects on AMP kinase, and indirectly, through reduction in circulating growth factors such as insulin. To examine the potential preventive effects of metformin, we analyzed MD, a known risk factor for breast cancer, in participants enrolled in the NCIC MA.32 trial, a randomized, placebo-controlled trial investigating the effects of 5 years of metformin on invasive disease-free survival in women with early breast cancer. The primary aim of this companion to MA.32 (Alliance A211201) was to examine the effects of 1 year of metformin vs placebo on MD in women treated for estrogen receptor negative (ER-) breast cancer. Methods: Eligible women were enrolled on MA.32, had ER- breast cancer, had >25% MD at baseline, and had an unaffected contralateral breast. MD was calculated on the contralateral breast using Cumulus software and analyzed at baseline, 1 and 2 years. The aim was to achieve a sample size of 274 (137 per arm), which would have at least 85% power to detect a 4% difference between study arms, i.e., 5% change from baseline to one year of treatment for metformin vs. 1% for placebo, using a two-sided two-sample t-test at a significance level of 5%. The current study was activated in the United States on 8/22/2012. Enrollment was halted on 10/13/2017 with 177 participants enrolled. Results: 146 women were evaluable (66 on metformin and 80 on placebo). The mean age of evaluable women was 53.6 years (standard deviation [SD] 9.4), 88.4% were white, 31.5% had a body mass index greater than or equal to 30 kg/m², 99.3% of participants received chemotherapy. The proportions of white women and HER2+ participants in the metformin group were higher compared to placebo ([white 92.4% vs. 85.0%] and [HER2+ 21.2% vs. 11.3%], respectively). The mean MD at baseline was 22.7% (SD 14.1%) for the group as a whole (24.5% (SD 14.5%) for the metformin group and 21.2% (SD 13.6%) for the placebo group. This is lower than the eligibility criteria (>25%MD), due to difference in methods of calculation of MD (BIRADs for eligibility and Cumulus for study analysis). At 1 year, MD decreased by 0.7% (SD 7.6%) in the metformin group and 0.2% (SD 7.3%) in the placebo group (p = 0.58). At 2 years, MD decreased by 0.7% (SD 8.0%) in the metformin group and 0.8% (SD 7.1%) in the placebo group (p=0.60). Conclusions: In a cohort of women with early ER- breast cancer, there was no significant difference in the change in MD in women treated with metformin vs. placebo. There was a non-significant trend toward decreased MD while on metformin after 1 year of treatment. The arms were well balanced, though with more white and HER2+ participants in the metformin arm. The sample size for this study was about half of what was intended thus limiting power to detect a significant difference. Additional limitations include low mean MD, which may have limited our ability to detect the benefit in those at the highest risk, and further, the Cumulus method of MD determination can be imprecise. Interestingly, trends toward decreased MD with metformin were seen in Alliance A211102, and it might be reasonable to consider further study of metformin in a select population (highest MD or atypical hyperplasia). Support: UG1CA189823, U10CA180863, CCS 707213; https://acknowledgments.alliancefound.org. Clinicaltrials.gov ID: NCT01666171.
Presentation numberPS3-02-01
Trends in the uptake of nipple-sparing mastectomy in BRCA1/2, PALB2 and other pathogenic variant carriers undergoing risk reducing mastectomy
Marya Alsuhaibani, McGill University, Montreal, QC, Canada
M. Alsuhaibani1, A. Melanson1, A. Viezel-Mathieu2, T. Dionisopoulos2, M. Basik1, J. Boileau1, M. Martel1, I. Prakash1, S. Meterissian1, J. Vorstenbosch2, W. Foulkes3, S. Wong1; 1General Surgery, McGill University, Montreal, QC, CANADA, 2Plastic Surgery, McGill University, Montreal, QC, CANADA, 3Oncology, McGill University, Montreal, QC, CANADA.
Introduction: Risk-reducing mastectomy (RRM) a well-established preventive intervention in germline pathogenic variants (GPVs) carriers. Historically, RRMs were performed as total mastectomy (TM) or skin-sparing mastectomy (SSM), although nipple-sparing mastectomy (NSM) has recently emerged as a RRM option. In this study, we assessed temporal trends in NSM uptake. Methods: This retrospective cohort study included all female patients with confirmed BRCA1, BRCA2, PALB2, or other GPVs carriers at two academic institutions in Montreal, Canada, between 2003 and 2024. The primary outcome was the proportion of GPV carriers NSM compared to SSM and TM over time as evaluated by Mantel Haenszel test for trend. Results: After exclusions, 441 GPV carriers were included in the cohort, 260 (59%) affected carriers and 181 (41%) unaffected carriers. The median age at RRM was 44 years old (range 22-76). Most were BRCA1 carriers (45.5%) or BRCA2 carriers (44.8%) while the remaining 9.8% were PALB2 and other GPV carriers. Overall, 269 (61%) underwent NSM, 118 (26.8%) underwent SSM, and 54 (12.2%) underwent TM. NSM uptake was highest among patients aged 40-49 years (68.6%) and lowest in those ≥60 years (45.8%, p=0.04). Relative to affected carriers, unaffected carriers were more likely to undergo NSM (72.4%, Vs.53.1% p<0.001). Prior radiation significantly decreased the likelihood of NSM (38.4% vs. 68.7%, p<0.001). A clear increase in NSM occurred over time, from 12.5% of GPV carriers pre-2012 to 77.5% after 2022 (p<0.001). Multivariable analysis adjusting for age, GPV, personal history of breast cancer and prior radiation identified year of surgery as the strongest predictor of NSM (OR 28.9, 95% CI 10.1-77.0) post-2019 compared to pre-2012 (p<0.001). Conclusion: There has been a significant increase in NSM uptake among GPV carriers over the last two decades. Understanding these trends supports informed decision-making and standardization of preventive care in high-risk populations.
Presentation numberPS3-02-02
Impact of COVID-19 on neoadjuvant chemotherapy efficacy in patients with breast cancer: an ambidirectional cohort study and Mendelian randomization analysis
Yali Wang, Fujian Medical University Union Hospital, Fuzhou, China
Y. Wang, J. Zhan, S. Liu, W. Cai, Y. Qiu, P. He, M. Chen, L. Chen, F. Fu, C. Wang; Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA.
Background: Patients with breast cancer are susceptible to coronavirus disease (COVID-19), which affects cancer treatment efficacy and prognosis. However, its effects on neoadjuvant chemotherapy (NAC) response and its genetic correlation with breast cancer remain unclear. We aimed to clarify these effects and investigate potential genetic associations and shared pathogenic mechanisms. Μethods: In this ambidirectional (retrospective and prospective) cohort study involving 856 breast cancer patients receiving NAC, with and without COVID-19, we evaluated the impact of COVID-19 on NAC response using multivariable logistic regression and three matching methods: propensity score matching, inverse probability of treatment weighting, and overlap weighting. Using single-cell and bulk transcriptome data from the Gene Expression Omnibus and genetic variants from the Genome-Wide Association Study databases, we conducted Mendelian randomization (MR) analysis to explore the core cellular subsets, shared genes, causal relationships, and common pathogenic mechanisms between COVID-19 and breast cancer. Results: Patients with breast cancer and COVID-19 showed poor NAC responses, including a low objective response rate and reduced likelihood of achieving RCB class 0-I in the HR+/HER2+ subgroup (OR=0.46, P=0.028, P for interaction=0.024). The consistent findings from the three matching models support these results. Integrating single-cell and bulk transcriptome data with MR analyses revealed genetic correlations between COVID-19 and breast cancer and identified ABLIM1 and GZMM as potential genes linked to NAC resistance. Conclusions: SARS-CoV-2 infection during NAC may compromise therapeutic efficacy in patients with breast cancer. ABLIM1 and GZMM may be causal genes in the genetic correlation between COVID-19 and breast cancer.
Presentation numberPS3-02-04
Omitting Oncotype Dx Genomic Testing in Ultra Low Risk Breast Cancer
Megha Schmalzle, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY
M. Schmalzle1, R. Radigan1, S. Singh2, W. Liu3, K. Deng2, M. Fabrikant2, R. Shah4, S. L. Fu2, J. Kao4; 1Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 2The Cancer Institute, Good Samaritan University Hospital, West Islip, NY, 3Department of Pathology, Good Samaritan University Hospital, Hauppauge, NY, 4Department of Radiation Oncology, Good Samaritan University Hospital, West Islip, NY.
Introduction: Older women with early-stage luminal A breast cancer tend to have an excellent prognosis with a 5-year relative survival exceeding 99%. Ongoing research on curtailing overdiagnosis and overtreatment have challenged long established standards of care by reducing the use of whole breast radiotherapy and selectively omitting sentinel lymph node biopsy. In light of emerging data suggesting that endocrine therapy can be safely omitted in older women with ultra-low risk luminal A breast cancer, the marginal utility of Oncotype Dx genomic testing was explored. Materials and Methods: We queried the Good Samaritan University Hospital Cancer Registry to identify consecutive breast cancer patients treated from 2014 through 2022. Based on extensive research on radiation omission and selecting patients for partial breast irradiation, we defined an ultra-low risk breast cancer population as age ≥60 years, estrogen receptor ≥1% positive, progesterone receptor >20% positive, HER2-negative, pathologic stage T1b-T2N0, well to moderately differentiated, no lymphovascular invasion, and Ki-67 ≤13.25%. The primary endpoint was the proportion of patients with low (0-18) or intermediate (19-25) risk Oncotype scores, for which chemotherapy is not indicated. Secondary endpoints included local recurrence and overall survival. Results: Out of 1711 patients, 91 met all ultra-low risk eligibility criteria. Median age was 70 years (IQR 65-74; range 60-85) and median breast tumor size was 10 mm (IQR 7-15). Most patients were diagnosed with invasive ductal carcinoma (71.4%) or lobular carcinoma (16.5%). All patients underwent surgical intervention, with 79.1% receiving a lumpectomy and 20.9% receiving a mastectomy. Sixty-nine (75.8%) patients received post-operative radiotherapy and no patients received chemotherapy. The median follow-up was 5.14 years. Only 1 of 91 patients (1.1%) had a high risk Oncotype Dx score of 26, while 12 (13.2%) had an Oncotype Dx score of 19 to 25 and 78 (85.7%) scored 0 to 18. Five-year local control and overall survival was 100% and 93%, respectively. Nine deaths occurred, none related to breast cancer, and no patients developed distant metastases. Conclusion: We describe a pragmatic method of using clinical and pathological features routinely available in a community hospital breast program to define an ultra-low risk cohort. Older luminal A patients with favorable pathology have a very low probability of a high Oncotype Dx genomic score and have an excellent prognosis with standard adjuvant therapy. Future research for ultra-low risk breast cancer patients should focus on treatment de-escalation beyond radiotherapy.
Presentation numberPS3-02-05
Shared Genetic Architecture between Breast Cancer and Psychiatric Disorders: Insights from Large-Scale Genome-wide Cross-trait Analysis
Canzhou Wang, First Hospital of Shanxi Medical University, Taiyuan, China
C. Wang1, L. Jing1, Y. Lei1, Y. Zhang1, X. Wang1, Y. Wang2, H. Jia1; 1Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, CHINA, 2Department of Basic Medicine, Shanxi Medical University, Taiyuan, CHINA.
Background: Breast cancer (BC) has been associated with psychiatric disorders. However, the shared genetic determinants underlying these associations remain unevaluated. This study aims to investigate the shared genetic architecture between psychiatric disorders and BC, and to identify the involved shared risk loci, potential key tissues, and underlying genetic mechanisms.Methods: Based on the summary statistics from genome-wide association studies (GWAS), the genetic correlation and overlap between psychiatric disorders and BC were investigated. Furthermore, shared pleiotropic loci and genes were identified through cross-trait analyses. Additionally, a series of functional annotations and tissue-specific enrichments were performed to determine potential associations between each trait pair. Pathway enrichment analysis was conducted to assess possible associated mechanisms. Finally, the causal relationship at the protein level was also evaluated.Results: The results demonstrated shared genetic mechanisms between eight psychiatric disorders and two BC subtypes. After Bonferroni correction,4 of the 16 trait pairs exhibited significant genetic correlations (p < 3.13 × 10⁻³). A total of 7,274 SNPs were identified at a genome-wide significance threshold of p < 5. 00 × 10⁻⁸ threshold. Furthermore, annotation analysis identified 156 genomic risk loci, of which 19 were analyzed using causal colocalization. Gene-level analysis revealed a series of pleiotropic genes, with MTRFA and FGFR2 being identified in the majority of trait pairs. Tissue enrichment analysis indicated that pleiotropic mechanisms play a significant role in the ovary, breast mammary tissue, uterus, and certain brain regions, including the caudate nucleus, cerebellar hemisphere, and putamen. Pathway analysis showed that most trait pairs were enriched for positive regulation of RNA metabolism. Additionally, protein-level causal correlation analysis identified 5 proteins significantly associated with BC risk and 36 proteins significantly associated with the risk of psychiatric disorders.Conclusion: This study explored the shared genetic architecture between psychiatric disorders and BC and identified several potential genetic mechanisms that are critical for the development of novel therapeutic strategies in clinical practice.
Presentation numberPS3-02-06
Stromal‑Fibroblastic remodelling and involution signatures in non‑tumoral tissue of postpartum breast cancer patients: implications for breast cancer risk
Juan de la Haba-Rodríguez, Reina Sofía University Hospital, Córdoba, Spain
J. de la Haba-Rodríguez1, R. Peña-Enríquez2, D. Ponferrada1, R. Bautista-Moreno3, S. Romero-Martín4, E. Contreras4, A. Díaz-Chacón2, C. Morales1, B. Rodríguez1, P. Sanchez-Maurinho1, A. Martinez-López5, C. León6, J. Cejas-Arjona6, P. Notario-Fernández6, P. Rioja-Torres6, J. Raya-Povedano4, M. Álvarez4, S. Guil-Luna2; 1Medical Oncology Department, Reina Sofía University Hospital, Córdoba, SPAIN, 2New therapies in cancer group (CG06), Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), University of Córdoba, Córdoba, SPAIN, 3Bioinformatics Unit, Supercomputing and Bioinformatics Centre (SCBI), University of Malaga, Malaga, SPAIN, 4Radiodiagnosis Department, Reina Sofía University Hospital, Córdoba, SPAIN, 5Anatomical Pathology Department, Reina Sofía University Hospital, Córdoba, SPAIN, 6Breast Surgery Department, Reina Sofía University Hospital, Córdoba, SPAIN.
Background: Breast cancer (BC) in young women under 45 is a growing public health concern, with around 50% of cases occurring in mothers within 5-10 years after childbirth. This entity, known as postpartum breast cancer (PPBC), is characterised by aggressive clinical behaviour and higher metastasis and mortality rates compared to age-matched BC patients. Despite its recognised poor prognosis, the molecular mechanisms driving PPBC remain poorly understood, and this knowledge gap limits the ability to identify women at higher risk and develop prevention strategies. This study aimed to characterize the transcriptomic alterations in both tumoral and histologically-normal adjacent breast tissue from PPBC patients, compared to non-PPBC and healthy controls, with a focus on identifying early risk signatures. Methods: RNA-seq analysis was performed on fresh breast tissue samples collected from 30 young women (aged ≤ 45 years) at Reina Sofia University Hospital of Córdoba: paired tumor and adjacent normal tissue from PPBC patients (n=12), tumor tissue from non-PPBC patients (n=8), and normal breast tissue from healthy nulliparous/parous women (n=10). Transcriptomic data were analysed for differential gene expression using DESeq2, and pathway enrichment analysis was performed using GSEA querying the Hallmarks (MSigDB) and 100 customized gene sets. Significantly enriched pathways were identified based on FDR q-values 1.5 and FDR p-value <0.05. The shared overexpressed genes in PPBC tumour and adjacent tissue versus healthy controls were evaluated as potential biomarkers using multivariate ROC analysis. Results: PPBC tumors exhibited a distinct, aggressive profile compared to non-PPBC patients with significant enrichment of cell cycle activation pathways (E2F, MYC targets and G2M checkpoint), reflecting high proliferative activity, as well as dysregulation of the WNT/β-catenin signalling pathway (FDR q-value < 0.1). Notably, adjacent histologically-normal tissue of PPBC patients also exhibited a unique transcriptomic profile, distinct from healthy breast tissue. This included enrichment of stromal-fibroblastic remodeling signatures, such as TGF-β signaling, epithelial-mesenchymal transition (EMT), and markers related with cancer-associated fibroblasts (CAFs) and endothelial cells. Interestingly, a robust post-lactational involution-like signature, characterized by upregulation of genes linked to mammary gland regression, late parity, and a transient postpartum immune state (FDR qvalue 0.9). These genes were predominantly involved in pathways related to profibrotic and cytokine signaling, with top gene markers such as SOCS4, TMED7 and DYNLT3. Conclusions: Our findings reveal that molecular alterations in PPBC extend beyond the tumor itself, implicating the surrounding normal tissue in a tumor-promoting, stromal fibroblastic remodeling and post-lactational microenvironment. This transcriptomic profile supports the hypothesis that PPBC may develop within a mammary microenvironment already altered by post-gestational involution and stromal processes that could favour normal cells to malignancy. These insights highlight the significant potential for improving risk stratification and guiding early intervention strategies in postpartum women.
Presentation numberPS3-02-07
Co-occurring pathogenic variants in patients with breast cancer.
Kallie Woods, Myriad Genetics, Inc., Salt Lake City, UT
K. Woods1, S. Cummings1, F. Ademuyiwa2; 1Oncology Medical Affairs, Myriad Genetics, Inc., Salt Lake City, UT, 2Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO.
Background: Numerous studies have examined germline variant rates in individuals with breast cancer (BC), consistently reporting that about 5-10% carry a germline pathogenic variant (PV). Although multiple PVs in a single patient have been documented, such cases are rare and reported in small cohorts. As a result, the clinical implications of multiple PVs remain poorly understood. To address this gap we analyzed a large dataset of patients with BC who underwent germline genetic testing. Methods: The Myriad Collaborative Research Registry, containing clinical, genetic, and genomic information on over 1.2 million individuals with cancer, was queried. The registry includes data on 795,165 patients with a BC diagnosis who completed testing at a commercial laboratory between 1996 and 2025. Data was assessed using descriptive statistics to understand the prevalence and characteristics of patients with multiple PVs. Results: Of the 795,165 patients with BC, 75,491 (9.5%) patients had one PV, 1,600 (0.2%) carried two PVs, and 34 patients carried more than 2 PVs. Of the 1,600 patients with two PVs, the most common second primary diagnoses included ovarian cancer (84), colon adenomas/polyps (68), breast cancer (45), and endometrial cancer (39). In patients with two PVs, the median age at cancer diagnosis was 49 years; most patients were female (1,561) and White/Non-Hispanic (1,197). Patients with two PVs were grouped by PV risk categories in BC: high-risk, moderate-risk, and other-risk. See Table 1 for patient groupings based on risk categories. The largest subgroup (28.3%) had one high-risk and one other-risk PV. The median age at diagnosis was 48 years across most groups, increasing to 54 in the other/other-risk group. BC was the most reported family history across all groups. In groups with at least one high-risk PV, ovarian cancer was the second most reported family history. Colon cancer was the second most reported family history in all groups with at least one other-risk PV. BRCA1/BRCA2 made up 73.7% of pairings in the high/high-risk group, and ATM/CHEK2 accounted for 69.2% in the moderate/moderate-risk group. The frequency of these pairs was markedly higher than the highest frequency pairings in other groups. Notably, one-third of all patients with two PVs had a PV in MUTYH. Conclusions: Assessing patients with two germline PVs presents clinical and interpretive challenges. This study identified two PVs at a frequency of 1,600 in 795,165 (0.2%) patients with BC and highlights patterns suggesting that detailed personal and family history may aid in understanding these complex genetic profiles. Continued data collection is essential to identify subtle trends and improve risk assessment. Future research is needed on specific gene pairings to clarify phenotypic differences and refine risk stratification. The registry enabled identification of a large cohort with two PVs, laying a foundation for deeper investigation into their clinical significance.
| Cohort Type |
Higha/
Higha Risk Gene
|
Higha/
Moderateb Risk Gene
|
Higha/
Otherc Risk Gene
|
Moderateb/
Moderateb Risk Gene
|
Moderateb/
Otherc Risk Gene
|
Otherc/
Otherc Risk Gene
|
| Number of Patients |
198
(12.4%)
|
279
(17.4%)
|
453
(28.3%)
|
91
(5.7%)
|
386
(24.1%)
|
193
(12.1%)
|
| Median Age of Cancer Diagnosis | 45 | 48 | 48 | 48 | 52 | 54 |
| Cancer Diagnoses or Polyp History Findings | Totald: 245 | Totald: 343 | Totald: 573 | Totald: 111 | Totald: 509 | Totald: 266 |
| Breast: 205 | Breast: 286 | Breast: 464 | Breast: 96 | Breast: 399 | Breast: 195 | |
| Ovary: 23 | Ovary: 14 | Ovary: 25 | Colon: 3 | Colon polyps: 29 | Endometrial: 11 | |
| Endometrial: 2 | Colon polyps: 9 | Colon polyps: 18 | Ovary: 3 | Melanoma: 13 | Colon: 10 | |
| Colon Polyps: 2 | Endometrial: 3 | Endometrial: 13 | Pancreas: 2 | Colon: 11 | Colon polyps: 9 | |
| Common Gene Pairings |
BRCA1/
BRCA2: 146 (73.7%)
|
BRCA2/
CHEK2: 67 (24.0%)
|
MUTYH/
BRCA2: 93 (20.5%)
|
ATM/
CHEK2: 63 (69.2%)
|
MUTYH/
CHEK2: 99 (25.6%)
|
MUTYH/
BRIP1: 19 (9.8%)
|
|
BRCA2/
PALB2: 12 (6.1%)
|
PALB2/
CHEK2: 43 (15.4%)
|
MUTYH/
BRCA1: 73 (16.1%)
|
ATM/
BARD1: 6 (6.6%)
|
MUTYH/
ATM: 60 (15.5%)
|
MUTYH/
MITF: 13 (6.7%)
|
|
|
BRCA1/
PALB2: 10 (5.1%)
|
BRCA1/
CHEK2: 36 (12.9%)
|
MUTYH/
PALB2: 43 (9.5%)
|
CHEK2/
RAD51C: 6 (6.6%)
|
CHEK2/
BRIP1: 16 (4.1%)
|
MUTYH/
NTHL1: 13 (6.7%)
|
|
| Family History Reported | All Cancer: 638 | All Cancer: 970 | All Cancer: 1571 | All Cancer: 312 | All Cancer: 1340 | All Cancer: 616 |
| Breast: 315 | Breast: 367 | Breast: 614 | Breast: 111 | Breast: 378 | Breast: 181 | |
| Ovary: 71 | Ovary: 75 | Colon: 139 | Prostate: 30 | Colon: 162 | Colon: 88 | |
| Prostate: 33 | Prostate: 69 | Prostate: 122 | Lung: 26 | Lung: 108 | Lung: 54 | |
| Pancreas: 28 | Colon: 54 | Ovary: 118 | Colon: 25 | Prostate: 99 | Prostate: 39 | |
| No Cancer: 26 | Lung: 51 | Lung: 100 | Pancreas: 18 | Ovary: 66 | Ovary: 25 | |
| Legend: | aHigh-Risk BC PVs – BRCA1, BRCA2, CDH1, PALB2, PTEN, STK11, TP53 | bModerate Risk BC PVs – ATM, BARD1, CHEK2, RAD51C, RAD51D | cOther Risk PVs – APC, BRIP1, CDKN2A P14ARF, CDKN2A P16, CTNNA1, EGFR, EPCAM, FH, FLCN, HOXB13, MITF, MLH1, MSH2, MSH3, MSH6, MUTYH, NTHL1, PMS2, RET, SDHA, SDHB, SDHC, SDHD, SMAD4, TERT, TSC2, VHL | dTotal number of diagnoses of cancer in the patient group |
Presentation numberPS3-02-08
Medium-chain fatty acid exposure in non-transformed mammary glands leads to pro-tumorigenic alterations associated with aging
Mariana Bustamante Eduardo, Northwestern University, Chicago, IL
M. Bustamante Eduardo1, A. B. Islam2, M. Zappia2, A. Z. Samuel3, C. W. McCloskey4, M. V. Frolov2, R. W. Khokha4, E. V. Benevolenskaya2, S. A. Khan1, S. E. Clare1; 1Department of Surgery, Northwestern University, Chicago, IL, 2Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, 3Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 4Department of Medical Biophysics, Princess Margaret Cancer Centre, Toronto, ON, CANADA.
Introduction. The local breast environment is an important source for identifying the etiological and biological factors that contribute to the development of breast cancer (BC). We identified a lipid metabolism gene signature associated with Estrogen Receptor negative BC (ERnegBC) in high-risk benign breast tissue. We then exposed non-transformed breast cells and breast microstructures in vitro and ex vivo to fatty acids (FA) and observed a metabolic shift toward the serine, one-carbon, glycine and methionine pathways, engendering epigenetic plasticity, increasing reactive oxygen species (ROS), and promoting cell survival. This promted the question: are there in vivo context(s) where lipid alterations resemble those of FA exposure in vitro/ex vivo? Since epigenetic plasticity and increased ROS are also reported in the aged mammary gland, we hypothesize that FA-induced metabolic reprogramming increases in the aged mammary gland, contributing to pro-tumorigenic alterations observed during aging. Methods. MCF-10A cells were exposed to the medium-chain FA octanoic acid (OA) for proteomics. Breast tissue-derived microstructures exposed to ± OA were utilized for single-cell RNA-seq. Breast microstructures and 3D mammary spheres (formed from mammary cells in Matrigel) were exposed to ± OA for 7 days, stained for luminal/basal markers, F-actin, and nuclei, and imaged using confocal microscopy. Raman spectroscopy was used to characterize the lipid content in pre- versus post-menopausal breast tissue. Results. In vitro and ex vivo data on FA exposure in non-transformed mammary cells and breast microstructures revealed striking similarities to the aged mammary gland: 1) Significant (p < 0.01) aging-related changes in gene expression upon OA exposure in epithelial and non-epithelial compartments. Notably, up-regulation of GDF15 (related to EMT, invasion, lipolysis and aging), MDK (involved in neurogenesis, cancer progression, and aging) and the downregulation of MMP7, a gene for which reduced expression contributes to the accelerated aging of mammary epithelial cells. Moreover, OA led to significant downregulation (p < 0.01) of lineage markers. 2) Significant (p < 0.01) upregulation of components of the Senescence-Associated Secretory Phenotype in both epithelial and non-epithelial subtypes. Including those that promote reprogramming (AREG), cancer progression (EREG), EMT (CCL20) and migration (ANGPTL4). 3) Altered inter-cellular communications upon OA exposure. Cell-cell communication analysis indicated increased secreted signaling in OA, with AREG, GDF15, and MDK among the strongest signals. 4) Decreased cell-cell junctions and extracellular matrix-receptor (ECM-R) interactions. Differential proteomics revealed downregulation of proteins involved in ECM-R interaction and focal adhesion. Cell-cell communication analysis also revealed decreased ECM-R interactions following OA exposure, alongside reduced cell-cell adhesions. 5) OA led to aging-associated pro-tumorigenic changes that may increase susceptibility to BC: Ex vivo culture of breast microstructures and 3D mammary spheres revealed that OA exposure alters cell-cell adhesions, compromising the basal barrier and leading to luminal cell dissemination. Conclusions. The proportion of breast adipocytes releasing free FAs increases with age. We show that FA exposure causes changes typical of the aged mammary gland, such as elevated ROS, epigenetic alterations, downregulation of cell-cell junctions, altered ECM-receptor interactions, and aging associated gene changes. We propose that the release of free FAs from an increased number of breast adipocytes (potentially due to elevated GDF15-induced lipolysis during aging) is a factor that induces mammary gland remodeling, increasing vulnerability to BC.
Presentation numberPS3-02-09
Recruitment Strategy Success and Challenges in WISDOM 1.0: A Nationwide Risk-Based Breast Cancer Screening Trial
Allison Stover Fiscalini, UCSF, San Francisco, CA
A. S. Fiscalini1, T. Glatt2, S. Goodman1, C. Kaplan3, A. Z. LaCroix4, L. van ‘t Veer5, M. Scheuner6, P. Sales7, A. Petruse8, A. Naeim9, A. Kaster10, J. Wernisch11, O. I. Olopade12, B. Gonzales13, H. L. Park14, A. D. Borowsky15, S. A. Raouf15, J. Atamer1, K. Leggat-Barr16, J. Esserman17, I. Cabaleiro17, R. Lancaster18, L. Anderson19, D. Heditsian20, S. Brain20, V. Lee20, D. Moorehead21, B. A. Parker22, A. Torres1, L. Sabacan1, L. Johansen8, X. Calderon23, A. Rocha8, A. Verma24, J. Cover25, T. Miller26, M. Che27, N. Kim28, T. Lewis27, S. Kapoor1, R. Soonavala29, D. Goodman-Gruen30, S. D. Stewart31, R. A. Hiatt32, N. Wenger33, H. Anton-Culver34, I. A. Soto1, M. Eklund35, K. F. Rhoads36, L. J. Esserman1; 1Surgery, UCSF, San Francisco, CA, 2School of Medicine, Tufts University, Boston, MA, 3Division of General Internal Medicine, Department of Medicine, UCSF, San Francisco, CA, 4Family and Preventive Medicine, UCSD, San Diego, CA, 5Lab Medicine, UCSF, San Francisco, CA, 6Department of Medicine, UCSF, San Francisco, CA, 7Medicine Service, Hematology-Oncology Section, San Francisco VA Health Care System, San Francisco, CA, 8Clinical and Translational Science Institute Office of Clinical Research, UCLA, Los Angeles, CA, 9Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, San Francisco, CA, 10Edith Sanford Breast Center, Sanford Health, Fargo, ND, 11Edith Sanford Breast Center, Sanford Health, Sioux Falls, SD, 12Surgery, University of Chicago, Chicago, IL, 13Department of Radiology, University of Chicago, Chicago, IL, 14Department of Pathology and Laboratory Medicine, UCI, Irvine, CA, 15Pathology, UC Davis, Sacramento, CA, 16School of Medicine, Tufts, Boston, MA, 17OB-GYN, Diagnostic Center of Miami, Miami, FL, 18Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 19Division of General and Internal Medicine and Population Sciences, University of Alabama at Birmingham, Birmingham, AL, 20Breast Science Advocacy Core, Breast Oncology Program, UCSF, San Francisco, CA, 21Community Engagement, Women’s Cancer Resource Center, Berkeley, CA, 22Moores Cancer Center, UCSD, San Francisco, CA, 23Surgery, UCSF, Los Angeles, CA, 24Pritzker School of Medicine, University of Chicago, Chicago, IL, 25Research Services, Northern California Institute for Research and Education, San Francisco, CA, 26Department of Veteran’s Affairs, San Francisco VA Health Care System, San Francisco, CA, 27School of Medicine, UCSF, San Francisco, CA, 28School of Medicine, Georgetown University, Washington, DC, 29David Geffen School of Medicine, UCLA, San Francisco, CA, 30Department of Epidemiology and Biostatistics, UC Irvine, Irvine, CA, 31Department of Pathology, School of Medicine, UC Irvine, Sacramento, CA, 32Helen Diller Family Comprehensive Cancer Center, Department of Epidemiology & Biostatistics, UCSF, San Francisco, CA, 33Division of General Internal Medicine and Health Services Research, UCLA, Los Angeles, CA, 34Department of Medicine, Genetic Epidemiology Research Institute, UCI, Irvine, CA, 35Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SWEDEN, 36Helen Diller Family Comprehensive Cancer Center, UCSF, San Francisco, CA.
BackgroundThe Women Informed to Screen Depending on Measures of risk (WISDOM) Study 1.0 (2016-2023; NCT02620852) is testing a novel, personalized approach to breast cancer screening compared to annual mammography. As a large-scale screening trial, WISDOM aimed to enrolled individuals ages 40-74 with no personal history of breast cancer using a preference tolerant randomized trial design that aimed to reflect the heterogeneity of the U.S. population. The goal of this analysis is to evaluate the recruitment strategies employed during WISDOM 1.0 and assess their effectiveness in building a geographically and racially diverse cohort of U.S. women for breast cancer screening research.MethodsWe analyzed enrollment and recruitment data sources including participant-reported responses to the ‘How You Heard’ (HYH) survey question administered at enrollment. Recruitment channels were grouped into categories (e.g., email from doctor, flyer/brochure, social media, friend/family referral), and stratified analyses were performed by recruitment site and self-reported race/ethnicity. We conducted analyses to evaluate volume and proportion of participants recruited by each strategy, rates by outreach method over time, differences by geography and participant demographics. ResultsBetween August 2016 and February 2023, WISDOM 1.0 registered 70575 women, consented 55284, and fully enrolled 46289 participants. Initial recruitment was limited to the Athena Breast Health Network within the University of California system (2016 -2017) but expanded through collaborations with external institutions and health systems and later to nationwide virtual enrollment.The recruitment method that yielded the most participants during the trial was emails from medical centers, MyChart, and physicians (40% of total enrollment). This was followed by family/friend referrals (10%) and emails from a Veteran’s Affairs (VA) recruitment partnership (6%). During the VA outreach, this strategy accounted for 25% of enrollment driving upward spikes in participation particularly in women of color. Other strategies included health insurance plan (4%), news article/radio/TV (4.5%) and social media (2.5%). WISDOM’s cohort diversity improved over the course of the trial, led by efforts from our Community Leadership Advisory Board, new recruitment centers, nationwide collaborations, and partnership with the VA. Over the course of the trial, there was a reduction in white, Non-Hispanic participants from 83% pre-community engagement efforts to 60% after new strategies were implemented (average 75%). The VA contributed to 26% of Black/African American participant enrollment, 10% of Hispanic participants, and 9% each for American Indian/Alaska Native (AI/AN) and Native Hawaiian/Pacific Islanders (NH/PI). Family/friend referrals accounted for over 7% of NH/PI but less than 4% for Black/AA, and only 1.5% for AI/AN. NH/PI participants were less likely to report physician/medical center (31%) and had the highest proportion of other options such as community organizations (9%) and health insurance plan (9%).DiscussionWISDOM 1.0 demonstrated the feasibility of recruiting a large, nationwide cohort for a personalized breast cancer screening trial using multi-modal, decentralized outreach. Physician emails and MyChart emerged as the highest-yield strategy, leveraging pre-existing patient trust and communication channels. The VA collaboration stands out as a highly scalable and demographically impactful approach while friend/family referrals offered downstream enrollment benefits. WISDOM 2.0 launched in June 2023 leveraging WISDOM 1.0’s recruitment approaches with an emphasis on refining high-yield, low-barrier strategies, amplifying trusted voices, and integrating culturally responsive approaches.
Presentation numberPS3-02-10
Pathologic Complete Response in Germline Pathogenic Variant Carriers: a Comparison Study Between a Safety-Net and Academic Population
Ethan Low, Baylor College of Medicine, Houston, TX
E. Low, N. Nyamongo, A. Lackan, C. Gonzalez, K. Hoffman, N. Kunta, S. Sarlin, S. Bulsara, S. Hilsenbeck, T. Snow, C. Sullivan, G. Garza, A. Elkhanany, N. Chen, K. Osborne, M. F. Rimawi, S. Shah, J. Nangia; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX.
Background: Pathologic complete response (pCR) after neoadjuvant chemotherapy in breast cancer (BC) has important prognostic implications. Understanding its associations with pathogenic/likely pathogenic variants (P/LPV) in hereditary cancer genes (HCG) adds further prognostic value. Minorities remain underrepresented in hereditary cancer research. In this retrospective chart review, we compare pCR rates among patients with P/LPV in HCG at a safety-net clinic comprised of majority Latino patients to those at an academic clinic with majority white patients. Methods: All patients at Baylor St. Luke’s Medical Center (BSLMC, academic clinic) and Smith Clinic (SC, safety-net clinic) with a diagnosis of stage I-III BC who were found to have a P/LPV in HCG from 10/1/09-9/30/19 and received neoadjuvant chemotherapy followed by surgery were included. Demographic, clinical characteristics, and pathologic data were collected. Results: 118 patients qualified for analysis (71 from SC, 47 from BSLMC). In SC 50 (70%) were Latino, 3 (4.2%) White, 11 (15.5%) Black, and 1 (1.4%) Asian. In BSLMC, 15 (32%) were Latino, 20 (42.6%) White, 8 (17%) Black, and 2 (4.3%) Asian. Median age was 42 for SC and 54 for BSLMC. SC comprised of 41 (58%) BRCA1+, 13 (18.3%) BRCA2+, and 12 (16.9%) PALB2+ patients. BSLMC comprised of 16 (34%) BRCA1+, 8 (17%) BRCA2+, 6 (12.8%) ATM+, 5 (10.6%) CHEK2, and 3 (6.4%) PALB2+. SC and BSLMC patients ranged from stage I-III, with 49% and 36.2% falling in stage III respectively. SC had 37 (52.1%) triple negative BC (TNBC) patients, 25 (35.2%) ER/PR+, HER2- (HR+), 7 (9.9%) ER+/HER2+, and 2 (2.8%) ER-/HER2+. BSLMC had 14 (29.8%) TNBC, 25 (53.2%) HR+, 7 (14.9%) ER+/HER2+, and 1 (2.1%) ER-/HER2+. 36 (51%) and 21 (44.7%) were treated with both anthracycline and taxanes in the SC and BSLMC populations respectively. In the SC population, pCR rates (detailed in Table 1) were highest among TNBC (16, 43.2%). Of the TNBC patients that had pCR, 15 (94%) had P/LPV in a high-risk HCG (BRCA1/2 or PALB2). Similarly, of the HR+ patients that had pCR, 6 (85%) had P/LPV in a high-risk HCG. In the BSLMC population, pCR was achieved in 4 (28.6%) TNBC patients. Of these patients, 4 (100%) carried P/LPV in a high-risk HCG. Of the HR+ patients that had pCR, 4 (80%) had P/LPV in a high-risk HCG. RCB class by hereditary P/LPV and biomarker status will be reported. Conclusions: Rates of P/LPV in high-risk HCG (BRCA 1/2 and PALB2) were higher in the SC population compared to BSLMC, which may be reflective of differences in ethnicities. Across both institutions, pCR rates were highest among patients with high-risk P/LPV, detailed in Table 1. When further divided by biomarker, HR+ patients had relatively high pCR rates. More data in patients with P/LPV in HCG may help us better determine prognosis and optimize treatment strategies. Given the high pCR rate in high-risk HCG, consider germline genetic testing in HR+ BC patients with pCR.
| Safety-net | Clinic | Academic | Clinic | |||
| Genes | pCR (n) | pCR% | pCR (n) | pCR% | ||
| ER-PR-HER2- | 16 (37) | 43.2% | 4 (14) | 28.6% | ||
| BRCA1 | 10 (25) | 40% | 4 (10) | 40% | ||
| BRCA2 | 3 (6) | 50% | 0 (0) | 0% | ||
| PALB2 | 2 (4) | 50% | 0 (0) | 0% | ||
| ATM | 0 (0) | 0% | 0 (0) | 0% | ||
| CHEK2 | 0 (0) | 0% | 0 (1) | 0% | ||
| Other | 1 (2) | 50% | 0 (4) | 0% | ||
| ER+ and/or PR+ (HER2-) | 7 (25) | 28% | 5 (25) | 20% | ||
| BRCA1 | 3 (12) | 25% | 3 (4) | 75% | ||
| BRCA2 | 2 (6) | 33.3% | 0 (5) | 0% | ||
| PALB2 | 1 (5) | 20% | 1 (3) | 33.3% | ||
| ATM | 0 (0) | 0% | 1 (5) | 20% | ||
| CHEK2 | 0 (1) | 0% | 0 (4) | 0% | ||
| Other | 1 (1) | 100% | 0 (7) | 0% | ||
| HER2+ | 1 (9) | 11.1% | 4 (8) | 50% | ||
| BRCA1 | 0 (3) | 0% | 0 (1) | 0% | ||
| BRCA2 | 0 (2) | 0% | 2 (3) | 66.7% | ||
| PALB2 | 1(3) | 33.3% | 0 (0) | 0% | ||
| ATM | 0 (0) | 0% | 0 (1) | 0% | ||
| CHEK2 | 0 (0) | 0% | 0 (0) | 0% | ||
| Other | 0(1) | 0% | 2 (3) | 66.7% |
Presentation numberPS3-02-11
Co-segregation of XAF1-E134and TP53-R337H in a brazilian cohort: clinical impact of a high-risk haplotype
Elisângella Paula Silveira-Lacerda, Universidade Federal de Goiás, Goiânia, Brazil
K. M. Veiga1, P. F. Silva2, W. D. Oliveira;3, D. C. Maia4, R. M. Rahal5, E. P. Silveira-Lacerda6; 1Centro de Genetica Humana – ICB, Universidade Federal de Goiás, Goiânia, BRAZIL, 2Centro de Genética Humana – CEGH – ICB, Universidade Federal de Goiás, Goiânia, BRAZIL, 3Genética, Hospital das CLínicas, Universidade Federal de Goiás, Goiânia, BRAZIL, 4Oncologia, Universidade Federal do Ceará, Fortaleza, BRAZIL, 5CORA, Clinical Hospital, Hospital das CLínicas, Universidade Federal de Goiás, Goiânia, BRAZIL, 6Genética, Universidade Federal de Goiás, Goiânia, BRAZIL.
Cancer remains the second leading cause of death worldwide, with 5-10% of cases attributed to hereditary syndromes. Among these, Li-Fraumeni Syndrome (LFS) is closely associated with pathogenic germline variants in the TP53 gene, a critical tumor suppressor responsible for maintaining genomic stability and regulating cell cycle and apoptosis. In Brazil, the TP53-R337H variant is highly prevalent due to a well-documented founder effect, particularly in the central and southern regions. Recent studies have revealed the co-segregation of the truncating variant XAF1-E134* with TP53-R337H, forming a functional haplotype associated with increased tumor aggressiveness and earlier onset (Jung et al., 2020; Fortes et al., 2021).This study aimed to evaluate the frequency and clinical impact of this haplotype in a cohort of Brazilian individuals with suspected hereditary cancer syndromes, recruited through the Brazilian Unified Health System (SUS). A total of 527 participants (probands and relatives) were referred to the Human Genetics Center at the Federal University of Goiás (UFG), based on NCCN 2025 criteria for Hereditary Breast and Ovarian Cancer Syndrome (HBOC) and LFS. After informed consent, venous blood samples were collected for DNA extraction. Next-generation sequencing (NGS) was used to analyze the coding region of TP53, and genotyping assays were performed to detect the R337H and XAF1-E134*variants in both probands and their family members.Among the 527 participants, 198 (37.56%) were probands and 329 (62.44%) were family members. Of the probands, 7.58% (15/198) carried pathogenic TP53 variants, with 93.33% (14/15) being R337H. Thirteen probands were tested for XAF1-E134*, and 76.92% (10/13) tested positive. Of these, 90% (9/10) had a confirmed cancer diagnosis, with a mean age at diagnosis of 40.7 years. Furthermore, 30% (3/10) presented with multiple primary tumors, including prostate, osteosarcoma, sarcoma, and adenocarcinoma. Metastasis was observed in 10% (1/10). Among family members, 14.29% (47/329) carried TP53 variants. Of the 35 relatives tested for XAF1-E134*, 91.43% (32/35) were positive.These results provide strong evidence for co-inheritance of TP53-R337H and XAF1-E134*, forming a high-risk haplotype associated with more aggressive cancer phenotypes. Notably, in families where probands were negative for TP53-R337H, relatives were also negative for both variants, supporting the hypothesis of joint transmission. The clinical manifestations observed among carriers reinforce the functional synergy between these two variants and their potential contribution to early-onset and multifocal malignancies.This study highlights the relevance of including XAF1-E134* in genetic screening panels, particularly in Brazilian regions affected by the R337H founder mutation. Incorporating haplotype-based analysis into clinical genetics can improve risk stratification, facilitate early diagnosis, and guide personalized prevention and surveillance strategies in high-risk families. Further studies are essential to clarify the molecular mechanisms underlying this haplotype and to define its role in cancer pathogenesis and clinical management.
Presentation numberPS3-02-12
Contralateral Breast Cancer Subtypes Mirror the Primary: A 23-Year Single-Center Cohort Study
Eunhye Kang, Seoul national university hospital, Seoul, Korea, Republic of
E. Kang, C. Lee, H. Cho, D. Shin, J. Jung, H. Kim, H. Lee, W. Han, H. Moon; General surgery, Seoul national university hospital, Seoul, KOREA, REPUBLIC OF.
BackgroundContralateral breast cancer (CBC) has traditionally been considered a newly developed, biologically independent tumor rather than a recurrence of the primary breast cancer. However, accumulating evidence suggests that some CBCs share molecular characteristics with the primary tumor, indicating possible biological relatedness. This study aimed to evaluate the biological association between primary and contralateral breast cancers by analyzing their subtype distribution and concordance patterns.MethodsWe analyzed 382 patients who underwent surgery for primary breast cancer at Seoul National University Hospital between 2001 and 2024 and subsequently developed contralateral breast cancer. Synchronous CBC was defined as contralateral breast cancer diagnosed simultaneously or within 6 months of primary invasive breast cancer diagnosis, while metachronous CBC was defined as contralateral breast cancer diagnosed thereafter. Patients with contralateral DCIS, unknown subtype, or initial distant metastasis were excluded. The study included 186 synchronous and 196 metachronous CBC cases. Subtype distributions of CBC were compared with those of sporadic breast cancers using multinomial exact tests. Additionally, subtype concordance between primary and contralateral tumors was evaluated using concordance rates.ResultsWe identified distinct clinical patterns between synchronous and metachronous CBCs. Synchronous CBCs were more frequently associated with hormone receptor (HR) positive and HER2 negative primary tumors, whereas metachronous CBCs were more often associated with primary triple-negative breast cancer (TNBC) and showed stronger associations with BRCA mutation status.The subtype distribution of both metachronous and synchronous CBCs differed significantly from that of sporadic breast cancers (p<0.001), with a relatively higher proportion of TNBC. Subtype concordance between primary and CBC tumors was significantly higher than expected under a model of random subtype distribution among all subtypes (p < 0.001, concordance rate vs. expected rate: 79.7% vs. 63.1%, 32.0% vs. 5.5%, 30.8% vs. 10.5%, and 61.7% vs. 20.9% for HR+/HER2-, HR+/HER2+, HR-/HER2+, and TNBC, respectively)ConclusionsContralateral breast cancer does not arise as a random second primary event but rather reflects subtype-specific characteristics of the primary tumor. These findings indicate that the pathogenesis of CBC might not be an independent one but rather be primary tumor-dependent or host factor-dependent which warrants further investigation.
Presentation numberPS3-02-13
Enhancing breast cancer risk assessment in a community imaging center to identify high-risk patients and guide screening and management
Tammy McKamie, Myriad Genetics, Inc., Salt Lake City, UT
T. McKamie1, T. Hudson2, L. Brzeskiewicz1, S. Cummings1; 1Oncology Medical Affairs, Myriad Genetics, Inc., Salt Lake City, UT, 2Imaging, Singing River Health System, Ocean Springs, MS.
Background: To evaluate the effectiveness of using Tyrer-Cuzick version 8 (TCv8, herein referred to as TC) alone compared to a combined approach of a family history assessment, germline testing, and TC with a breast cancer (BC) multiple ancestry polygenic risk score (MA-PRS), in identifying patients at high risk for breast and other cancers at imaging centers. Here we compared approaches for identifying high-risk patients to improve personalized screening and management. Methods: In November 2020, Singing River Health System launched a comprehensive breast program using the TC risk model and germline testing with MA-PRS to identify high-risk patients. During mammogram visits, patients completed the TC risk model and were assessed for genetic testing eligibility based on NCCN guidelines using a cancer history questionnaire. Eligible patients received a comprehensive family history intake and education by telephone from a genetic counselor and, if consented, had blood drawn for genetic testing the same day. The testing included a laboratory’s, a laboratory’s multi-gene panel and MA-PRS to determine BC risk and hereditary cancer syndromes. Post-testing, a nurse navigator reviewed the results and counseled patients on appropriate next steps. We evaluated results to identify guideline-recommended management changes and to determine if high-risk patients would not have been identified without germline testing and MA-PRS. Results: From November 2020 through December 2023, a total of 2,434 patients completed TC and if they met the criteria for genetic testing at the time of their mammogram, they had multi-gene panel testing and MA-PRS. Within this cohort, 677 patients had a TC <20% lifetime BC risk, yet were candidates for medical management change based on their genetic testing, MA-PRS, and/or comprehensive family history results. Specifically, 70 patients with TC <20% met the criteria for genetic testing and were found to carry pathogenic variants in genes associated with hereditary cancer syndromes. Additionally, 133 patients with TC 20% due to the incorporation of MA-PRS. Furthermore, 474 patients with TC <20% were appropriate for modification in management strategy based on family history information obtained through patient education intake. Conclusions: A detailed risk assessment and testing program involving TC, a polygenic risk score model, and germline testing at a community imaging center identified more high-risk individuals and suggested alterations in screening and management compared to using TC alone. Without this approach, patients at elevated cancer risk requiring management changes may not have been recognized.
Presentation numberPS3-02-15
L024, a promising candidate for the prevention and interception of triple negative breast cancer and its metastasis to the brain
Atieh Hajirahimkhan, Northwestern University, Chicago, IL
A. Hajirahimkhan1, E. T. Bartom2, S. M. Roy3, A. M. Eremin1, R. Chen4, D. M. Watterson3, S. E. Clare1, S. A. Khan1; 1Surgery, Northwestern University, Chicago, IL, 2Biochemistry and Molecular Genetics, Northwestern University, Chicago, IL, 3Pharmacology, Northwestern University, Chicago, IL, 4Preventive Medicine, Northwestern University, Chicago, IL.
L024, a promising candidate for the prevention and interception of triple negative breast cancer and its metastasis to the brainIntroduction: Estrogen receptor negative (ER-neg) breast cancer (BC) disproportionately affects younger women, women of African descent, and socioeconomically disadvantaged populations. Excluding high-risk groups, e.g. BRCA1 carriers, for whom there are surgical risk reduction strategies, no approved preventive agents exist for this subtype. ER-neg BCs, including triple-negative (TNBC) and HER2+ tumors, often metastasize to the brain at early stages, yet remain clinically undetected until neurologic symptoms appear, by which time prognosis is poor. Current therapies, including adjuvant drugs for ER-neg BC, offer limited efficacy against brain metastases due to poor brain penetration. Radio-surgical approaches to treat brain metastases carry significantly debilitating side effects with no significant enhancement in survival. There is an urgent need for safe, non-hormonal risk-reducing medications that can prevent both ER-neg BC and its dissemination to the brain.We have identified L024, a promising first-in-its-class, non-endocrine agent which shows strong efficacy against ER-neg BC. Mechanistic studies demonstrate that L024 acts through inhibition of SREBP1-driven lipogenesis, suppression of the PI3K-AKT axis, and downregulation of NF-κB-mediated inflammation, key pathways in ER-neg BC progression and metastasis. However, its potential to intercept or prevent metastatic dissemination, particularly to the brain, remains unexplored. We hypothesize that L024 suppresses ER-neg tumor growth and hinders early metastatic spread to the brain, representing a novel preventive and interceptive approach.Methods: We evaluated the antiproliferative effects of L024 across five ER-neg BC cell lines: MDA-MB-231 (human TNBC), HCC-1937 and HCC-3153 (BRCA1-mutant TNBC), 4T1 (mouse TNBC with high metastasis potential), and MDA-MB-231-Br (human brain-tropic TNBC) using IncuCyte under single and repeated dosing. We developed xenograft tumors of MDA-MB-231 cells in nude mice and treated the animals with daily doses of 80 mg/kg of L024 when the tumors reached a threshold size. We monitored changes in tumor size over 28 days and conducted RNA sequencing on the tumors upon the completion of the studies. Invasion and migration were assessed using Transwell assays with or without extracellular matrix following sublethal L024 treatment (5 μM, 2.5 μM). In silico ADME studies were conducted using ADC Lab software. Pharmacokinetics (PK) were evaluated in female CD-1 mice dosed with L024 (50 mg/kg). Drug levels in plasma, mammary, and brain tissue were measured and modeled using SAAM II.Results: L024 significantly inhibited malignant cell proliferation at concentrations dose-dependently, with effects sustained up to six days post-treatment. It also significantly reduced tumor growth and epithelial to mesenchymal transition in xenograft tumors, suggesting anticancer and antimetastatic effects. Sublethal doses reduced invasion and migration of 4T1 and MDA-MB-231-Br cells, suggesting further validating anti-metastatic potential. In silico profiling indicated favorable CNS penetration: low molecular weight (1 between 1–8 h post-dosing and clearance by 24 hours, indicating suitability for prevention and therapy. Conclusions: L024 reprograms metabolic and inflammatory pathways essential for ER-neg BC progression and early brain metastasis. CNS pharmacokinetics support further evaluation in immunocompetent models of TNBC with brain metastatic potential.
Presentation numberPS3-02-16
Delays in breast cancer diagnosis and treatment in young women
Bessie X Zhang, Brigham and Women’s Hospital, Boston, MA
B. X. Zhang1, T. Sella2, Y. Zheng3, G. J. Kirkner4, K. J. Ruddy5, S. M. Rosenberg6, V. F. Borges7, L. Schapira8, S. E. Come9, J. Peppercorn10, A. H. Partridge4, K. D. Brantley4; 1Department of Medicine, Brigham and Women’s Hospital, Boston, MA, 2Department of Oncology, Sheba Medical Center, Ramat Gan, ISRAEL, 3Department of Biostatistics, Dana-Farber Cancer Institute, Boston, MA, 4Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 5Department of Medical Oncology, Mayo Clinic, Rochester, MN, 6Division of Epidemiology, Weill Cornell Medicine, New York, NY, 7Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO, 8Division of Medical Oncology, Stanford University, Stanford, CA, 9Department of Medical Oncology, Beth Israel Deaconness Medical Center, Boston, MA, 10Department of Medical Oncology, Massachusetts General Hospital, Boston, MA.
Background: Women younger than 40 generally fall outside the routine screening population for breast cancer (BC). We aimed to understand factors influencing diagnosis and treatment delays in these patients, and the impact of delays on disease-free survival (DFS). Methods: We included women aged ≤ 40 years at diagnosis of stage 0-IV BC enrolled in the Young Women’s BC Study from 2006-2016. Sociodemographics, initial BC symptoms, and disease status were collected via annual surveys. Medical record review was used to capture tumor characteristics and recurrence status. We used logistic regression to estimate associations between patient and tumor factors and three delay types: (1) self-delay (≥ 90 days from first symptom to presentation to a provider), (2) diagnosis delay (≥ 90 days from healthcare visit to diagnosis), and (3) treatment delay (≥ 60 days from diagnosis to first treatment). Odds ratios (OR) and 95 % confidence intervals (CI) were estimated in multivariable models after choosing factors via backwards selection (p≤0.05 retained). Hazard ratios (HR) and 95% CIs for the association between delays and DFS, overall and by stage, were estimated via Cox proportional hazard models, adjusted for tumor and treatment factors. Time to event was measured from diagnosis or treatment to recurrence, death, or last follow-up, whichever occurred first. Results: Of 1,297 patients, 1,071 who completed the long-form baseline survey were included. Median time from first symptom to presentation was 14 days (interquartile range (IQR)=2-45), presentation to diagnosis was 14 days (IQR=7-30), and diagnosis to treatment was 41 days (IQR=25-57). Overall, 14% (n=153) had a self-delay and 11% (n=120) had a diagnosis delay. Of 839 patients with self-detected tumors, 18% had a self-delay and 13% had a diagnosis delay. Among those with Stage 1-3 BC (N=928), 22% (n=205) had a treatment delay. In univariable models, income <50K (p=0.01), lower financial comfort (p=0.01), and not living with a partner (p=0.03) were associated with higher odds of self-delay, while pregnancy within 1 year pre-diagnosis (p=0.05) and first-degree family history of any cancer (p=0.04) or BC (p=0.03) were associated with lower odds. Self-delays were more likely for women diagnosed with stage 4 BC (p=0.004) or with HER2+ tumors (p=0.03) and less likely for those with triple negative (TN) BC (p<0.001). In the multivariable model, family history of BC (OR=0.40, 95% CI=0.17-0.80), income <50K (OR=1.97, 95% CI=1.22-3.13) and TN subtype (OR v. luminal A=0.30, 95% CI=0.13-0.60) remained associated. Odds of diagnosis delay were higher among women who were younger (p=0.01) or with Stage 4 BC (p=0.004), and lower for those with luminal B (p=0.04), TN (p=0.01), or high grade tumors (p=0.03). In the multivariable model, younger age (OR per 1-year=0.94, 95% CI=0.89-0.99), and subtype (luminal B v. A OR=0.60, 95% CI=0.38-0.95; TN v. luminal A OR=0.35, 95% CI=0.16-0.70) remained associated. Multivariable models for self-and diagnosis delays remained similar when restricted to self-detected patients.Treatment delays were less likely among unemployed individuals (p=0.001), and women with a positive germline BC risk variant (p=0.002), luminal B (p=0.005) or TN (p<0.001) subtypes, and high-grade tumors (p=0.03). Only tumor factors remained significant in the multivariable model, including subtype (luminal B v. A OR=0.59, 95% CI=0.41-0.86, TN v. luminal A OR=0.25, 95% CI=0.13-0.43), and stage (stage 3 v. 1 OR=0.37, 95% CI=0.20-0.65). Overall and by stage at diagnosis, delays were not statistically significantly associated with DFS before or after adjusting for tumor and treatment factors. Conclusions: Delays in breast cancer care are influenced by multiple psychosocial and clinical factors, and disparities exist by socioeconomic status. It is critical to decrease self- and diagnosis delays to prevent stage 4 breast cancer.
Presentation numberPS3-02-17
Barriers and Facilitators to Implementing a Successful Hereditary Breast Cancer Genetic Testing Program for Women Undergoing Mobile Mammography Screening at Safety-Net Clinics in Southeast Texas
Kelly Meza, Baylor College of Medicine, Houston, TX
K. Meza1, B. Arun2, A. Ruiz Cuevas2, K. Lee3, C. Amaram4, S. Shanker4, A. Vara4, D. Kizub2; 1Department of Medicine, Baylor College of Medicine, Houston, TX, 2Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Department of Breast Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Department of Clinical Cancer Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX.
Introduction: Breast cancer (BC) remains the leading cause of cancer-related death among Hispanic women in Texas. While 5-10% of BC cases are linked to hereditary cancer syndromes, the uptake of genetic testing (GT) among eligible individuals remains low, especially in underserved populations. In our prior study using our validated one-page Cancer Genetic Risk Assessment (CGRA) administered at mammography visits, only 10% of 150 eligible underserved women completed GT. To address this gap, we implemented a 12-month, multi-level program in Southeast Texas that included: (1) culturally tailored genetic education, (2) on-site CGRA screening and GT, (3) financial navigation, and (4) telegenetic counseling. Here, we present barriers and facilitators to program implementation. Methods: This 12-month prospective program was conducted in Harris County (2023-2024) within mobile and safety-net clinics of the VALET mammography program. The primary outcome was GT completion. Women undergoing screening completed our validated one-page CGRA. If eligible, saliva-based GT kits were provided onsite or mailed. A bilingual coordinator assisted with consent and financial paperwork. Participants with pathogenic variants (PVs) or variants of uncertain significance (VUS) received telegenetic counseling; others were informed of negative results. From 2022-2023, 870 women were reached; 590 (87%) completed CGRA, 537 (91.0%) identified as Hispanic. Ninety-nine (15.8%) met criteria for GT, and 54.8% completed testing. completed it. 35 (64.8%) tested negative, 14 (25.9%) had a variant of unknown significance, and 5 (9.5%) had a pathogenic mutation in MUTYH, NF1, CHEK2, MSH3 genes. Program implementation was evaluated through semi-structured interviews guided by the RE-AIM (Reach, Effectiveness, Adoption, Implementation, Maintenance) framework. Interviews were transcribed, double-coded, and thematically analyzed. Results: In-depth interviews were conducted with 40 participants (34 patients, 6 staff). Among patients, program participation was motivated by a family history of cancer (73.7%), desire for greater knowledge (28.9%), commitment to one’s health (28.9%) and psychological relief (28.9%). Key facilitators of GT included the ease of testing (71.1%), prior GT knowledge (15.8%), and prior conversation with PCP (13.2%). Barriers included fear and uncertainty after a positive result (68.4%) and financial constraints (34.2%). Patients recommended expanding telehealth (10.5%), reducing costs (7.9%), increasing education (36.8%), and community outreach (21.1%). After participation, patients reported improved screening adherence (68.4%), encouraging family GT (31.6%), and lifestyle changes (18.4%). Notably, 18.4% said their PCP never discussed GT, even with strong family history. Staff were motivated by their desire to improve community health (100%). Facilitators included integration into clinic workflow and provider education (67%). Staff recommended more education (100%), cost reductions (50%), and provider follow-up (33.3%). Barriers included staff shortages (100%) and time constraints (75%). Conclusions: The qualitative data revealed that emotional factors, prior awareness, and ease of access significantly influenced participation. Patient narratives highlighted the importance of culturally responsive education and logistical support in promoting uptake. Staff insights underscored the value of integrating services into existing workflows and the need for sustained provider engagement. These findings offer actionable strategies to improve the design, delivery, and scalability of community-based genetic risk assessment programs.
Presentation numberPS3-02-19
Primary Care Provider Input On Personalized Breast Cancer Screening Recommendations in WISDOM 1.0: A Nationwide Risk-Based Breast Cancer Screening Trial
Arash Naeim, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
A. Naeim1, A. Stanton2, K. Sepucha3, S. Vangala4, A. Petruse5, L. Madlensky6, A. S. Fiscalini7, L. J. Esserman7, M. Eklund8, K. Ross9, D. Goodman-Gruen10, J. Tice11, E. Ziv12, Y. Shieh13, L. van ‘t Veer14, A. Kaster15, A. M. Blanco9, O. I. Olopade16, M. Scheuner17, B. Tong18, L. Sabacan19, J. Atamer7, H. Harvey20, R. A. Hiatt21, A. D. Borowsky22, J. Esserman23, H. Anton-Culver24, A. LaCroix25, H. L. Park26, B. A. Parker27, R. Lancaster28, WISDOM Investigators and Advocate Partners, N. Wenger29; 1Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 2Department of Psychology,, UCLA, Los Angeles, CA, 3Division of General Internal Medicine, Mass General, Boston, MA, 4Division of General Internal Medicine and Health Services Research, UCLA, Los Angeles, CA, 5Clinical and Translational Science Institute Office of Clinical Research, University of California Los Angeles, Los Angeles, CA, 6Department of Medicine,, University of California, San Diego,, San Diego, CA, 7Department of Surgery, UCSF, San Francisco, CA, 8Department of Medical Epidemiology and Biostatistics,, Karolinska Institutet, Stockholm, SWEDEN, 9Cancer Genetics and Prevention Program, University of California San Francisco, San Francisco, CA, 10Department of Epidemiology and Biostatistics, UC Irvine, Irvine, CA, 11General Internal Medicine, School of Medicine, University of California San Francisco, San Francisco, CA, 12Department of General and Internal Medicine, University of California San Francisco, San Francisco, CA, 13Department of Population Health Sciences,, Weill Cornell Medicine, New York, NY, 14Lab Medicine, UCSF, San Francisco, CA, 15Edith Sanford Breast Center, Sanford Health, Fargo, ND, 16Center for Clinical Cancer Genetics and Global Health, Department of Medicine,, University of Chicago, Chicago, IL, 17Department of Medicine, University of California San Francisco and San Francisco VA Health Care System, San Francisco, CA, 18Cancer Genetics and Prevention Program, UCSF, San Francisco, CA, 19Department of Surgery, University of California San Francisco, San Francisco, CA, 20Edith Sanford Breast Center, Sanford Health, Sioux Falls, SD, 21UCSF Helen Diller Family Comprehensive Cancer Center, Department of Epidemiology & Biostatistics,, UCSF, San Francisco, CA, 22Department of Pathology, School of Medicine, UC Davis, Sacramento, CA, 23OB-GYN, Diagnostic Center of Miami, Miami, FL, 24Department of Medicine, Genetic Epidemiology Research Institute, UCI, Irvine, CA, 25Department of Family and Preventive Medicine, Division of Epidemiology, UCSD, San Diego, CA, 26Department of Pathology and Laboratory Medicine, Department of Epidemiology, UC Irvine School of Medicine, Irvine, CA, 27Moores Cancer Center, University of California San Diego, San Diego, CA, 28Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 29Division of General Internal Medicine and Health Services Research,, University of California Los Angeles, Los Angeles, CA.
BackgroundThe Women Informed to Screen Depending on Measures of risk (WISDOM) Study 1.0 (2016-2023; NCT02620852) is testing a novel, personalized approach to breast cancer screening compared to annual mammography. WISDOM, a large-scale screening trial, enrolled women ages 40-74 with no personal history of breast cancer using a randomized, preference tolerant trial design, reflecting the heterogeneity of the U.S. population. The goal of this analysis is to evaluate whether women discussed WISDOM screening recommendations with their primary care provider, factors associated with discussion, and whether primary care providers (PCPS) agreed or disagreed with the screening recommendation.MethodsWe analyzed participant-reported responses among the first 15,000 WISDOM participants who completed patient-centered outcomes surveys at baseline, at the time of they received their screening recommendation, and at first annual follow-up. The annual survey asked about discussing the trial screening assignment with their PCPS and the reaction of their PCPS to the screening assignment (Likert 5 scale from Strongly Disagreed to Strongly Agreed). These items were compared between the randomized arms (annual versus personal risk-based) and across the study breast cancer screening assignments (6 months with MRI, Yearly with Risk-Reduction, Yearly, Two Years, Delayed Screening).ResultsParticipants in the risk-based screening arm were significantly more likely to report discussing the screening recommendation with their PCP (35.6%, 2708/7610) compared to those in the annual arm (19.0%, 1308/6888; p < 0.001). PCPS agreement with the screening recommendation also varied by arm. In the annual group, 75.7% of participants reported their PCPS agreed or strongly agreed with the recommendation and only 2.5% disagreed, In the risk-based group, 58.6% agreed and 16% disagreed. Among participants in the risk-based arm, discussion rates with PCPS and level of PCPS agreement differed by screening recommendation. The highest rate of discussion occurred in those assigned to 6 months with MRI (71.1%), followed by Yearly with risk-reduction (50.3%), Two Years (34.6%), Yearly (29.6%) and Delayed (32.4%). Disagreement was highest in those identified at lowest risk, Delayed Screening (24.2%) and those with the Two Year (16.1%) and disagreement was lowest with the higher screening recommendations wherein Yearly (1%) and 6 Month with MRI (5.6%) disagreed. Conclusion/DiscussionWomen often discuss breast cancer screening recommendations with their primary care providers, particularly risk-based recommendations. Data from WISDOM 1.0 show PCPS had stronger agreement more frequent screening recommendations and disagreement with less frequent intervals. These findings suggest that education to increase primary care physician familiarity with personalized screening approaches may enhance the implementation and acceptance of risk-based strategies in clinical practice.
Presentation numberPS3-02-22
De Madres a Hijas: A Culturally Tailored Intervention to Increase Knowledge on Hereditary Breast Cancer Risk among Latina Mothers & Their Daughters
Laura Ann Logie, Nueva Vida, Alexandria, VA
L. A. Logie1, A. Serrano1, C. Campos Galván2, A. Hurtado de Mendoza3, M. De Jesus4, A. Jimenez1; 1N/A, Nueva Vida, Alexandria, VA, 2Community Outreach and Engagement, George Washington University, District of Columbia, DC, 3Cancer Prevention and Control at Lombardi Comprehensive Cancer Center, Georgetown University, District of Columbia, DC, 4SIS | Environment, Development & Health, American University, District of Columbia, DC.
Background: An important step toward understanding the role of BRCA and other cancer genes in Latinas is to increase the number of individuals who receive genetic testing and counseling (GTC). Latinas have the 2nd highest prevalence of BRCA gene mutations after women of Ashkenazi Jewish ancestry and the lowest level of awareness about GTC for inherited cancer risk than all other racial/ethnic groups, and are 4-5 times less likely to get tested compared to Whites. Recognizing a lack of knowledge among Latina breast cancer survivors (BCS) and their daughters, Nueva Vida, developed an intergenerational intervention as an effective method to leverage cultural values and combat mistrust, laying the groundwork for learning and preventive action. The aim of De Madres a Hijas is to increase Latina mothers/daughters’ knowledge about Hereditary Breast Cancer (HBC) risk, enhance their talking about familial HBC risk with relatives, and develop an “action plan” to make educated decisions on their own health.Methods: We used an explanatory pre-post mixed methods design to assess how the intervention contributes to an increase in Latina mothers/daughters’ knowledge about HBC risk and promotes intent to participate in genetic counseling and testing. We used descriptive statistics to characterize the study sample of Latina BCS and their daughters (n=48). Participants were administered a 13-item pre-post survey to assess knowledge (i.e., having a genetic mutation does not mean you will get cancer T/F). An expanded Consolidated Framework for Implementation Research (CFIR) comprised of 15 domains (e.g., Difficulty Understanding Genetics, Actions to Learn about Family History) guided the qualitative data analysis. Four Coders utilized template analysis to analyze the interviews. Results: This first-of-its-kind, community-based initiative leverages a one-day, workshop to educate BCS and their daughters about HBC, their risks, preventive behaviors and GTC. These workshops show 91% of daughters improved understanding of genetic cancer, and 83% reported behavior changes toward healthier daily activities and prevention-focused habits. As stated by a participant, “We were told to be aware if we feel pain… I did my self-exam and I didn’t find anything, but I felt pain right next to my armpit and I felt more confident to make a follow up appointment and they found it (a lump).” Lastly, to extend the reach of the program, we have developed the Toolkit—a manual for other Latino-serving organizations hoping to facilitate these life-saving conversations in their communities. Discussion: Culturally tailored interventions are vital for ensuring that GTC services are accessible, effective, and equitable for all individuals, regardless of their ethnicity. Education plays a pivotal role in closing the gaps in GTC. Creating the opportunity for mothers and daughters to share this experience, provides not only knowledge building about GTC, but also a special bonding by listening and understanding each other’s lived experience. Second generation Latinas show better literacy levels than immigrant Latinas who deal even with their own language reading and writing barriers. For the Latina community, it’s crucial that we not only offer HBC education but do so in a way that resonates with their unique cultural perspectives.
Presentation numberPS3-02-23
Hereditary Breast Cancer (BC) in the Moderate-Risk Spectrum – Surgical Practices and Risk of Second Primary Tumors
Andre Luiz Cicilini, A.C. Camargo Cancer Center, São Paulo, Brazil
A. L. Cicilini1, J. C. Casali-da-Rocha2, F. B. Makdissi3, S. M. Sanches4, S. M. Volc2, R. Cagnacci-Neto3, F. P. Leite3, S. M. Castro3, M. Sonagli3, D. G. Rodrigues de Souza1, P. V. de-Cerqueira1, V. P. de Souza1, L. R. Soares1, V. C. Cordeiro de Lima4, A. A. Balieiro4, S. M. Caputo5, E. Santana dos Santos4; 1Clinical Oncology, A.C. Camargo Cancer Center, São Paulo, BRAZIL, 2Oncogenetic Reference Center, A.C. Camargo Cancer Center, São Paulo, BRAZIL, 3Mastology- Breast Reference Center, A.C. Camargo Cancer Center, São Paulo, BRAZIL, 4Clinical Oncology – Breast Reference Center, A.C. Camargo Cancer Center, São Paulo, BRAZIL, 5Genetic Service, Institut Curie, Paris, FRANCE.
Background: Hereditary breast cancer represents 5-10% of all cases. While BRCA1/2 pathogenic variants (VP) are well-established high-risk factors, clinical implications of moderate-penetrance gene VP (PALB2, CHEK2, ATM, RAD51C, RAD51D, BARD1, BRIP1, FANCM) remains less explored. Global prevalence estimates for moderate-penetrance genes such as PALB2, CHEK2 and ATM range between 1-3%, underscoring the clinical relevance of tailored management. Definition of optimal surgical strategies and surveillance, particularly based on the prevalence of second primary tumors, are needed. We aimed to evaluate surgical management strategies and the prevalence of second primary tumors in patients with hereditary breast cancer associated with moderate-penetrance gene mutations, to inform clinical decision-making and surveillance recommendations. Methods: A retrospective cohort study was conducted at A.C. Camargo Cancer Center including women diagnosed with BC and VP in moderate-penetrance genes between 2016-2025. Descriptive statistics summarized clinical-pathological data. Cumulative incidence of second primary tumors and breast cancer (BC) recurrence was calculated. Five-year overall survival (OS) was estimated by the Kaplan-Meier method. Moderate-risk classification was defined according to current NCCN guidelines and a comprehensive hereditary cancer NGS panel. Results: Among 1,429 patients tested, 120 (8%) had VP in moderate-penetrance genes: 28 PALB2, 28 CHEK2, 28 ATM, 15 RAD51C, 4 RAD51D, 6 FANCM, 8 BRIP1, 3 BARD1. They were all female. Family history of BC was present in 73%. Most were diagnosed at stage I (54%). Predominant histology was invasive ductal carcinoma (79%), with SBR grade 3 in 63%. Subtypes included: Luminal B (24%), HER2-low (23%), HER2-overexpressing Luminal B (19%), TNBC (18%), Luminal A (10%) and HER2 -overexpressing (4%). Contralateral synchronous BC occurred in 3 patients (2%). Unilateral adenomastectomy was the most common surgical approach (51%), followed by segmental resection (45%). Four percent of patients underwent bilateral adenomastectomy. Sentinel lymph node biopsy was performed in 67%; axillary dissection in 33%. Over a median follow-up of 5 years, second primary tumors were observed in 16 patients (13.2%): 7 papillary thyroid carcinomas (5 CHEK2, 2 ATM), 6 ovarian cancers (1 ATM, 5 RAD51C), and single cases of melanoma, endometrial, lung, and colorectal cancer. BC recurrence occurred in 8 patients (6.6%). Prophylactic salpingo-oophorectomy was performed in 17%. The 5-year OS was 96.8%. Conclusion: Unilateral mastectomy with sentinel lymph node biopsy was the most frequent surgical strategy for moderate penetrance gene VP carriers. Despite excellent OS, patients with VP in moderate-penetrance genes—especially CHEK2, ATM, and RAD51C —faced a significant cumulative incidence of second primary tumors (13.2%), with thyroid and ovarian cancers being the most prevalent. These findings highlight the need for tailored surveillance strategies beyond breast-focused care in this genetically distinct population. This highlights the need for a multidisciplinary approach involving breast surgeons, genetic counselors, gynecologists, and endocrinologists to optimize long-term surveillance beyond breast-focused care. Breast surgeons should also consider bilateral prophylactic mastectomy in highly selected moderate-risk cases to further reduce long-term risk.
Presentation numberPS3-02-24
Completion Rates of Risk Counseling Consultations For Women at High-Risk for Breast Cancer within the WISDOM 1.0 Pragmatic Screening Trial
Jackelyn Moya, UCSF, San Francisco, CA
J. Moya1, K. Ross2, D. Goodman-Gruen3, L. Madlensky4, A. Blanco2, B. Tong2, H. Harvey5, A. S. Fiscalini1, A. Kaster6, M. Scheuner7, L. Sabacan1, J. Atamer1, L. van ‘t Veer8, O. I. Olopade9, J. Tice10, E. Ziv11, S. Kapoor1, Y. Shieh12, A. D. Borowsky13, A. Naeim14, H. L. Park15, A. LaCroix16, WISDOM Investigators and Advocate Partners, L. J. Esserman1; 1Department of Surgery, UCSF, San Francisco, CA, 2Cancer Genetics and Prevention Program, UCSF, San Francisco, CA, 3Department of Epidemiology and Biostatistics,, UC Irvine, Irvine, CA, 4Department of Medicine, UCSD, San Diego, CA, 5Edith Sanford Breast Center, Sanford Health, Sioux Falls, CA, 6Edith Sanford Breast Center, Sanford Health, Fargo, ND, 7Department of Medicine, University of California, San Francisco and San Francisco VA Health Care Sys, UCSF, San Francisco, CA, 8Department of Lab Medicine, UCSF, San Francisco, CA, 9Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, IL, 10General Internal Medicine, School of Medicine, UCSF, San Francisco, CA, 11Department of General and Internal Medicine, UCSF, San Francisco, CA, 12Department of Surgery, Weill Cornell Medicine, New York, NY, 13Department of Pathology, School of Medicine, UC Davis, Sacramento, CA, 14Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 15Department of Pathology and Laboratory Medicine, Department of Epidemiology,, UCI, Irvine, CA, 16Department of Family and Preventive Medicine, Division of Epidemiology, UCSD, San Diego, CA.
TITLE: Completion Rates of Risk Counseling Consultations for Participants Identified as High Risk for Breast Cancer within the WISDOM 1.0 Pragmatic Screening Trial BACKGROUND: The WISDOM study is innovative in that it is the first, prospective large-scale trial of assessing risk-based mammographic screening recommendations versus annual screening. Women identified as high-risk were invited to complete a Breast Health Specialist consultation, which included use of an automated educational tool, Breast Health Decisions (BHD) tool, identifying their specific breast cancer risk factors, and how to promote uptake of risk-reducing options. During initial rollout, the preliminary data obtained demonstrated that racial and ethnic minority populations were underrepresented in the study demographics, thus, we created a program to incorporate a more diverse participant pool. We sought to understand whether completion rates of consultations with trained breast health specialist (BHS) varied among race/ethnicity, geography, and age. METHODS: We analyzed data from WISDOM 1.0 participants that completed the BHD tool from 2015-2023 that were considered being in the highest 2.5% breast cancer risk based on the Breast Cancer Surveillance Consortium. Of those participants, we compared completion of BHS consultations by comparing them by age (40-49, 50-59, 60-69, 70-79), race/ethnicity (White, Asian, Black, Hispanic, Other/Mixed Race), and geography (Northeast, Midwest, South, West). We compared each category by proportions of total completed versus not completed and obtained percentages of each. RESULTS: There were a total of 2883 participants identified as high-risk and offered a BHS consultation. Most of all participants were White (84%) and from the West region of the United States (66%). Completion rates were similar across age groups (52-71%), but lower for women ages 70-79 (52%). Within the race/ethnicity category, the proportions that completed consultations were: Other/Mixed Race 105/137 (77%); White 1666/2434 (68%); Hispanic 76/122 (62%); Asian 42/78 (54%); and Black 46/112 (41%). Lastly, the geographic region with the highest completion rate was the Midwest at 521/623 (84%) and the lowest in the South at 157/306 (51%). CONCLUSION: WISDOM identified women at high risk and provided virtual clinical consultations with breast health specialists to review their personalized risk and discuss breast cancer prevention strategies. There were notable differences in completion of clinician consultations rates seen across categories of age, geography, and race/ethnicity. Further assessment of scheduling availability, structural barriers, participant preferences, clinician contact methods, and other contributing factors is required to understand why completion rates differed so much across demographic groups. Based on these results and participant feedback, the WISDOM 2.0 study has changed consultation workflows, improved clinician availability and scheduling methods. We also plan to increase community engagement, tailor outreach methods based on community member feedback, and offer more options for participants to receive the personalized risk information than were offered in WISDOM 1.0. As a pragmatic and iteratively improving study, WISDOM 2.0 has been funded to create and implement these changes with the goal of providing a more equitable method of providing specialty consultations to high-risk participants.
Presentation numberPS3-02-25
A blood-based lipid panel for risk determination of harboring breast cancer among women at elevated risk
Ehsan Irajizad, MD Anderson Cancer Center, Houston, TX
E. Irajizad1, S. Hanash2, J. Fahrmann2, A. Brewster2; 1Biostatistics, MD Anderson Cancer Center, Houston, TX, 2Clinical Cancer Prevention, MD Anderson Cancer Center, Houston, TX.
There remains a need to establish tools to better tailor screening for women at elevated risk of breast cancer (BrCa). Alterations in metabolism are inherently intertwined with breast cancer development and progression. In the current study, we performed multi-assay metabolomic analyses on plasma samples collected from a total of 438 breast cancer cases and 311 female controls from two independent cohorts of high-risk women. Samples from one cohort were used for discovery and samples from the second cohort were used for initial pre-validation. Machine learning was applied to establish a 12-lipid marker panel and tertile-based thresholds for low-, intermediate-, and high-risk of harboring breast cancer subsequently established. The lipid panel and corresponding risk thresholds were further validated in an independent nested case-control cohort of women at elevated risk, using plasma samples prospectively collected from 81 women who developed invasive breast cancer and 191 age-matched female controls. The lipid panel achieved ORs [per unit increase] of 2.31, 2.37, 1.48, in the respective Discovery, Pre-Validation, and Validation Sets. In the combined datasets, the lipid panel yielded an overall OR of 2.02 and AUC of 0.67 for BrCa. Compared to women in the low-risk strata, women in the high-risk group based on the lipid panel were 3.48-times more likely to have BrCa. The lipid panel provides a potential tool for stratifying women at highest risk of developing or harboring breast cancer who would benefit from more intensive screening and interception strategies from those at lowest risk who may be spared overdiagnosis and overtreatment.
Presentation numberPS3-02-26
Using a plain language tool to overcome barriers to research participation by people with inherited cancer risk
Susan Joy Friedman, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL
S. J. Friedman1, D. B. Rose2, J. D. Rogers1, K. N. Owens3, P. L. Welcsh3, M. Dean4, R. H. Pugh Yi5; 1Research and Education, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 2Research, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 3Education, Facing Our Risk of Cancer Empowered (FORCE), Tampa, FL, 4Department of Communication, University of South Florida, Tampa, FL, 5Program Evaluation, Akeso Consulting, LLC, Vienna, VA.
Background: Only 7% of adult cancer patients enroll in treatment-related clinical trials, with even lower rates for genetics and quality-of-life studies.1 People with germline mutations are interested in participating in hereditary cancer research, but few are being offered the opportunity by their healthcare team. In a FORCE survey, only 14% of people at high risk for breast cancer reported hearing about clinical research opportunities from their healthcare providers, while 75% expressed interest.2 Dedicated efforts are needed to ensure that the hereditary cancer community is aware of clinical research opportunities. In 2020 FORCE surveys, 46% of oncology nurses and 55% of certified genetic counselors identified research jargon and health literacy as major barriers to clinical trial referral.3,4 FORCE and other members of the “Consistent Testing Terminology Working Group” found large gaps in patient understanding of commonly-used cancer research terms.5 In focus groups, FORCE asked women with or at high risk for hereditary breast cancer to review a cancer study listing on ClinicalTrials.gov. All agreed that the study description was difficult to understand due to complexity and jargon and that Clinicaltrials.gov required a lot of practice to use.6 Methods: In 2014, FORCE built a tool to help hereditary cancer patients find, understand, and enroll in relevant studies. The tool consists of a custom database with plain-language summaries of research studies relevant for people with inherited cancer risk. Users can search for studies listed in the FORCE database as well as on ClinicalTrials.gov by study type, cancer type, gene or biomarker, study location and key word. FORCE evaluated the tool through focus groups and a user survey. Focus group participants were asked to compare the ClinicalTrials.gov listing with the FORCE plain-language version of the same study. Results: Focus group participants all agreed that the FORCE listing was clearer and easier to understand than ClinicalTrials.gov.6 Key user survey results are included in Table 1. Of the respondents, 79% found the tool easy to use, 84% would use it again, 79% would refer others to the tool. Of respondents, 36% enrolled, and 43% planned to enroll in a research study. Conclusions: People with inherited cancer risk are highly motivated to participate in clinical research—but most are never informed about studies by their healthcare team. ClinicalTrials.gov is not patient friendly, and hereditary cancer patients find the site difficult to use and understand. FORCE’s “Search and Enroll Tool” bridges this gap by offering accessible, accurate, and easy-to-understand listings of studies enrolling hereditary cancer patients. Results from our focus groups and survey demonstrate that the tool meets a critical need. Participants reported high user satisfaction, comprehension, and intention to utilize the tool to enroll in future studies.
| Agree | Neutral | Disagree | |
| I found a study that interested me | 72% | 19% | 9% |
| I found a study for which I was eligible. | 53% | 23.5% | 23.5% |
| I found a study for which a friend or relative was eligible. | 45% | 32% | 23% |
| I understood the study goals. | 85% | 13% | 2% |
| I was able to understand what was required of participants. | 86% | 12% | 2% |
| I was able to understand who was eligible. | 86% | 10% | 4% |
Presentation numberPS3-02-27
A Screening Paradox in TNBC: Exploring communities with High Late-stage Diagnosis Despite High Screening Uptake
Courtney Pisano, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH
C. Pisano1, R. Abou Zeidane1, F. Hussain2, W. Dong3, T. Lal4, L. Vu5, N. Mehta6, C. Speers7, S. M. Koroukian8, J. Rose9; 1Radiation Oncology, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, 2Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 3Department of Population and Quantitative Health Sciences , Case Western Reserve University, School of Medicine, Cleveland, OH, 4Surgery, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, 5Population Cancer Analytics Shared Resource, Case Western Reserve University, School of Medicine, Cleveland, OH, 6Preventive Medicine, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH, 7Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 8Department of Population and Quantitative Health Sciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 9Population Cancer Analytics Shared Resource, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, OH.
Introduction: Screening mammography has significantly reduced breast cancer mortality by enabling earlier diagnosis. Triple negative breast cancer (TNBC) is an aggressive subtype disproportionately affecting Black women. Given its aggressive nature and tendency to occur in younger women, understanding the role of screening in early detection of TNBC is critical. Accordingly, we sought to determine if common traits characterized communities where, despite high screening uptake, the proportion of patients presenting with late-stage TNBC (defined as presenting with regional or distant disease) remained elevated. By understanding characteristics of communities that appear to benefit less from screening, we hope to identify opportunities for intervention around improving the earlier diagnosis of TNBC. Methods: We identified women diagnosed with TNBC from 2010 to 2021 using SEER 22-Registry data (excluding Alaska). We calculated the proportion “late stage” in each county of the SEER regions. To ensure adequate sample sizes, we used the Max-P regionalization method to combine adjacent similar sociodemographic counties (determined by Area Deprivation Index) with ≤10 late-stage cases, creating composite counties (CC) as our unit of analysis. For each CC, we estimated the proportion of women up to date on mammography using CDC PLACES. We estimated community-level characteristics using the 2014-2018 American Community Survey 5-year data and CDC PLACES data including smoking, binge drinking, education, obesity. We used Classification and Regression Tree (CART) analysis to identify combinations of community characteristics associated with having a high (above median) proportion of late-stage TNBC despite having high (above median) mammography uptake. Results: The CART analysis delineated four distinct community profiles with varying rates of the outcome of interest. Communities with high proportions of Black residents (>25.8%) showed the highest rate of women with high screening uptake yet high late-stage TNBC at 63.2%. In contrast, communities with lower rates of binge drinking and lower Black population percentages showed the lowest rates (7.1%) of the same outcome. Conclusions: The study reveals that high screening uptake alone does not uniformly reduce late-stage TNBC, particularly in communities with higher percentages of Black residents or those with high-risk health behaviors (e.g. binge drinking). Future studies should assess the role that both system factors and racial differences in tumor biology play in this finding.
| Profile (n=485) | Characteristics | % of Composite Counties with high screening uptake but high late-stage TNBC |
| 1 (n=226) | ≤ 21.8% binge drinking; ≤ 6.4% Black | 7.1% |
| 2 (n=138) | ≤ 21.8% binge drinking; > 6.4% Black | 22.5% |
| 3 (n=47) | > 21.8% binge drinking; ≤25.8 % Black | 38.3% |
| 4 (n=68) | > 21.8% binge drinking; > 25.8% Black | 63.2% |
Presentation numberPS3-02-28
Emergency Department Breast Cancer Screening Pilot: Improving Access to Screening and Diagnosis for Underserved Populations
Cammeo Mauntel-Medici, University of Illinois at Chicago- UI Health, Chicago, IL
A. Maheswaran1, C. Mauntel-Medici1, J. Green1, R. Hunt1, N. Awati2, S. Hay1, T. Schmidlen3, E. Simmons4, M. Snir5, J. Lin1; 1Emergency Medicine, University of Illinois at Chicago- UI Health, Chicago, IL, 2Internal Medicine, University of Illinois at Chicago- UI Health, Chicago, IL, 3Clinical Development, Nest Genomics, Henderson, NV, 4Product Development, Nest Genomics, Henderson, NV, 5CEO, Nest Genomics, Henderson, NV.
Objectives: Breast cancer is the most common cancer in women, and those diagnosed with breast cancer early have a 93% higher 5-year survival rate compared to those diagnosed at a later stage. Presently, biennial mammogram screenings are recommended for women 40 yrs or older. In 2022, 67.8% of U.S. women aged 40 or older reported having had a mammogram in the past 2 years. However, mammography rates were lowest among women with no insurance or access to primary care. Emergency departments (EDs), which serve as a safety net for populations with poor access to care, offer a unique opportunity to reach these underserved groups with preventive health services. An urban ED in Chicago piloted an Electronic Health Record (EHR)-driven breast cancer screening initiative targeting females aged 40 years or older to improve access to preventive health services for underserved populations. Through a partnership with Nest Genomics, the program implemented a customized digital navigator, built on the Nest platform, to facilitate rapid breast symptom screening and hereditary cancer risk assessment (HCRA). The initiative used a multi-modal outreach strategy to engage with patients in-person, by phone, and via text and email. Two patient navigators contacted patients identified by the EHR algorithm and offered breast cancer education, the opportunity to complete the Nest symptom screener and HCRA, and linkage to screening mammography. Referrals for breast Magnetic Resonance Imaging (MRI), genetic testing, and/or chemoprevention were provided to qualifying patients. This retrospective quality improvement study evaluated outcomes from this initiative between 4/1/2024 and 6/30/2025. Methods: Descriptive statistics were used to summarize patient characteristics and the primary outcomes, including completion of risk assessment and linkage to mammogram. Results: Between 4/1/2024 and 06/30/2025, 303 patients aged 40 or above (mean age = 51) were reached (45% via phone outreach, 37% via text and email, and 18% via in-person outreach) through the ED Breast Cancer Screening initiative. Of those reached, the majority were Non-Latinx Black (n= 199, 66%) and insured through Medicaid (n= 136, 45%). Screening mammogram orders were placed for 65 patients, 56 screening mammograms were scheduled, and 26 screening mammograms were performed. For half (n=13, 50%) of the 26 patients completing a screening mammogram, it was their first ever mammogram. Among the 303 patients reached, 63% completed the HCRA (n= 191), 73 patients met national guidelines-based criteria for genetic testing, 13 met criteria for breast chemoprevention, and 11 met criteria for supplemental screening with Breast MRI. During the study period, 11 of 73 patients who met genetic testing criteria were scheduled for a genetic counseling appointment and no patients had yet attended the appointment. Two of the 13 patients eligible for chemoprevention were scheduled with the high-risk clinic, and 1 had attended and received a prescription. Four of the 11 patients eligible for supplemental breast MRI were referred to their PCP and 1 patient had a breast MRI ordered. Conclusions: Our ED initiative identified an underserved patient population that had not yet received recommended breast cancer screening in other venues. EHR-driven screening initiatives that leverage multi-modal outreach strategies and digital patient navigation can improve access to preventive health screenings for vulnerable patients seen in EDs. Further work is needed to address challenges in ensuring timely and effective linkage to recommended follow-up services (e.g. mammography, genetic counseling, high-risk clinic).
Presentation numberPS3-02-29
Reevaluation of Hereditary Cancer Screening and Testing at an Urban Breast Cancer Survivorship Program in the Bronx, NY – A Pilot Study
Aushna Saleem, Albert Einstein College of Medicine, Bronx, NY
A. Saleem1, A. Sanchez2, E. Suskin2, M. Lalla3, A. Moadel-Robblee4, D. Makower3, S. Klugman2; 1Medicine, Albert Einstein College of Medicine, Bronx, NY, 2Department of Obstetrics and Gynecology and Women’s Health, Division of Reproductive and Medical Gen, Montefiore Medical Center, Bronx, NY, 3Montefiore Einstein Comprehensive Cancer Center, Montefiore Medical Center, Bronx, NY, 4Medicine, Albert Einstein College of Medicine / Montefiore Einstein Comprehensive Cancer Center, Bronx, NY.
Introduction Current national guidelines recommend consideration of genetic testing for all newly diagnosed breast cancer (BC) patients (pts). However, hereditary cancer predisposition genetic testing has been historically underutilized for many reasons, including limitations of prior National Comprehensive Cancer Network (NCCN) guidelines, insurance/financial barriers, and emotional burden of diagnosis and testing. Thus, many BC survivors did not receive current standard of care genetic testing at diagnosis. Benefits of germline genetic testing and identification of pathogenic variants (PV) in late BC survivorship include eligibility for enhanced screening for other malignancies and potential cascade testing of family members. We conducted a pilot study evaluating interest in and uptake of genetic testing among BC survivors. Methods Charts of pts seen at Montefiore Einstein Comprehensive Cancer Center BC survivorship program from January 1, 2023 to December 31, 2023 (n=551) were reviewed. All pts were at least 5 years from diagnosis and completed BC treatment. Pts who had not previously received multi-gene panel testing were contacted by telephone to determine interest in genetic counseling and testing. The benefits of genetic counseling were formally discussed with pts by either the clinician caring for them at their annual survivorship visit or by person distributing survey. Results 312 (57%) pts seen in survivorship had not previously completed genetic testing. 34 (6%) had been previously referred for counseling, but did not complete testing due to financial barriers, personal reasons, or guideline exclusion. Of pts who did not undergo testing, 95 (30%) met 2024 NCCN criteria for testing: 47 by age at diagnosis, 23 by diagnosis of triple negative BC, 12 by family history (FH) of ovarian cancer, 6 by FH of pancreatic cancer, and 7 by FH of BC and/or prostate cancer. 130 patients who had previously not completed testing were called for telephone survey, prioritizing those previously referred but not tested, and those who met current NCCN guidelines for testing. 20 pts declined the survey. Reasons for declining survey included other medical concerns (n=4), scheduling issues (n=10), and lack of interest (n=6). 64 pts were unable to be reached by telephone. 46 pts completed the survey (44 pts surveyed in English, 2 in Spanish). Of those surveyed, 29 (63%) expressed interest in genetic testing. Reasons for interest included obtaining information for family (n=2); obtaining information about prior diagnosis and cancer risk (n=3); reducing anxiety (n=2). Reasons for declining counseling included not desiring further invasive tests (n=3). Most pts did not endorse a reason for agreeing or declining counseling. 11 pts (38%) have completed testing to date. Results of testing include BRCA1 PV (n=1), TP53 PV (n=1), variants of uncertain significance (n=2), and negative results (n=7). 19 pts have been referred for genetic consultation and are awaiting testing. Conclusions While more than half of BC patients eligible for genetic testing at diagnosis did not receive it, 63% of those surveyed in survivorship expressed interest in undergoing testing, and 38% of those expressing interest have completed testing to date. Our study demonstrates feasibility of and interest in germline genetic testing in late BC survivorship. Clinicians caring for BC survivors who have not undergone genetic testing should discuss benefits of testing for patients and their families, as well as changes in guidelines and insurance coverage. Next steps include completing genetic consultation and testing for referred patients and continuing to reach out to all pts without prior genetic testing or with outdated testing.
Presentation numberPS3-02-30
Ancestry-specific prevalence of pathogenic variants among patients with breast cancer who do not meet guidelines for genetic testing
Timothy Simmons, Myriad Genetics, Inc., Salt Lake City, UT
R. Bernhisel1, M. Kucera1, S. Cummings2, E. Smith2, T. Simmons1, E. Hughes1; 1Biostatistics, Myriad Genetics, Inc., Salt Lake City, UT, 2Medical Affairs, Myriad Genetics, Inc., Salt Lake City, UT.
Background Patients with breast cancer (BC) who harbor germline pathogenic variants (PVs) in hereditary cancer genes have improved survival when their surgical and treatment decisions are tailored to their specific genetic alterations. Additionally, the identification of germline PVs is crucial for family members, as it allows for interventions that can decrease cancer-related morbidity and mortality. Commonly utilized guidelines recommend genetic testing only under specific conditions: if cancer is diagnosed at a young age, typically at or before the age of 50; if there is a strong family history (FH) of cancer; or if there are specific tumor characteristics. Several studies have demonstrated that many patients who do not meet these criteria unknowingly carry PVs and may, therefore, be receiving suboptimal care. In the United States, Black and Hispanic women face disproportionately high rates of BC mortality, highlighting the importance of genetic testing in these populations. It is unclear whether guideline-based restrictions differentially impact women of certain ancestral backgrounds. Here, we explore this question by estimating PV prevalence among BC patients of Asian, Black/African, Hispanic, Multiple, and White/non-Hispanic ancestries who do not meet guidelines for genetic testing. Methods Multivariable logistic regression models were constructed to analyze trends in the prevalence of PVs based on age, personal/family history, and ancestry in a consecutive cohort of patients referred for hereditary cancer testing with a multigene panel. We report ancestry-specific model-based estimates of prevalence for patients diagnosed with BC at older ages with no FH and no prior personal history of cancer of any type. Prevalence is summarized overall for 25-48 hereditary cancer genes with guideline-based medical management recommendations, as well as for the genes most frequently implicated in BC. Results The study cohort comprised 245,669 female patients affected by BC. There were 8,025 (3.3%) Asian, 25,900 (10.5%) Black/African, 20,022 (8.1%) Hispanic, 141,279 (57.5%) White/non-Hispanic patients, and 16,689 (6.8%) patients with multiple self-reported ancestries. Estimates of PV prevalence for a patient with BC with no FH and no prior personal history of cancer of any type were similar across ancestries, with the highest prevalence among Hispanic women (Table). Overall prevalence estimates were 6.3% for those diagnosed at age 50 and 3.6% among those diagnosed at age 80. PVs were most frequently identified in BRCA1, BRCA2, CHEK2, ATM, and PALB2 (Table). Conclusions Among patients with BC of all ancestries, a substantial fraction of patients who do not meet guidelines for genetic testing may unknowingly carry PVs. Elimination of age-based restrictions on genetic testing could improve the survival of patients with BC and positively impact their family members.
| Any gene* | Age 30-34 | Age 40-44 | Age 50-54 | Age 60-64 | Age 70-74 | Age 80-84 |
| All patients | 15.0% | 8.4% | 6.3% | 4.9% | 4.0% | 3.6% |
| Asian | 13.9% | 7.8% | 5.8% | 4.5% | 3.7% | 3.4% |
| Black/African | 14.2% | 8.0% | 6.0% | 4.6% | 3.8% | 3.5% |
| Hispanic | 16.6% | 9.4% | 7.1% | 5.5% | 4.5% | 4.1% |
| Multiple | 14.1% | 7.9% | 5.9% | 4.6% | 3.7% | 3.4% |
| White/Non-Hispanic | 15.1% | 8.5% | 6.4% | 5.0% | 4.0% | 3.7% |
| BRCA1, BRCA2, CHEK2, ATM, or PALB2 | ||||||
| All patients | 13.8% | 7.1% | 4.9% | 3.5% | 2.4% | 2.0% |
| Asian | 13.8% | 7.1% | 4.9% | 3.5% | 2.4% | 2.0% |
| Black/African | 13.0% | 6.7% | 4.6% | 3.3% | 2.3% | 1.8% |
| Hispanic | 16.8% | 8.8% | 6.1% | 4.4% | 3.0% | 2.5% |
| Multiple | 13.5% | 6.9% | 4.8% | 3.4% | 2.4% | 1.9% |
| White/Non-Hispanic | 13.7% | 7.0% | 4.8% | 3.5% | 2.4% | 1.9% |
| BRCA1 or BRCA2 | ||||||
| All patients | 9.5% | 3.9% | 2.1% | 1.2% | 0.7% | 0.5% |
| Asian | 11.7% | 4.8% | 2.7% | 1.5% | 0.9% | 0.7% |
| Black/African | 10.7% | 4.4% | 2.4% | 1.4% | 0.8% | 0.6% |
| Hispanic | 12.9% | 5.4% | 3.0% | 1.7% | 1.0% | 0.7% |
| Multiple | 9.5% | 3.9% | 2.1% | 1.2% | 0.7% | 0.5% |
| White/Non-Hispanic | 8.7% | 3.5% | 1.9% | 1.1% | 0.7% | 0.5% |
|
*Any PV in 25-48 hereditary cancer genes with guideline-based medical management recommendations |
Presentation numberPS3-03-01
Male Breast Cancer: Genomic Landscape, Epidemiological Patterns, and Clinical Outcomes in The United States
lama kamal, Menoufia University, Shebin el-kom- Menoufia, Egypt
l. kamal, E. Belal, a. Beddah, H. Shaheen; Clinical Oncology Department, Menoufia University, Shebin el-kom- Menoufia, EGYPT.
Male Breast Cancer: Genomic Landscape, Epidemiological Patterns, and Clinical Outcomes in The United States Background:Male breast cancer is a rare form of breast cancer that has its distinct biological characteristics. Because of the disease rarity, most data on genomic landscape, epidemiological patterns, and clinical outcomes originate from small studies with limited number of patients. In this study, we aimed to leverage the publicly available genomic and epidemiological datasets to provide the largest to date analysis of clinic-genomic characteristics of male breast cancer in the United States.Methods: We utilized the Surveillance, Epidemiology, and End Results (SEER) database to extract data on patients diagnosed with male breast cancer from the SEER 17 Reg. Nov 2024 submission dataset (including patients diagnosed 2000-2022). SEER collects cancer incidence data from population-based cancer registries covering approximately 45.9 percent of the U.S. population. We used the AACR project GENIE v17.0 database to extract data on genomics of patients with male breast cancer who had samples sequenced and results accessible as part of AACR project GENIE. To avoid duplication, only patients with one profiled sample were included in the analysis. Only genes which were sequenced in at least 50 samples were included in frequency calculations.Results: The epidemiology analysis set included 10,284 patients with male breast cancer. The median age was 68 (interquartile range, 18). Most patients were white (80.3%, n=8,256) and non-Hispanic non-Latino (92.7%, n=9,529). Almost 51.7% (n=5,314) had left-sided breast cancer. In patients with known receptor status, the majority had ER-positive (96.2%, n=8,908), PR-positive (88.2%, n=8,063), and HER2 negative disease (88.1%, n=5,218). Most cases had HR+/HER2- disease subtype (85.9%, n=5,073). Lower proportions had HR+/HER2+, HR-/HER-, or HR-/HER2+ disease (11.2%, n=662; 2.2%, n=128; and 0.8%, n=41; respectively). The age-adjusted incidence rate was 1.2 per 100,000 and the median OS in the overall cohort was 109 months (95% CI: 105.1-112.9).The genomic analysis set included 144 patients. The most frequently mutated genes were PIK3CA (38.2%, n=55/144), GATA3 (21.6%, n=29/134 profiled), KMT2C (14.1%, n=14/99), TP53 (13.9%, n=20/144), and BRCA2 (10.4%, n=14/135). Structural variants were most frequently observed in INPPL1 (3.4%, n=2/58), CDK12 (1.7%, n=2/121), FGFR2 (1.6%, n=2/128), FGFR1 (1.6%, n=2/128), and NSD3 genes (1.2%, n=1/81). The most frequent copy number alterations were amplifications in FGF19 (19.4%, n=14/72), FGF4 (19.4%, n=14/72), FGF3 (19.4%, n=14/72), CCND1 (17.8%, n=18/101), and FGFR1 (14.9%, n=15/101).Conclusions: Male breast cancer occurs at a rate of 1.2 per 100,000 and has a median OS of 9 years. Patients mostly present with ER+ HER- disease and a molecular profile is quite different from female breast cancer.
Presentation numberPS3-03-02
Factors associated with uptake of endocrine prevention in patients with atypical hyperplasia and lobular carcinoma in situ
Hamda Almarzooqi, McGill university, Montreal, QC, Canada
H. Almarzooqi1, N. Cuillerier1, S. Wong1, V. Villareal-Corpuz2, M. Basik1; 1General Surgery, McGill university, Montreal, QC, CANADA, 2Stroll Cancer Prevention Centre, Jewish General Hospital, Montreal, QC, CANADA.
INTRODUCTION: Women diagnosed with atypical hyperplasia (AH) and lobular carcinoma in situ (LCIS) are at higher risk of developing invasive breast cancer. This risk may be reduced by endocrine prevention, though patient uptake and compliance are low. We sought to evaluate factors associated with the uptake and initiation of endocrine prevention therapy among women with AH and LCIS within a real-world setting.METHODS: This retrospective cohort study included women with high-risk lesions (HRL) diagnosed on biopsies performed between 2014 and 2025 and referred to the Jewish Stroll Cancer Prevention Centre and Stroll Cancer Prevention Centre, including women with AH and classical LCIS. The primary outcome was uptake of endocrine prevention, defined as receipt of a prescription and endocrine prevention initiation in eligible women.RESULTS: Endocrine prevention was prescribed to 106 (56.4%) of 188 eligible patients, and 71 (67.0%) started treatment. The regimen most frequently prescribed was low-dose tamoxifen at 64.2%, Raloxifene at 19.8%, and standard-dose tamoxifen at 11.3%. The highest uptake was 73.4% for women aged 51-60 years compared to 56.5% of women aged 41- 50 years, and 41.5% of women aged above 60 years (p < 0.001). A high uptake rate was also observed in parous women (64.6% vs 44.0%, p=0.005) and in women with a history of oral contraceptive use (69.5% vs. 46.2%, p=0.001). Before 2019, prescription rates were similar between Raloxifene and standard dose tamoxifen (47.1% each). However, low-dose tamoxifen became the most commonly prescribed regimen after 2019 (76.4%, p<0.001).CONCLUSION: After 2019, low-dose tamoxifen became the most commonly prescribed endocrine prevention regimen in HRL patients who accepted a prescription.
Presentation numberPS3-03-03
Evaluating the recommendations of community-based surgeons for germline genetic testing in patients with newly diagnosed localized breast cancer
Neil Love, Research To Practice, Key Biscayne, FL
N. Love1, K. S. Hughes2, M. Robson3, G. Kelly1, T. Wallace1, T. Cruse1, K. Miller1, K. A. Ziel1, K. H. Pang1, D. Paley1; 1Clinical Content Department, Research To Practice, Key Biscayne, FL, 2Department of Surgery, Medical University of South Carolina, Charleston, SC, 3Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY.
BACKGROUND: Germline genetic testing has important implications for both the patient with breast cancer and family members. The first physician with the opportunity to consider this option is usually the surgeon. Current practice guidelines commonly consulted on this issue from the American Society of Clinical Oncology (ASCO), the National Comprehensive Cancer Network (NCCN) and the American Society of Breast Surgeons (ASBrS) provide differing recommendations regarding which patients should be tested. We sought to determine how age, ER/PR and HER2 status and other variables factor into surgeons’ recommendations for testing. METHODS: From February to March 2025, 50 US-based surgeons in community practice were recruited to complete a survey designed to identify clinical situations when germline genetic testing is recommended. For each case-based question, a number of factors (age, ER/PR and HER2 status, family history, gender) were varied. Ten surgeons at academic centers were also recruited to complete the survey. A modest honorarium was provided. RESULTS: For a 30-year-old patient without a family history (all 3 guidelines recommend testing), almost all respondents recommend testing regardless of ER/PR/HER2 status (Table). For a similar 55-year-old patient, almost all participants recommended testing in patients with TNBC (as is consistent with guidelines), but less than half would test patients with ER/PR-positive tumors regardless of HER2 status (testing recommended by ASCO and ASBrS and a consideration per NCCN). For a 70-year-old patient (only ASBrS recommending testing), again almost all respondents recommended testing for patients with TNBC, but far fewer respondents would test patients with ER/PR-positive tumors regardless of HER2 status. There was also agreement that all men with localized breast cancer should be tested. Interestingly, the responses from the community-based cohort largely aligned with those of their academic counterparts. CONCLUSION: The findings from this survey demonstrate that whether a patient with newly diagnosed localized breast cancer receives a recommendation for germline genetic testing depends largely on the practice pattern of the surgeon initially managing the case as many do not initiate testing for patients 55 years and older with ER/PR-positive tumors. While other physicians, including medical oncologists, may seek out this information at a later time as they weigh adjuvant therapy options, it is important to consider that the main downside of genetic testing is financial. Given the important implications for family members and the overall survival benefit associated with olaparib in the OlympiA trial, perhaps in the future genetic testing will join ER and HER2 assessment as a standard for all patients with localized disease.
| Tumor Histology |
30-year-old |
55-year-old |
70-year-old |
| ER/PR-positive
HER2-negative
|
47/50 (94%) | 16/50 (32%) | 12/50 (24%) |
|
ER/PR-negative
HER2-negative |
49/50 (98%) | 45/50 (90%) | 38/50 (76%) |
|
ER/PR-positive
HER2-positive |
47/50 (94%) | 24/50 (48%) | 18/50 (36%) |
Presentation numberPS3-03-04
Predictive Factors of High Risk Were Indirectly Compared in Node-negative Stage I-II Breast Cancer Patients Examined Oncotype Dx or EndoPredict: The BRAIN Study
Jee Hyun Ahn, Yonsei university, Seoul, Korea, Republic of
J. Ahn; General surgery, Yonsei university, Seoul, KOREA, REPUBLIC OF.
Purpose There have been reported different characteristics of multigene assays (MGAs) to discriminate high and low risk groups in estrogen receptor (ER)-positive/HER2-negative breast cancer patients. In real world data, however, direct comparison between MGAs is not easy because clinical guidelines recommended against using two or more MGAs in a patient simultaneously. In this study, clinicopathological characteristics and results of Oncotype Dx (ODx) and EndoPredict (EP) were indirectly compared to investigate characteristics of each assay. Methods A total of 1,782 node-negative stage I/II patients with ER-positive/HER2-negative breast cancer were retrospectively selected between 2013 and 2025. Of them, 1,537 (86.3%) patients performed ODx and 245 (13.7%) examined EP. High risk of ODx was defined considering clinical risk and recurrence score. EPclin was used to determine high risk of EP. Clinicopathological characteristics and assay results were compared using logistic regression models. After 1:1 propensity score matching (PSM) adjusting clinicopathological characteristics, predictive factors for high risk assay result were re-analyzed. Results High risk of ODx and EP was 25.4% and 24.5%, respectively. EP patients showed older age, more obese, and higher mixed/lobular/special histology. ODx patients demonstrated larger tumor size, higher grade II/III disease, and higher Ki-67 levels. Allred score of ER and progesterone receptor (PR) and HER2 immunohistochemical score were not different. Correlation analysis showed ODx recurrence score was significantly associated with ER (Spearman’s rho, -0.079) and PR (Spearman’s rho, -0.425) Allred score negatively and with Ki-67 levels (Spearman’s rho, 0.251) positively. EP 12-gene molecular score was negatively associated with PR Allred score (Spearman’s rho, -0.290) and positively related to Ki-67 values (Spearman’s rho, 0.516). Multivariate analysis adjusting clinicopathological characteristics showed that EP had a higher probability of high risk [Hazard ratio, 1.783 (95% confidence interval, 1.330-2.390); p<0.001]. Subset analysis by each assay showed younger age, lower ER and PR Allred score, and HER2 2+ immunohistochemical score were major determinants of high risk in ODx patients. But larger T2-3 stage, histologic grade II/III, and Ki-67 ≥20% were significant factors of high risk in EP cases. After 1:1 PSM, assay method was not significant [Hazard ratio, 1.286 (95% confidence interval, 0.761-2.174); p=0.348] but T2-3 stage, grade II/III, low PR, and higher Ki-67 were independently significant predictors of high risk result. Conclusion Each MGA showed different features of major determinants for high risk group. Although definition of high risk considering clinical risk factors and gene score might be partly related, ODx was strongly dependent on ER-related signatures and EP was significantly determined by proliferative features of histologic grade and Ki-67 levels. Primary physician and patient could select one among available MGAs based on understanding of its distinct characteristics.
Presentation numberPS3-03-05
High-prevalence of NF1 pathogenic variants in cancer patients without classic features of Neurofibromatosis type 1
Dimitrios Lachanas, Genekor Medical SA, Attica, Greece
A. Koutras1, K. Papazisis2, I. Boukovinas3, R. Iosifidou4, I. Intzidis4, K. Potska5, C. Dogka5, A. Katseli5, D. Paranou5, A. Karavaggeli5, A. Meintani5, D. Bouzarelou5, D. Lachanas5, G. Rigas6, M. Kanara7, M. Müslümanoglu8, I. Bobolaki9, E. Biziota10, E. Papadopoulou5, G. Nasioulas5; 1Medical School, University of Patras, Patras, GREECE, 23rd Dpt of Oncology, Interbalcan European Medical Centre, Thessaloniki, GREECE, 3Oncology unit, Bioclinic, Thessaloniki, GREECE, 4Breast Surgical Oncology Clinic, Theageneio Anticancer Hospital, Thessaloniki, GREECE, 5Molecular Oncology, Genekor Medical SA, Attica, GREECE, 6Breast Clinic, Agios Savvas Cancer Hospital, Attica, GREECE, 7Breast Clinic, General Hospital of Trikala, Trikala, GREECE, 8Breast Clinic, Istanbul University School of Medicine, Instabul, TURKEY, 9Oncology Department, General Hospital of Chania, Crete, GREECE, 10Oncology Department,, Universal Hospital of Alexandroupoli, Alexandroupoli, GREECE.
Background: Neurofibromatosis type 1 (NF1), is a classic example of a complete penetrance disease with an estimated prevalence of 1 in 3000. Βroader application of multigene panel testing in candidate hereditary cancer predisposition cases, reveals pathogenic NF1 variants in Breast/ovarian/colorectal cancer patients without a clinical NF1 diagnosis. The scope of this study is to enlighten the contribution of NF1 to cancer risk in general population and the need of medical management guidelines in NF1 breast/ ovarian cancer patients lacking the relevant clinical phenotype. Methods: This retrospective observational study included patients with mutated germline NF1, referred at GENEKOR Medical SA, Athens, Greece between the period of 1st Jan 2020 to 31st Oct 2024. Ηigh risk cancer patients, with no neural crest derivant malignancies, underwent germline mutation testing, with a 52 gene panel. All the NF1 mutations were crosschecked via the ClinVar mutation database to be classified as pathogenic. Results: This retrospective observational study included 15 patients with incidentally identified mutated germline NF1, who were referred due to breast cancer (5/15, 33%), ovarian cancer (4/15,27 %), colorectal cancer (2/15,13%), multiple myeloma (1/15,7%) and glioblastoma (1/15,7%). Among the 12782 cancer patients, with no neural crest derivant malignancies, who underwent germline high-risk cancer gene susceptibility testing, 2616 (20.5%) cases had pathogenic mutations and 15 out of them (0.6%) were NF1 mutated. We identified an unexpectedly high prevalence (1 out of 852), of PVs in the NF1 gene, more than 3 times the rate expected given the reported prevalence of NF1. We identified 13 pLOF variants, 1 missense, 1 single amino acid deletion and 1 deletion involving the entire NF1 gene, all in heterozygosity, with the exception of 2 samples with blood mosaicism. Physical exam, medical and family history revealed no features consistent with NF1 diagnosis contrary to the complete penetrance of the disease. In 3 samples, additional PVs were identified, in BRCA2 (ovarian cancer), ATM and NTHL1 (2 colorectal cancer cases) genes. Conclusions: This study highlights that NF1 PVs are more frequent than previously estimated and often discovered in unaffected individuals. Incomplete penetrance and missed diagnoses probably explain this. The aforementioned data presented provides insights into the need of establishment of screening and management strategies in patients with incidental findings.
Presentation numberPS3-03-06
Hepatosteatosis as a risk factor for development of breast cancer: A single center experience
Bulent Citgez, Halic University, Memorial Hospital, Istanbul, Turkey
B. Citgez1, B. Yigit2, E. Namal3; 1General Surgery, Halic University, Memorial Hospital, Istanbul, TURKEY, 2General Surgery, Bagcilar Training and Research Hospital, Istanbul, TURKEY, 3Oncology, Memorial Hospital, Istanbul, TURKEY.
Background: Hepatosteatosis is frequently observed in breast cancer patients due to shared risk factors such as obesity, metabolic syndrome, and advanced age. The aim of this study is to quantitatively evaluate the association between hepatosteatosis and early-stage breast cancer by examining routine abdominopelvic computed tomography images obtained for staging purposes in breast cancer patients.Methods: The imaging and histopathology reports of patients who presented to Memorial Hospital for breast screening or diagnostic imaging were reviewed retrospectively. Patients with advanced-stage breast cancer, additional malignancies, or significant comorbidities were excluded from the study. A total of 129 patients (Group 1) who underwent surgery for early-stage breast cancer between January 2017 and January 2025 were included in this retrospective analysis. A control group (Group 2) was formed from 103 patients without a history of cancer or significant comorbidities. Liver attenuation values on abdominopelvic computed tomography, as well as serum transaminases (AST, ALT), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP) and total bilirubin levels were recorded and compared with those of the control group. Hepatosteatosis was evaluated using a four-point scale ranging from 0 to 3, where higher grades reflect greater degrees of lipid accumulation in the liver tissue.Results: There were no statistically significant differences between Group 1 and Group 2 in terms of age, AST, ALT, GGT, ALP, or total bilirubin levels (all p > 0.05) (Table 1). The rate of patients with moderate to severe hepatosteatosis (grades 2-3) was significantly higher in Group 1 than in Group 2, whereas the rate of patients with grade 0-1 hepatosteatosis was significantly higher in Group 2 than in Group 1 (18.6% vs. 8.74% and 91.26% vs. 8.74%, respectively; p= 0.033).Conclusion: The study results indicated an increased prevalence of hepatosteatosis in early-stage breast cancer patients when compared to control group. Lifestyle modifications, weight loss, increased regular physical activity, and a healthy diet help reduce hepatosteatosis and insulin resistance, thereby preventing the progression of non-alcoholic fatty liver disease. Considering the evidence suggesting that hepatosteatosis may increase the risk of breast cancer development, clinicians should keep in mind the importance of planning and implementing preventive strategies in this patient group.
| Group 1 | Group 2 | p | ||
| Patients | n(%) | 129 | 103 | |
| Age | Mean±Sd, Median (Min-Max) | 54.36±13.02, 53 (27-88) | 53.36± 10.89, 54 (27-85) | 0.529 |
| AST (IU/ml) | Mean±Sd, Median (Min-Max) | 18.51±6.82, 17 (8-53) | 19.19±7.57, 17 (10-69) | 0.471 |
| ALT (IU/ml) | Mean±Sd, Median (Min-Max) | 17.32±10.35, 14 (4-77) | 18.84±11.8, 15 (7-84) | 0.286 |
| GGT (IU/ml) | Mean±Sd, Median (Min-Max) | 25.48±48.55, 16 (7-536) | 22.06±21.5, 16 (5-159) | 0.729 |
| ALP (IU/ml) | Mean±Sd, Median (Min-Max) | 81.24±34.67, 78 (39-346) | 75.07±26.61, 68 (32-187) | 0.100 |
| Total Bilirubin (mg/dL) | Mean±Sd, Median (Min-Max) | 0.5±0.26, 0.45 (0.14-1.52) | 0.48±0.26, 0.44 (0.14-1.44) | 0.308 |
Presentation numberPS3-03-07
Breast and Ovarian Cancer Genomic Risk Assessment: 10-Year Experience from a High-Risk Program
Pamela Mora, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
P. Mora1, M. Acosta1, J. Herzog2, N. Valdiviezo3, L. Reynaga4, J. Abugattas5, Y. Sullcahuaman1, T. Vidaurre3, S. Neciosup3, J. Dunstan5, H. Gómez6, I. Otoya3, Z. Morante3, V. Acuña7, A. López8, S. Gruber4; 1Unidad Funcional de genética y biología molecular, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 2Division of Clinical Cancer Genomics Molecular Laboratory, City of Hope National Medical Center, Duarte, CA, 3Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 4Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, 5Cirugía de Mamas y Tejidos Blandos, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 6Instituto de Investigaciones de Ciencias Biomédicas, Universidad Ricardo Palma, Lima, PERU, 7Facultad de Farmacia y Bioquímica, Universidad Nacional Mayor de San Marcos, Lima, PERU, 8Cirugía Ginecológica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU.
Breast cancer (BC) is one of the most common cancers among women in both Peru and worldwide. Ovarian cancer (OC), due to the difficulty of early detection, also represents a significant public health concern in the country. In recent years, genetic testing has become essential for identifying individuals at high risk for hereditary cancer predisposition syndromes, guiding decisions related to prevention, early detection, and treatment. In Peru, the use of genetic testing is still developing. This work presents an inter-institutional collaborative experience that has facilitated access to genetic testing and contributed to the implementation of a multidisciplinary care system for managing high-risk patients. The main objective of this study is to identify the genomic profile of patients with breast and/or ovarian cancer and to evaluate the clinical impact of genetic testing on cancer management. Methods: This study includes individuals diagnosed with breast and/or ovarian cancer who were referred for Medical Genetics evaluation at the Instituto Nacional de Enfermedades Neoplásicas (INEN) between August 2013 and March 2020. Participants met criteria for inclusion in the inter-institutional protocol INEN13-48 on hereditary cancer and underwent genetic testing at City of Hope (IRB #96144).Results and Discussion: We recruited 1577 patients; 99.3% were females (n=1566/1577) with the initial diagnosis of BC (89.4%. n=1401/1566), OC (8.5%. n=133/1566) and BC/OC (2%. n=32/1566); 0.7% were males (n=11/1577) all BC. Of all the individuals, 46.8% (n=738/1577) had the first diagnosis under 40yo; 36.1% (n=570/1577) was between 41-50yo. 13.4% (n=212/1577) had a LP/P variant in an actionable gene with management guidelines for at least one cancer type; 0.8% (n=12/1577) had a LP/P variant in an non actionable gene; 34.5% (n=545/1577) had a BRCA1/BRCA2 panel with no variant detection and 51.2% (n=808/1577) had an extended panel with no variant detection. The most frequent LP/P variants in actionable genes were 53.8% (n=114/212) in BRCA1 and 24% (n=51/212) in BRCA2; 1% (n=2/212) had more than one LP/P variant (BRCA1/BRIP1 and BRCA1/BRCA2). The detection rate of LP/P variant in BC was 12% (n=170/1401) in BC, 19.5%(n=26/133) in OC and 50% (n=16/32) in B/OC. The most frequent LP/P variants were BRCA1(NM_007294.4):c.68_69del (p.Glu23ValfsTer17) (6%: n=13/212) and BRCA2(NM_000059.4):c.1219C>T (p.Gln407Ter) (4% n=8/212). 9% (n=1/11) of the male patients, had a variant in BRCA2 gene. All 212 patients were evaluated in clinical genetics. Over 10 years of follow-up, 72% (n=152) received recommendations for high-risk breast cancer (BC) management and 54% (n=115) for ovarian cancer (OC); the rest did not due to disease progression or geographic barriers. Among those at high BC risk, 34% (n=60) began imaging-based surveillance, 26% (n=40) underwent unilateral/bilateral risk-reducing mastectomy (RRM)—with 10% (n=4) diagnosed with new BC—and 34% (n=52) declined recommendations. In the high OC risk group, 18% (n=21) had prior ovarian surgery, 49% (n=56) underwent risk-reducing salpingo-oophorectomy (RRSO)—with 2 OC diagnoses—and 23% (n=27) declined. As of now, 36.7% (n=78) have died and 17% (n=37) are lost to follow-up.Conclusion: The importance of ensuring access to genetic studies, in addition to the recognition of genetic predisposition syndromes to cancer is paramount. In this cohort we identified that 13% of individuals had an LP/P in an actionable gene, similar to other cohorts. This information support the needs of long-term financing, not only to access to genetic testing, but helps us to improve the access to multidisciplinary high-risk management in our institution considering the geographical and economic barriers in our country.
Presentation numberPS3-03-09
Charting the shared genetic architecture between breast cancer and major depressive disorder based on genome-wide association study
Yilong Lin, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
Y. Lin1, J. She1, R. Zhao1, Y. Zhang2, A. Qiu1, L. Zhang1, Q. Yang1; 1Department of Breast Surgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, CHINA, 2Medical college, Guangxi University, Nanning, CHINA.
Background: An increasing body of epidemiological evidence has suggested that breast cancer (BC) and major depressive disorder (MDD) are frequently comorbid, with MDD being particularly prevalent among BC patients during diagnosis and treatment. However, whether this relationship reflects a shared genetic predisposition or a causal biological link remains unclear. Here, we aimed to comprehensively investigate the shared genetic architecture and potential causal associations between BC and MDD using large-scale genome-wide association study (GWAS) data. Methods: We obtained publicly available GWAS summary statistics for BC and MDD. First, linkage disequilibrium score regression (LDSC) was used to estimate the genome-wide genetic correlation between the two traits. To identify shared genetic variants (pleiotropic loci), we conducted multiple cross-trait GWAS analyses including multi-trait analysis of GWAS (MTAG), cross-phenotype association (CPASSOC), and pleiotropic analysis under composite null hypothesis (PLACO). We then functionally annotated PLACO-identified SNPs using Functional Mapping and Annotation (FUMA) to identify lead variants and genomic risk loci. Gene-level associations were further evaluated using MAGMA to uncover shared susceptibility genes. Finally, we used both Generalized Summary-data-based Mendelian Randomization (GSMR) and two-sample Mendelian Randomization (TSMR) approaches to assess the direction and magnitude of potential causal effects between MDD and BC. Results: LDSC revealed a significant positive genetic correlation between BC and MDD (rg = 0.0545, SE = 0.0151, P = 3.0 × 10⁻⁴), indicating a modest but meaningful shared genetic basis. MTAG analysis identified 17 pleiotropic SNPs reaching genome-wide significance. CPASSOC and PLACO analyses revealed 16,470 and 775 pleiotropic SNPs, respectively, suggesting widespread cross-trait genetic overlap. FUMA-based annotation of PLACO hits yielded 22 independent genomic loci and MAGMA further identified 130 pleiotropic genes, highlighting biological convergence between psychiatric and oncologic pathways. GSMR (OR = 1.27, 95% CI: 1.11-1.46, P = 5.90 × 10⁻⁴) and TSMR (OR = 1.12, 95% CI: 1.02-1.22, P = 1.43 × 10⁻2) demonstrated significant causal effects of MDD on increased BC risk. The sensitivity analysis suggested that the results of TSMR were not affected by horizontal pleiotropy (P = 0.75). Conclusions: Our study charted the shared genetic architecture between BC and MDD and explored their potential causal relationship. The findings enhanced the understanding of the genetic pathways commonly involved in both BC and MDD, highlighting their complex interconnections. This research provided valuable insights for experimental studies aimed at developing new preventive measures and therapeutic interventions for BC and MDD.
Presentation numberPS3-03-10
Whole exome sequencing reveals germline pathogenic variant landscape in multi-center Taiwanese breast cancer patient cohort at moderate to high hereditary genetic risks
Wen-Ling Kuo, Chang Gung Memorial Hospital, Linkou, Tauoyuan, Taiwan
W. Kuo1, W. Shen2, H. Chou1, M. Peng3, S. Huang4, S. Chen5, Y. Lin3; 1Breast Cancer Center, Chang Gung Memorial Hospital, Linkou, Tauoyuan, TAIWAN, 22.Medical Oncology, Chang Gung Memorial Hospital, Linkou, Tauoyuan, TAIWAN, 3Medical Oncology, Chang Gung Memorial Hospital, Linkou, Tauoyuan, TAIWAN, 4Medical Oncology, Chang Gung Memorial Hospital, Kaohsiung, Tauoyuan, TAIWAN, 5General and Breast Surgery, Chang Gung Memorial Hospital, Linkou, Tauoyuan, TAIWAN.
Background: The incidence of breast cancer in East Asia has been steadily increasing, with cases presenting approximately a decade earlier than those observed in Western populations. This study utilizes whole-exome sequencing (WES) to investigate the prevalence of hereditary germline pathogenic or likely pathogenic variants (PV) in Taiwanese breast cancer patients identified as having moderate to high hereditary genetic risk. Method: Breast cancer patients with age at diagnosis less than 40 years, positive familial cancer history, bilateral cancer or multiple cancers, and triple negative breast cancer (TNBC) were enrolled from the Linkou, Taipei, and Kaohsiung hospital branches of Chang Gung Medical Foundation. BRCA1/2 likelihood was evaluated with the pathology-adjusted Manchester Scoring System (MSS3). (1) Whole-exome sequencing (WES) was conducted using the IDT Lotus DNA Library Prep Kit and xGen Exome Research Panel v2, with sequencing on the NovaSeq 6000 platform. Reads were aligned to the GRCh38 genome reference using DRAGEN v.3.9.3 with stringent quality metrics. Variants were filtered against databases such as dbSNP version 150 and Taiwan Biobank, annotated via wANNOVAR, and BRCA1/2 PVs were confirmed using Sanger sequencing. Results: A cohort of 525 breast cancer patients were enrolled, revealing 36 individuals (6.9%) carring confirmed germline BRCA1/2 PVs and another 28 patients (5.3%) carrying PVs in other non-BRCA hereditary breast and ovarian cancer (HBOC)-related genes (ATM, BARD1, BRIP1, CHEK2, MUTYH, PALB2, PMS2, RAD50, RAD51D, and TP53) (Table1). Among the risk groups classified by the Manchester Scoring System, 72.8% were categorized as low (20%) groups. The detection rate of BRCA PV was 1.6% (6/382), 11.3% (9/80), and 33.3% (21/63), respectively, in low to high-risk Manchester categories. For patients diagnosed with breast cancer under age 40 (n= 291,55%), the prevalence of BRCA1/2 PV was observed at 7.2% (21/291). Among patients with TNBC (n=79), the proportion of patients with BRCA1/2 PVs was notably higher, at 16.5% (n=13). In cases of bilateral breast cancer (n=47) (synchronous or metachronous), 12.8% (n=6) had BRCA1/2 PVs. Among the 16 male breast cancer patients, no BRCA 1/2 PVs were detected. For individuals with a high Manchester Score but no detected BRCA1/2 PVs through sequencing, further analysis using the MLPA assay remains ongoing.Conclusion: Whole-exome sequencing has provided valuable insights into the prevalence and distribution of germline pathogenic variants in HBOC-related genes among Taiwanese breast cancer patients with moderate to high hereditary risk. (2) The occurrence rates of BRCA1/2 pathogenic variants in the overall cohort and within triple-negative breast cancer patients align with trends observed in other breast cancer populations. (3) 1. Evans DG, Harkness EF, Plaskocinska I, Wallace AJ, Clancy T, Woodward ER, et al. Pathology update to the Manchester Scoring System based on testing in over 4000 families. (1468-6244 (Electronic)).2. Felicio PS, Grasel RS, Campacci N, de Paula AE, Galvão HCR, Torrezan GT, et al. Whole-exome sequencing of non-BRCA1/BRCA2 mutation carrier cases at high-risk for hereditary breast/ovarian cancer. Hum Mutat. 2021;42(3):290-9.3. Lee NY, Hum M, Amali AA, Lim WK, Wong M, Myint MK, et al. Whole-exome sequencing of BRCA-negative breast cancer patients and case-control analyses identify variants associated with breast cancer susceptibility. Hum Genomics. 2022;16(1):61.
Presentation numberPS3-03-11
Hereditary Breast Cancer Beyond BRCA1/2: Real-World Value of Cascade Testing in Southern Europe cohort
Juncal CLAROS, HOSPITAL UNIVERSITARIO SEVERO OCHOA, MADRID, Spain
J. CLAROS1, I. SISO1, M. ECHARRI1, D. MORCHON2, L. PIZARROSO1, J. MESA1, C. ORTEGA1, A. GARRIDO1, C. PERNAUT1, H. FRAILE1, A. PEÑALVER1, B. ALONSO3, A. LOPEZ1; 1MEDICAL ONCOLOGY, HOSPITAL UNIVERSITARIO SEVERO OCHOA, MADRID, SPAIN, 2MEDICAL ONCOLOGY, COMPLEJO ASISTENCIAL UNIVERSITARIO DE SALAMANCA, SALAMANCA, SPAIN, 3INTERNAL MEDICINE, HOSPITAL UNIVERSITARIO SEVERO OCHOA, MADRID, SPAIN.
Background:Multi-gene panel testing has expanded the identification of hereditary breast cancer syndromes. ESMO Precision Oncology Working Group recently published recommendations advising against routine testing of certain genes suggesting minimal mortality benefit (ATM, CHEK2). We present the germline findings from a large, single institution cohort in Southern Europe, focusing on the diagnostic yield of high- and moderate-penetrance genes and the clinical context of variant identification.Methods:We conducted a retrospective analysis of patients (pts) referred to our Hereditary Cancer Genetic Counseling Unit between May 2023 and April 2025. Variant classification followed American College of Medical Genetics (ACMG) criteria.Results:From a total of 518 pts referred for counseling, 387 were tested and included in the analysis (median age 57). Overall, 91 pts (23.5%) carried a pathogenic/likely pathogenic (P/LP) variant (Class 4/5) and 34 (8.8%) had a variant of uncertain significance (VUS). Among the 151 (39%) patients with a personal history of BC, the specific diagnostic yield for a P/LP variant was 14.56% (n=22), while the VUS rate was 15.2% (n=23). Among P/LP carriers, 51 (56%) were in high-penetrance breast cancer genes: BRCA1 (n=24), BRCA2 (n=24) and PALB2, PTEN, CDH1 (n=1 each). 36 of 51 (70.5%) of these carriers were identified via cascade testing of healthy relatives. 18 of 91 pts (19.7%) P/LP variants were found in moderate-penetrance genes: ATM (n=9), CHEK2 (n=6), RAD51C (n=2), BRIP1 (n=1). Of note, 11 variants (61%) were identified through cascade testing rather than personal history of cancer. Conclusion:In this real-world cohort, nearly a quarter of individuals undergoing testing carried a clinically significant germline variant. While BRCA1/2 remain dominant, a substantial number of variants in genes like ATM, CHEK2, and RAD51C were discovered through cascade testing, mainly in unaffected carriers. This highlights the clinical challenges surrounding the moderate penetrance genes, including appropriate follow-up and counseling strategies.
| GENE | TOTAL-CARRIERS(n) | BCcases(n) | MULTIGENE-PANEL(n) | CASCADE-TESTING(n)(F;M) |
| BRCA1 | 24 | 8 | 8 | 16(10;6) |
| BRCA2 | 24 | 7 | 5 | 19(14;5) |
| PALB2 | 1 | 0 | 1 | 0 |
| PTEN | 1 | 0 | 1 | 0 |
| CDH1 | 1 | 0 | 0 | 1(0;1) |
| ATM | 9 | 3 | 1 | 8(7;1) |
| CHEK2 | 6 | 3 | 3 | 3(3;0) |
| RAD51C | 2 | 0 | 0 | 2(2;0) |
| BRIP1 | 1 | 1 | 1 | 0 |
| TOTAL | 69 | 22 | 20 | 49 |
Presentation numberPS3-03-12
From Pathogenic Variant to Action: Genetic Risk Identification and Preventive Oncology in Brazil’s Public Health System
Elisângela P Silveira-Lacerda, Universidade Federal de Goiás, Goiânia, Brazil
L. P. Lacerda1, P. F. Silva2, K. M. Veiga2, B. F. Nascimento2, I. M. Calassa2, W. D. Oliveira3, A. S. Limongi4, R. L. Mesquita5, K. J. Monteiro6, R. R. Santos Junior5, R. Taminato7, R. M. Rahal8, E. P. Silveira-Lacerda9; 1Medicine, Universidade do Planalto Central, Brasília, BRAZIL, 2Genética, Universidade Federal de Goiás, Goiânia, BRAZIL, 3Genetic, Universidade Federal de Goiás, Goiânia, BRAZIL, 4Superintendência de Políticas e Atenção Integral à Saúde, Secretaria Estadual de Saúde, Goiânia, BRAZIL, 5SPAIS – Superintendência de Políticas e Atenção Integral à Saúde, SES – Secretaria Estadual de Saúde de Goiás, Goiânia, BRAZIL, 6Superintendência de Políticas e Atenção Integral à Saúde, Secretaria Estadual de Saúde de Goiás, Goiânia, BRAZIL, 7Farmácia, Hospital Araújo Jorge, Goiânia, BRAZIL, 8Centro Avançado de Diagnóstico de Mama, Universidade Federal de Goiás, Goiânia, BRAZIL, 9Centro de Genética Humana – ICB, Universidade Federal de Goiás, Goiânia, BRAZIL.
Approximately 10% of breast and ovarian cancer cases are hereditary. Identifying germline pathogenic variants and acting on this information through preventive strategies is a key pillar of precision oncology. In Brazil, the public health system (SUS) in Goiás has incorporated genetic risk screening and management, setting a precedent for accessible, population-level genomic medicine.This study aimed to evaluate the frequency of germline pathogenic variants in patients at risk for hereditary breast and ovarian cancer, and to track these variants among their relatives for early diagnosis and prevention. We evaluated 268 patients who met the 2025 National Comprehensive Cancer Network (NCCN) criteria for hereditary breast and ovarian cancer syndromes, alongside 268 parents. All participants were referred to the Human Genetics Center at UFG through the SUS network.Following pre-test counseling and informed consent, blood samples were collected and analyzed using next-generation sequencing (NGS) with the Oncomine™ BRCA Expanded Panel (Ion Torrent platform). Among the 268 index patients, 14% (38/268) had pathogenic germline variants. The distribution was: BRCA1 (50%, 19/38), TP53 (21%, 8/38), PALB2 (10.5%, 4/38), CHEK2 (2.9%), BRCA2 (5.3%), and ATM (5.3%).Further familial testing was conducted in 30 families. Among eight families with the TP53 c.1010G>A variant, 36 of 99 tested relatives (36.4%) were positive. In two BRCA2-positive families, 9 of 18 relatives (50%) carried the same variant. Altogether, 45 of 117 relatives (38.46%) tested positive for the familial variant.Crucially, all relatives with positive results were evaluated by a medical geneticist and referred for cancer surveillance and risk-reducing strategies according to NCCN 2025 guidelines. This included MRI, mammography, and risk-reducing surgery when indicated, ensuring translation from genetic finding to clinical action.The project established a structured, replicable workflow for managing hereditary cancer risk in families. This includes genetic counseling, testing, cascade screening, and follow-up within the public healthcare system. By integrating genetic risk management into routine SUS care, the program enables early-stage diagnosis and reduces cancer-related mortality, particularly in underserved populations.This initiative demonstrates that comprehensive genomic care and precision prevention are feasible within a public system and scalable to other regions. It highlights how transforming a genetic finding into preventive action can generate real-world impact bridging the gap between variant and value.
Presentation numberPS3-03-13
Global Ancestry Shapes Clinical and Genetic Profiles in an Admixed Colombian Breast Cancer Cohort.
William A Mantilla, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center, Bogota, Colombia
W. A. Mantilla1, Y. T. Zambrano-O2, L. Rey-Vargas2, D. C. Sierra-Diaz3, M. Lema4, D. Y. Fonseca-Mendoza3, S. J. Serrano-Gomez5, C. M. Restrepo3, M. C. Sanabria-Salas5; 1Breast Cancer Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center, Bogota, COLOMBIA, 2Grupo de Investigacion Biologia del Cancer, Instituto Nacional de Cancerologia, Bogota, COLOMBIA, 3School of Medicine and Health Sciences, Center for Research in Genetics and Genomics (CIGGUR), Universidad del Rosario, Bogota, COLOMBIA, 4Medical Department, Clinica de Oncologia Astorga, Medellin, COLOMBIA, 5NA, NA, Bogota, COLOMBIA.
Background: Breast cancer (BC) susceptibility is driven by a complex interplay of modifiable and non-modifiable risk factors. While the impact of BRCA1 and BRCA2 pathogenic/likely pathogenic variants (PVs) is well documented, the contribution of genetic ancestry to the distribution of PVs in BRCA1/2 and other homologous recombination repair (HRR) genes remains understudied in genetically admixed populations such as Colombians. Methods: Whole-exome sequencing data from 358 unselected Colombian women with BC were analyzed for genetic ancestry. Among them, 24 individuals were identified as carriers of PVs in BRCA1 (n=3), BRCA2 (n=10), PALB2 (n=2), ATM (n=5), CHEK2 (n=2), BARD1 (n=1), and RAD51D (n=1), as classified by ACMG/AMP criteria. Raw BAM files underwent preprocessing, quality control, and joint genotyping, yielding 14.9 million variants. Quality filtering using PLINK v1.9 included genotype call rate ( 1e-6), minor allele frequency (>0.1), and linkage disequilibrium pruning. Samples with >25% missingness were excluded, resulting in a final dataset of 344 individuals and 95,671 variants (genotyping rate: 99.93%). Global ancestry proportions were estimated using principal component analysis with reference populations from the 1000 Genomes and Human Genome Diversity Projects, specifically African (AFR), European (EUR), and Native American (NAM) populations. Statistical comparisons of ancestry proportions were performed across clinical variables (e.g., age at diagnosis, family history), genetic status (PVs carriers vs non-carriers), and tumor subtypes. Results: Global ancestry was predominantly EUR (mean ~53%), followed by NAM (~36%) and AFR (~10%). Significant differences in ancestry distribution were observed across clinical subgroups. AFR ancestry was higher among cases born in the Caribbean and Pacific coast regions (18% and 11%, respectively; p < 0.001), while EUR ancestry was higher among those from the Andean and Plains regions (54% and 60%, respectively; p < 0.001). The NAM component was more homogeneous across regions (34-38%). Higher EUR ancestry was associated with lower rates of breastfeeding and higher reported oral contraceptive use (p < 0.05 for both). EUR ancestry was higher in individuals with ER-positive tumors compared to ER-negative tumors (54% vs. 51%, respectively, p < 0.05), whereas AFR ancestry was higher in ER-negative (8% vs. 6% in ER-positive, p < 0.05) and PR-negative tumors (8% vs. 5% in PR-positive, p < 0.05). A higher EUR component was observed among those with a positive family history (55%) (p = 0.057) and PVs carriers in BC-related genes (58%). Two cases with CHEK2PVs [c.349A>G and c.1100delC] had 71% EUR ancestry, significantly higher than non-carriers (54%, p < 0.05), consistent with their Northern/Central European founder origins. Among the 24 PV carriers, luminal subtypes predominated (63%), particularly in ATM (100%) and BRCA2 (80%) carriers. Four of five ATM PV carriers with ancestry estimates showed predominantly EUR ancestry (49-59%). The ATM splice-site variant c.5496+2_5496+5delTAAG was identified in three unrelated individuals from the Andean region—two of whom have predominantly European ancestry and one with missing ancestry data. Conclusions: Our findings demonstrate significant associations between global ancestry proportions and key clinical and genetic features in an admixed Colombian BC cohort. To further elucidate the genetic and demographic factors influencing the frequency, distribution, origin, and potential impact of PVs identified in this cohort, we have initiated ongoing local ancestry estimations and haplotype analyses. This study highlights the importance of ancestry-informed analyses to improve risk stratification and guide precision medicine approaches in admixed populations.
Presentation numberPS3-03-14
Knowledge, Attitude and Practice of Gynecological Oncologists on Breast Cancer-related Risks in BRCA1/2 Germline Mutation-related Ovarian Cancer Patients
Fengxia Gan, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
F. Gan, Y. Yang, Q. Liu; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA.
Background: BRCA1/2 germline mutations (gBRCAm) increase the risk of breast and ovarian cancers. Multidisciplinary collaboration between gynecological oncologists and breast specialists is crucial for the comprehensive assessment and personalized treatment of patients. Breast specialists often recommend gBRCAm breast cancer patients consult gynecological oncologists for ovarian cancer issues, but they rarely receive referrals from gynecologists for gBRCAm ovarian cancer patients to consult about breast cancer. This study aimed to investigate gynecological oncologists’ knowledge, attitudes, and practices (KAP) regarding breast cancer-related risks in ovarian cancer patients with gBRCAm. Methods: A cross-sectional online survey was conducted among gynecological oncologists from November 19 to December 7, 2024. The data were collected using a self-designed electronic questionnaire assessing general characteristics of respondents, KAP regarding breast cancer risks in ovarian cancer with gBRCAm. A total of 625 valid questionnaires were collected. Descriptive analysis and logistic regression analysis were performed on the collected data. Results: The participants’ mean age was 44.04 years, with 85.6% being female, and 64.5% held senior professional titles. Regarding KAP on breast cancer risks in ovarian cancer with gBRCAm, 11.4% of participants reported being unfamiliarity of the breast cancer risk; 88.0% believed it necessary to refer ovarian cancer patients with gBRCAm to breast specialists; 61.6% had previously referred such patients. Primary reasons for non-referral included: prioritizing ovarian cancer treatment due to its high lethality (46.4%), lack of focus on breast cancer risk (43.4%), and concurrent breast evaluation during ovarian cancer management (5.4%). The annual number of ovarian cancer patients treated was ≤10 for 310 participants (49.6%), 11-50 for 243 (38.9%), and >50 for 72 (11.5%). 67.7% had treated ovarian cancer patients carrying gBRCAm. 334 participants (53.4%) had been consulted by breast cancer patients carrying gBRCAm regarding ovarian cancer risks. Multivariate logistic regression revealed that understanding the breast cancer risk (P10 ovarian cancer patients annually (11-50 patients: P=0.041; >50 patients: P=0.003), having treated ovarian cancer patients with gBRCAm (P<0.001), and having been consulted by breast cancer patients carrying gBRCAm (P<0.001) were significantly associated with higher referral rates to breast specialists. Regarding opportunistic salpingectomy, 71.7% of participants agreed that all women should consider prophylactic salpingectomy concurrent with other pelvic surgery after completing childbearing. 70.6% agreed that prophylactic salpingectomy (preserving ovaries) could potentially reduce cancer risk while preserving ovarian function, and would consider this procedure for young carriers (≤40 years) with gBRCAm who had completed childbearing. Conclusion: Gynecologic oncologists’ knowledge regarding breast cancer risk in ovarian cancer patients with gBRCAm and their referral rates of these patients to breast specialists need improvement. Enhancing gynecologic oncologists’ understanding of BRCA1/2-related breast cancer risks will help increase referral rates to breast specialists, and promote multidisciplinary collaboration, and facilitate holistic healthcare for patients carrying gBRCAm.
Presentation numberPS3-03-15
Germline Genetic Testing Access and Survival Outcomes in Male Breast Cancer: A Veterans Affairs Cohort Analysis
Sarah V Colonna, Huntsman Cancer Institute / George E Wahlen VA / National Oncology Program, VA, Salt Lake City, UT
C. Li1, V. Patel1, R. McShinsky2, Z. Burningham2, A. Chiba3, S. Bushan4, S. Trehan5, M. Kelley6, A. Halwani7, S. V. Colonna7; 1Epidemiology, University of Utah / George E Wahlen VA, Salt Lake City, UT, 2Medical Oncology, University of Utah / George E Wahlen VA, Salt Lake City, UT, 3Surgical Oncology, Duke University / Durham VAMC / National Oncology Program, VA, Durham, NC, 4Medical Oncology, Baylor University / Michael E DeBakey VA / National Oncology Program, VA, Houston, TX, 5Medical Oncology, Tampa VA / National Oncology Program, VA, Tampa, FL, 6Medical Oncology, Duke University / Durham VAMC / National Oncology Program, VA, Durham, NC, 7Medical Oncology, Huntsman Cancer Institute / George E Wahlen VA / National Oncology Program, VA, Salt Lake City, UT.
Background Male breast cancer (MBC) accounts for ~1%of breast cancers, with ~2,800 new US cases in 2025. MBC has a higher germline mutation frequency than female breast cancer, mostly the result of a higher frequency of BRCA2 mutations in males (10-15%) compared to females (5-10%). NCCN guidelines and VA clinical pathways recommend genetic testing for all men with breast cancer given high mutation frequency and treatment implications. However, real-world genetic testing access and survival outcomes in veterans remain understudied. With >90% male patients, the Veteran Affairs (VA) represents the largest MBC cohort nationwide, providing a unique opportunity to evaluate genetic testing utilization and survival outcomes. Methods Veterans with MBC (2019-2025) were identified using VA Cancer Registry (site code C50). Patients receiving VA care were defined as having ≥2 visits with oncology services. Clinical notes were analyzed using natural language processing and large language model (Llama 3.3 70B) to extract germline genetic testing (GGT) access. GGT access proportion was compared for year 2019-2021 vs. 2022-2024. Overall survival (OS) analysis was conducted and outcomes by GGT access were compared. Results 523 Veterans with male breast cancer were identified. Mean age was 72 years, 61% were White, 28% were Black and 4% were Hispanic. 75% lived in urban areas. 520 (99.43%) were ER/PR+, 46 (8.8%) HER2+, and 3 (57%) triple negative. Of 523 veterans, 334 (63.9%) had access to germline genetic testing at VA over the course of their disease. Patient characteristics were similar between GGT groups. Five-year OS was 70.9% overall, with significant difference by GGT access (78.6% vs. 59.3%, p<0.001). Genetic testing access rates significantly increased from 56.5% (134/237) in 2019-2021 to 70.6% (192/272) in 2022-2024, representing a 14.1 percentage point improvement (p < 0.001, χ² test). Conclusion The vast majority of veterans with MBC receive GGT, with increasing access over time. GGT access is associated with improved overall survival, suggesting potential benefits of access to comprehensive genetic evaluation. A more detailed covariate analysis is needed to further explore.
Presentation numberPS3-03-16
Distribution of Molecular Subtypes of Hereditary Breast Cancer in Brazilian Patients according to Pathogenic or Likely Pathogenic Germline Genetic Variants
Cesar Cabello, State University of campinas – UNICAMP, campinas, Brazil
A. R. cabello, C. Cabello, S. Ramalho, C. E. Alem, S. R. Teixeira, L. R. Silva, G. S. Paiva, B. N. Duarte, M. Souza, T. cabello, D. I. Silva, A. Viaro, T. Gaspar, L. C. Zeferino; Obstetric and Gynecology, State University of campinas – UNICAMP, campinas, BRAZIL.
Distribution of Molecular Subtypes of Hereditary Breast Cancer in Brazilian Patients According to Pathogenic or Likely Pathogenic Germline Genetic Variants. Ana Elisa Ribeiro da Silva Cabello 1, Cesar Cabello 1, Bárbara Narciso Duarte 1; Christine Elisabete Rubio Alem 1; Sandra Regina Campos Teixeira; 1 Leonardo Roberto da Silva 1, Geisilene Russano Paiva 1, Susana Oliveira Botelho Ramalho 1, Thiago Cabello1 ,Márcio Lopes de Souza 1, Thiago Gaspar, 1 Daniel Imbassahy SBC Silva 1, Ana Lidia Viaro 1, Luiz Carlos Zeferino 1 1.State University of Campinas (UNICAMP)Corresponding author: cesar cabelloEmail: cabello@unicamp.brAddress: Alexander Fleming, 101-Cidade Universitaria, City: Campinas State; São Paulo, ZIP Code: 13083-881 Objective: To assess the distribution of molecular subtypes of hereditary breast cancers, according to the presence of pathogenic or likely pathogenic germline genetic variants detected by multigene panel testing. Methods: A prospective cohort study was conducted from November 2021 to October 2022, involving women under follow-up at the high-risk outpatient clinic at the University of Campinas (UNICAMP). Patients were required to meet the following criteria: a personal history of luminal or HER2-positive breast cancer diagnosed before age 45, or triple-negative breast cancer diagnosed before age 60, along with at least one first- or second-degree relative with breast, ovarian, and/or prostate cancer, or who met the NCCN high-risk criteria (NCCN version 3.2021). Patients completed a questionnaire, after which they were referred for genetic testing. Following the test results, they received counseling based on the findings. Results: A total of 184 patients and 198 breasts were studied. Pathogenic variants (PV) or likely pathogenic variants (LPV) were detected in 36% (67/184) of the patients. Approximately 28% (51/184) were under 40 years of age, 30% (74/184) were non- Caucasian, and 49% (90/184) were premenopausal. General Analysis: The molecular subtypes most associated with pathogenic/probably pathogenic variants (PV/LPV) were Luminal B HER2-negative “like” (OR=11.76; CI 4.19-33.01) and triple-negative (OR=8.4; CI 3.45-20.43). These associations remained consistent even when the analysis was restricted to BRCA1 and BRCA2 genes only (OR=9.33; CI 2.00-29.99) (OR=9.03; CI 3.37-24.20) or to non-BRCA genes (OR=7.75;CI 2.96-29.44) (OR=3.48; CI 1.32-9.20) for Luminal B HER2-negative “like” triple-negative subtypes. Conclusion: Two molecular subtypes were associated with the presence of pathogenic or likely pathogenic variants in genes associated with hereditary breast cancer: Luminal B HER2-negative and triple-negative subtypes. Keywords: High risk; Germline variants; Genetic testing; Breast cancer; Molecular subtypes
Presentation numberPS3-03-17
Analysis of second breast cancers in BRCA1/2 and other predisposing gene carriers at the Reggio Emilia Hereditary Cancer Center, Italy
Elisa Gasparini, Comprehensive Cancer Centre, AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
E. Gasparini1, M. Rotolo1, G. Moretti1, A. Bologna1, R. Di Cicilia1, C. Casartelli1, L. Mangone2, F. Leoni2, C. Pinto1, M. Dominici3, L. Cortesi3; 1Department of oncology and advanced technologies, Comprehensive Cancer Centre, AUSL-IRCCS Reggio Emilia, Reggio Emilia, ITALY, 2Epidemiology Unit, Comprehensive Cancer Centre, AUSL-IRCCS Reggio Emilia, Reggio Emilia, ITALY, 3Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, ITALY.
Background Hereditary breast cancer (HBC) accounts for 5-10% of all breast cancers (BC). Many high and moderate penetrance genes are responsible for HBC. The identification of HBC led to the development of preventive strategies for BC and ovarian cancer (OC). In recent years BRCA1/2 germline pathogenic variants (gPV) have been used as predictive role for PARP inhibitors treatment of BC, OC and prostate cancer improving overall survival. So far, criteria for selecting patients to test remain fundamental to identify mutation carriers to offer adequate treatment. Among those, second BC represents one of the typical characteristics to take in account, mostly in case of metachronous development. Our study aimed to analyze differences in second BC between BRCA1/2 and other mutation carriers, in terms of age, clinical characteristics and prognosis at the Reggio Emilia Hereditary Center. Methods The Reggio Emilia Hereditary Cancer Center was established in 2012, identifying patients at high HBC risk, based on personal/family history and Tyrer-Cuzick likelihood model. Among criteria utilized for multigene panel testing (MGPT), two BC arisen until 50 years alone, or at any age associated to first-degree BC, were considered sufficient to be analyzed with Sophia Genetic Hereditary Cancer Syndrome (HCS) panel, including 27 predisposing genes. From the clinical point of view, patients and BC characteristics were registered in hospital-based record charts. The record chart registered height, weight, BMI, smoking habit, hormonal status, pregnancies, vital status, histology, clinical and pathological status, grading, hormonal receptors, proliferative rate, HER2 expression, type of surgery, type of neoadjuvant/adjuvant treatments, other tumors, prophylactic surgeries and gPV in different genes. The comparisons between BRCA1/2 and other gPV carriers were made by Fisher’ exact test or by Chi-squared test when more than two variables were analyzed. Loco-regional breast cancer-free survival (LRBCFS) was defined as the time from diagnosis of BC to the date of the first local recurrence (including diagnosis of second BC and regional relapse). Overall survival (OS) was defined as the time from diagnosis of BC to death or last follow-up. OS and LRBCFS curves were calculated using Kaplan-Meier method. Results Among 220 multiple BC patients analyzed by MGPT, 44 (20%) resulted gPV carriers in predisposing genes (26 in BRCA1/2 genes and 18 in other ones). Out of 18 not BRCA1/2 genes, 5 MUTYH, 3 CHEK2, 3 PALB2, 3 Mismatch Repair genes, 2 ATM, 1 RAD50 and 1 TP53 gPV carriers were identified. Regarding the first BC, BRCA1/2 carriers showed a younger age at onset (45 vs. 53 years), more advanced disease, grading III and triple negative BC (TNBC) phenotype. The median LRBCFS was equal to 11.2 years (12.6 vs 10.9 in BRCA1/2 and other carriers, respectively). Of interest, 95% of BRCA1/2 carriers had contralateral BC, whereas 27% of other carriers had ipsilateral BC. The second BC characteristics reflect those of the first BC. Other gPV carriers had more non-second BC than BRCA1/2 (17.6% vs.9.5%). The bilateral salpingo-oophorectomy was performed in 62% of BRCA1/2 vs. 12% of not BRCA1/2. Finally, 92% of all patients were alive at 17.5 median follow-up years. Conclusions Second BC represents an important feature to consider for eligibility to MGPT. A high rate of gPV in predisposing genes (20%) was found in patients with two BC. More TNBC were shown, as expected, in BRCA1/2 gPV carriers, underlying the need to treat those patients with more aggressive regimens. The older age at the first BC onset and the lower rate of contralateral BC in not BRCA1/2 gPV carriers do not justify a bilateral mastectomy at the surgical time. However, the OS curves do not differ between the two groups suggesting that the second BC do not impact on the patients’ outcome
Presentation numberPS3-03-18
Guidelines for Breast Cancer Index Test before and after Epic software enhancement
Cindy Chau Tran, City of Hope, Upland, CA
C. Tran1, A. Deshmukh2, T. Doan3, N. Kethireddy3, M. Elia3, S. Bhardwaj3, I. Kang3, J. Mortimer3; 1Medical Oncology, City of Hope, Upland, CA, 2Breast Health, Hologic, San Diego, CA, 3Medical Oncology, City of Hope, Duarte, CA.
Background: The Breast Cancer Index® or BCI™ test is a gene-based test that analyzes two key aspects of early-stage, hormone receptor-positive breast cancer. In a first aspect, the HOXB13:IL17BR ratio (H/I) assesses how well the cancer is likely to respond to continued hormone therapy. In a second aspect, the molecular grade index (MGI) predicts the likelihood of recurrence. This combination of features helps inform the appropriate duration of endocrine therapy beyond traditional tests. Importantly, the BCI test is effective for both lymph node-negative and lymph node-positive patients. The BCI test was incorporated into the NCCN Clinical Practice Guidelines in Oncology (NCCN® Guidelines) in January 2021 and the ASCO® Clinical Practice Guideline (ASCO Guideline) in April 2022. Additionally, Epic® medical record software was recently updated to allow users to order the BCI test. We examined the frequency of BCI test orders as a quality improvement metric, both at the main campus of the City of Hope Cancer Center and our community network. Enhancing guidelines for Breast Cancer Index (BCI) testing and improvement is essential for elevating its quality and effectiveness in clinical settings. Methods: Data was obtained from Biotheranostics, Inc., for the period January 2020 to May 31,2025. We analyzed the data for testing rates with the BCI test over four time periods: pre-NCCN Guidelines (Jan 2020-Jan 2021), post-NCCN Guidelines (Jan 2021- April 2022), post- ASCO Guideline (April 2022-May 2024) and post-Epic medical record software integration (June 2024 – May 2025). Results: A total of 1456 women with early-stage breast cancer underwent testing with the BCI test during the study period. Following the NCCN Guidelines update in January 2021, there was a notable increase in adoption of testing with the BCI test, with utilization increasing by 83% (p=0.026). Following endorsement of the BCI test by the ASCO Guideline update in April 2022, there was a further increase in test ordering by 30% (p=0.22). Increased utilization of the BCI test was comparable at the main campus and our community network. The test was ordered comparably for all age groups, LN- and LN+ disease, HER2- and HER2+ patients, and pre- and post-menopausal status. The Epic software enhancement, implemented one year ago, has led to a significant year-over-year increase in test ordering: 38% on the main campus and 110% at our community site. Conclusion: The implementation of NCCN and ASCO Guidelines significantly increased BCI test utilization across all City of Hope sites. Furthermore, an Epic software enhancement dramatically improved adherence to these guidelines, leading to an even greater uptake of BCI testing.
Presentation numberPS3-03-19
Single Cell Proteomic Phenotyping Reveals Mitigation of Abrupt Involution-Induced Alteration in Mammary Epithelium Upon Tamoxifen Treatment
Sarmila Majumder, The Ohio State University, Coulmbus, OH
S. Majumder1, K. G. Gray2, N. Shinde1, R. Mawalkar1, A. Zhang1, M. Basree1, K. Ormiston1, X. Zhang3, R. Ganju4, G. Sizemore5, B. Ramaswamy1, D. G. Stover1; 1Internal Medicine, The Ohio State University, Coulmbus, OH, 2Internal Medicine, Dana Farber Cancer Institute, Boston, MA, 3Center for Biostatistics, The Ohio State University, Coulmbus, OH, 4Pathology, The Ohio State University, Coulmbus, OH, 5Radiation Oncology, The Ohio State University, Coulmbus, OH.
Background: Epidemiological studies link lack of or short-term breast feeding to increased risk of breast cancer (BC), including triple negative breast cancer (TNBC). However, few models allow interrogation of the underlying biology. To decipher this link, we developed a model of short-term breastfeeding (BF) leading to abrupt involution (AI) versus long-term BF leading to gradual involution (GI) in mice. In our prior work, we showed that AI led to significant increase in estrogen signaling and precancerous changes (hyperplasia, squamous metaplasia) in mammary glands (MG) (Basree, et al Br Can Res 2019). Further, AI glands are characterized by a notable increase in alveolar progenitor (AP) population, the cells of origin of TNBC. In this study, we addressed the hypothesis that the anti-estrogen tamoxifen (TAM) could revert the adverse effects of abrupt involution/lack of breast feeding by deep interrogation of shifts in cellular phenotypes through CyTOF single cell proteomics. Methods: FVB/N female mice (~8 week old) were bred and allowed to nurse six pups/dam at partum. All pups were removed on postpartum (PP) day 7 to induce AI. On PP day 8, 5mg-60day sustained release tamoxifen pellet or placebo pellet (PLAC) was implanted in the subscapular region of the AI mice. MGs were harvested on PP day 28 and 120 from three groups: 1) TAM-treated AI; 2) PLAC-treated AI; 3) age matched nulliparous mice. FFPE sections were used for histology and immunohistochemistry. Immune cell composition and epithelial subpopulations from PP day 28 and day 120 MGs were evaluated by FACS and subjected to CyTOF using an established 40-antibody murine mammary targeted proteome panel (Gray, et al Cell Rep 2024). The Ingest algorithm was used to integrate and analyze mass cytometry datasets. Results: Overall, TAM treatment for 60 days completely abrogated the hyperplastic changes induced by AI in MGs, including reduction in proliferation index, collagen deposition, macrophages, and alveolar AP cells in the AI glands. As seen in our prior work, at day 120, abrupt involution-placebo glands (relative to nulliparous MGs) demonstrated marked enrichment of AP (1.5-fold) and basal (BA 1.6-fold) cells at the expense of hormone sensing (HS) cells. Analyses of MGs from the abrupt involution-tamoxifen group at day 120 revealed a significant increase (3-fold) in hormone sensing (HS) epithelial cells and an attendant decrease in AP cells compared to placebo. Further, abrupt involution-placebo glands show upregulation of AP markers (CD61, TSPAN8, SSEA1, CD14) and basal markers (CD54, CD47, CD104, CD49F) in the AP cells and higher expression of BA markers and reduced expression of HS markers in the HS cells. TAM treatment reversed these aberrant changes in abrupt involution-placebo glands in fact inducing a shift in HS cells to an involution-like state. Integration of the AI-PLAC/TAM data with lineage trajectory of these three cell types across the gestation-lactation-involution cycle revealed that AI-AP cells exhibited a mix of post-involutional and gestational states at day 120. TAM led to a 4.1-fold higher proportion of post-involutional-like AP cells and a 10.2-fold lower proportion of the aberrant gestational-like cells at day 120. Similar changes were observed in BA cells but not in HS cells upon TAM treatment. Conclusion: Further understanding of the biological impact of short/no breast feeding is critical and this work suggests that TAM treatment offers marked protection against AI-induced precancerous changes in a murine model. TAM restored long-term lineage balance after AI, altering AP differentiation towards a typical post-involution state as opposed to AI-induced hyperplasia. The corrective effect of TAM provides a rationale for short-term TAM treatment for women who are unable to BF to reduce risk of BC.
Presentation numberPS3-03-20
Trends in Hormonal Contraceptive Use Among Premenopausal Women in the United States, 1999-2020
Lin Yang, University of Calgary, Calgary, AB, Canada
L. Yang1, H. Harsanyi2, I. Bacha3, S. Uttamapinan3, A. Toriola3; 1Oncology, University of Calgary, Calgary, AB, CANADA, 2Cancer epidemiology and prevention research, Alberta Health Services, Calgary, AB, CANADA, 3Surgery, Washington University in St. Louis, St. Louis, MO.
Background: Hormonal contraceptive use in premenopausal women has been associated with a small but meaningfully higher risk of breast cancer. However, contemporary patterns of hormonal contraceptive use among premenopausal women in the United States (US) are unclear. Objective: To evaluate trends in and correlates of hormonal contraceptive use among premenopausal women in the US. Methods: Serial, cross-sectional analyses of hormonal contraceptive use using US nationally representative data from the National Health and Nutrition Examination Survey (NHANES) were conducted. Participants included noninstitutionalized premenopausal women from ten NHANES study cycles (1999-2000 to 2017-March 2020 pre-pandemic). The prevalence of hormonal contraceptive use was extracted from prescription medication data collected during household interviews. Hormonal contraceptive formulations were determined by hormone type. Trends in the prevalence of hormonal contraceptive use across study cycles were examined using multivariable logistic regressions, adjusting for sociodemographic factors. Results: Data on 15531 premenopausal women from ten NHANES cycles were analysed. From 1999 to 2020, the prevalence (95% CI) of hormonal contraceptive use significant increased from 5.2% (2.2 to 11.5%) to 15.5% (11.2 to 21.1%) for young women aged 15-<20 years, was stable among women aged 20-<40 years (from 16.9% [13.3 to 21.3%] to 15.8% [13.0 to 19.0%]), and remained stably low among those aged 40-<60 years (from 3.8% [1.5 to 9.3%] to 7.0% [4.0 to 12.1%]). Compared to non-Hispanic White women, non-Hispanic Black and Hispanic women consistently had lower prevalence of use but exhibited greater relative increases (prevalence ratio [PR]=3, 95% CI: 1.0 to 8.5, PR=2, 95% CI: 0.9 to 4.1, respectively) from 1999 to 2020. Combined estrogen and progestin formulation accounted for over 95% of the hormonal contraceptive prescriptions in 1999-2020, yet progestin formulation increased approximately five-fold, accounting for 16.3% (10.8 to 2.8%) of all prescriptions in 2017-2020. The prevalence of contraceptive use varied by race/ethnicity and family income to poverty ratio in all age groups, as well as by educational attainment, health insurance coverage, marital status, and weight status among women 20-<40 years. Conclusions and Relevance: In the past two decades, hormonal contraceptive use in the US significantly increased among young women aged 15-<20 years and among non-Hispanic Black premenopausal women of all age groups. Combined estrogen and progestin formulations account for the majority of hormonal contraceptive use; however, prescriptions for progestin formulations are increasing. Future investigations are needed to elucidate the implications of these changes in hormonal contraceptive use on the incidence of breast cancer.
Presentation numberPS3-03-22
Brca1/2-tested breast and ovarian cancer patients in peru: clinical and genomic features
Pamela Mora, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru
P. Mora1, N. Valdiviezo2, M. Acosta1, J. Herzog3, L. Reynaga3, L. Taxa4, T. Vidaurre2, Y. Sullcahuaman1, C. Castañeda2, C. Munive2, B. Muñante2, J. Abugattas5, H. Guerra6, I. Otoya2, V. Acuña7, S. Gruber3; 1Unidad Funcional de genética y biología molecular, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 2Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 3Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, 4Equipo Funcional de Patología Quirúrgica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 5Cirugía de Mamas y Tejidos Blandos, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, 6Anatomía Patológica y Patología Oncológica, Patólogos Especializados SAC, Lima, PERU, 7Facultad de Farmacia y Bioquímica, Universidad Nacional Mayor de San Marcos, Lima, PERU.
Pathogenic and likely pathogenic (P/LP) variants in BRCA1/2 are key markers of hereditary cancer, guiding clinical and preventive care. In Peru, late-stage diagnoses and limited genetic services make breast and ovarian cancer major public health issues. This study aimed to describe the clinical and genomic characteristics of Peruvian patients with breast and/or ovarian cancer who underwent BRCA1/2 testing between 2013 and 2020. Ten-year overall survival was analyzed using univariate and multivariate Cox regression, with left truncation to minimize immortal time bias. METHODS This study includes individuals diagnosed with breast and/or ovarian cancer referred to a Medical Genetics evaluation at the Instituto Nacional de Enfermedades Neoplásicas (INEN) during the period of August 2013 to March 2020. Individuals who met clinical criteria to be included in the inter-institutional protocol INEN13-48: “Estudio del Cáncer Hereditario y Familiar en Población Peruana” as part of the “Molecular Genetic Studies in Cancer patients and their relatives”; IRB #96144 City of Hope (COH); accessed genetic testing (carried out at COH). In patients diagnosed with both breast and ovarian cancer, only the first diagnosed cancer was considered for classification. RESULTS A total of 1,576 patients were included in the study, with a median age at diagnosis of 41 years. The vast majority were female (99.3%), and only 0.7% (n = 11) were male. Family history of cancer was absent in 61% of cases; 18% reported first-degree relatives with breast and/or ovarian cancer, while 20% reported other types of cancer. Breast cancer was diagnosed in 89.5% of patients (n = 1,410), ovarian cancer in 8.5% (n = 134), and both in 2% (n = 32). In breast cancer cases, the most frequent tumor subtype was HR+/HER2− (39.6%), followed by triple-negative (31.3%) and HER2+ (28.1%). Overall, we identified pathogenic variants in BRCA1 were identified in 7.2% (n = 114) of the whole population, and in BRCA2 in 3.2% (n = 51). Specifically, 6% of breast cancer patients carried a BRCA1 pathogenic variant, and 3% had a BRCA2 variant. BRCA1-positive breast tumors were predominantly of the triple-negative subtype (85.4%), whereas HR+/HER2- (47.6%) tumors were the most frequent among BRCA2-positive cases. In patients with ovarian cancer, 82% were BRCA-negative, while 14% carried BRCA1 and 2.8% BRCA2 pathogenic variants. Clinical stage II was the most common at presentation (39.3%), followed by stage III (38.8%), stage I (11%), and stage IV (10%). Additionally, 15% of patients developed a second primary malignancy, most commonly a new breast cancer (52%), followed by other cancers (15%), thyroid (12%), ovarian (8%), and colorectal cancer (6%). Notably, ovarian cancer diagnosis, advanced clinical stage, and triple-negative breast cancer subtype were significantly associated with poorer overall survival (p < 0.001 for all). CONCLUSIONSPatients carrying pathogenic BRCA variants were found to have a higher risk of developing a second primary cancer, mainly a new breast cancer, reinforcing the need for long-term surveillance in this population. In the subgroup of breast cancer patients with BRCA1 mutations, a higher incidence of triple-negative tumors was observed, whereas BRCA2-mutated patients more commonly exhibited luminal subtypes. These findings underscore the importance of strengthening genetic counseling services and implementing risk-reduction strategies as well as precision oncology programs. Given the higher mortality observed in patients with BRCA1 mutations, it is essential to understand the clinical and genomic profiles of these individuals to optimize cancer care for high-risk populations in the country.
Presentation numberPS3-03-23
Effects of hormonal contraceptive on tamoxifen efficacy to prevent chemically induced invasive cancers in rats
Oukseub Lee, Feinberg School of Medicine, Northwestern University, Chicago, IL
O. Lee, A. Eremin, S. A. Khan; Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL.
Background: Tamoxifen (TAM) is the only anti-estrogen approved for women with functioning ovaries who are advised to avoid pregnancy due to risk to the fetus but are cautioned against using hormonal contraceptives (HCs). This reluctance is based on concerns that HCs may counteract the efficacy of TAM and increase adverse effects when used concomitantly. However, these concerns lack direct supporting evidence, and the possibility of a neutral or even beneficial interaction has not been explored. We hypothesized that the most widely used combined hormonal contraceptive (17α-ethinylestradiol + levonorgestrel) would not reduce the tumor-preventive efficacy of oral TAM in a hormonally sensitive, ER-positive mammary tumorigenesis model in rats. Methods: Female Sprague Dawley rats (4-5 weeks old) received N-Methyl nitrosourea (50 mg/kg i.p.) to induce mammary tumors. Three weeks later, they were randomized (n=12-13 per group) to control, HC alone, TAM alone, or TAM+HC. HC pellets (17α-ethinylestradiol 0.06 mg + levonorgestrel 0.28 mg, 90-day release; Innovative Research of America) were implanted subcutaneously in the dorsal area and replaced at 90 days. TAM was incorporated into the diet (1.03 mg/kg/day in rats, equivalent to daily 7 mg in humans; Inotiv Inc.). Rats were monitored for 6 months for tumor formation. Tumor incidence, latency, weights, and volumes were measured. Wet uterine weight was recorded, adjusted for body weight, and processed for histology. Tumor-free survival was analyzed with the log-rank test; other comparisons used Kruskal-Wallis with Dunn’s post hoc tests. Results: In the control group, median tumor latency was 3 months with a final incidence of 92%, consistent with published studies. Tumor incidence was higher in the HC alone group (46%) than in the TAM alone (17%) and TAM+HC (15%) groups. Tumor weights and volumes were significantly lower in both TAM-treated groups compared to controls (all adjusted p 0.99). TAM significantly reduced body weight versus control (median difference = 30 g, adjusted p = 0.001), with no difference between TAM alone and TAM+HC groups (median difference = 16 g, adjusted p = 0.73). HC alone had no significant effect on uterine weight (adjusted p > 0.99). TAM significantly reduced uterine weight relative to controls (adjusted p < 0.001), with no difference between TAM alone and TAM+HC groups (adjusted p = 0.97). Histopathological assessment of mammary glands, uterus, and endometrial thickness is ongoing. Conclusions: Combined HC did not reduce the tumor-preventive efficacy of oral TAM in this ER-positive mammary tumorigenesis model. Ongoing analyses will assess uterine safety with this combination.
Presentation numberPS3-03-24
Patterns and Prevalence of Pathogenic Germline Mutations Among Patients with Invasive Lobular Breast Cancer
Hikmat Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
H. Abdel-Razeq, F. Tamimi, B. Sharaf, S. Abdel-Razeq, O. El Khatib, A. Sharaf, H. Bani Hani, A. Al-Atary, L. El Saket, Y. Talab, H. Khalil, M. Harb, S. Yousef, A. Nofal, A. Alghrabli, M. Abunasser; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: Invasive lobular carcinoma (ILC) is the second most common histological subtype of breast cancer after invasive ductal cancer (IDC). Previous studies have shown that 5-10% of all breast cancers are associated with cancer-predisposing germline mutation. The frequency of these mutations in ILC is scarce. Here, we report on the pattern and frequencies of germline genetic mutations among Arab patients with ILC. Methods: In this retrospective observational study, electronic medical records were reviewed for all breast cancer patients diagnosed at King Hussein Cancer Center (KHCC) from January 2016 to May 2024. Included patients had histologically confirmed ILC and underwent germline genetic testing using restricted or expanded panels. Descriptive analyses were performed, with categorical variables reported as percentages. Chi-squared and Fisher’s exact tests were used to compare proportions of categorical variables between patients with pathogenic or likely pathogenic (P/LP) mutations and those without, as well as between BRCA and non-BRCA patients. Results: Out of the 6,539 patients who met the inclusion criteria, 477 (7.3%) had pure ILC histology. The median age at diagnosis was 49 years and only 13% were ≥ 65 years. Except for 2, all were female. Most patients (95%) were ER-positive, 92% PR-positive and 91% HER2 negative. Multicentric disease was observed in 43% of patients, bilateral disease in 14%, and 19% presented with metastatic disease. A family history of breast cancer was reported in 39% of patients, including 25% who had a first-degree relative with the disease. The most common additional malignancy observed in these patients were breast cancer (9.2%), with colorectal and endometrial cancers each at 0.8%, and ovarian cancer in only one patient. Regarding genetic testing, 12.6% had limited gene panels and 87.4% had extended panels. Overall, 47 (9.9%) carried a P/LP variant, most frequently in BRCA2 (n=25, 53.2%), followed by ATM and PALB2 (n=6 each, 12.8%). BRCA1 pathogenic variant was detected in only one patient. BRCA1/2 mutations were significantly more common in patients younger than 40 years; 17.0% compared to only 4.2% in older patients, p<0.001. Patients with P/LP variants presented at younger ages, had more frequent first-degree breast cancer family history (p<0.001), and were more likely to undergo risk-reduction surgery (p<0.001). Both male patients also carried P/LP variants. Conclusions: We found that many Jordanian patients with invasive lobular breast cancer had germline mutations, especially in BRCA2. Younger age at diagnosis and having a first-degree relative with breast cancer, were linked to a higher chance of having a pathogenic mutation. These results show the importance of genetic testing to guide preventive measures and treatment personalization. Larger future studies are needed to improve genetic testing guidelines and outcomes for these patients.
Presentation numberPS3-03-25
Alteration in estrogen receptor status in metachronous contralateral breast cancer among unilateral early breast cancer patients with BRCA 1/2 mutations
Ki Jo Kim, Samsung medical center, Seoul, Korea, Republic of
K. Kim1, E. Kang2, S. Baek3, H. Lee2, S. Ahn4, W. Park1, D. Shin2, H. Lee5, S. Nam1, S. Kim1, J. Yu1, B. Chae1, L. Se Kyung1, J. Lee1, J. Ryu1; 1Division of Breast Surgery, Department of Surgery, Samsung medical center, Seoul, KOREA, REPUBLIC OF, 2Department of Surgery, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF, 3Department of Surgery, Yongin Severance Hospital, Yongin, KOREA, REPUBLIC OF, 4Department of Surgery, Gangnam Severance Hospital, Seoul, KOREA, REPUBLIC OF, 5Department of Pathology and Translational Genomics, Samsung medical center, Seoul, KOREA, REPUBLIC OF.
Background BRCA1/2 mutations are known to significantly increase the risk of contralateral breast cancer (CBC). However, their influence on changes in estrogen receptor (ER) status between primary breast cancer and metachronous CBC remains unclear. This study investigates ER status alterations in CBC patients with BRCA1/2 mutations to provide insights into their clinical implications.Methods This multicenter retrospective study was conducted between 2004 and 2020. Patients with unilateral early breast cancer who underwent BRCA1/2 testing and developed metachronous CBC were grouped by BRCA1/2 mutation status. ER status of primary and CBC tumors was compared using Fisher’s exact test and logistic regression to assess the relationship between BRCA1/2 mutations and ER alterations.Results Among 423 CBC patients, those with BRCA1 mutation had a higher likelihood of ER negative primary tumors transitioning to ER negative CBC (70.7%) compared to BRCA1/2 negative patients (odds ratio [OR] 3.8, p < 0.001). Similarly, ER-negative primary tumors were more likely to remain ER negative in CBC among BRCA1 or BRCA2 mutation carriers (64.8%, OR 2.9, p = 0.002). These findings demonstrate a strong association between BRCA1 mutations and the development of ER negative CBC in ER negative primary breast cancer. Of the patients with primary ER-negative breast cancer, those who received adjuvant chemotherapy for both primary and contralateral breast cancers showed a significantly higher rate of CBC with ER-negative alteration (OR 4.23, p=0.001).Conclusion BRCA1 mutation carriers face a significantly higher risk of developing ER negative metachronous CBC, in ER negative primary tumor. These findings highlight the importance of genetic counseling and risk-reducing strategies, including prophylactic mastectomy, for BRCA1 mutation carriers, which typically requires more aggressive treatments.
Presentation numberPS3-03-26
Guideline concordant surgical management of hereditary variants of uncertain significance: outcomes from a safety-net hospital
Karina P Verma, Boston Medical Center, Boston, MA
K. P. Verma1, L. J. Oshry1, M. Rudderman1, I. Arshad2, B. Moy3, L. Wu1; 1Department of Medicine, Section of Hematology and Oncology, Boston Medical Center, Boston, MA, 2Boston University School of Public Health, Boston Medical Center, Boston, MA, 3Department of Medicine, Division of Hematology and Oncology, Massachusetts General Hospital, Boston, MA.
Background: Germline genetic testing is recommended for individuals with personal or family history of certain cancers, as pathogenic variants (PV) can guide cancer screening and management. Variants of uncertain significance (VUS) present clinical challenges as guidelines recommend that VUS alone should not determine cancer management or decisions about risk-reducing surgeries. VUS are disproportionately reported in non-white individuals, reflecting the historic underrepresentation in genetic reference databases. These disparities complicate genetic counseling and exacerbate inequities in cancer care. Methods: The High Risk Cancer Genetics Clinic (HRC) was established at Boston Medical Center, the region’s largest safety net hospital, to improve access to cancer screening and surveillance. We conducted a retrospective cohort study of patients with a VUS in hereditary breast and ovarian cancer (HBOC) genes and compared surgical decision-making among patients found to have VUS two years before and after HRC establishment from May 2021 to April 2025. Male patients and those with metastatic disease were excluded. Results: There were 438 patients with at least one VUS in BRCA1, BRCA2, BRIP1, TP53, PALB2, PTEN, CDH1, STK11, RAD51C, or RAD51D genes, for which PV can inform risk-reducing bilateral mastectomy (BLMa) and/or bilateral salpingo-oophorectomy (BSO). A total of 158 patients were included, and 69% had Medicaid, Medicare, or were uninsured, 78% self-identified as non-white, and 46% spoke a language other than English. After HRC, significantly more patients with HBOC VUS were referred to see genetic counselors (87% vs 96%; p=0.05) and HRC physicians (0 vs 10%, p=0.006). Before and after HRC establishment, there were no cases of BLMa among patients with a VUS who did not have breast cancer. Similarly, patients with a VUS who did not have ovarian cancer did not undergo BSO. A small proportion of breast cancer patients with a VUS had BLMa before and after HRC (5% vs 12%, p=0.64). Two patients with BRCA1 and PTEN VUS were reclassified as likely PV, and the HRC facilitated updated screening guidance that led to timely olaparib initiation and detection of localized breast cancer, respectively. Conclusion: At our safety-net hospital serving a diverse community, the management of patients with HBOC VUS has remained guideline concordant, with no risk-reducing surgeries performed solely based on a VUS result. Among breast cancer patients with an HBOC VUS who elected BLMa, all had documented counseling regarding the lack of survival benefit compared to breast conserving surgery, but chose BLMa for cosmetic or personal reasons. The integration of HRC has enabled timely re-evaluation and clinical action when a VUS is upgraded to PV. This demonstrates the value of multidisciplinary risk assessment in improving diagnostic and therapeutic outcomes for underserved populations.
| Patients with HBOC VUS | Before HRC | After HRC | P-value |
| Clinic visit with genetic counselor |
56/64 (87%) |
91/94 (96%) |
0.05 |
| Clinic visit with HRC MD | 0/64 |
10/94 (10%) |
0.006 |
| Non-breast cancer patients who underwent bilateral mastectomy (BLMa) | 0/45 | 0/62 | – |
| Non-ovarian cancer patients who underwent bilateral salpingo-oophorectomy | 0/64 | 0/90 | – |
| Breast cancer patients who underwent BLMa |
1/19 (5%) |
4/32 (12%) |
0.64 |
Presentation numberPS3-03-27
Potential genetic targets of recurrence at different time points in Asia breast cancer with NanoString and multigene prognostic test
Ming Feng Hou, Kaohsiung Medical University Hopsital, Kaohsiung, Taiwan
M. Hou1, F. Chen1, C. Li1, C. Chiang2; 1Division of Breast Oncology and Surgery, Kaohsiung Medical University Hopsital, Kaohsiung, TAIWAN, 2Department of Biomedical Sciences, Chung Shan Medical University, Taichung, TAIWAN.
BackgroundMost breast cancer-related death is caused by recurrence and metastasis. Approximately 30% to 40% of patients will experience recurrence during their lifetime. Although the promising effect of endocrine therapy in ER-positive breast cancer, there are still about 30% of patients who ultimately die due to recurrence. In contrast to ER-negative, more than 50% of recurrences occur beyond five years (late recurrence, LR) in ER-positive breast cancer, which represents the proportion of early (ER, recurrence occur within five years) or even very early recurrence (VER, recurrence occur within one year in our preliminary results) are rare. In comparison with LR, the prognosis of ER and VER is poor and more aggressive; however, most studies focused on LR, including mechanisms, treatment, and genetic/immune profiles, few studies focused on ER, and even No studies concerned what we named VER. On the other hand, the introduction of a multigene prognostic test (MPT) can provide the dual role of prognostic and predictive to fulfill and strengthen the information in treatment decisions. However, there still existed some defects in the current MPT, including high cost, ethnic difference, and specificity for different time points of recurrence type. Thus, there is a huge gap in ER or even VER, and a critical unmet need to identify who is prone to recurrence for Asia breast cancer.MethodsFifty ER-positive patients with disease-free (DF, average follow-up 48 months), and 14 ER-positive patients with recurrence were enrolled in this study. Initially, three patients from 50 patients with DF and nine patients from 14 patients with recurrence were selected. In total of 12 patients (three patients for each group, including DF, VER, ER, and LR) were selected, and residual RNA samples from EndoPredict (EP) were analyzed by using the NanoString technique. Finally, raw data processing, normalization, and analysis were performed in the nSolver analysis software. Appropriate statistical analyses were performed using GraphPad Prism 9. ResultsIn our preliminary results, we collected the data with EndoPredict and long-term follow-up records. By using the NanoString technique, we found some novel and unique target genes in different groups of recurrence. The expression of SERPINA1 was specific to disease-free (DF, average follow-up 48 months), CEACAM6 was overexpressed in LR group (average PFS 45 months), TGFB2 was only observed in VER group (average PFS 8 months), and AREG was co-existed in the group of ER (average PFS 23 months) and VER. Moreover, we compared the scores from a low-cost platform of immunohistochemical 4 (IHC4) and EndoPredict within 64 patients (average follow-up 48 months) and found a positive correlation between the IHC4 risk score and EPClin score in prognosis prediction.ConclusionTo our knowledge, we provide the first preliminary results of novel gene targets at different time points of recurrence, especially for VER. We also found a positive correlation between the IHC4 risk score and EPClin score in prognosis prediction. The ability of prediction and mechanisms in recurrent breast cancer from these target genes will be validated in a large cohort and requires further investigations in the future.
Presentation numberPS3-03-28
Recurrent, Cryptic BRCA1 Complex Rearrangement by Whole-genome Sequencing in Patients with Breast Cancer
Erin Connolly-Strong, Inocras Inc, San Diego, CA
B. Oh1, S. Choi2, S. Seo3, H. Park4, E. Connolly-Strong1, T. Kim5, Y. Ju6, J. Hwang7, Y. Park8; 1Medical Center, Inocras Inc,, San Diego, CA, 2Department of Internal Medicine, Division of Clinical Genomic Medicine(Hematology & Medical Oncology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Gyeonggi-do, KOREA, REPUBLIC OF, 3Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Gyeonggi-do, KOREA, REPUBLIC OF, 4Genome Science, Bioinformatics, Inocras Inc,, San Diego, CA, 5Biology, Johns Hopkins University, Baltimore, MD, 6Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, KOREA, REPUBLIC OF, 7Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, KOREA, REPUBLIC OF, 8Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, KOREA, REPUBLIC OF.
Introduction: Detecting pathogenic germline variants in BRCA1/2 is crucial for hereditary cancer risk assessment and guiding targeted therapies. While targeted NGS panels are effective for SNVs and indels, they often miss large structural variations (SVs), leading to false negatives. Whole-genome sequencing (WGS) offers a comprehensive solution by providing uniform, genome-wide coverage for robust SV detection. This report presents two clinical cases where WGS identified an identical, complex pathogenic BRCA1 SV missed by conventional panel testing.Methods: DNA was extracted from fresh-frozen tumor tissue and matched normal blood samples from two patients: a 40-year-old premenopausal female with breast and ovarian double primary cancers and a female in her 60s with metastatic pancreatic cancer (history of breast cancer). WGS analysis was performed using the CancerVision™ system.Results: Both patients initially had non-decisive hereditary cancer panel results. WGS revealed an identical, complex pathogenic germline BRCA1 variant in both: a large deletion of exons 12-14 coupled with an adjacent inversion. This BRCA1 deletion was successfully orthogonally confirmed by MLPA. The first patient’s breast tumor showed BRCA1 loss of heterozygosity (LOH), a high HRD score (0.93), 40.0% of base substitutions attributed to SBS3 (indicating flawed homologous recombination-based DNA repair), and a high prevalence of SV duplications (45.8%), suggesting a complete BRCA1 defect. The second patient’s tumor also showed BRCA1 LOH, 33.7% of base substitutions attributed to SBS3, and high SV duplications (33.7%), collectively suggesting impaired BRCA1 function. This patient achieved and maintained a complete response to platinum-based chemotherapy.Discussion: This study demonstrates WGS’s transformative power in resolving clinically important cases missed by conventional genomic approaches. The key finding is the identification of an identical, complex recurrent BRCA1 SV in two unrelated patients, providing definitive molecular diagnoses that significantly impacted clinical management. This recurrence suggests a potential underdiagnosed founder mutation. WGS’s unparalleled advantage lies in its uniform, genome-wide coverage, enabling simultaneous detection of diverse genomic variations, including complex SVs, ensuring a comprehensive and unbiased genetic landscape elucidation. WGS provides precise genomic breakpoint locations, allowing unequivocal determination of variant identity across patients and facilitating tracking of origins and population-specific prevalence. These cases underscore WGS’s critical role in identifying pathogenic SVs undetectable by standard methods, advocating for its integration into clinical diagnostics for high-risk patients to enhance accuracy and support personalized treatment strategies.
Presentation numberPS3-03-30
Association between allostatic load and clinical and sociodemographic risk factors for breast cancer among diverse women undergoing screening mammography
Vicky Ro, Columbia University Irving Medical Center, New York, NY
V. Ro1, J. B. Gibbons1, C. N. Ta2, C. Liu2, W. K. Chung3, C. Weng2, K. D. Crew1; 1Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, 2Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, 3Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA.
Introduction: Allostatic load (AL) is an index that accounts for cumulative stress on the body and is associated with increased breast cancer risk and breast cancer mortality. We aimed to determine the association between demographic, clinical and lifestyle factors and AL among women undergoing screening mammography.Methods: Patients were enrolled in the Electronic Medical Records and Genomics (eMERGE4) study at Columbia University Irving Medical Center and completed surveys on demographic, clinical and lifestyle factors, provided samples for germline BRCA1/2 and PALB2 genetic testing and polygenic risk score (PRS) for breast cancer (308-SNP model), and allowed access to electronic medical record (EMR) data. Women with at least one screening mammogram were included. To calculate AL score, we extracted the most recent EMR data on 9 biometrics representing major regulatory systems: cardiovascular (pulse, systolic and diastolic blood pressure), metabolic (albumin, alkaline phosphatase, glucose), renal (blood urea nitrogen, creatinine), and immune (white blood cells). One point was assigned to each biometric in the highest quartile except albumin which was the lowest quartile. Patients with at least 5 biometrics were included; missing values were imputed. High AL was defined as AL score above the cohort median. Demographic, clinical, and lifestyle factors from pre-test survey data included age, race/ethnicity, highest educational level, annual household income, health insurance, disability, body mass index (BMI), smoking and alcohol use, and physical activity level. We extracted breast density (4-category BIRADS classification) and prior breast biopsy results from the EMR. Univariable and multivariable logistic regression analyses were conducted to evaluate the association between high AL and demographic, clinical, and lifestyle factors.Results: Among 955 evaluable women, the mean age was 54.8 years (standard deviation [SD] 11.4), with 48.2% Hispanic, 34.9% non-Hispanic White, 9.1% non-Hispanic Black, and 7.9% Asian/other. Median AL score was 2 (range, 0-8), and 418 patients had high AL. Mean BMI was 27.7 kg/m2 (SD 6.3). A total of 10.4% of the patients had a prior history of breast cancer. Most patients (61.7%) reported 0 days of vigorous exercise a week. Over half (54.6%) have a bachelor’s or advanced degree. Almost half (45.5%) of those with high AL have Medicaid versus 25.1% of those with low AL. In univariable analysis, older age, Black race, Hispanic ethnicity, higher breast cancer PRS, lower breast density, having a disability, lower educational attainment, lower income, public health insurance, higher BMI, less alcohol use, and less physical activity were associated with high AL. In multivariable analysis, older age (odds ratio [OR]=1.03, 95% confidence interval [CI]=1.02-1.05), higher BMI (OR=1.09, 95% CI=1.06-1.12), and having Medicaid vs. private health insurance (OR=2.21, 95% CI=1.30-3.75) were significantly associated with high AL score.Conclusions: In our diverse population of women undergoing screening mammography, we found a significant association between clinical, demographic, and socioeconomic factors and AL, which is a well-established breast cancer risk factor. In addition to age and BMI, having Medicaid compared to private health insurance was associated with a greater than 2-fold likelihood of having high AL. Although protective against breast cancer risk, lower breast density and less alcohol use were also associated with higher AL, warranting further investigation. These findings highlight the importance of addressing modifiable risk factors such as obesity for reducing AL and breast cancer risk and the need to address psychosocial stressors in vulnerable populations with Medicaid.
Presentation numberPS3-05-01
Risk factor profile in male breast cancer: a cohort-based assessment from a breast cancer institute in northeastern México
Luis F Martinez-Caudillo, Tecnológico de Monterrey, Monterrey, Mexico
L. F. Martinez-Caudillo, M. S. Guzman-Garcia, V. Leitzelar-Bueso, J. E. Guzman-Garcia, D. Aguilar-Y-Mendez; Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey, MEXICO.
Introduction: Male breast cancer (MBC) accounts for 1.8% of all breast cancer cases worldwide. Compared to women, men tend to have poorer prognosis, mainly due to delayed diagnosis at an older age, when comorbidities are more prevalent. Risk factors can be divided into modifiable and non-modifiable. Among the non-modifiable, genetic predisposition -mutations in BRCA1 and BRCA2- play a major role. However others include: family history, Klinefelter Syndrome, and advanced age (>65 years). In contrast, modifiable risk factors include tobacco use, obesity, and alcohol. The mechanism for the latter remains unclear, but is found to be widely associated with MBC. Understanding key risk factors may aid in early diagnosis and diminish mortality rates. Given its low incidence, characterization of cohorts may help to identify relevant risk factors, improve early detection strategies and contribute to a better understanding of MBC. Methods: We conducted an observational retrospective study including eleven male patients diagnosed with breast cancer between 2016 and 2025 at the Breast Cancer Institute of the Zambrano Hellion Hospital, in northeastern Mexico. The following risk factors were evaluated, non-modifiable factors included: Klinefelter syndrome, advanced age, family history of breast cancer, breast cancer susceptibility genes (BRCA1, BRCA2, among others); modifiable: obesity (BMI > 30), alcohol and/or tobacco use, exogenous estrogen or testosterone use, ionizing radiation exposure, gynecomastia, bone fracture after the age of 45, cryptorchidism. Descriptive statistical analysis was applied. Results: Eleven male patients diagnosed with MBC were included in the analysis. Median age at diagnosis was 54 years with a SD of 13.92 and a range varying from 34-74. The most prevalent modifiable risk factors were alcohol consumption (75%) and tobacco use (63%), while obesity was present in 25% of patients. Among non-modifiable risk factors breast cancer susceptibility genes and advanced age prevailed with 38% and 27% respectively. Family history of cancer was found in 45%, more information in Table 1. Conclusions: This cohort-based analysis highlights important deviations from existing literature, particularly regarding age at diagnosis, having a median of 54 and cases as early as 34 years; despite the known association of MBC with advanced age (>65). Modifiable risk factors such as alcohol consumption and tobacco use were highly prevalent, detailing potential prevention opportunities. Moreover, a notable proportion presented family history of cancer, reinforcing the need for thorough familial assessment in clinical practice. Although limited by cohort size, this study contributes valuable insights to the scarce literature on MBC, and supports the need for larger, population-based research to better characterize risk profiles and detail early detection strategies.
| Patient ID | Patient’s age at cancer diagnosis | Degree of relatedness | Relative | Cancer Type |
| 1 | 38 | Second | Uncle | Breast |
| 2 | 58 | First | Father | Melanoma |
| 2 | 58 | First | Mother | Pancreas |
| 2 | 58 | Third | Paternal cousin | Breast |
| 3 | 74 | First | Daughter | Breast |
| 3 | 74 | First | Brother | Colon |
| 3 | 74 | Third | Cousin | Breast |
| 4 | 66 | First | Father | Prostate |
| 4 | 66 | Third | Paternal cousin | Breast |
| 7 | 63 | First | Sister | Breast |
| 7 | 63 | First | Brother | Stomach |
| 9 | 34 | Second | Uncle | Testicle |
Presentation numberPS3-05-02
Prevalence and re-classification Variants of Uncertain Significance (VUS): experience of a breast cancer center in northeastern Mexico.
Maria S Guzman-Garcia, Tecnologico de Monterrey, Monterrey, Mexico
M. S. Guzman-Garcia1, J. E. Guzman-Garcia1, V. Leitzelar-Bueso1, L. F. Martinez-Caudillo1, D. Aguilar-Y-Mendez2, C. Villarreal-Garza2; 1Escuela de Medicina y Ciencias de la Salud del Tecnologico de Monterrey, Tecnologico de Monterrey, Monterrey, MEXICO, 2Breast Cancer Institute, Hospital Zambrano Hellion, San Pedro Garza Garcia, MEXICO.
Introduction: Variants of uncertain significance (VUS) are variants in the genetic code for which the impact on the protein function is unclear. VUS estimated probability in genes associated with hereditary breast and ovarian cancer (HBOC) is 23%, and the rate of reclassification varies from 19-27% depending on the population studied. It represents a challenge to medical management and surveillance decisions. Methods: From 2018 to 2024 a total of 334 VUS have been reported among 1014 breast cancer patients at the Breast Cancer Institute of Zambrano Hellion Hospital. A retrospective clinical-genomic review was conducted, including only patients with breast cancer.Data were extracted from electronic medical records, including: age at diagnosis, tumor laterality, immunohistochemical (IHC) profile, VUS-related gene(s), and family history of cancer (e.g., breast, ovarian, pancreas). For each case, we documented the gene in which the VUS was identified, whether it underwent reclassification, the final classification (pathogenic, likely pathogenic, or benign), and the time elapsed (in months) until reclassification. Results: 19 VUS were identified across multiple cancer susceptibility genes (Table 1). Of these, 9 variants were reclassified: 55% as benign and 45% as likely pathogenic. The mean time to reclassification was 26.44 ± 12.6 months (range 7-52).Table 1 summarizes the number of VUS per patient and their corresponding reclassification time. A total of 9 female breast cancer patients carriers of at least one of these VUS reclassified with or without a pathogenic variant. The average age at diagnosis was 40.3 years (SD ±5.32). All BC diagnoses were unilateral. Four patients (44%) presented with triple-negative breast cancer (TNBC) by immunohistochemistry (IHC). The most frequently involved gene was NF1, accounting for 5 variants (55%), of which 4 were reclassified as benign and 1 as likely pathogenic. BRCA2 presented 2 variants (22%), with one benign and one likely pathogenic outcome. BRCA1 and POLE each had 1 variant (11%), both of which were reclassified as likely pathogenic. Conclusions: The observed rate of reclassification may be influenced by the underrepresentation of Hispanic populations in genetic databases and limited access to genetic testing. Given the potential clinical implications of VUS in HBOC-associated genes, further investigation is essential to improve variant classification and optimize genetic counseling. These variants are increasingly reclassified through the integration of population-specific data, functional assays, co-occurrence with known pathogenic mutations, and other lines of evidence.
| Patient ID | Identified VUS (gene) | Time to reclassification (months) | Reclassification outcome |
| 1 | BRCA1 | 56 | Pathogenic |
| 1 | RET | 81 | Bening |
| 2 | NF1 | 21 | Likely pathogenic |
| 2 | BRCA1 | 21 | VUS |
| 3 | NF1 | 52 | Bening |
| 4 | NF1 | 15 | Bening |
| 5 | NF1 | 18 | Bening |
| 5 | BRCA1 | 18 | VUS |
| 6 | BRCA2 | 30 | Likely pathogenic |
| 6 | DICER1 | 30 | VUS |
| 7 | POLE | 26 | Likely pathogenic |
| 7 | ATM | 26 | VUS |
| 8 | NF1 | 38 | Bening |
| 9 | BRCA2 | 7 | Bening |
| 9 | ATM | 7 | VUS |
| 9 | BRCA1 | 7 | VUS |
| 9 | BMPR1A | 7 | VUS |
| 9 | MEN1 | 7 | VUS |
| 9 | TMEM127 | 7 | VUS |
Presentation numberPS3-05-03
Clinicopathologic and Molecular Characteristics of Middle-Eastern Breast Cancer Patients with Pathogenic or Likely Pathogenic ATM Variants: A Comprehensive Cohort Analysis
Hikmat Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
H. Abdel-Razeq, L. El Saket, H. Bani Hani, S. Abdel-Razeq, F. Tamimi, B. Sharaf, Y. Talab, A. Al-Atary, M. El-Atrash, T. Al-Batsh, M. Horani, O. Othman, Y. Al-Masri, O. El Khatib, M. Abunasser; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: ATM gene alterations have increasingly been recognized for their role in breast cancer susceptibility and therapeutic implications. However, the phenotypic and molecular features of ATM-mutant breast cancers remain under-characterized in Middle Eastern populations. This study provides a comprehensive analysis of breast cancer patients harboring pathogenic or likely pathogenic (P/LP) ATM variants treated in Jordan, with emphasis on clinicopathologic features, genetic context, and therapeutic outcomes. While ATM mutations confer a moderate lifetime breast cancer risk (25%), their impact on treatment decisions remains under investigation. Methods: We retrospectively reviewed data on 52 female patients with breast cancer and confirmed pathogenic or likely pathogenic ATM mutations. Parameters analyzed included age at diagnosis, family history, histopathology, stage, hormone receptor status, Ki-67 proliferation index, and genetic testing eligibility based on NCCN/ASCO guidelines. Co-occurring variants in other genes and variant types (missense, nonsense, frameshift, splice site) were compiled. Results: The median (range) age at diagnosis was 48 (27-79) years, with 32 (61.5%) diagnosed before age 50. Invasive ductal carcinoma (IDC) was the predominant histologic subtype (n=41, 78.8%), followed by invasive lobular carcinoma (ILC). Hormone receptor (HR) positivity was high: ER-positive (92.3%), PR-positive (90.4%), and HER2-negative in most cases (n=36, 70.6%). High-grade tumors (GIII) comprised 39.2% of the cohort. The majority of ATM variants were deletions (frameshift, 46.2%), followed by nonsense (17.3%), missense (15.4%), and splice site alterations (11.5%). Common hotspots included c.2284_2285del and c.5755C>T. Co-occurrence with other susceptibility genes was notable; 6 (11.5%) had additional pathogenic variants in other genes: 3 with BRCA2, 1 with RAD50, 1 with MUTYH, and 1 with a second distinct ATM mutation. Furthermore, 16 of the 52 patients (30.8%) harbored additional variants of uncertain significance (VUS). Despite a high prevalence of early-stage disease 39 (75.0%), lymph node involvement was observed in 27 (51.9%) cases. Neoadjuvant chemotherapy was utilized in 16 (32.7%) patients with a modest rate of pathologic complete response (12.2%). Notably, 5 (9.6%) of patients did not meet testing criteria per conventional guidelines, suggesting gaps in current eligibility frameworks, particularly for patients under universal genetic testing. Conclusions: ATM-mutant breast cancers in this Jordanian cohort exhibited a predominance of HR-positive, high-grade tumors with frequent lymph node involvement. The diversity in variant types and co-existing mutations underlines the clinical complexity of these cases. A proportion of patients would have been missed by existing genetic testing guidelines, emphasizing the utility of universal testing in this setting. These findings advocate for broader germline screening in Middle Eastern populations to inform precision oncology strategies.
Presentation numberPS3-05-04
From Variant to Vigilance: Clinical Implications of CHEK2 Mutation-Associated Breast Cancer and Second Malignancies in a Non-Westrern Cohort
Hikmat Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
H. Abdel-Razeq, H. Bani Hani, F. Tamimi, L. El Saket, S. Abdel-Razeq, B. Sharaf, H. Abu-Jaish, Y. Talab, A. Al-Atary, M. El-Atrash, T. Al-Batsh, M. Horani, L. Barakat, Y. Al-Atary, O. El Khatib; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: The CHEK2 (Checkpoint Kinase 2) gene plays a central role in DNA damage repair and cell cycle regulation. Germline pathogenic or likely pathogenic (P/LP) CHEK2 variants confer a moderate risk of breast cancer, especially estrogen receptor (ER)-positive subtypes, and have been associated with other malignancies, including colorectal and thyroid cancers. Despite CHEK2 being one of the most frequently mutated genes identified in hereditary cancer panels, clinical data on variant distribution, tumor characteristics, and associated malignancies remain underreported in many populations. This analysis is the largest of its kind in a Middle Eastern population. Methods: We conducted a retrospective review of breast cancer patients with germline P/LP CHEK2 mutations identified through multigene panel testing (20-gene or 84-gene panels). Testing was done at one of three accredited international reference genomic labs. Clinical variables included demographics, tumor histology and grade, hormone receptor status, treatment modalities, family history, and the presence of second primary cancers. Genetic variant analysis and co-occurrence with additional mutations were also documented. Results: Fifty-nine female CHEK2-positive breast cancer patients were identified. Median age at diagnosis was 48 years (range: 28-77), with 59.3% diagnosed at or before age 50. The majority had invasive ductal carcinoma (n=48, 81.4%) and grade 1 or grade 2 tumors (73.1%). Hormone receptor status showed that all patients were estrogen receptor (ER)-positive, while progesterone receptor (PR) positivity was observed in 86.2% of cases (n=50). HER2 overexpression was observed in 23.2% of cases. Neoadjuvant therapy was administered in 48.1% of patients, and 52.5% underwent mastectomy. Family history of cancer was reported by 33 (91.5%) patients; 81.5% had at least one relative with breast cancer. The most frequent CHEK2 variant was c.592+3A>T (39.0%), followed by c.499G>A (20.3%). Notably, 5 patients (8.5%) carried additional germline P/LP variants: APC I1307K (n=3), BRCA2 (n=1), and co-occurring PALB2 + RAD51D (n=1). Second primary cancers were identified in 16 (27.1%) patients and mostly were breast cancer (n=12, 20.3%) and one case each of colorectal, endometrial, pancreatic and lymphoma. Among those who underwent screening colonoscopy (n=25), 3 patients (12.0%) were diagnosed with colorectal cancer, and 3 others (12.0%) had tubular adenomas and hyperplastic polyps. Conclusions: This study provides insight into the clinical and molecular landscape of CHEK2-positive breast cancer patients in a non-Western cohort. The high prevalence of the c.592+3A>T variant may suggest regional founder effects. The identification of additional actionable and the high rate of second primary cancers support the use of comprehensive multigene panels in hereditary cancer testing. These findings underscore the importance of tailored surveillance and risk-reduction strategies, including screening for second primary malignancies, in CHEK2 mutation carriers.
Presentation numberPS3-05-05
High-risk cancer susceptibility genes mutation carriers’ compliance with surgical risk reduction for breast and ovarian cancer.
Konstantinos Papazisis, Interbalcan European Medical Centre, Thessaloniki, Greece
K. Papazisis1, M. Paraskeva2, S. Giassas3, M. Skondra4, R. Iosifidou5, C. Tolis6, G. Kesisis7, E. Zairi7, V. Venizelos8, C. Markopoulos9, I. Natsiopoulos10, E. Bleka11, I. Xanthakis10, A. Ananiadis7, D. Matheos12, D. Stefanou13, A. Adamidis10, A. Meintani14, G. Kapetsis14, D. Bouzarelou14, E. Papadopoulou14, G. Nasioulas14; 13rd Dpt of Oncology, Interbalcan European Medical Centre, Thessaloniki, GREECE, 21st Department of Oncology, Rhodes General Hospital, Rhodes, GREECE, 3Dpt of Oncology, IASO Hospital, Athens, GREECE, 4Dpt of Oncology, Henry Dunant Hospital, Athens, GREECE, 5reast Surgical Oncology Clinic, Theageneio Anticancer Hospital, Thessaloniki, GREECE, 6Private practice, Private practice, Ioannina, GREECE, 7Dpt of Oncology, Agios Loukas Hospital, Thessaloniki, GREECE, 8Dpt of Oncology, Metropolitan Hospital – Persefs Ygionomiki Merimna AE, Piraeus, GREECE, 9Dpt of Oncology, Athens Medical center, Athens, GREECE, 10Dpt of Oncology, Interbalcan European Medical Centre, Thessaloniki, GREECE, 11Dpt of Oncology, Euromedica Kyanous Stavros, Thessaloniki, GREECE, 12Dpt of Oncology, General Hospital of Alexandroupoli, Alexandroupoli, GREECE, 13Dpt of Oncology, Laiko Hospital, Athens, GREECE, 14Molecular Oncology, GENEKOR MEDICAL SA, Athens, GREECE.
Background Identifying pathogenic/likely pathogenic variants in high-risk cancer genes is key to cancer risk management.Prophylactic surgeries like RRM and RRSO are guideline-recommended options. This study aims to provide real-world evidence on their clinical implementation. Methods Between 2020 and 2025, 203 women referred for genetic testing at Genekor Medical S.A., were detected with P/LP variants in cancer susceptibility genes, for which risk-reducing surgical options are either recommended or considered for discussion. Referring clinicians were invited to complete a questionnaire to assess whether prophylactic surgical options had been discussed, if the patients had agreed to undergo such procedures, and whether the surgeries have already been performed or postponed. Results Among the individuals referred for genetic testing, 168(83%) were referred following a cancer diagnosis, while 35(17%) were unaffected individuals referred due to a family history. 153 individuals (76%) harbored BRCA1/2, 31(15%) harbored PALB2, PTEN, TP53 and 19 (9%) harbored BRIP1,RAD51C, RAD51D P/LP variants. RRSO was discussed with 78/153 (51%) of BRCA1/2 carriers and agreed in 60/78 (77%) cases. Addition of hysterectomy to RRSO was discussed in 16/78 (21%) of cases and agreed in 10/16 (63%). RRM was discussed with 113/203 (56%) of patients and agreed in 73/113(65%). In PALB2, PTEN, TP53 carriers, RRM was discussed in 21/31 (68%) cases, and accepted in 9/21 (43%), while in 5 cases both RRM/RRSO were discussed with 3 women agreeing. Finally, for 19 BRIP1,RAD51C, RAD51D carriers, in 9 cases the physician discussed RRSO as an option, with the examinees agreeing in 7 of them and in 5 cases RRM was discussed with agreement to proceed in 2 of them. In all cases, 22 individuals although agreed to proceed with RRM and/or RRSO, decided to postpone the procedure. Conclusions This study highlights the high awareness and acceptance of surgical management demonstrating the adherence to the recommendations. Age under 35 years and receiving therapy at the time of genetic testing were significant factors in lowering risk reduction RRSO and hysterectomy performance rates. Stage IV cancer was the main reason for not discussing surgical intervention.
Presentation numberPS3-05-06
Landscape of PIK3CA Mutations in Chinese Patients with Breast Cancer: A Multi-Center Real-World Study
Fei Xu, Sun Yat-sen University Cancer Center, Guangzhou, China
F. Xu1, K. Jiang1, R. Chen2, H. Wu1, Y. Zhong1, H. Liu2, W. Lyu2, S. Mo1, H. Chen1; 1Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, CHINA, 2Breast Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, CHINA.
Background: PIK3CA mutations are established therapeutic targets in breast cancer (BC). The population-level prevalence has been explored, but the distribution across key clinicopathological strata in Chinese patients (pts) remains to be elucidated. Methods: This retrospective study enrolled BC pts diagnosed between January 1997 and February 2025 with available PIK3CA testing (next-generation sequencing) results. Pts were divided based on PIK3CA mutation status into mutated and wild-type (WT) groups. Mutation status was defined as any positive test result during follow-up. Clinicopathological characteristics and prognosis were compared between the two groups. Results: This study enrolled 577 pts, divided into PIK3CA mutated group (n=226) and the WT group (n=351). PIK3CA mutation frequencies varied across molecular subtypes: highest in Luminal A (LA) (56.4%), followed in frequency by LB1 (45.8%), HER2-positive (45.5%), and Luminal B2 (LB2) (40.5%), but lower in triple-negative breast cancer (TNBC) (21.4%). Among mutated tumors, H1047R was predominant (42.5%), followed by E545K (17.7%) and E542K (11.5%). H1047R mutations showed higher prevalence in HER2+ (22.7% of HER2+ tumors) and LA (21.8%) subtypes compared to TNBC (11.7%). E545K mutations were enriched in LB2 (11.9%) and LA (11.5%) subtypes, while being rare in TNBC (2.8%). BRCA mutations were significantly less frequent in PIK3CA-mutated pts (mutated vs. WT: 4.0% vs. 16.2%, P0.05) (Table 1). After a median follow-up of 38.2 months, no significant survival difference was observed. Five-year overall survival (OS) rates were 93.1% (95%CI: 88.9-97.5%) for the PIK3CA mutated group compared to 90.2% (95%CI: 85.9-94.8%) for the PIK3CA WT group. A molecular subtype-stratified survival analysis revealed that pts with the LA subtype harboring PIK3CA mutations had a 5-year OS rate of 92.5% (95% CI: 82.9-100.0%), while PIK3CA wild-type LA patients achieved a 5-year OS rate of 100.0% (95% CI: 100.0-100.0%) (P=0.035). In the stratification analysis of E545K, the PIK3CA WT group had a 5-year OS rate of 90.2% (95% CI: 85.9-94.8%). In contrast, pts with other PIK3CA mutations (non-E545K, n=186) achieved a 5-year OS rate of 95.3% (95% CI: 91.6-99.1%). The E545K mutation group (n=40) showed the lowest 5-year OS rate at 84.4% (95% CI: 71.1-100.0%). Conclusions: This real-world study identifies distinct clinicopathological profiles linked to PIK3CA mutations in Chinese BC pts, characterized by LA subtype enrichment and H1047R dominance. Notably, PIK3CA mutations are associated with poor survival in LA tumors and E545K mutations. These findings underscore the significant impact of PIK3CA mutations on the survival of Chinese BC pts.
| Variables | Total (N=577) | PIK3CA mutated (n=266) | PIK3CA-WT (n=351) | P-value | |
| Clinicopathological characteristics | Age at diagnosis, years, median (Q1, Q3) | 45.0 (37.0, 53.0) | 46.0 (38.0, 54.0) | 45.00 (37.0, 52.0) | 0.149 |
| Clinical stage at diagnosis, n (%) | 0.119 | ||||
| I | 55 (9.5) | 17 (7.5) | 38 (10.8) | ||
| II | 152 (26.3) | 57 (25.2) | 95 (27.1) | ||
| III | 153 (26.5) | 64 (28.3) | 89 (25.4) | ||
| IV | 68 (11.8) | 35 (15.5) | 33 (9.4) | ||
| Missing | 149 (25.8) | 53 (23.5) | 96 (27.4) | ||
| Bilateral Breast Cancer, n (%) | 35 (6.1) | 14 (6.2) | 21 (6.0) | 0.917 | |
| Molecular subtype, n (%) | <0.001 | ||||
| HER2-positive | 44 (7.6) | 20 (8.8) | 24 (6.8) | ||
| Luminal A | 78 (13.5) | 44 (19.5) | 34 (9.7) | ||
| Luminal B1 | 236 (40.9) | 108 (47.8) | 128 (36.5) | ||
| Luminal B2 | 42 (7.3) | 17 (7.5) | 25 (7.1) | ||
| Triple-negative | 145 (25.1) | 31 (13.7) | 114 (32.5) | ||
| Other | 30 (5.2) | 5 (2.2) | 25 (7.1) | ||
| Missing | 2 (0.3) | 1 (0.4) | 1 (0.3) | ||
| Outcome | Death events, n (%) | 38 (6.6) | 17 (7.5) | 21 (6.0) | 0.467 |
| 5-year OS rates, % (95% CI) | 91.5 (88.4, 94.6) | 93.1 (88.9, 97.5) | 90.2 (85.9, 94.8) | ||
| 5-year OS rates by molecular subtype, % (95% CI) | |||||
| HER2-positive | 94.2 (86.7, 100.0) | 94.1 (83.6, 100.0) | 94.4 (84.4, 100.0) | ||
| Luminal A | 96.0 (90.8, 100.0) | 92.5 (82.9, 100.0) | 100.0 (100.0, 100.0) | ||
| Luminal B1 | 90.8 (85.8, 99.2) | 89.6 (82.3, 97.6) | 92.0 (85.3, 99.2) | ||
| Luminal B2 | 96.4 (89.8, 100.0) | 100.0 (100.0, 100.0) | 93.3 (81.5, 100.0) | ||
| Triple-negative | 86.5 (78.0, 96.0) | 100.0 (100.0, 100.0) | 81.4 (70.0, 94.7) |
Presentation numberPS3-05-07
Behind the Stethoscope: Breast Cancer Awareness Gaps Among Healthcare Workers : A Wake Up Call from Coastal South India
Smitha S Rao, KS Hegde Medical Academy, Mangalore, India
S. S. Rao1, V. V. Shetty2, V. Rajendra3; 1Endocrine & Breast Surgery, Oncology, KS Hegde Medical Academy, Mangalore, INDIA, 2Medical Oncology, KS Hegde Medical Academy, Mangalore, INDIA, 3Surgical Oncology, KS Hegde Medical Academy, Mangalore, INDIA.
Background:Healthcare workers (HCWs) play a pivotal role in early detection, treatment, prevention and education strategies for breast cancer. However, there is limited data assessing their own levels of awareness and knowledge in resource-constraint settings. This survey aims to identify the gaps in breast cancer awareness among HCWs in Mangalore, a prominent healthcare hub in Southern India. Methods:A cross-sectional, questionnaire-based study was conducted from June to July 2025 among 45 HCWs, including doctors from non-oncological specialties, nurses, paramedical staff, interns, and resident doctors, across four tertiary care hospitals in Mangalore. The validated questionnaire assessed knowledge of breast cancer risk factors, symptoms, available treatment options and attitudes toward breast self-examination (BSE) and clinical breast examination (CBE). Data were analysed using SPSS v28; Chi-square tests determined statistical significance. Results:Majority of the participants were final year students and residents (65%). None of them were breast cancer survivors. Few of them reported family history of breast cancer (6.5%). In first and second degree relatives (6%). Surprisingly, only 11% of participants correctly identified all major risk factors, while 60% of them correctly understood the signs and symptoms. Only 40% of the participants in the survey were aware of all treatment modalities and 60% of them knew the routine investigations for follow up. Almost 88.8% reported to be aware of BSE monthly, and 35.5% of them believed mammogram could cause radiation exposure. Awareness scores were significantly higher among doctors compared to nursing and allied staff with the bias of difference in participation numbers(p<0.01). Conclusion:This study clearly demonstrates a critical and concerning gap in breast cancer awareness among healthcare providers — a gap that directly undermines early detection efforts and compromises patient outcomes. As frontline influencers of public health behavior, healthcare workers must not only advocate for cancer awareness but embody it. It is imperative that institutions implement structured breast health education, mandate annual clinical breast examinations (CBEs) for staff, and integrate awareness assessments into regular training programs. Investing in HCW education is investing in community cancer control.
Presentation numberPS3-05-08
Exploring Germline Genetic Testing Across a Diverse Ethnic Group with Triple-Negative Breast Cancer
Hikmat Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
H. Abdel-Razeq, T. Al-Batsh, F. Tamimi, B. Sharaf, H. Bani Hani, A. Al-Atary, L. El Saket, H. Khalil, Y. Talab, A. Issa, Z. Muhanna, O. Almuhaisen; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Introduction: Triple-Negative Breast Cancer (TNBC) is the most aggressive form of breast cancer. Despite advancements in medical treatments, TNBC continues to be associated with a high likelihood of recurrence and poor outcomes. Gaining a deeper understanding of genetic profiles of TNBC is essential for identifying potential biomarkers, improving prognostic tools, and developing targeted therapies. This study aims to explore the baseline characteristics, survival outcomes, and genetic alterations in TNBC among a population with distinct ethnic backgrounds. Methods: Data on TNBC patients diagnosed and treated at the King Hussein Cancer Center who underwent germline genetic testing between 2014-2024 were retrospectively collected. Testing was performed on peripheral blood, and sequencing was done at international reference labs. Based on the date of testing, some patients underwent limited testing with only BRCA1/2 ± PALB2 (n=150), or an expanded multigene panel (n=432). Mutations were classified as benign/likely benign (negative), pathogenic/likely pathogenic (P/LP) (positive), or variant of uncertain significance (VUS). Event-Free Survival (EFS) and Overall Survival (OS) were estimated using the Kaplan-Meier method to evaluate patient outcomes. Results: A total of 582 patients were included in the study, predominantly Jordanian (521, 89.5%), with a median age of 45 years (range: 37-54), and majority (548, 94.2%) presented with non-metastatic disease. Genetic testing revealed that 124 patients (21.3%) had P/LP variants, and 141 (24.2%) had VUS. The most frequent variants were BRCA1 (82, 66.1%) and BRCA2 (32, 22.8%). Other variants encountered (10, 8.1%) included PALB2, RAD51D, NF1, TP53, RAD51C, MLH1, BARD1, BRIP1, and MSH2. Rate of P/LP mutations were highest among younger patients aged 65 (3/36, 8.3%), p <0.001. Among the 124 patients with P/LP variants, 119 (96.0%) had a positive family history of cancer in a close relative. Of the 434 patients who underwent multigene panel testing, only 10 (2.3%) had mutations beyond BRCA1 and BRCA2, with just two in high-penetrance breast cancer-related genes (TP53 and PALB2). Risk-reducing contralateral mastectomy was performed by 79 (65.8%) patients with P/LP variants, while bilateral salpingo-oophorectomy was performed by 63 (52.5%). During follow-up, 28 patients (4.8%) experienced recurrence, and another breast cancer was diagnosed in 40 patients (6.8%), with a significantly higher rate observed among those with germline mutations (12.5% vs. 5.1%, p = 0.004). Conclusion: This study highlights the genetic characteristics of TNBC in an Arab population. The notably high prevalence of BRCA1 and BRCA2 pathogenic variants reinforces their central role in TNBC tumorigenesis. In resource-constrained settings, prioritizing genetic testing for these two genes offers a practical and cost-effective strategy. Although the prevalence of P/LP variants is significantly lower in patients over 65 years of age, it remains sufficiently substantial to justify continued testing in this group. Importantly, over half of mutation carriers opted for risk-reducing surgical interventions, reflecting the actionable value of genetic findings in clinical decision-making.
Presentation numberPS3-05-09
Approaches to Breast Cancer Risk Management in Ovarian Cancer (OC) Patients Harboring Germline BRCA1/2 Mutations (gBRCA1/2mut).
Débora Guilherme de Albuquerque e Rodrigues de Sousa, A. C. Camargo Cancer Center, São Paulo, Brazil
D. Guilherme de Albuquerque e Rodrigues de Sousa1, L. Soares1, T. Saruwatari1, A. Cicilini1, J. Casali da Rocha1, S. Sanches1, A. Gadelha1, F. Makdissi2, G. Baiochi2, B. Tirapelli1, F. Leite2, M. Sonagli2, R. Cagnacci2, S. Caputo Durand3, A. Favarin4, B. Rossi4, G. Molin4, A. da Costa1, E. dos Santos1; 1Clinical Oncology, A. C. Camargo Cancer Center, São Paulo, BRAZIL, 2Surgical oncology, A. C. Camargo Cancer Center, São Paulo, BRAZIL, 3Clinical Oncology, Institut Curie – Ensemble Hospitalier, Paris, FRANCE, 4Clinical Oncology, Hospital Beneficência Portuguesa, São Paulo, BRAZIL.
Introduction: Patients with ovarian cancer carrying germline BRCA1/2 mutations or pathogenic variants (gBRCA1/2mut) have a significantly increased lifetime risk of developing breast cancer. While the treatment of ovarian cancer is usually the primary clinical focus, proactive strategies to reduce future breast cancer risk – including intensified surveillance, risk-reducing mastectomy, and chemoprevention – are essential components of comprehensive care. Given the evolving therapeutic landscape and improving survival outcomes in ovarian cancer, individualized approaches to breast cancer risk management are becoming increasingly relevant in this high-risk population. Our objective was to evaluate and describe breast cancer risk management strategies in OC patients carrying gBRCA1/2mut. Methods: A retrospective review of medical records was conducted for patients with OC and gBRCA1/2mut, focusing on breast cancer risk – reduction strategies. Results: A total of 88 patients with OC and gBRCA1/2mut were identified. Among them, 66 (75%) had BRCA1mutations and 22 (25%) had BRCA2 mutations. Twenty-three patients (26%) had a previous history of breast cancer prior to the BRCA1/2 mutation diagnosis; of these, 16 (18%) were treated with lumpectomy, 6 (26%) with mastectomy, and 1 (4%) with adenomastectomy. Regarding axillary management, 11 (48%) underwent sentinel lymph node biopsy, 9 (39%) axillary dissection, and in 3 cases (13%) axillary status was unknown. Fifty-one patients (58%) had a family history of breast cancer.After a median follow-up time of 70 months, only 12 patients (13%) underwent risk-reducing breast surgery: 8 (67%) underwent bilateral adenomastectomy, 3 (25%) bilateral mastectomy, and for 1 patient the chosen procedure was not specified. Among 13/88 patients with stage I-II ovarian cancer, 5 underwent prophylactic mastectomy, compared to 7 out of 75/88 patients with stage III-IV disease. Of the 12 patients who underwent prophylactic mastectomy, only 3 had relapsed ovarian disease – all of whom had advanced-stage cancer at diagnosis. Overall survival from the time of ovarian cancer diagnosis was 94.2 months. Most patients (65%) opted for active surveillance every 6 months, with magnetic resonance imaging (MRI), ultrasound, and mammography. Imaging findings classified as BI-RADS 3 were identified in 9 patients (16%), and BI-RADS 4/5 in 8 patients (14%). A total of 6 patients (10%) were ultimately diagnosed with breast cancer, half of which were luminal subtype tumors. Among these 6 cases, half were clinical stage IA, two were stage IIA, and one was stage IIIA breast cancer. Conclusion: In this cohort of OC patients with gBRCA1/2mut, the majority underwent imaging-based surveillance, with a relevant proportion presenting with suspicious findings and subsequent breast cancer diagnoses. These findings highlight the importance of individualized counseling and the integration of breast cancer prevention into the long-term management of ovarian cancer survivors with gBRCA1/2 mutations, particularly in the context of improving survival outcomes.
Presentation numberPS3-05-10
The Self-Declared White Ethnicity is Associated with the Presence of Pathogenic Variants in Brazilian Public Health Patients Undergoing Extended Multigene Panel Testing
Cesar Cabello, State University of campinas – UNICAMP, campinas, Brazil
C. Alem, A. Cabello, b. N. Duarte, S. R. teixeira, T. Gaspar, A. viaro, D. I. Silva, M. L. Sousa, T. Cabello, S. Ramalho, L. R. Silva, C. Cabello; Obstetric and Gynecology, State University of campinas – UNICAMP, campinas, BRAZIL.
The Self-Declared White Ethnicity is Associated with the Presence of Pathogenic Variants in Brazilian Public Health Patients Undergoing Extended Multigene Panel Testing Christine Elisabete Rubio Alem 1,, Ana Elisa Ribeiro da Silva Cabello1, Bárbara Narciso Duarte1; Sandra Regina Campos Teixeira; 1 Leonardo Roberto da Silva 1, Susana Oliveira Botelho Ramalho 1, Thiago Cabello 1 , Márcio Lopes de Souza 1, Thiago Gaspar, 1 Daniel Imbassahy SBC Silva 1, Ana Lidia Viaro 1, Cesar Cabello 11.State University of Campinas (UNICAMP)Corresponding author: cesar cabelloEmail: cabello@unicamp.brAddress: Alexander Fleming, 101- Cidade Universitaria, City: Campinas State; São Paulo, ZIP Code: 13083-881 Introduction: Breast cancer is the most common cancer among women in Brazil, with high mortality rates, especially in younger women. Hereditary factors, including mutations in BRCA1 and BRCA2, are responsible for a significant number of cases. The use of multigene panels for genetic testing has increased, though data on ethnically admixed populations remain limited. Objective: To evaluate the prevalence of pathogenic variants in a Brazilian public health population and their association with epidemiological variables. Methodology: This study involved women with a personal or family history of breast cancer. Blood samples were collected for genetic sequencing of 144 genes associated with hereditary syndromes. The study took place at the High-Risk sector of the Hospital da Mulher Prof. Dr. José Aristodemo Pinotti – CAISM, UNICAMP, between November 2021 and October 2022. Data were analyzed using next-generation sequencing (NGS) and statistical tests (Chi-Square, Fisher’s Exact, Kruskal-Wallis, Bowker, McNemar, logistic regression) with significance set at p < 0.05. Results: Of the 364 participants, 85.9% had genetic variants, with 29.7% having pathogenic or likely pathogenic variants. White women (OR: 2.1; CI 1.31-3.36) and those with a personal history of breast cancer (OR: 1.90; CI 1.16-3.10) had significantly higher risks for these variants. The most commonly affected genes were BRCA1, BRCA2, TP53, PALB2, and MUTYH. Conclusion: The study revealed a high prevalence of pathogenic variants, highlighting the role of race and personal history in genetic predisposition to breast cancer.
Presentation numberPS3-05-11
Is Breast Cancer After Kidney Transplant More Common in Men?
Matthew Sporn, NYU Langone, New York, NY
M. Sporn1, C. Marsh1, A. Sharma1, A. Marmer1, B. Greenfield1, V. Tatapudi2, B. Lonze2, M. Gemignani1, F. Schnabel1, C. DiMaggio2; 1Department of Surgery, NYU Langone, New York, NY, 2Department of Transplant, NYU Langone, New York, NY.
Introduction:Within the general population, male breast cancer (BC) is exceedingly rare, with approximately one case for every 100 female cases. However, as a high-volume transplant center, we have anecdotally observed several cases of male BC following kidney transplant. Despite this, the gender-specific BC incidence in solid organ transplant remains a gap in the current literature. We therefore sought to examine the gender-specific incidence of BC among kidney transplant recipients. Methods:Retrospective analysis of data from 1987-2022 from the Scientific Registry of Transplant Recipients (SRTR) database was used to identify patients who developed BC following kidney transplant. Clinical, pathological, treatment, and follow-up data were collected. Using R statistical software program comparisons of continuous variables were based on t tests and chi squared tests were used to compare categorical variables. Measures of association were based on odds ratios from multivariable logistic regression for dichotomous outcomes. Survival analyses were conducted using Kaplan-Meier curves and proportional hazard modeling for time to event outcomes.Results:Within the study period a total of 214 patients with BC were identified, with 42 developing BC in the 1,267 post kidney transplant recipients. Among the 484 women post kidney transplant, 15 (3%) developed BC compared to 27 of 783 men (3%). The majority of BC was identified in men 27 of 42 (64%). The incidence of BC in women was 0.08 per 1,000 transplants vs. 0.09 per 1,000 transplants in men. The odds ratio for association of BC with male gender was OR=1.1. (95% CI 0.6, 2.2) with no significant difference noted.The median time to mortality among post-transplant patients with BC was 144 months (95% CI, 87, NA) compared to 198 months for patients without BC (95% CI 179, 252). This difference was not statistically significantly different on Cox Proportional Hazard modeling controlling for gender (HR=1.1, 95% CI 0.6, 2.1).Controlling for gender, patients with post-transplant tumors were nearly 3 times more likely to have died. (OR=2.9, 95% CI 2.8, 2.9). There wasn’t a statistically significant association of BC with reported post-transplant mortality when controlling gender (OR = 1.0, 95% CI 0.5, 2.0).Conclusion:We observed a three-fold increase in mortality for patients who had BC after transplant surgery, regardless of gender. Additionally, we interestingly found an unexpected shift in the gender distribution of BC cases among patients who underwent kidney transplant. Unlike the typical pattern seen in the general population, male transplant recipients exhibited a slightly higher risk of developing breast cancer compared to their female counterparts. This striking reversal of the usual gender disparity in BC represents a highly significant finding. Further investigation into the factors affecting the internal hormonal milieu in these patients may shed light on the physiologic basis of this heightened risk. Understanding the risk of BC in well-defined subsets of post-transplant populations may help guide screening approaches with hopes of lowering morbidity and mortality.
Presentation numberPS3-05-12
Genetic Risk Stratification for Breast Cancer in Patients Considering or Undergoing Hormone Replacement Therapy
Rodrigo Santa Cruz Guindalini, Oncology D’Or, São Paulo, Brazil
J. T. Santos1, D. B. Macedo1, P. B. Porto2, I. B. Rocha2, R. S. Guindalini1; 1Oncogenetics, Oncology D’Or, São Paulo, BRAZIL, 2Endocrinology, All Clinic, Rio de Janeiro, BRAZIL.
Introduction: Hormone replacement therapy (HRT) is a widely recommended strategy for relieving menopausal symptoms, particularly for healthy women under 60 years of age or within 10 years of menopause. However, HRT has been excessively used in Brazil, including less studied compounds and delivery methods, such as subcutaneous implants of gestrinone. Although HRT provides symptomatic benefits, the increased risk of breast cancer has already been confirmed with certain types of hormone therapy. While numerous risk factors—such as age at menarche, menopause, parity, and obesity—are well established, 5-10% of breast cancer cases are linked to hereditary genetic predisposition. In this context, tools such as CanRisk, when integrated with genetic counseling (GC) and germline testing, enable personalized risk assessment and inform clinical decision-making. Objective: To investigate how the integration of GC and CanRisk (v3.0.0) influences BC risk stratification and modifies screening and prevention recommendations in women using or planning to initiate HRT. Methods: This exploratory, quantitative, retrospective study analyzed electronic medical records of women referred for BC risk assessment at a private oncogenetics service in Brazil between September 2022 and May 2025. The primary reason for referral was to discuss HRT initiation or continuation. Testing indications followed the 2025 NCCN guidelines. Risk management recommendations were based on CanRisk estimates: breast MRI for lifetime BC risk >20%; chemoprevention for 5-year BC risk >1,7% or 10-year BC risk >5%; and risk-reducing surgery for carriers of high-penetrance pathogenic variants (PVs). Results: A total of 144 women were included (mean age: 51 years). Of these, 100 (69,4%) met NCCN criteria for genetic testing, and 95 (94,1%) proceeded with testing. Among the 44 women (30,6%) who did not meet criteria, 40 (90,9%) opted for testing after a shared decision-making process, totaling 135 tested individuals. The integration of CanRisk and GC resulted in a change in clinical management for 29 women (21,5%). Regarding chemoprevention, 117 women (86,7%) were initially eligible, and after genetic testing, 9 (6,7%) lost this indication, while 4 (3,0%) gained it due to findings of PVs in BC susceptibility genes. For MRI surveillance, 15 patients (11,1%) were initially eligible; 5 (3,7%) lost and 8 (5,9%) gained the indication based on revised lifetime risk estimates. A total of 14 pathogenic variant (PV) carriers (10.4%) were identified. Among these, nine women (6.7%) received new surgical recommendations due to high-penetrance PVs: four women with BRCA1 mutations were advised to undergo bilateral risk-reducing mastectomy and bilateral salpingo-oophorectomy; two women with BRCA2 mutations were recommended bilateral mastectomy—one had already undergone oophorectomy, while the other was additionally advised salpingo-oophorectomy. One woman with a PALB2 mutation was advised bilateral mastectomy. Two women with RAD51C and RAD51D mutations were recommended bilateral salpingo-oophorectomy. Notably, one patient was diagnosed with ductal carcinoma in situ after undergoing a risk-reducing mastectomy. Discussion and Conclusion: Integrating CanRisk and GC significantly impacted risk stratification, with the new BC risk estimates resulting in reclassification in over one-fifth of the cohort (21,5%). Importantly, genetic testing should not be universally indicated for all women considering or using HRT; however, this context provides a valuable window of opportunity to identify patients who already meet criteria for genetic counseling and testing but had not yet been referred. This proactive approach supports precise cancer prevention, better clinical safety, and informed, shared decision-making for HRT use
Presentation numberPS3-05-13
The impact of family history on breast cancer stage: data from a Brazilian breast cancer cohort without inherited predispositions.
Tatiana Strava Correa, Hospital Sírio Libanes, Brasília, Brazil
B. Resendes, R. Sandoval, T. Correa, A. Castro, Z. Souza, I. Guttieres; Centro de Oncologia Clínica, Hospital Sírio Libanes, Brasília, BRAZIL.
Introduction: Family history (FH) of breast cancer (BC) is an independent risk factor for BC, regardless the presence of a germline mutation in a BC predisposition gene. Women with a FH of BC could be diagnosed at earlier stages due to increased awareness and screening. Nevertheless, other factors could be associated with diagnosis at later stages such as early onset, fear, and misinformation. Objective: To determine if women with a family history of breast cancer present earlier stages of disease. Methods: This single institution study was conducted at the Oncogenetics Department of Hospital Sirio-Libanês (Brasília DF, Brazil). Eligibility included patients with a personal history of BC who received genetic counseling between 2017 and 2025, and a genetic testing result without a pathogenic/probably pathogenic germline variant in a BC gene. Lack of FH data was an exclusion criteria. Data were collected retrospectivelly from medical charts. FH of BC was defined as a BC cancer diagnosis in 1st, 2nd, and/or 3rd degree relatives (defined as close relatives). For patients who received neoadjuvant therapy, BC stage was based on clinical staging and pathological or clinical staging were used for the remaining cases. Chi-square test was used for comparison of patients with and without a FH of BC. Results: Among 161 patients, 155 met study criteria. Median age of BC diagnosis was 49 years (23.0-87.0), 6 patients were diagnosed during pregnancy or breastfeeding, 6 had bilateral synchronous BC and 7 metastatic de novo disease. Out of 155 patients, 88 (56.7%) had at least one close relative with BC (FH+), including 37 (23.9%) with a family member affected before 50 years and 7 (4.5%) with bilateral BC. In total, 68 (43.8%) patients were diagnosed due to symptoms, 68 (43.8%) through screening, 3 incidentally found during mamoplasty and in 16 cases presentation was not specified. Among those screen-detected cancers (n=68), 22 (32.3%) were detected by ultrasound, 35 (51.5%) by mammogram, 7 (10.3%) by MRI, and in 4 cases the diagnostic modality was unavailable. There were no differences between groups (FH+ vs FH-) regarding age of BC diagnosis, menopausal status, BC presentation, and BC stage. Conclusion: In this BC Brazilian cohort from a private health setting without a cancer predisposition syndrome, a family history of BC did not impact BC stage at diagnosis. Cancer risk assessment based on risk models such as Tyrer Cuzick could inform the benefit of BC screening with MRI in this scenario.
Across all variables tested, no statistically significant association was found with family history status (FH+ vs FH-) at the conventional p < 0.05 threshold.
Presentation numberPS3-05-14
Clinical significance of pathogenic variants in breast cancer patients at risk for hereditary breast cancer
Xiaying Kuang, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
X. Kuang, Y. Lin, N. Shao; Breast Cancer, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, CHINA.
Background: Approximately 5-10% of breast cancers (BC) are attributed to inherited germline pathogenic variants, primarily in genes involved in homologous recombination repair (HRR), most notably BRCA1/2. This study aimed to identify clinical risk factors associated with pathogenic variants in patients with a high genetic risk of breast cancer and evaluate the utility of multigene panel testing. Methods: A total of 533 high-risk BC patients underwent next-generation sequencing (NGS)-based germline multigene panel testing covering 61 HRR-related genes. Eligibility was defined by the presence of at least one of the following: triple-negative breast cancer (TNBC), early-onset disease (≤50 years), a family history of malignancy, or multiple primary tumors (including bilateral BC). Targeted capture-based sequencing was performed using DNA extracted from peripheral blood leukocytes. Results: The median age at diagnosis was 40.6 years old. All patients had ductal carcinoma. Pathogenic HRR variants were detected in 120 patients (22.5%), with 123 total variants identified; one patient (HR-positive/HER2-negative) carried two variants and another (TNBC) carried three. Pathogenic BRCA1 and BRCA2 variants were observed in 36 (6.75%) and 37 (6.94%) patients, respectively. Non-BRCA HRR pathogenic variants were present in 47 patients (8.82%), with PALB2 being the most frequently mutated non-BRCA gene (n=19, 3.56%). TNBC patients (43/145, 29.66%; P=0.012) and those with ≥2 clinical risk factors (84/329, 25.53%; P=0.021) had a significantly higher prevalence of pathogenic variants. A trend toward increased mutation frequency was noted in younger age groups (≤30y: 27.54%, 31-40y: 23.61%, 41-50y: 20.96%, ≥51y: 17.19%), although the difference was not statistically significant (P=0.284). Among HR-positive/HER2-negative and HR-negative/HER2-positive patients, pathogenic variant prevalence was 20.65% and 18.75%, respectively. Conclusions: This study highlights the clinical relevance of germline HRR gene testing in high-risk BC patients. Multigene panel testing should be considered routine for patients with TNBC or those presenting with multiple risk factors to guide personalized management and familial risk assessment.
Presentation numberPS3-05-15
Targeting Breast Cancer via Dual Antagonism of Estrogen Receptor α and GPER
SHWETA SINGH, University of Texas Rio Grande Valley, McAllen, TX
S. SINGH, S. Vidaurri, M. Noorani, S. Goyal, A. Dhasmana, S. Dhasmana, M. Yallapu, S. Fofana, S. C. Chauhan, D. Nguyen, S. Khan; School of Microbiology and Immunology, University of Texas Rio Grande Valley, McAllen, TX.
Targeting Breast Cancer via Dual Antagonism of Estrogen Receptor α and GPERBackgroundEstrogen’s diverse biological effects are primarily driven by the classical receptors ERα and ERβ,which function as ligand-activated transcription factors. Among these, ERα plays a pivotal role inbreast cancer development, making it a key target for therapies such as tamoxifen. However,tamoxifen’s clinical success is often compromised due to both inherent and acquired resistance.Notably, tamoxifen inhibits ERα but paradoxically acts as an agonist for GPER/GPR30, estrogenreceptor that mediates non-classical estrogen signaling in normal and cancerous cells. Activationof GPER can promote gene expression and cell proliferation, contributing to tamoxifen resistancein hormone-sensitive tumors. Our study addresses this challenge by focusing on a dual-targetingstrategy that simultaneously inhibits both ERα and GPER, aiming to overcome resistance andimprove therapeutic outcomes in breast cancer treatment.MethodsGPER expression was evaluated in breast cancer cell lines and tumor tissues using immunoblotting,qRT-PCR, and immunohistochemistry. Cell proliferation was assessed via MTT and colonyformation assays. Migration and invasion capabilities were measured using wound healing andtranswell invasion assays. Intracellular calcium signaling was monitored using fluorescence-basedcalcium imaging following GPER stimulation. Metagenomic analysis of Hispanic breast cancertissues were performed using 16S rRNA sequencing to identify microbial populations correlatedwith GPER expression and tumor subtypes. Statistical analyses included correlation tests andsubtype stratification.ResultsGPER expression was detected in both breast cancer cell lines and tumor tissues. Our resultsdemonstrate that GPER is significantly overexpressed in ERα-positive breast tumors, with aunique correlation observed in a Hispanic breast cancer patient population (P < 0.001). GPERexpression was also observed in Triple-Negative subtypes. The correlation of GPER expressionwas analyzed across different breast cancer subtypes, classified as Luminal A (HR+/HER2−),Luminal B (HR+/HER2+), HER2-Enriched (HR−/HER2+), and Triple-Negative (HR−/HER2−).In vitro studies confirmed the presence of GPER protein and RNA in breast cancer cells of varioussubtypes. Furthermore, this study identifies Ormeloxifene (ORM), a selective estrogen receptormodulator (SERM), as a dual antagonist of ERα and GPER. GPER expression was associated withincreased cancer aggressiveness, migration, and invasion, all of which were effectively inhibitedby Ormeloxifene. ORM acts as an antagonist ligand for both ERα and GPER by suppressing genetranscription and inhibiting growth signaling pathways in breast cancer cells. Mechanistically,Ormeloxifene suppresses GPER-induced calcium signaling and downstream activation of EGFR,ERK, YAP, and TAZ pathways, thereby inhibiting gene transcription and tumor growth. Additionally, metagenomic analysis of Hispanic breast cancer tissues revealed specific microbialpopulations associated with GPER expression and tumor subtypes.ConclusionsGPER is significantly overexpressed in ERα-positive and some Triple-Negative breast cancers,especially within the Hispanic population. The exclusive antagonistic activity exerted byOrmeloxifene on both ERα and GPER represents an innovative pharmacological approach fortargeting breast carcinomas expressing one or both receptors at diagnosis or during progression.Simultaneous inhibition of ERα and GPER may offer greater therapeutic benefits compared toselective estrogen receptor antagonists alone. Additionally, the association of GPER expressionwith specific microbial populations suggests new directions for personalized breast cancertreatment.
Presentation numberPS3-05-16
Recurrence Following Initiation of Low Dose Tamoxifen for Breast Cancer Prevention: A Real-World Study
Karishma Vijay Rupani, Icahn School of Medicine at Mount Sinai, New York, NY
K. Rupani1, E. Moshier2, C. Jin3, D. Jaber4, N. Casasanta5, C. Wu6, A. Tiersten*7, R. Patel*7; 1Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 2Division of Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, 3Department of Pathology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, 4Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 5Division of Hematology and Medical Oncology, Department of Medicine, Yale-New Haven Hospital, New Haven, CT, 6Department of Anesthesiology, Perioperative and Pain Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 7*Denotes co-final authors, Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: Patients diagnosed with non-invasive breast lesions such as DCIS, LCIS, ADH, and ALH are at increased risk of developing invasive breast cancer (BC). The TAM-01 study found that low-dose tamoxifen (LDT) for chemoprevention resulted in an approximate 50% risk reduction in BC events compared to placebo. A recent study at our institution (PMID: 39837695) showed that the uptake of LDT was 19%. The objective of our present study was to evaluate the recurrence patterns for patients who initiated LDT. Methods: We performed a retrospective review of patients with DCIS, LCIS, ADH, and/or ALH who started LDT from January 2019 to December 2021. We evaluated rates of LDT completion and recurrence characteristics. A time-dependent Cox regression model was used to evaluate the association between early discontinuation of LDT and risk of recurrence, accounting for changes in LDT use over time. Results: 88 eligible patients were identified and 7 were excluded due to lack of follow up. Of the remaining 81 patients, 67% (N=54) completed 3 years of LDT and 30% (N=24) stopped early due to adverse events (AEs); the remaining 3% stopped due to recurrence (N=1), to attempt pregnancy (N=1), and diagnosis of another malignancy (N=1). Overall, the mean age of our population was 54 years (SD 12) and 39% (N=32) were postmenopausal. 39% (N=32) of patients had DCIS, and 61% (N=49) had either LCIS, ADH, and/or ALH. At a median follow up time of 4.74 years, the overall recurrence rate was 10% (N=8). Among the 27 patients who discontinued LDT early, 19% (N=5) recurred: 4 after discontinuing LDT due to AEs and 1 recurred while on LDT. Among 54 patients who completed LDT, 6% (N=3) recurred: 1 during treatment and 2 after 3 years of LDT (Table 1). Based on the time-dependent Cox model, the estimated 5-year probability of recurrence was 8% for patients who completed LDT and 15% for those who discontinued early. Early discontinuation was associated with nearly a two-fold increased risk of recurrence, although this was not statistically significant due to the low event rate [HR: 1.94; 95% CI: 0.42-9.07; p=0.40]. All recurrences were in patients who had LCIS, ADH, and/or ALH. Most recurrences (75%) were in premenopausal patients, and the 3 patients who recurred after completing 3 years of LDT were all premenopausal. Except for 1 patient who developed HR-, HER2+ IDC and DCIS while on LDT, all other recurrences, irrespective of completion of 3 years of LDT, were atypia or LCIS. Conclusions: In patients initiated on LDT for chemoprevention, at a median follow up of 5 years, the estimated probability of recurrence was 8% in those who completed 3 years of LDT and 15% in those who stopped early. Most recurrences were seen in premenopausal patients consistent with a subgroup analysis of the TAM-01 study suggesting decreased efficacy of LDT in premenopausal women.
| Initial Diagnosis | Menopausal Status |
Completed 3 years of LDT?
Reason for discontinuation |
Type of Recurrence | Recurrence Treatment |
| LCIS | Pre-menopausal | Yes | Contralateral ADH | Continued tamoxifen 5 mg |
| LCIS | Pre-menopausal | Yes | Ipsilateral ALH | Surveillance |
| ALH | Pre-menopausal | Yes | Contralateral ADH and ALH | Lumpectomy |
| LCIS and ADH | Pre-menopausal |
No
Back pain, headaches, ocular issues, mood changes |
Ipsilateral LCIS and ADH | Surveillance |
| LCIS and ADH | Pre-menopausal |
No
Severe migraines |
Ipsilateral LCIS, ADH and ALH | Lumpectomy |
| LCIS and ADH | Pre-menopausal |
No
Mood changes, bloating |
Ipsilateral LCIS and ADH | Surveillance |
| LCIS, ADH and ALH | Post-menopausal |
No
Hot flashes, weight gain |
Ipsilateral LCIS | Surveillance |
| ADH and ALH | Post-menopausal |
No
Developed recurrence while on LDT |
Ipsilateral HR-, HER2+ IDC, DCIS | Lost to follow up |
Presentation numberPS3-05-17
Prevalence of BRCA 1/2 mutation among breast cancer patients in Kenya
Abeid Athman Omar, KUTRRH, Nairobi, Kenya
A. Athman Omar1, R. Mukadam2, A. Kalebi3; 1Oncology, KUTRRH, Nairobi, KENYA, 2Pathology, Dr. Kalebi Lab – DKL, Nairobi, KENYA, 3Pathology, DKL, Nairobi, KENYA.
Background: Breast cancer (BC) is the most common cancer in Kenya, with patients being diagnosed a decade earlier than in the USA. Patients harbouring BRCA1/2 mutations have aggressive disease, but paradoxically respond well to tailored treatments such as poly ADP-ribose polymerase (PARP) inhibitors. However, BRCA testing is not accessible for most patients in Africa; thus, the prevalence largely remains unknown, with very scant published literature from Sub-Saharan Africa showing a wide variation from 2% up to 17%. We hereby report the prevalence of BRCA1/2 mutations for patients screened in the first six months in a recently opened independent referral lab in Kenya. Methods: The BC patients with high-risk clinical features such as young age, triple-negative breast cancer (TNBC), bilateral BC, and familial history of breast cancer who had been referred for germline (gBRCA 1/2) or somatic (sBRCA 1/2) testing were screened. After consent, patients’ samples, either peripheral blood or Formalin-Fixed Paraffin-Embedded, were tested for BRCA 1/2 mutations using the Oncomine BRCA Research Assay. Gene variant ID, variant class, and variant allele frequency, among others, were reported. Only patients with a confirmed BC diagnosis and de-identified data were included in this analysis. Results: In the six months, 42 patients were screened for BRCA 1/2 mutation. Of which 14 females had confirmed BC diagnosis, and their median age is 47 years (range: 29 – 70). The majority (10 out of 14) had triple-negative breast cancer, one with bilateral breast cancer, while another had metastasis to the ovary. The family history was not provided for any of these patients. The prevalence of BRCA 1/2 mutations was 21% among the patients who underwent germline testing (n=12). Two had pathogenic variants in BRCA1, while one in BRCA2. The most common mutation was indels. Moreover, three patients had a BRCA 2 variant of uncertain significance (VUS). The most common mutation was missense. Conclusion: In this study, the prevalence of BRCA 1/2 mutations among BC patients in Kenya is 20%. To our knowledge, this is among the first works on BRCA testing in BC reported from Kenya, and increased availability and accessibility to oncogenetic testing will not only lay a foundation for future research work but also facilitate better management of BC patients with PARP inhibitors.
Presentation numberPS3-05-18
Non-breast cancer related germline mutations in breast cancer patients: a single institution experience.
Maha Zafar, Arkansas College of Osteopathic Medicine, Mercy Hospital Fort Smith, Fort Smith, AR
M. Zafar1, M. Krishnakumar2, A. Reddy3; 1Department of Internal Medicine, Arkansas College of Osteopathic Medicine, Mercy Hospital Fort Smith, Fort Smith, AR, 2Department of Internal Medicine, SRM Medical College Hospital and Research Center, Chennai, INDIA, 3Department of Hematology Oncology, Mercy Clinic, Fort Smith, AR.
Background: Breast cancer is the most common non-cutaneous malignancy among women, with around 10% of cases involving germline mutations in DNA repair genes. Germline mutations in BRCA1/2 genes significantly increase breast cancer risk. Other relevant genes to increase the risk of breast cancer include TP53, PTEN, PALB2, CHEK2, and ATM. Multigene panel testing can sequence many genes, sometimes identifying non-disease-related genes and variants of unknown significance. Methods: We reviewed patients with newly diagnosed breast cancer diagnosis from 2022 to 2024 in our institution. Germline genetic testing was offered to all newly diagnosed breast cancer patients at the time of diagnosis irrespective of their family history. Testing was offered to patients with invasive breast cancer and ductal carcinoma in situ. The goal of this study was to identify the prevalence of genes not attributed to increasing the risk of breast cancer and the implications of those results. Results: A total of 478 patients (Invasive cancer and DCIS) were seen in our oncology clinic from January 2022 to December 2024. The median age at diagnosis was 66 years (range 28-91). Invasive ductal cancer (IDC) was the most common subtype (66%), followed by DCIS (18%). Most patients had estrogen-positive cancer (81%), and approximately 14% had HER2-positive disease (62% HER2 low). We found 27 patients with pathogenic germline mutations which are not related to breast cancer risk. The most common mutation was MUYTH (7 patients), followed by FH gene (3 patients). MSH3, LZTR1, and HOXB13 genes were each found in 2 patients. APC, BAP1, BLM, BRIP1, NTHL1, SDHA, SDHB, MSH6, MITF, and FLCN genes were each identified in one patient. All patients with MUYTH mutations had hormone-positive (HER 2 negative) disease contrary to the limited observation of triple-negative breast cancer related to the MUYTH gene in the available literature. Patients with positive mutations were offered genetic counseling and referred to a high-risk genetics’ clinic. FANCM was observed in one patient which has rarely been associated with an increased risk of triple-negative breast cancer. Conclusion: We present our single institution’s experience of germline mutations in breast cancer patients. Previously, we observed CHEK2 being the most common breast cancer-related germline mutation in our patient population. This analysis focuses on identifying germline mutations without an increased risk of breast cancer. MUTYH was the most common pathogenic mutation identified. Further studies are needed to determine the actual risk percentage of breast cancer associated with the MUTYH gene. Universal testing for all breast cancer patients is feasible with the lower cost of multi-gene panels. However, identifying other non-disease-related genes, as observed in our study, has implications. Patients must be informed about the consequences of a positive result, including genetic counseling, additional screening tests, and testing of immediate family members.
Presentation numberPS3-05-19
Black women perceptions regarding use of biomarker screening for breast cancer prevention and treatment.
Renisha Campbell, University of CA, San Francisco, San Francisco, CA
R. Campbell1, L. Houghton2, D. Walker3; 1School of Nursing- Community Health Systems, University of CA, San Francisco, San Francisco, CA, 2Mailman School of Public Health, Columbia University, New York, NY, 3Medical Humanities and Ethics, Columbia University, New York, NY.
Introduction/Background: Early-onset breast cancer remains a critical public health concern. Despite advances in medical research, young women continue to account for approximately 15% of breast cancer (BC)-related deaths. Among these, Black young women have double the mortality rate at 12.1 per 100,000 compared to young White women. Black young women face significant disparities in both access to comprehensive BC preventive care, and the personalization of BC treatments. There is a crucial need for improved primary prevention strategies to reduce BC incidence and ensure access to personalized treatment upon diagnosis. Biomarker-based preventative BC screening, and monitoring post-diagnosis offers a promising avenue to combat these disparities. These approaches have the potential to provide individualized risk assessments and individualized BC treatment. The purpose of this study was to explore the perspectives of Black women under the age of 50 living with BC on the acceptability of biomarker-based primary BC prevention screening, as well as biomarker’s role post BC diagnosis. Study Aims: (1) Examine the perspectives of Black women living with BC on the acceptability of preventative BC biomarkers, and use of biomarker monitoring after BC diagnosis; (2) Understand the breast health and BC risk information available to young Black women prior to their diagnosis; (3) Explore the pathways leading to BC diagnosis of these women; and (4) Assess the extent to which biomarker monitoring and genomic testing was received after their BC diagnosis. Methods: Nineteen Black women living with breast cancer were invited to participate in virtual focus groups (FG) via the Zoom platform in November 2025. Each FG session was preceded by an educational PowerPoint presentation detailing the role of primary preventive BC biomarker screening, and biomarker and genomic screening following a BC diagnosis. Using a critical lens informed by Intersectionality Theory and the Embodied Health Movement Theory, Reflective Thematic Analysis was employed to analyze FG transcripts, extracting both articulated and emergent themes. Results: Participants generally expressed openness to the idea of preventive BC biomarker screening, recognizing its potential benefits. However, concerns arose regarding the financial burden of screening, insurance coverage, and fear of how test results might impact access to job benefits. Two participants reported a comprehensive breast assessment due to a known family genetic BC mutation. While 17 participants reported receiving minimal breast health education, no comprehensive BC risk assessments prior to diagnosis, and identified their breast change(s) themselves, which lead to BC diagnosis. Regarding the use of biomarkers in post-diagnosis treatment, responses varied widely. Access to biomarker monitoring was influenced by factors such as the time since diagnosis, disease aggressiveness, and physician preference. Several emergent themes surfaced during the discussions, including the need for self-advocacy both before and after diagnosis, the racialized experience of BC, and the inadequate marketing of BC prevention. Conclusion and Future Research: Biomarkers for BC prevention and use post- diagnosis are novel but promise more personalized BC risk assessments, individualized BC treatment, and improved disease monitoring. Future research should prioritize randomized controlled trials evaluating preventive BC biomarkers, and biomarker monitoring post-diagnosis among diverse young women.
Presentation numberPS3-05-20
Genetic Mutations in a Young Population in Chile with Breast Cancer
Maria Trinidad Esperanza Gonzalez, Clinica Alemana, Santiago, Chile
M. E. Gonzalez1, M. Mullins2, C. Barriga2, M. Bravo2, D. Carvajal2, J. Camacho2; 1Surgery, Clinica Alemana, Santiago, CHILE, 2Oncology, Clinica Alemana, Santiago, CHILE.
IntroductionApproximately 5-10% of breast cancer cases are linked to genetic predisposition, primarily involving pathogenic germline variants in BRCA1 and BRCA2, which confer a lifetime risk of 80-90% and 60-85%, respectively. Other genes like CHEK2, BRIP1, and PALB2 are associated with moderate risks (20-40%). While pathogenic variants can predispose to triple-negative breast cancer, most cases occur without these mutations. High-risk patients may opt for genetic screening and prophylactic mastectomy, which reduces breast cancer risk and improves disease-free survival, though it does not eliminate risk or increase overall survival. ObjectiveThis study aims to analyze genetic mutations and variants of uncertain significance in breast cancer cases treated at Clínica Alemana, Santiago, over the past ten years, and compare them with global data. It also examines how these findings influence clinical management, including prophylactic mastectomy. Methods and materialsRetrospective descriptive study of genetic mutations in 200 women aged ≤40 with breast cancer over 10 years at Clínica Alemana, Santiago, Chile. ResultsIn this study, 47.4% of patients received genetic counseling and all underwent testing. Pathogenic mutations were found in 18 patients, mostly in BRCA1/2, while 28 had variants of uncertain significance, primarily in CHEK2. DiscussionGenetics are implicated in about 10% of breast cancer cases, consistent with our finding of 9.4% positive genetic tests. Variants of uncertain significance (VUS) occur in 2-7% of BRCA1/2-only tests and up to 10-41% in multigene panels; our study found 14.6% VUS after multigene testing. BRCA1/2 pathogenic variants were present in 4.2% of cases, aligning with reported rates of 3-4%. Prophylactic mastectomy rates vary between 40-80% in carriers of high-penetrance mutations, and are lower for moderate-penetrance genes. In our cohort, 63% of BRCA1/2 carriers and 10% with moderate-risk gene mutations underwent prophylactic mastectomy.
| Pathogenic Mutation | Population | Prophylactic mastectomy |
| BRCA 1 | 5 | 3 |
| BRCA 2 | 3 | 2 |
| PALB2 | 2 | 1 |
| MUTYH | 2 | 0 |
| ATM | 2 | 0 |
| APC | 1 | 0 |
| MSH2 | 1 | 0 |
| LIN | 1 | 0 |
| TP53 | 1 | 0 |
| None | 174 | 18 |
| Pathogenic Mutations | 18 (19.8%) |
| Uncertain variant | 28 (30.8%) |
| No genetic alterations | 45 (49.4%) |
Presentation numberPS3-05-21
Pathogenic germline variants associated with different HER2 expression among breast cancer patients
Ning Liao, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
N. Liao1, J. Wu2, G. Zhang1, J. Weitzel3, C. Balch4; 1Department of Breast Cancer, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, CHINA, 2Clinical Research Devision, Kanghui Biotech, Beijing, CHINA, 3Department, The University of Kansas Comprehensive Cancer Center, Leawood, KS, 4Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Purpose: Germline mutations were evaluated in 530 Chinese breast cancer (BC)patients with different HER2 expression status plus 98 patients with atypical ductalhyperplasia or a breast cancer family history.Methods: DNA extracted from blood samples was analyzed with a next generationsequencing based multigene panel, with reporting of likely pathogenic and pathogenicvariants (PV) of 102 cancer associated genes, and correlation betweenclinicopathologic characteristics and known BC-associated genes. Results: The 71 identified PVs were categorized into five distinct pathway clusters:BRCA/FANC, DDR, HRR, FANC, and Other. The distribution of PVs enriched withinthese clusters differed significantly between BC patients and unaffected individuals(p=0.031). This difference was primarily driven by the BRCA/FANC, FANC, and DDRclusters, with enrichment percentages of 57.7% vs. 16.7% (BRCA/FANC), 7.0% vs.25.0% (FANC), and 15.5% vs. 41.7% (DDR) in BC vs. unaffected groups, respectively.Notably, the three BC groups stratified by HER2 expression level exhibited distinct PVdistributions. Both the HER2-low and HER2-zero BC groups showed significantlydifferent distributions compared to unaffected individuals (p=0.0132 and p=0.0081,respectively). The BRCA/FANC cluster was the predominant pathway enriched in bothHER2-low (59.6%) and HER2-zero (81.8%) groups. Furthermore, the PV distributionin the HER2-zero group was significantly different from that in the HER2-high group(p=0.0028). In contrast, the distribution in the HER2-high group resembled thatobserved in non-BC individuals. These findings indicate that BC patients with differentHER2 expression statuses harbor distinct germline PV signatures, which correlate withdifferential clinical outcomes following neoadjuvant systemic therapy.Discussion: Chinese breast cancer patients with different HER2 expression statusdemonstrated different pathogenic germline variant, which correlated with differentclinical outcome after neoadjuvant therapy. Multi-gene genetic test for pathogenicgermline variant should be offered to breast cancer patients as well as high-riskindividuals at risk for future breast cancer who would benefit from prevention and earlydetection programs.
Presentation numberPS3-05-22
Real-world adherence to germline genetic testing guidelines in breast cancer: patterns and disparities in a community oncology setting
Lyeba Shahid, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA
L. Shahid1, A. J. Patev2, M. A. Danso3, R. A. Burke4, N. Balanchivadze3; 1Internal Medicine, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, 2Research and Infrastructure Service Enterprise, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA, 3Hematology/Oncology, Virginia Oncology Associates, Norfolk, VA, 4Surgical Oncology, Macon & Joan Brock Virginia Health Sciences at Old Dominion University, Norfolk, VA.
Title: Real-world adherence to germline genetic testing guidelines in breast cancer: patterns and disparities in a community oncology setting Authors: Lyeba Shahid, DO; Alison J. Patev, PhD; Michael Danso, MD; Rachel Burke, MD; Nina Balanchivadze, MD Objectives Germline genetic testing is a critical component of personalized care in breast cancer, informing surveillance, treatment, and risk-reduction strategies for patients and their families. National Comprehensive Cancer Network (NCCN) recommend testing patients with early breast cancer and specific high-risk features. However, adherence to these guidelines in real-world community settings, and the role of social determinants such as insurance and geography, remains incompletely understood. Methods: We retrospectively reviewed charts of patients who were diagnosed with breast cancer at Sentara Norfolk General Hospital between June 1, 2019 to June 1, 2024. The sample included 89 patients, between 36 to 65 years old (M = 55.53), with 88 female patients and 1 male patient. Patients were deemed eligible for genetic screening in accordance NCCN Guidelines Version 1.2025, which included: 1) diagnosis of breast cancer at < 50 years old, or 2) triple-negative breast cancer, multiple primary breast cancers, or male breast cancer, or 3) a family history of one or more close blood relative with breast cancer at or before age 50, pancreatic cancer, male breast cancer, ovarian cancer, prostate cancer with high-risk features, or three or greater breast and/or prostate cancer diagnoses on the same side of the family [1]. Demographic variables included race, marital status, insurance, and social vulnerability, which was derived from zip code. Results: Genetic testing was ordered for 75% of patients (n=67). Of the total sample, 65 out of 89 patients had testing completed. Fisher’s exact tests explored relationships between social determinants and testing. For eligible patients, no significant relationships were found. However, examination of subgroup testing completion rates showed some group differences. Nearly 92% of White eligible patients received testing, compared to 77% of Black eligible patients. Married patients completed testing at a rate of almost 87%, while 70% of single people completed testing. Additionally, ninety-two percent of eligible patients with a qualifying family history were tested, compared to 83% based on age and 80% based on cancer type. Discussion: Despite high overall adherence to NCCN guidelines for germline genetic testing in this community-based breast cancer cohort, disparities emerged across several social dimensions. While statistical significance was not reached, subgroup analyses revealed meaningful trends. Black patients were less likely to complete testing compared to White patients, highlighting potential racial disparities in access, communication, or trust. Similarly, single patients had lower testing completion rates than married patients, suggesting that social support may influence engagement with genetic services. Overall, these findings suggest that even in a high-performing clinical setting, social determinants of health may influence the delivery of precision oncology, and targeted interventions are needed to ensure equitable access to genetic testing for all eligible patients. References:– [1] National Comprehensive Cancer Network. Genetic/Familial High-Risk Assessment: Breast, Ovarian, and Pancreatic. Version 1.2025. https://www.nccn.org/professionals/physician_gls/pdf/genetics_bop.pdf.
Presentation numberPS3-05-23
Characteristics and Outcomes of Breast Cancers Diagnosed in Patients with Germline Hereditary Mutations – The BC Cancer Hereditary Cancer Program High Risk Clinic Experience
Zahi Mitri, BC Cancer Vancouver, Vancouver, BC, Canada
Z. Mitri1, K. Preston2, A. Harrison1, A. Smith1, T. Nghiem1, R. Cheifetz1; 1Medical Oncology, BC Cancer Vancouver, Vancouver, BC, CANADA, 2Medicine, University of British Columbia, Vancouver, BC, CANADA.
Background. Individuals with germline hereditary mutations in breast cancer (BCa) predisposition genes are at significantly higher risk of developing BCa during their lifetime compared to the general population. In British Columbia, patients identified as carrying a deleterious mutation are followed by the Hereditary Cancer Program (HCP) High Risk Clinic (HRC) for management of their increased BCa risk. This study evaluates the clinical and pathologic features, and outcomes for BCa diagnosed in hereditary mutation carriers under the HRC.Methods. Patients followed in HRC with a confirmed deleterious hereditary mutation, and at least one BCa diagnosis after HCP enrollment were included in this study. Patient and tumor characteristics, detection method, treatment, and survival outcomes were collected. BCa diagnoses were categorized as prevalent (detected on first screening after HCP enrollment); incident (detected on routine imaging beyond first screening). Prevalent or incident BCa detected within 6 months of a normal screening and outside of the usual screening schedule were characterized as interval cancers. Results. Clinico-Pathologic Characteristics. Between 1997-2023, 166 patients were diagnosed with BCa out of 2011 patients (166/2011, 8.2%) seen in HRC in that period. Median age of BCa diagnosis was 45 (IQR 39-57). The most common deleterious mutations were BRCA1 (44.6%) and BRCA2 (40.4%). Other mutations included CHEK2 (4.2%), PALB2 (4.2%), TP53 (3.6%), ATM (2.4%), and NF1 (0.6%). In our cohort, 39 (23.5%) BCa were classified as prevalent, 127 (76.5%) as incident. Twenty-four cases (14.5%) met the criteria for interval BCa. MRI was the most common detection modality (50.6%), followed by mammography (26.5%), clinical exam (10.8%), and ultrasound (2.4%). Of note, 16 (9.6%) cases were diagnosed at time of prophylactic surgery and were not identified on prior imaging. Among all BCa diagnoses, 37 cases (22.3%) were in-situ disease only, and 129 (77.7%) had invasive disease on final pathology. Within invasive tumors, 66.7% had stage 1, 26.4% stage 2, 6.2% stage 3, and one patient had de-novo stage 4 disease at diagnosis. Incident cancers were more likely to be diagnosed at stage 1 compared to prevalent cancers (71% vs 48%, p=0.015). Invasive BCa subtype distribution was 51.9% hormone receptor positive HER2-negative (HR+HER2-), 32.6% triple negative (TNBC), and 15.5% HER2-positive. Of note, BRCA1 tumors were more likely TNBC (52.2%, p<0.001) and BRCA2 tumors were more likely HR+HER2- (60.3%, p<0.001). Treatment. Most patients (153/166, 92.2%) underwent bilateral mastectomies as the surgical treatment of choice. Among patients with invasive BCa, 79 (47.6%) received chemotherapy either in the neo-adjuvant or adjuvant setting. There was no statistically significant association between incident or prevalent BCa and receipt of chemotherapy. There is a marginally statistically significant association between interval cancer and receipt of chemotherapy (p =0.067). Approximately 20% of all patients received adjuvant radiation therapy, including 26/153 (17%) who had bilateral mastectomies; 114/166 (68.7%) underwent prophylactic risk-reducing bilateral salpingo-oophorectomy. Outcomes. At time of last follow up, there were 8 deaths in our cohort, 5 due to metastatic BCa, and 3 due to ovarian cancer. An additional 10 patients were diagnosed with recurrent BCa and undergoing treatment. Three and 5-year overall survival rates in our cohort were 98% and 95.2%.Conclusions. Patients diagnosed with BCa under the HCP program have excellent outcomes. BRCA mutations are the most common identified alteration, and BRCA1 is enriched for TNBC. Further work is needed to identify clinical and biological features of interval cancers in this population.
Presentation numberPS3-04-01
Prediction of pathologic complete response from histopathology images of HER2+ breast cancer using an AI foundation model
Reva Basho, Ellison Medical Institute, Los Angeles, CA
R. Basho1, E. Heredia2, H. McArthur3, Z. Fang2, V. Narumi4, A. LeVee5, T. Jonsson6, F. Dadmanesh7, S. Bose7, S. Patre2, T. Keller2, J. Paine2, B. Tran4, B. Simental4, O. Castellanos4, J. Lee8, S. Shiao9, D. Agus1, N. Matasci2; 1Medicine, Ellison Medical Institute, Los Angeles, CA, 2AI and Advanced Molecular Medicine, Ellison Medical Institute, Los Angeles, CA, 3Internal Medicine, UT Southwestern, Dallas, TX, 4Clinical Research, Ellison Medical Institute, Los Angeles, CA, 5Medicine, University of California Los Angeles, Santa Monica, CA, 6Program Management, Ellison Medical Institute, Los Angeles, CA, 7Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, 8Molecular Carcinogenesis, Ellison Medical Institute, Los Angeles, CA, 9Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA.
BackgroundNeoadjuvant taxane-based chemotherapy in combination with anti-HER2 therapy is standard in patients with locally advanced HER2-overexpressing (HER2+) breast cancer. Pathologic complete response (pCR) after combination therapy is associated with improved long-term outcomes and occurs less frequently in patients whose disease is also estrogen and/or progesterone receptor positive (HR+) compared to patients with estrogen and progesterone receptor negative (HR-) disease. In patients with HER2+, HR+ disease, improved patient selection for standard neoadjuvant therapy is needed to optimize outcomes and minimize toxicity in patients unlikely to achieve pCR. Here we sought to utilize digital pathology and artificial intelligence/machine learning (AI/ML) algorithms on baseline biopsy tissue of newly diagnosed patients with operable HER2+ breast cancer to predict likelihood of pCR after standard neoadjuvant therapy, with a particular focus on HR status-specific differences.Methods Digitized H&E-stained baseline biopsy slides from patients with HER2+ operable breast cancer treated neoadjuvantly with taxane-based chemotherapy plus anti-HER2 therapy with pathologic response data available were identified from 2 publicly available datasets and an institutional cohort. Utilizing CanvOI, a pre-trained computational pathology foundation model, we developed an AI/ML model for predicting pCR from whole-slide images (WSIs). For processing, WSIs were divided into 380×380 pixel tiles. CanvOI’s tile encoder converted tile image data into numeric vectors, and subsequently, CanvOI’s slide encoder converted tile vectors into an aggregated slide vector. The slide vectors were used as predictors of pCR for HR+ and HR- (ER and PR ≤ 10%) cohorts independently, using logistic regression as a vector-data classification method. Model performance and generalizability were evaluated using 4×4 cross-validation, consisting of 4-fold cross-validation repeated 4 times with randomly mixed data. In each iteration, 75% of the data was used for model training and 25% for evaluation. Multivariable logistic regression analysis was conducted in each cohort to evaluate the model and pathological features associated with pCR. To address bias due to the naturally occurring imbalance of pCR rates within the HR+ and HR- cohorts, we implemented sample trimming in the dominant class within each cohort.ResultsA total of 231 patients had adequate baseline tissue for analysis, N=119 HR+ and N=112 HR-. The rates of pCR for these patients was 40.4% for HR+ disease and 64.1% for HR- disease. For prediction of pCR status, the model achieved a mean Area Under the Curve (AUC) of 0.69 (95% CI: 0.66-0.73) and Matthews Correlation Coefficient (MCC) of 0.304 in HER2+/HR+ patients, and a mean AUC of 0.65 (95% CI: 0.61-0.69) and MCC of 0.213 in HER2+/HR- patients. After multivariable analysis evaluating the ML model, tumor grade (1-2 vs 3) and HER2 immunohistochemistry status (2+ vs 3+) for patients in whom the pathological data was available (N=70 HR+ and N=73 HR-), the ML model remained a significant predictor of pCR in both HR+ and HR- cohorts. Applying data balancing techniques improved model performance, with a mean AUC of 0.70 (95% CI: 0.67-0.73) and MCC of 0.318 in HER2+/HR+ patients, and a mean AUC of 0.67 (95% CI: 0.63-0.71) and MCC of 0.181 in HER2+/HR-.Conclusions These findings demonstrate the potential of a digital pathology-based AI/ML model to capture information about tumor sensitivity to therapeutic agents that are not provided by current clinical parameters. In patients with HER2+, HR+ operable breast cancer, where rates of pCR remain low, such a tool has the potential to guide the evaluation of novel therapeutic strategies in appropriately selected patients. Larger datasets are needed to build upon these findings.
Presentation numberPS3-04-02
A Novel Non-invasive Machine Learning Model for Predicting Tertiary Lymphoid Structures and Treatment Response to Neoadjuvant Therapy in Triple-Negative Breast Cancer: A Multicenter Retrospective Study
Yidan Lin, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
Y. Lin1, Y. Yu1, Q. Wang1, K. Huang2, S. Tang3, J. Yuan4, C. song1; 1Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, CHINA, 2Department of Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, CHINA, 3Department of Breast Surgery, Yunnan Cancer Hospital, Kunming, CHINA, 4Department of Medical Imaging, Henan Cancer Hospital, Zhengzhou, CHINA.
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) and neoadjuvant therapy (NAT) has become an essential treatment strategy. Recent studies have showed tertiary lymphoid structures (TLSs) is associated with better treatment response to NAT in BC. However, there is still a lack of non-invasive biomarkers to predict the presence of TLSs in TNBC. This study aimed to develop a novel non-invasive machine learning model for early identification and prediction of the presence of TLSs and treatment response to NAT in TNBC, which were essential for timely adjustments in treatment strategies for TNBC. Methods: In this study, 698 patients from multicenter were divided into a training cohort (n = 137), the TLSs validation cohort (n = 63) and the NAT response validation cohorts (n = 561). A total of 4788 radiomic features per patient were extracted from the intratumoral and peritumoral regions of DCE-MRI. After extracting the optimal features, five machine learning models were developed to predict the presence of TLSs, including K-Nearest Neighbors (KNN), Extreme Gradient Boosting (XGBoost), Light Gradient Boosting Machine (LightGBM), Support Vector Machine (SVM) and Multilayer Perceptron (MLP) and were subsequently applied to predict the treatment response to NAT in NAT response validation cohorts. The performance of the models was assessed by accuracy, sensitivity, specificity, negative predictive value (NPV), positive predictive value (PPV), and area under curve values (AUC), and prognostic analysis were performed to evaluate its predictive value. Then,the rTLS predictive model was interpreted by SHapley Additive Explanations (SHAP). Finally, the correlation between key radiomic features extracted from H&E-stained histological slide images using CellProfiler and key pathomic features was further analyzed to reveal the tumor heterogeneity of TNBC from a pathological perspective.Results: The XGBoost model, which outperformed all other predictive models, was selected as the radiomics-based TLS (rTLS) predictive model. It achieved AUCs of 0.922 in the training cohort, 0.852 in the TLS validation cohort, 0.724 in the Chinese-TNBC cohort, 0.919 in the DUKE cohort, and 0.883 in the I-SPY2 cohort. The rTLS predictive model demonstrated robust predictive performance, including across various patient subgroups defined by age, menopausal status, Ki67 level, cT stage, cN stage, and MammaPrint gene status. In YNCH cohort and DUKE cohort, prognostic analysis showed that low rTLS predictive score was significantly correlated with better disease-free survival in TNBC receiving NAT, and Cox regression analysis also confirmed the rTLS predictive score was a strong independent prognostic factor. H&E-stained tumor slides of patients with high predictive score revealed the presence of TLSs, and these patients achieved pathological remission after NAT, resulting in a favorable outcome. Pathomic features further explained the pathological heterogeneity of TNBC with different responses to NAT. Conclusions: The rTLS predictive model, which accurately predicted the presence of TLSs and treatment response to NAT in TNBC, held promise for future clinical application in formulating personalized and effective treatment strategies for TNBC, ultimately improving prognosis.
Presentation numberPS3-04-03
Inter-chromosomal focal amplifications frequently co-carry multiple oncogenes in aggressive breast cancer, accumulating oncogenic elements during breast cancer progression
Ji-Yeon Kim, Samsung Medical Center, Seoul, Korea, Republic of
J. Kim1, C. HyeongJin2, H. Lee3, E. Cho3, J. Lee4, S. Kim4, Y. Park1, H. Kim2; 1Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 2School of Pharmacy, Sungkyunkwan University, Suwan, KOREA, REPUBLIC OF, 3Department of Pathology, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 4Department of Surgery, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF.
BACKGROUND: Focal copy number amplification is known to play a crucial role in oncogenesis of breast cancer (BC). Especially, the amplification of interchromosomal oncogenic translocation such as t(8;11), t(8,17) and t(11;17) is known to be the main feature of BCs. However, there is a lack of clinical evidence that such interchromosomal focal amplifications promotes tumor progression. Here, we investigated the prevalence of interchromosomal focal amplifications in primary and metastatic BCs collected at a single hospital. METHODS: 65 primary tumors were collected from BC surgical specimens obtained after curative surgery following NAC (neoadjuvant chemotherapy), and 10 metastatic BC tumors from post-adjuvant biopsies. Whole genome sequencing (mean coverage 62X, range 51~74X for tumors; mean coverage 33X, range 30~40X for patient-matching normal bloods) was generated and analyzed to profile focal amplifications and genomic characteristics. BCs were categorized into three prognostic groups: primary with dismal prognosis (TP-D; total N = 34; HER2+ N = 12), primary with good prognosis (TP-ND, total N = 31; HER2+ N = 6), and metastatic (TM; total N = 10; HER2+ N = 6). Genomic characteristics including focal amplifications were compared between these three prognostic groups. RESULTS: Interchromosomal focal amplifications co-carrying ERBB2/CDK12/C17orf37 were highly prevalent in HER2+ TM (5/6: 83.3%) and TP-D (10/12: 83.3%) patients compared to HER2+ TP-ND (1/6: 16.7%) (P = 0.040 in TM vs TP-ND; P = 0.013 in TP-D vs TP-ND; one-sided Fisher’s exact tests), indicating that those amplifications may be early events during aggressive tumor progression in HER2+ BCs. Interestingly, we also found that interchromosomal oncogenic focal amplifications cargo more oncogenes as the patients carrying those amplifications have worse prognosis (TP-ND: mean 6.5 oncogenes; TP-D: mean 15.3 oncogenes; TM: mean 21.4 oncogenes present in interchromosomal oncogenic focal amplification), which was statistically significant (P = 0.0043 in TM vs. TP-ND; P = 0.034 in TP-D vs TP-ND; wilcox ranksum test). CONCLUSION: The high prevalence of ERBB2/CDK12/C17orf37 co-amplified interchromosomal focal amplifications in HER2+ poor prognosis groups (TM and TP-D) indicate a role for these amplicons in tumor progression. Additionally, the correlation between an increasing number of oncogenes in interchromosomal focal amplifications and poor prognosis suggests that these amplifications may evolve by accumulating oncogenic elements during tumor progression, thereby promoting tumor aggressiveness.
Presentation numberPS3-04-04
Ai predicts response to neoadjuvant therapy in breast cancer across diverse cohorts
Frederick Howard, University of Chicago, Chicago, IL
J. Park1, F. Howard2, P. Wysocki3, A. Przywara3, K. Zeng1, J. Cappadona1, B. Machura1, J. Szpor4, C. Bifulco5, B. Piening5, V. Reid6, J. Witowski1, F. Esteva7, K. J. Geras1; 1AI, Ataraxis AI, New York, NY, 2Department of Medicine, University of Chicago, Chicago, IL, 3Department of Oncology, Jagiellonian University Medical College, Kraków, POLAND, 4Department of Pathomorphology, Jagiellonian University Medical College, Kraków, POLAND, 5Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, 6Hall Perrine Cancer Center, Mercy Medical Center, Cedar Rapids, IA, 7Division of Hematology/Oncology, Northwell Health Cancer Institute, Lenox Hill Hospital, New York, NY.
Background: Neoadjuvant therapy (NAT) is standard of care for many early-stage breast cancer patients. Traditional clinical characteristics such as ER and HER2 receptor status provide useful prognostic information, but lack sufficient precision when used in isolation. Basic histological features, such as tumor infiltrating lymphocytes, are also associated with NAT response. With the advent of computational pathology and machine learning, it is now possible to extract complex spatial and morphological predictive patterns from digitized slides that are not apparent to the human eye. Integrating histopathologic features with clinical variables holds promise for building robust predictive models that can guide personalized treatment planning, reduce overtreatment, and improve survival outcomes in breast cancer patients undergoing neoadjuvant therapy. Ataraxis Breast Neo (ATX-N) is a multi-modal artificial intelligence test that integrates morphological features extracted from standard H&E-stained slides with baseline clinical data (ER/PR/HER2 status, staging, age, IDC/ILC presence). Here we validate the accuracy of ATX-N in diverse patient populations. Methods: Whole slide images of H&E-stained biopsy specimens and corresponding clinical variables were collected from 563 patients who have undergone neoadjuvant therapy across UChicago (n=257), Providence Health (n=65), Jagiellonian University (n=115), and public IMPRESS dataset (n=126) cohorts. Across the cohorts, there are 186 TNBC patients, 201 HER2+ patients and 164 HR+HER2- patients. The remaining 12 patients had HER2 equivocal status or the information was missing. ATX-N scores were generated using a locked model. Performance of ATX-N was assessed for pathologic complete response (pCR) AUC. Reported AUCs are pooled using random effects models. Results: The overall pCR rate was 40.1%. ATX-N achieved a pooled AUC of 0.721 (95% CI: 0.646-0.796) across four independent cohorts, with cohort-level AUCs ranging from 0.644 to 0.804 (Table 1). Specifically, ATX-N achieved an AUC of 0.644 (CI: 0.577-0.716) on UChicago cohort, 0.734 (CI: 0.598-0.846) on Providence cohort, 0.804 (CI: 0.717-0.881) on Jagiellonian cohort, and 0.714 (CI: 0.624-0.797) on IMPRESS cohort. Notably, the confidence intervals for all cohorts did not overlap with 0.5, indicating that the model’s discriminative performance was better than random classification. In stratified analyses across molecular subtypes, pooled AUCs for TNBC, HER2+, and HR+HER2- patients are 0.675 (CI: 0.597-0.752), 0.633 (CI: 0.488-0.778), and 0.689 (CI: 0.578-0.799), respectively. When stratifying all patients based on ATX-N predictions, the bottom quarter had 15.6% pCR rate whereas the top quarter had 63.8% pCR rate. Conclusions: ATX-N demonstrates promising performance in prediction of pCR in breast cancer across diverse datasets. Larger studies are needed to confirm its efficacy and utility.
| Dataset | N total patients in the cohort | pCR rate, N (%) | AUC (95% CI) |
| UChicago | 257 | 94/257 (36.6%) | 0.644 (0.577-0.716) |
| Providence | 65 | 30/65 (46.2%) | 0.734 (0.598-0.846) |
| Jagiellonian | 115 | 37/115 (32.2%) | 0.804 (0.717-0.881) |
| IMPRESS | 126 | 65/126 (51.6%) | 0.714 (0.624-0.797) |
| Pooled – All | 563 | 226/563 (40.1%) | 0.721 (0.646-0.796) |
| Pooled – TNBC | 190 | 84/190 (44.2%) | 0.675 (0.597-0.752) |
| Pooled – HER2+ | 197 | 106/197 (53.8%) | 0.633 (0.488-0.778) |
| Pooled – HR+HER2- | 164 | 34/164 (20.7%) | 0.689 (0.578-0.799) |
Presentation numberPS3-04-05
Classifying Germline BRCA Status from Unstructured Electronic Health Record Notes: A Systematic Prompt Engineering Approach
Wenjie Zhu, MD Anderson Cancer Center, Houston, TX
W. Zhu, A. Gutierrez, L. Hsu, D. Tripathy, J. Litton, B. Arun, C. Barcenas,, A. Singareeka Raghavendra; Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX.
Background: Vast critical precision oncology data, including results for actionable findings like germline BRCA (gBRCA) mutations, remain hidden in unstructured electronic health record (EHR) notes. We sought to develop and evaluate a systematic, zero-shot large language model (LLM) framework to accurately classify gBRCA mutation status in patients using real-world EHR clinical note narratives. Methods: In this retrospective study at a large single academic cancer center, we utilized two distinct cohorts randomly sampled from a population of metastatic breast cancer patients whose genetic data were confined to unstructured notes. The development cohort (n=439) was used for model optimization, and an independent validation cohort (n=510) was used for objective performance evaluation. Our framework involved selecting an optimal base model by evaluating three LLMs with standardized prompts to ensure an unbiased comparison. The selected model that demonstrated the best performance then underwent further optimization of its input data and iterative prompt engineering to enhance performance on a granular 11-class schema. Key performance metrics included classification accuracy and F1-score (the harmonic mean of precision and recall). Results: Our final optimized framework achieved an accuracy of 86.1% and a weighted F1-score of 0.88 on the 11-class task in our development cohort. Crucially, the model demonstrated robust performance and generalization on our validation cohort with an accuracy of 82.4% and weighted F1 score of 0.81. A qualitative error analysis of the test cohort set clarified that many discrepancies were attributable to heterogenous clinical annotation criteria between the two cohorts. Significant performance gains on the development cohort set were attributed to prompt engineering, which improved accuracy by 7.5%: from 78.6% to 86.1%. Conclusion: Zero-shot LLMs, when applied within a systematic framework characterized by its multi-phase optimization of inputs and iterative engineering of prompts that enforce step-by-step reasoning, can accurately and reliably extract critical genetic data from unstructured EHR notes. This approach provides a reproducible and scalable blueprint for developing trustworthy clinical artificial intelligence methodology to accelerate research and precision oncology.
Presentation numberPS3-04-06
Benchmarking Large Language Models for Clinical Decision Support in Breast Cancer Care: A Multi-Institutional Expert Evaluation
Zunairah Shah, Roswell Park Cancer Center, Buffalo, NY
Z. Shah1, S. S. Afridi2, M. Ombada1, A. M. Roy3, A. LeVee4, S. Premji5, V. Gupta1, N. M. Lopetegui3, D. M. Quiroga6, R. L. Sacks7, S. Shaikh8, Y. Abdou9, H. Yu10, A. Madabhushi11, R. Parikh7, L. N. Chaudhary12, E. Levine1, M. Lambertini13, K. Kalinsky7, S. Kabraji1, S. Gandhi7; 1Medicine, Roswell Park Cancer Center, Buffalo, NY, 2Medicine, SUNY Upstate Medical University, Buffalo, NY, 3Medicine, OSU Comprehensive Cancer Center, Colombus, OH, 4Medicine, UCLA Medical Cente, Los Anjeles, CA, 5Medicine, Sarah Cannon Cancer Institute, Nashville, TN, 6Medicine, Roswell Park Cancer Center, Colombus, OH, 7Medicine, Emory Winship Cancer Institute, Atlanta, GA, 8Medicine, UT Health San Antonio, Mays Cancer Center , San Antonio, TX, 9Medicine, UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 10Biostatistics & Bioinformatics, Roswell Park Cancer Center, Buffalo, NY, 11Radiology & Imaging Sciences, Biomedical Informatics, Emory, Atlanta, GA, 12Medicine, Medical College of Wisconsin, Milwaukee, WI, 13Medicine, University of Genoa, Genova, ITALY.
Background: Artificial intelligence (AI) and large language models (LLMs) are increasingly explored as tools to support clinical decision-making in oncology. However, evidence validating their performance in complex breast cancer clinical scenarios (BCCS) remains limited. Given breast cancer’s diverse subtypes, evolving standards of care, and the need for nuanced, personalized treatment, we compared three LLMs for treatment decision-making to assess their capabilities and determine their readiness for integration into real-world breast oncology clinics. Methods: Ten breast cancer cases mimicking real-world scenarios were posed to three LLMs: ChatGPT-4o (GPT), DeepSeek-R1 (DS), and OpenEvidence (OE). 16 breast medical oncologists (BMOs; assistant to full professors) from 9 academic centers graded responses using a 5-point Likert scale (1 = poor, 5 = excellent) for clinical accuracy, clarity, relevance, and usability. Questions(Q) 1-9 assessed treatment decision-making cases; Q10 tested multimodal image interpretation (MMI) skills. Repeated measures ANOVA evaluated model differences, followed by Tukey’s post hoc comparisons. Results: On BCCS Q1-9 spanning all major breast cancer subtypes and treatment settings, including early-stage, metastatic, neoadjuvant, and adjuvant, OE achieved the highest mean score (3.91 ± 0.48; 2.57-4.43), significantly outperforming both GPT (3.19 ± 0.67; 2.36-3.93) and DS (2.93 ± 0.54; 1.50-3.86) in overall performance (p < 0.0001), with large effect sizes (Cohen’s d = 1.12 vs. GPT; d = 2.84 vs. DS). Repeated measures ANOVA identified significant differences among models in 5/9 (56%) Q (p < 0.05), as shown in the Table. Pairwise comparisons showed OE outperformed GPT in 4/5 (80%) and DS in 5/5 (100%) significant BCCS. GPT modestly outperformed DS in 2/5 (40%) (Cohen’s d = 0.41). GPT showed the highest inter-reviewer variability (SD = 0.67 vs. 0.48-0.54), indicating less agreement among BMOs on its responses. OE provided well-supported treatment recommendations and recurrence risk assessments with citations, though it lacked MMI. DS relied on optical character recognition in Q10 (inflammatory breast cancer image; 2.93 ± 1.33), limiting its utility in image-based BCCS. GPT, the only vision-enabled model, scored highest (4.29 ± 1.14), highlighting its strong potential for MMI integration in breast cancer diagnostic workflows. Conclusions: This is the first comparison of GPT, DS, and OE in BCCS. OE generated the most guideline-concordant treatment choices across BCCS, showing strong potential as a clinical decision support tool, though its verbosity may require streamlining. GPT showed moderate performance, while DS lagged in clinical relevance and accuracy. These findings highlight the promise of LLMs in breast oncology and the need for further refinement to ensure reliability and real-world applicability.
| Question | OpenEvidence (SD) | ChatGPT (SD) | Deep Seek (SD) | P value(ANOVA) | |||||
| 1 | 4.36 (0.63) | 3.07 (1.07) | 1.79 (0.8) | 0.0000 | |||||
| 2 | 4.21 (0.7) | 2.36 (1.08) | 1.5 (0.65) | 0.0000 | |||||
| 3 | 4.14 (1.03) | 2.57 (1.02) | 2.57 (1.22) | 0.0005 | |||||
| 4 | 2.57 (1.34) | 3.0 (1.47) | 3.64 (1.08) | 0.0929 | |||||
| 5 | 4.43 (1.09) | 2.5 (1.34) | 2.43 (1.34) | 0.0007 | |||||
| 6 | 3.14 (0.95) | 3.79 (1.05) | 3.36 (1.08) | 0.0821 | |||||
| 7 | 4.14 (0.86) | 3.93 (0.73) | 3.46 (0.88) | 0.0847 | |||||
| 8 | 4.43 (0.65) | 3.79 (0.89) | 3.86 (0.77) | 0.0218 | |||||
| 9 | 3.79 (0.8) | 3.71 (0.99) | 3.86 (0.86) | 0.7331 | |||||
| 10 | NA | 4.29 (1.14) | 2.93 (1.33) | 0.0216 |
Presentation numberPS3-04-07
Anisotropic active dense tissue: a strong image-based cancer risk predictor
Andre Khalil, University of Maine, Orono, ME
A. Khalil1, J. Juybari2, J. Hamilton3; 1CompuMAINE Lab, Chemical and Biomedical Engineering, University of Maine, Orono, ME, 2CompuMAINE Lab, Electrical and Computer Engineering, University of Maine, Orono, ME, 3CompuMAINE Lab, Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME.
Introduction: Mammography-based risk prediction models show promise, with top AI models achieving AUCs near 0.801, potentially outperforming traditional clinical models (TC8, BCSC, BRCAPRO, Gail) whose AUCs are ~0.60-0.652. However, AI models face limitations in widespread use due to issues with generalizability, calibration, and interpretability. Building on the understanding that tumor microenvironment changes promote breast tumorigenesis3,4 and tissue organization influences tumor invasion5, our patented interpretable computational biophysics approach6,7 separates mammographic dense tissue into “active” (structurally reorganizing) and “passive” subtypes8,9. We found that active dense tissue associates with early tumor onset10, that our method is agnostic to processing vs. presentation mammograms11, and our measurements synergize with volumetric breast density (VBD) and fibro-glandular volume (FGV) for a 3-yr risk AUC of 0.82 [under review]. We present a retrospective case-control analysis demonstrating how a novel morphological metric, called anisotropic mass, augments the predictive power of our active dense tissue measurement. Methods: Using the OPTIMAM database12,13, bilateral mammographic screenings were acquired on Hologic’s Selenia units ~3 years prior to diagnosis for n=197 cases (mean age = 59.4), or the last screening on record for n=3712 controls (mean age = 59.8). We calculated areas and anisotropic mass of fatty, active dense, and passive dense tissues. Anisotropy measures directional preference by quantifying departure from uniform directional gradients14 and anisotropic mass is the sum of anisotropy across an area. VBD and FGV were obtained from Volpara® (Lunit Inc.) software. An interaction generalized linear model (GLM) predicted cancer vs. control status using VBD, FGV, patient age, amounts of fatty, passive dense, and active dense tissue, our mammographic percent density, and active dense tissue’s anisotropic mass. AUC and 95% confidence intervals were calculated from bootstrapped ROC curves.The images and data used in this publication are derived from the OPTIMAM imaging database (https://medphys.royalsurrey.nhs.uk/omidb/about-omi-db/)12,13. We acknowledge the OPTIMAM project team and staff at the Royal Surrey NHS Foundation Trust who developed the OPTIMAM database, and Cancer Research UK who funded the creation and maintenance of the database. Results: Our model outputs an AUC of 0.85, with (0.84-0.86 95% CI), a very significant improvement (p-value < 10-7) over our prior model (AUC = 0.82), validating anisotropy as a complementary measure to active dense tissue. Conclusions: Our biophysics-based approach offers a significant advantage in breast cancer risk prediction. It generates interpretable measurements that match or exceed deep learning models’ performance while overcoming their drawbacks in generalizability, calibration, and interpretability. References:1. IBDW 2025, https://breastdensityworkshop.org/wp-content/uploads/2025/06/IBDW2025_program_online_6-2-2025.pdf.2. McCarthy et al. J Natl Cancer Inst, 2020. 112(5): p. 489-497.3. Bissel et al. Nat Med, 2011. 17(3): p. 320-9.4. Tanner et al. Proc Natl Acad Sci USA, 2012. 109(6): p. 1973-8.5. Nazari et al. Breast Cancer, 2018. 25(3): p. 259-267.6. Khalil & Batchelder U.S. Patent 10,467,755 B2, 2019.7. Khalil & Batchelder EU Patent 2,988,659 B1, 2022.8. Marin et al. Med Phys, 2017. 44(4): p. 1324-36.9. Gerasimova-Chechkina et al. Front. in Physiol., 2021. 12(595).10. Batchelder et al. medRxiv 2024: p. 2024.02.17.24302978.11. Khalil & Hamilton IBDW2025 poster #70 2025.12. Halling-Brown et al. Radiol Artif Intel, 2021. 3(1): p. e200103.13. OPTIMAM (OMI-DB) project: https://medphys.royalsurrey.nhs.uk/omidb/about-omi-db/.14. Hamilton et al. Front. in Oncol., 2022. 12: p. 991850.
Presentation numberPS3-04-08
Independent Validation of a Pathology-Based Multimodal Artificial Intelligence Biomarker for Predicting Risk of Distant Metastasis in Postmenopausal, Estrogen Receptor-Positive, Early-Stage Breast Cancer Patients: Analysis of the ABCSG Trial 8
Martin Filipits, Medical University of Vienna and ABCSG, Vienna, Austria
M. Filipits1, D. Hlauschek2, J. Zhang3, M. Balic4, R. Kates5, R. Greil6, F. Fitzal7, N. Toro-Bauer8, G. Rinnerthaler9, H. Pinckaers10, K. Sotlar11, Z. Bago-Horvath12, W. Hulla13, P. Regitnig14, A. Piehler15, C. E. Geyer4, H. Kreipe16, J. Griffin17, N. Harbeck18, N. Wolmark19, M. Gnant20; 1Center for Cancer Research, Medical University of Vienna and ABCSG, Vienna, AUSTRIA, 2Statistics, ABCSG, Vienna, AUSTRIA, 3Biostatistics, Artera, Inc, Mountain View, CA, 4Department of Medicine, NSABP and the University of Pittsburgh, Pittsburgh, PA, 5Statistics, WSG, Moenchengladbach, GERMANY, 6Center for Clinical Cancer and Immunology Trials, Salzburg Cancer Research Institute, Salzburg, AUSTRIA, 7Department of Surgery, Hanusch Hospital Vienna, Vienna, AUSTRIA, 8Department of Surgery, Hospital Wiener Neustadt, Wiener Neustadt, AUSTRIA, 9Division of Clinical Oncology, Department of Internal Medicine, Medical University of Graz, Graz, AUSTRIA, 10AI, Artera, Inc, Mountain View, CA, 11Institute of Pathology, Paracelsus Medical University Salzburg, Salzburg, AUSTRIA, 12Department of Pathology, Medical University of Vienna, Vienna, AUSTRIA, 13Institute of Clinical and Molecular Pathology, Hospital Wiener Neustadt, Wiener Neustadt, AUSTRIA, 14Diagnostic- and Research Institute of Pathology, Medical University of Graz, Graz, AUSTRIA, 15Biostatistics, Artera Inc., Mountain View, CA, 16Pathology, WSG, Hannover, GERMANY, 17Clinical Development, Artera, Inc, Mountain View, CA, 18LMU Breast Center, WSG and LMU Munich, Munich, GERMANY, 19Department of Surgery, NSABP and the University of Pittsburgh, Pittsburgh, PA, 20Comprehensive Cancer Center, Medical University of Vienna and ABCSG, Vienna, AUSTRIA.
Background: Genomic assays are currently used to determine prognosis and predict treatment outcome in patients with estrogen receptor-positive (ER+), HER2-negative early breast cancer (EBC). However, their use is expensive, time-consuming and not available in many clinical facilities. We previously developed a multimodal artificial intelligence (MMAI) model using clinical and histopathological data from more than 12,000 patients from six phase III trials, offering a faster, non-genomic alternative for personalized risk stratification. We present the first external independent validation of this MMAI model using the ABCSG trial 8 – a large, prospective phase III trial of a low- to moderate-risk cohort of ER+ postmenopausal breast cancer patients receiving endocrine therapy (ET) only – to assess its prognostic performance for distant metastasis (DM). Methods: ER+ HER2- patients from the prospective-randomized trial ABCSG 8 with digitized baseline H&E-stained sections, complete clinical data (age, tumor size and nodal status), and long-term follow up were included in this pivotal validation study. The locked MMAI model includes a continuous score and predefined risk groups. Prognostic performance was assessed using univariable and multivariable Cox proportional hazard models, with hazard ratios (HR) and corresponding 95% confidence intervals (CI). Results: MMAI raw scores were generated for 2,109 patients. The median follow-up time was 9.5 years. Despite the overall lower risk profile (G1 and G2 only) of the ET-only population, the MMAI classified 77% patients as low risk, 9% as intermediate risk, and 14% as high risk. The estimated 10-year DM-free rates were 94.7% (93.3% – 95.8%) for low-risk patients, 89.1% (82.5% – 93.3%) for intermediate-risk patients, and 77.3% (71.3% – 82.2%) for high-risk patients. In univariable analysis (UVA), both the continuous MMAI raw score and MMAI risk groups were significantly associated with risk of DM: MMAI raw score (HR [95% CI] = 2.22 [1.91 – 2.58], p<0.001) and MMAI risk group (intermediate vs. low risk HR [95% CI] = 2.19 [1.34 – 3.57], p=0.002; high vs. low risk HR [95% CI] = 4.45 [3.19 – 6.19], p<0.001). In multivariable analysis (MVA) adjusting for age, tumor size, nodal status, both continuous MMAI raw score (p<0.001) and MMAI risk group (p<0.001) remained independently associated with DM. The MMAI raw score remained significantly associated with the risk of DM across clinically meaningful subgroups, including lymph node status (N0 and N1), tumor grade (Grade 1 and 2), histology (invasive ductal carcinoma and invasive lobular carcinoma), and trial-defined Ki67 levels (≤5%, 6-29%, ≥30%). Additionally, the image-only component of the MMAI was independently associated with risk of DM (p<0.001 in both UVA and MVA). Conclusion: This study validates the MMAI model, originally developed for HR+ HER2- EBC, as a strong prognostic tool for DM in lower-risk postmenopausal women with ER+ HER2- EBC. This MMAI technology promises to improve personalized risk stratification and adjuvant treatment decisions in ER+ HER2- EBC as a cost-effective, non-tissue consuming, and faster alternative to genomic assays.
Presentation numberPS3-04-09
Using a Real-Time Artificial Intelligence Ultrasound System with Computer-Aided Detection and Diagnosis to Distinguish Ductal Carcinoma In Situ and Invasive Ductal Carcinoma
Joon Suk Moon, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea, Republic of
J. Moon1, J. Lee1, B. Kang1, W. Kim2, J. Kim3, J. Baek2, H. Park1, H. Kim2, G. Baek4; 1Department of Surgery, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF, 2Department of Radiology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, KOREA, REPUBLIC OF, 3School of Computer Science and Engineering, Kyungpook National University, Daegu, KOREA, REPUBLIC OF, 4Research and Development, BeamWorks Inc., Daegu, KOREA, REPUBLIC OF.
Using a Real-Time Artificial Intelligence Ultrasound System with Computer-Aided Detection and Diagnosis to Distinguish Ductal Carcinoma In Situ and Invasive Ductal Carcinoma Background: Real-time artificial intelligence-based computer-aided detection/diagnosis (AI-CAD) for breast ultrasound enables immediate assessment of suspicious lesions with probability of malignancy (POM) outputs, potentially aiding in the differentiation between ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC). Identifying ultrasonographic features with significant POM differences between DCIS and IDC may refine clinical triage. Methods: We retrospectively analyzed 34 cases (DCIS=12, IDC=22) assessed by a real-time AI solution (CadAI-B for Breast cancer) during breast ultrasound at a tertiary center. POM outputs and BI-RADS categorization were compared across ultrasonographic features including shape, margin, echo pattern, and orientation. Features demonstrating significant POM differences between DCIS and IDC were identified, and comparative analyses were performed to evaluate CadAI-B’s differential diagnostic utility. Results: Overall, IDC demonstrated a higher mean POM (0.365 ± 0.306) compared to DCIS (0.126 ± 0.196, p<0.001). Ultrasonographic features with significant POM differences included irregular shape (IDC: 0.344 vs DCIS: 0.135, p=0.002), indistinct margins (IDC: 0.318 vs DCIS: 0.012, p=0.014), microlobulated margins (IDC: 0.419 vs DCIS: 0.132, p=0.031), hypoechoic pattern (IDC: 0.345 vs DCIS: 0.135, p=0.018), and parallel orientation (IDC: 0.362 vs DCIS: 0.044, p=0.003). These features were associated with larger POM differentials between IDC and DCIS, suggesting enhanced discrimination capacity. Under CadAI-B, IDC cases were more frequently categorized into higher BI-RADS categories, with IDC cases showing a substantial shift towards BI-RADS 4B, 4C, and 5 with high POM outputs. Notably, DCIS cases retained lower POM even when categorized into BI-RADS 4A and 4B. Conclusions: CadAI-B may help distinguish IDC from DCIS during real-time breast ultrasound examinations by reflecting differences in probability of malignancy. This may assist clinical decision-making during scanning by providing consistent, objective data when evaluating suspicious breast lesions. Further studies are needed to confirm its diagnostic value in broader clinical settings. Keywords: AI-CAD, Breast Ultrasound, DCIS, IDC, Probability of Malignancy
Presentation numberPS3-04-10
Development and Validation of a Living Decision Support Tool (Living-DST) in Oncology Using Agentic AI-Augmented Systematic Literature Review (SLR)
Rozee Liu, Eviviz, Vancouver, BC, Canada
L. Schwartzberg1, H. S. Rugo2, A. Forsythe3, D. Flora4, S. Glück5, S. Grieve6, R. Campden6, R. Liu6, J. M. Rege3, P. A. Kaufman7; 1Medical Oncology and Hematology, Renown Health-William N. Pennington Cancer Institute, Reno, NV, 2Duarte Cancer Center, City of Hope, Duarte, CA, 3Executive, Oncoscope-AI, Miami, FL, 4Yung Family Cancer Center, St. Elizabeth Healthcare, Edgewood, KY, 5Consultant, Oncology Immunotherapy Biotech and AI, Miami Beach, FL, 6Data management, Eviviz, Vancouver, BC, CANADA, 7Oncologist, The University of Vermont Medical Center, Burlington, VT.
Background: Oncologists face growing difficulty in staying current with rapidly evolving data from congresses, journals, guidelines, and regulatory updates. Creating a well-organized, annotated clinical trial (CT) data library in an easily digestible format is time- and labor-intensive. To address this, we developed and validated a living-DST for breast cancer (BC), powered by an agentic AI system that supports daily human conducted / AI augmented SLR integrated with guidelines, regulatory approvals and ongoing CT results. Methods: An agentic AI system using GPT-4.1 & o3 (OpenAI), and Claude Sonnet-4 (Anthropic) was designed to emulate expert-led Cochrane-compliant SLR processes. The system follows an annotation manual, decomposes tasks into subtasks, and self-debugs and validates. The annotation manual was constructed by human scientists manually annotating 29,236 clinical trial abstracts across BC, lung, and prostate cancer from 2019-current. Each study was annotated with 4 review variables (population, intervention/comparator, reported outcomes, study design) and 32 extraction variables: i.e. TNM staging, histology, biomarkers, risk factors, treatment line, intervention, study design, size, follow-up, outcomes (overall and progression/disease-free survival, response, QoL), subgroup data, and toxicity. Structured data were integrated with national guideline-based treatment pathways, including new evidence beyond guidelines, forming a real-time, evidence-linked DST. Accuracy was assessed vs. 1,997 human annotations as validation set. DST evidence precision was evaluated against the AI chatbots: ChatGPT, Perplexity, and Consensus using 6 criteria for 8 cases on treatment options in BC with different treatment path, subpopulation, and biomarker variables. Accuracy for each criterion was defined as the percentage of questions evaluated as positive over all eight questions. Results: Accuracy across review variables ranged from 95.1-97.2%. For extraction variables, accuracy exceeded 90% for all variables (91.5%-99.1%) with 50% of variables above 95%. Agentic AI annotated 1,997 publications in 7.25 hours vs. an estimated 727.4 hours required by human experts—yielding 99% of time savings. When compared to other AI tools, our DST outperformed by providing more complete and accurate evidence, retrieving recently published, potentially practice-changing pivotal trials (23% of all evidence), reporting all available clinical evidence, and linking to original citations and FDA labels where available (Table). Conclusion: By integrating human-conducted, AI-augmented daily SLR integrated with guidelines and regulatory approvals, our Living-DST delivers real-time, clinically actionable decision support. This system offers oncologists precise, patient-specific insights and may improve cancer treatment outcomes.
| Living-DST | ChatGPT | Perplexity | Consensus | |
| Listed all available treatments | 100% | 38% | 25% | 50% |
| Cited guideline recommendations for each treatment if available | 100% | 0% | 0% | 0% |
| Included treatments with positive results not yet included in guidelines if available | 100% | 38% | 25% | 38% |
| Presented efficacy outcomes (PFS and OS) | 100% | 0% | 25% (partial data only) | 38% (partial data only) |
| Cited the original reference | 100% | 0% | 0% (no primary references) | 0% (no primary references) |
| Cited FDA approval where available | 100% | 0% | 0% | 0% |
Presentation numberPS3-04-11
Explainable machine learning reveals hidden hereditary risk of breast cancer beyond TP53: insights from Brazilian families with high prevalence of the p.R337H mutation
Jose Claudio Casali da Rocha, A.C.Camargo Cancer Center, São Paulo, Brazil
J. Casali da Rocha1, A. C. Ricciardi2, G. B. Pinheiro1, D. S. Pegos1, R. A. Romero3; 1Oncogenetics, A.C.Camargo Cancer Center, São Paulo, BRAZIL, 2Department of Computer Science, Institute of Mathematics and Computer Sciences (ICMC), University of São Paulo, São Carlos – SP, BRAZIL, 3Department of Computer Science, Institute of Mathematics and Computer Sciences (ICMC), University of São Paulo, São Carlos, BRAZIL.
Background: Clinical criteria, such as Chompret 2015, guide genetic testing for Li-Fraumeni Syndrome (LFS) and TP53 mutations. However, these guidelines may overlook hereditary breast cancer risk in genetically diverse populations. In Brazil, the founder mutation TP53 p.R337H is highly prevalent, presenting with atypical tumor patterns and age of onset, often leading to underdiagnosis by conventional criteria. This study aims to assess whether machine learning (ML) and explainable artificial intelligence (XAI) can enhance the prediction of hereditary breast cancer subtypes, particularly TP53-related, in a real-world Brazilian cohort.Methods: We curated a dataset of 576 probands (515 with complete cancer history data) subjected to genetic testing and with structured clinical and family information.Three multiclass machine learning models were trained to predict three groups: TP53-negative, TP53 R337H, and other TP53 mutations. Performance was evaluated using ROC-AUC, and SHAP values were employed to interpret variable importance. Among the tested algorithms, CatBoost achieved the highest ROC-AUC values, although Random Forest was selected for interpretability. Kaplan-Meier analysis assessed age at diagnosis across groups.Results: Among 515 probands, 70.1% had a personal cancer history. Among those with cancer, 32.7% carried pathogenic mutations, with TP53 variants being the most frequent: 7.8% for R337H and 6.1% for other TP53 mutations, totaling 13.8%. Other recurrent mutations included BRCA2 (3.0%), BRCA1 (2.5%), CHEK2 (1.4%), ATM (1.1%), and NF1 (0.6%). The remaining 67.3% had no identifiable pathogenic variant. The model achieved robust discrimination (AUC: 0.73 for TP53 R337H, 0.84 for other TP53, 0.85 for TP53-negative), outperforming the Chompret clinical criteria (AUC = 0.67). Unlike Chompret, the ML model distinguished between R337H and other TP53 mutations, offering a more tailored and data-driven approach to hereditary cancer risk stratification in genetically diverse populations. SHAP analysis identified key predictors for TP53 p.R337H, including early-onset maternal cancer, proband’s breast cancer, and early-onset brain tumors. Notably, TP53 p.R337H cases showed a significantly earlier median age at diagnosis (43 years) compared to TP53-negative cases, yet later than classical TP53 mutations. The model also accurately revealed that several TP53-negative individuals carried pathogenic variants in BRCA1, BRCA2, and ATM, with early-onset breast cancer mirroring TP53 carriers, highlighting the limitations of TP53-centric screening.Conclusion: Our findings reveal that Chompret criteria may miss substantial hereditary breast cancer risk associated with LFS, especially in populations with founder mutations like TP53 p.R337H. Explainable ML models integrating comprehensive clinical and familial data provide accurate and interpretable risk stratification, demonstrating superior performance in identifying diverse hereditary cancer patterns. These insights support the use of multigene panel testing and population-tailored triage strategies for improved genetic counseling and surveillance in hereditary breast cancer care, particularly in regions with high genetic heterogeneity.
Presentation numberPS3-04-12
Robust prediction of patients’ response to neoadjuvant therapy across breast cancer subtypes using transcriptomics and histopathology
Thomas Cantore, National Institute of Health (NIH), Bethesda, MD
T. Cantore1, D. Hoang1, L. R. Pal1, A. Stemmer1, S. Dhruba1, T. Chang1, S. Sammut2, S. Lipkowitz3, R. S. Padma1, C. Caldas4, N. Ulhas Nair1, E. Ruppin1; 1Cancer Data Science Laboratory (CDSL), Center for Cancer Research (CCR), National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, 2Division of Breast Cancer Research, Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK., London, UNITED KINGDOM, 3Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institute of Health (NIH), Bethesda, MD, 4Department of Clinical Biochemistry and Institute of Metabolic Science, School of Clinical Medicine, University of Cambridge, Cambridge, UK, Cambridge, UNITED KINGDOM.
BACKGROUND:Neoadjuvant therapy is central to breast cancer treatment, yet response rates vary widely, and robust biomarkers to predict pathological complete response (pCR) are lacking. While expression-based signatures such as Oncotype DX and MammaPrint routinely guide adjuvant treatment decisions, no tools are approved for the neoadjuvant setting. Although some predictive markers have been explored for HR-positive and HER2-positive disease, triple-negative breast cancer (TNBC) lacks reliable predictors despite its aggressive nature and unmet clinical need. Recent single-cell transcriptomic studies reveal that multiple molecular subtypes can coexist within individual tumors, challenging the conventional one-subtype-per-patient paradigm. We hypothesized that modeling tumors as compositions of intra-tumoral subtypes would improve neoadjuvant therapy response prediction. METHODS: We developed BRIDGE (Breast Intratumoral Deconvolution of Gene Expression), the first computational framework that deconvolves bulk breast cancer transcriptomes to quantify the relative abundance of malignant subtype populations within each tumor and predict neo-adjuvant therapy response. BRIDGE involves three key steps. (I) Subtype Deconvolution: We constructed a reference matrix of ‘pure’ molecular subtype profiles from an integrated single-cell RNA-seq compendium and applied support vector regression to infer subtype abundances per tumor. (II) Response prediction from bulk expression: We trained logistic regression models to predict pCR taking as input subtype compositions using 33 datasets from 17 independent neoadjuvant cohorts. These included 11 HR-positive, 10 TNBC chemotherapy, and 12 anti-HER2 datasets. For each treatment, models were trained on the three largest datasets and validated on the remaining 24. (III) Response prediction from histopathology: we integrated BRIDGE with Path2Omics, a deep learning framework that infers gene expression from histopathology slides, enabling transcriptome-free prediction in six datasets with available image data. RESULTS:BRIDGE accurately estimated subtype composition, achieving a mean Spearman correlation of 0.80 on simulated pseudo-bulk data. For HR-positive chemotherapy response prediction, BRIDGE achieved a mean ROC-AUC of 0.78 and an odds ratio (OR) of 7.8 – outperforming existing biomarkers. For anti-HER2 therapy, BRIDGE reached a mean ROC-AUC of 0.77 and OR of 7.75, substantially outperforming the use of HER2-enriched subtype classification alone (OR = 3.1). For TNBC chemotherapy-treated cohorts, BRIDGE achieved a mean ROC-AUC of 0.71 and an OR of 4.7, addressing a major clinical gap due to the lack of established predictors. When applied to AI-inferred transcriptomes from H&E slides, BRIDGE maintained high accuracy (AUC > 0.85 for chemotherapy and > 0.70 for anti-HER2), with response scores highly concordant with those derived from true transcriptomic data (correlation > 0.7). CONCLUSION: BRIDGE is the first robust and interpretable framework for deconvolving malignant subtypes to predict neoadjuvant therapy response across diverse breast cancer subtypes. By modeling intra-tumoral heterogeneity and leveraging histopathology images, BRIDGE bridges the gap between expression-based profiling and routine pathology. Its low-cost, image-based approach supports personalized, subtype-informed treatment strategies and helps democratize precision oncology in the neoadjuvant setting.
Presentation numberPS3-04-13
Identifying recurrent gene amplifications in breast cancer for antibody drug conjugate development
Hao-Kuen Lin, Danbury Hospital, Danbury, CT
H. Lin1, Y. Wang2, J. Dai2, L. Pusztai2; 1Internal Medicine, Danbury Hospital, Danbury, CT, 2Yale Cancer Center, Yale School of Medicine, New Haven, CT.
Identifying recurrent gene amplifications in breast cancer for antibody-drug-conjugate development Hao-Kuen Lin 1, Yueyue Wang 2, Jiawei Dai 2, Lajos Pusztai 2* 1Danbury Hospital, Danbury, CT 2Yale School of Medicine, Yale Cancer Center, New Haven, CT Background: We hypothesized genes with frequent copy number amplifications in breast cancer resulting in overexpression of membrane proteins could serve as potential targets for novel antibody drug conjugates (ADC). Methods: DNA copy number aberrations (CNA) and mRNA expression data was retrieved from cBioPortal (https://www.cbioportal.org/) for 3,970 patients with breast cancer. We defined gene amplification as CNA value >2. Chi-square or Fisher’s Exact test was used to test for significant difference in amplification frequency between 3 clinical subtypes ER+ (n=2474), HER2+ (n=712), and TNBC (n=632). Wilcoxon signed-rank test was used to test if gene mRNA expression is higher in cancers with amplification compared to wild type. We shortlisted amplified genes as candidates if amplification has led to overexpression and the gene encoded a membrane protein based on information in the Protein Data Bank (https://www.rcsb.org/) and Human Protein Atlas (https://www.proteinatlas.org/). We retrieved normal tissue mRNA expression data from GTEx (https://www.gtexportal.org/home/) covering 21 different tissue types and compared candidate ADC target expression in cancer versus normal tissues using DESeq2. We prioritized ADC candidates by the following criteria (i) CNA frequency > 15% across all breast cancers or within a subtype (FDR<.0001) (ii) higher expression in amplified cancers (FDR<0.001), (iii) mean Log2 Fold expression > 1.5 in cancer compared to each normal tissue one by ones, and (iv) known membrane protein that is primarily located in the cell membrane and ability for internalization. Results: 963 unique genes were recurrently amplified and showed higher gene expression in amplified cancers across all breast cancers (722 genes) and/or significantly amplified only in ER+ (206 4 genes), HER2+ (216 200 genes), and TNBC (114 37 genes), and. how many were amplified in all subtypes?. Of the amplified and overexpressed genes, ⋯N=?…154 genes were membrane proteins. were membrane proteins. Among these membrane membrane proteins, 9 genes had higher expression in breast cancer than in normal tissues and primary located on cell membrane with ability for internalization. They (including HER2) are highlighted in Table 1 met all 4 criteria as potential novel ADC targets⋯N=?… had higher expression in breast cancer than in normal tissues. Nine genes (including HER2) highlighted in bold in Table 1 met all 4 criteria as potential novel ADC targets. Conclusion: We have identified several novel potential targets for ADC development in breast cancer.
| Gene | ER+ frequency (significant difference compared to non-ER+ subtype) | HER2+ frequency (significant difference compared to non-HER2+ subtype) | TNBC frequency (significant difference compared to non-TNBC subtype) | Amplification Frequency (all breast) | Mean log2 expressionFold Change (breast cancer expression compared to normal tissue) | ||||||
| MUC1 | 0.2 | 0.17 | 0.21 | 0.2 | 4.09 | ||||||
| ERBB2 | 0.19 | 0.60 (sig) | 0.06 | 0.16 | 2.95 | ||||||
| ANO1 | 0.19 | 0.23 | 0.05 | 0.18 | 2.91 | ||||||
| LY6K | 0.19 | 0.26 | 0.27 | 0.21 | 2.67 | ||||||
| RNF43 | 0.08 | 0.17 (sig) | 0.02 | 0.09 | 2.3 | ||||||
| PSCA | 0.19 | 0.26 | 0.27 | 0.21 | 2.22 | ||||||
| F11R | 0.2 | 0.16 | 0.2 | 0.2 | 1.77 | ||||||
| VANGL2 | 0.19 | 0.15 | 0.2 | 0.19 | 1.72 | ||||||
| KCNK1 | 0.2 | 0.16 | 0.17 | 0.18 | 1.66 |
Presentation numberPS3-04-14
Artificial intelligence (AI) as a decision support tool for practicing oncologists: breast cancer cases
Mohammad Jahanzeb, Florida Atlantic University Schmidt College of Medicine, Boca Raton, FL
M. Jahanzeb1, K. Haines2, T. Buchholz3, R. Butler4, W. J. Gradishar5, S. A. Hurvitz6, T. A. King7, R. L. Mahtani8, T. Mamounas9, H. McArthur10, M. Morrow11, A. P. O’Dea12, J. O’Shaughnessy13, M. D. Pegram14, H. S. Rugo15, C. Shah16, F. Vicini17, N. Wolmark18; 1Medical Oncology, Florida Atlantic University Schmidt College of Medicine, Boca Raton, FL, 2Medical Oncololgy, OncAdvisor, Delray Beach, FL, 3Radiation Oncology, Scripps MD Anderson Cancer Center, San Diego, CA, 4Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, 5Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, 6Medical Oncology, Fred Hutch Cancer Center, Seattle, WA, 7Surgical Oncology, Emory Winship Cancer Institute, Atlanta, GA, 8Medical Oncology, Miami Cancer Institute, Miami, FL, 9Surgical Oncology, AdventHealth Cancer Institute, Orlando, FL, 10Medical Oncology, UT Southwestern Medical Center, Dallas, TX, 11Surgical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 12Medical Oncology, University of Kansas Medical Center, Westwood, KS, 13Medical Oncology, Baylor University Texas Oncology, Dallas, TX, 14Medical Oncology, Stanford University School of Medicine, Standford, CA, 15Medical Oncology, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 16Radiation Oncology, AHN Cancer Institute, Pittsburgh, PA, 17Radiation Oncology, MHP Radiation Oncology Institute, Farmington Hills, MI, 18Surgical Oncology, University of Pittsburgh Hillman Cancer Center, Pittsburgh, PA.
Background: Second opinions and multidisciplinary review (MDR) can inform and impact the management of cancer patients, yet these resources are not universally accessible. We previously presented over 400 anonymized complex cancer scenarios (cases) via video conference to MDR panel experts—including specialists in medical oncology, surgical oncology, radiation oncology, radiology, and a moderator—across 5 major tumor types and tracked their treatment recommendations. In this study, we focus on breast cancer cases to compare how recommendations from 3 leading AI models compare to those of MDR panel experts. Methods: We selected 50 complex breast cancer scenarios previously adjudicated by MDR panels from a larger database of over 400 anonymized cancer cases collected between 2020 and 2021. These cases were analyzed by 3 different foundational AI models (OpenAI’s ChatGPT 4.5, Anthropic’s Claude Opus 4 and Google’s Gemini Ultra) using PrecisCa’s proprietary method for prompts. The AI recommendations from each system were scored on a scale of 1-5 (5 being the highest) for completeness, reasoning, clarity, menu of options, recency, and relevance as compared to the MDR panel recommendations. The maximum possible competence score was 30 points per scenario and an aggregate score of 1500. Final AI recommendations were also compared with current National Comprehensive Cancer Network (NCCN) guidelines for serious omissions. Comparison in reverse (additional AI options that the experts missed) was not performed due to interval changes in treatment recommendations over the past 4 years. Results: Patient characteristics and aggregate concordance and competence scores are listed in Table 1 below. While there was some variation in the concordance and competence rates of the 3 AI models, there was excellent concordance with the expert opinion for all of them. Discordant cases were reviewed and primarily involved minor differences that would not have altered the patients’ management significantly. Relatively speaking within the 6 categories listed above, AI systems excelled in clarity and reasoning and less so in relevance and menu of options. Conclusions: This study demonstrates a high degree of concordance between 3 leading AI models and expert MDR panels for common yet complex breast cancer clinical scenarios. These findings suggest that AI tools have matured sufficiently to serve as valuable decision support aids in oncology practice, particularly where expert review may be limited or unavailable. Careful human oversight remains essential to ensure safe and personalized cancer care.
| Characteristic | Breast Cancer (n=50) | ||||
| Median Age (Range) | 55 (27-83) | ||||
| Stage | |||||
| I | 7 (14%) | ||||
| II | 11 (22%) | ||||
| III | 8 (16%) | ||||
| IV | 24 (48%) | ||||
| Histology | |||||
| Ductal Carcinoma | 46 (92%) | ||||
| Lobular Carcinoma | 4 (8%) | ||||
| Biomarker Status | |||||
| HR+/HER2- | 25 (50%) | ||||
| HR-/HER2- | 11 (22%) | ||||
| HR+/HER2+ | 11 (22%) | ||||
| HR-/HER2+ | 3 (6%) | ||||
| Aggregate / Median Competence Score (Range) | |||||
| OpenAI 4.5 | 1311 / 26.5 (23-30) | ||||
| Claude Opus 4 | 1408 / 27 (24-30) | ||||
| Gemini Ultra | 1423 / 28 (26-30) |
Presentation numberPS3-04-15
Merlin: dissecting therapeutically relevant elements of the oncogenic microenvironment using multi-modal deep learning
Albert E Kim, Mass General Hospital, Boston, MA
T. Goncalves1, D. Pulido-Arias1, C. Perrino2, M. Cleveland1, E. Gerstner3, J. Kalpathy-Cramer4, K. Flaherty3, V. Zanfagnin5, A. Iafrate5, S. Sirintrapun5, A. Parwani6, G. Tozbikian6, J. Cardoso7, D. Sgroi5, C. Bridge1, A. E. Kim3; 1Athinoula A. Martinos Center for Biomedical Imaging, Mass General Hospital, Boston, MA, 2Department of Pathology, Lahey Hospital & Medical Center, Burlington, MA, 3Cancer Center, Mass General Hospital, Boston, MA, 4Division of Artificial Medical Intelligence, University of Colorado, Aurora, CO, 5Department of Pathology, Mass General Hospital, Boston, MA, 6James Cancer Center, Ohio State University, Columbus, OH, 7Faculty of Engineering, University of Porto, Porto, PORTUGAL.
Background: At present, clinical decision making and drug discovery are based on an incomplete picture of tumor biology. Deep learning (DL) has demonstrated the ability to infer oncogenic molecular signatures from data acquired during standard oncologic care (e.g., histopathology [H&E]). To advance the paradigm of DL from a tool that merely predicts outcomes into an engine that uncovers new therapeutic insights, we present MERLIN: Multimodal Evaluation of therapeutic Resistance through deep Learning INtegrating multimodal data. Methods: To develop MERLIN, we curated a dataset of 3111 H&E whole slide images (WSIs) and patient-matched clinico-genomic data from The Cancer Genome Atlas-Breast Cancer dataset and cBioPortal. This dataset was split into training (70%), validation (15%), and testing (15%) sets. Using attention-based multiple instance learning and several multimodal fusion techniques, we trained uni-modal (input: H&E) and multimodal (input: H&E and clinical data) models that quantify a patient’s tumor microenvironmental (TME) phenotypes. The ground truth for training these models was defined by each patient’s bulk RNA-sequencing data and Gene Set Enrichment Analysis protocols. Consistent with our prior work, image patching, feature extraction, and data augmentation strategies were applied during model training. Results: MERLIN achieved robust performance in deriving TME insights using only a patient’s H&E WSI and clinical metadata (Table 1). Using attention maps, MERLIN correctly identified morphology on H&E that governs tumor and immune function. A board-certified pathologist (C.M.P.) verified that high-attention areas, as determined by MERLIN, consistently identified spatial patterns of cell populations (e.g. cluster of lymphocytes) that matched with the phenotype of interest (e.g. T-cell cytotoxicity). Additional validation with tissue-matched immunofluorescence is ongoing. To demonstrate MERLIN’s ability to guide clinical decision making, we curated a multi-institutional dataset of 232 H&E WSIs of core biopsies from stage 2 & 3 triple negative breast cancer patients. Consistent with a known link between tumor-infiltrating lymphocyte activity and checkpoint inhibitor response, we identified correlations between the patient’s likelihood of achieving a pathologic complete response to neoadjuvant Keynote-522 and increased expression of T-cell cytotoxicity (p=0.017) as determined by MERLIN’s analysis of a patient’s H&E WSI. Conclusion: MERLIN has illustrated promise in obtaining TME-derived insights and identifying therapeutically relevant spatial patterns of cell populations from H&E and clinical metadata. We are now applying MERLIN to large datasets of clinically acquired data to identify new insights for cancer therapeutics without known a priori mechanisms of resistance.
| Immune phenotypes | |||
| H&E-only model (PCC – Pearson’s correlation coefficient with 95% CI) | Cross-attention fusion of H&E with clinical metadata (PCC) | Late fusion of H&E with clinical metadata (PCC) | |
| B-cell proliferation | 0.8117 (0.7432, 0.8572) | 0.5953 (0.4649, 0.7060) | 0.8441 (0.7731, 0.8910) |
| T-cell mediated cytotoxicity | 0.7602 (0.6913, 0.8135) | 0.5606 (0.4508, 0.6545) | 0.8137 (0.7321, 0.8674) |
| FoxP3-mediated immunosuppression | 0.8208 (0.7615, 0.8671) | 0.6233 (0.5191, 0.7094) | 0.8735 (0.8217, 0.9105) |
| Antigen processing and presentation | 0.7653 (0.7115, 0.8185) | 0.5531 (0.4552, 0.6402) | 0.7904 (0.7061, 0.8531) |
| Tumor phenotypes | |||
| Angiogenesis | 0.6444 (0.5229, 0.7416) | 0.5069 (0.3907, 0.6201) | 0.5422 (0.3985, 0.6607) |
| Epithelial-mesenchymal transition | 0.7849 (0.7290, 0.8325) | 0.7166 (0.6409, 0.7791) | 0.7879 (0.7089, 0.8497) |
| Cell cycling | 0.7799 (0.7325, 0.8288) | 0.7896 (0.7371, 0.8334) | 0.8131 (0.7551, 0.8604) |
| Metabolic phenotypes | |||
| Fatty acid metabolism | 0.7380 (0.6468, 0.8003) | 0.5141 (0.3872, 0.6229) | 0.7538 (0.6397, 0.8377) |
| Glycolysis | 0.7519 (0.6836, 0.8083) | 0.6897 (0.6083, 0.7580) | 0.7681 (0.6854, 0.8348) |
| Oxidative phosphorylation | 0.7316 (0.6477, 0.7976) | 0.6065 (0.3686, 0.7300) | 0.7401 (0.5973, 0.8278) |
Presentation numberPS3-04-16
Decoding resistance to trastuzumab deruxtecan in metastatic breast cancer: S100P signaling and immune-niche barriers
Glori Das, Houston Methodist Research Institute, Houston, TX
G. Das1, S. Wang1, C. Xue1, M. Vasquez1, W. Yang2, L. Xiaoxian2, H. Zhao1, W. Dong1, S. T. Wong1; 1Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX, 2Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA.
Decoding resistance to trastuzumab deruxtecan in metastatic breast cancer: S100P signaling and immune-niche barriersGlori Das1,2, Shiruo Wang1, Catherine Xue1, Matthew Vasquez1,3, Wei Yang4, Xiaoxian Li4, Hong Zhao1,3,5,*, Wenjuan Dong1,*, Stephen T.C. Wong1,2,3,5,*1 Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Weill Cornell Medical College, Houston, TX 770302 Department of Biomedical Engineering, Texas A&M University College of Medicine, College Station, TX, 778433 Advanced Cellular and Tissue Microscopy Shared Resource, Houston Methodist Research Institute and Houston Methodist Neal Cancer Center, Houston, TX 770304 Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 303225 Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030*Corresponding authors Background: Trastuzumab deruxtecan (T-DXd) has shown strong clinical efficacy in HER2-high and HER2-low metastatic breast cancer (MBC). However, many patients develop intrinsic or acquired resistance, particularly within sanctuary sites like the brain and bone. These niche-specific mechanisms remain poorly understood, limiting durable treatment responses and the design of effective combination therapies.Methods: We analyzed 109 MBC patients treated with T-DXd, stratifying response using clinical and radiographic criteria. Pre-treatment biopsies from 31 patients underwent spatial immune profiling using GeoMX Digital Spatial Profiler. Eight brain and bone metastases were profiled using a 570-plex antibody panel with spatial transcriptomics. Single-cell spatial transcriptomics (CosMX) was performed on 6 brain metastases. Functional validation included CRISPR-Cas9–mediated knockout and lentiviral overexpression of S100P, RAGE inhibition with azeliragon, and in vivo xenograft models.Results: T-DXd response was not associated with HER2 (p=0.128), ER (p=0.832), PR (p=0.617), or Ki-67 (p=0.212). Prior T-DM1 exposure predicted improved response (OR=5.27, p=0.048). Spatial profiling identified enhanced immune activation (CD3, CD8α, FABP4) in responders, especially in bone lesions. In contrast, brain metastases displayed conserved immunosuppressive profiles regardless of response. S100P was the most upregulated protein in resistant tumors (6.8-fold, p=0.0018), with marked spatial heterogeneity. S100P knockout sensitized HER2+ cells to T-DXd, while overexpression induced resistance. RAGE inhibition enhanced T-DXd cytotoxicity (~49-fold reduction in EC50). Single-cell analysis of resistant brain metastases revealed astrocytic upregulation of CAT and FN1, and macrophage expression of DR5 and CD163, consistent with a neuroprotective, immune-silent niche impeding ADC activity.Conclusions: Resistance to T-DXd is driven by tumor-intrinsic S100P-RAGE signaling and reinforced by niche-specific microenvironments—particularly in the brain—characterized by immunosuppressive astrocyte and macrophage programs. These findings nominate S100P, RAGE, and brain niche-derived stromal pathways as actionable targets to overcome resistance and extend the efficacy of T-DXd in MBC.
Presentation numberPS3-04-17
Deep Learning Classification of Inflammatory Breast Cancer Using Multiparametric MRI: A Multi-Sequence Analysis
Saleh Ramezani, University of Texas MD Anderson Cancer Center, Houston, TX
S. Ramezani, H. T. Le-Petross, M. Naser, R. V. Young, T. K. Morris, M. Kai, M. C. Stauder, B. Lim, L. Anthony, MDACC-IBC, C. D. Fuller, W. Woodward, C. R. Goodman; University of Texas MD Anderson Cancer Center, Houston, TX
Background: The diagnosis of inflammatory breast cancer (IBC) depends upon subjective clinical criteria, often leading to misdiagnosis and delayed treatment. While multiparametric MRI (mpMRI) is sensitive to characteristic IBC features such as global skin thickening, chest wall edema, and non-mass enhancement, its interpretation remains subjective and prone to inter-observer variability. An objective, automated clinical decision support system has the potential to improve patient outcomes enabling earlier and more accurate identification of IBC. Methods: DenseNet121-based deep learning models were developed for the automated classification of IBC using mpMRI sequences from a prospective institutional patient registry. Four separate datasets were analyzed based on sequence availability: T1-weighted without contrast (T1; N=791), diffusion weighted imaging (DWI; N=611), dynamic contrast-enhanced (DCE) imaging (N=484), and T2-weighted with contrast (T2c; N=267). Models for each sequence were evaluated using both midline-cropped single-breast (ipsilateral) images and bilateral full-breast images. For each sequence, stage-aware stratified splitting allocated 80% of patients for training/validation and 20% for testing. Image preprocessing included resizing to 144³ voxels, intensity normalization, and augmentation with random rotations and flips. Binary classification models were trained using 5-fold cross-validation and performance was evaluated using area under the receiver operating characteristic curve (AUC-ROC) on ensemble predictions. Results: Deep learning classification performance demonstrated strong overall discriminative ability across all sequences. DWI achieved the highest ensemble performance (AUC=0.96, mean fold AUC=0.92±0.02), followed by T2c (ensemble AUC=0.96, mean fold AUC=0.89±0.04), T1 (ensemble AUC=0.89, mean fold AUC=0.84±0.04), and DCE (ensemble AUC=0.80, mean fold AUC=0.84±0.02). Stage-specific analysis revealed high accuracy for early-stage disease (cT1-cT3: 92-97%) and classic IBC (cT4d: 94-95% accuracy), but reduced performance in distinguishing cT4b from cT4d cases (40-42% accuracy). Cross-validation demonstrated robust model stability with minimal variance across folds. The ensemble consistently outperformed individual fold averages, with DWI and T2c achieving clinically significant discrimination for overall IBC detection. A bilateral imaging approach did not significantly improve performance compared to analysis of the involved breast alone. Conclusion: Automated deep learning classification of IBC using mpMRI achieved excellent diagnostic performance, with DWI demonstrating superior discriminative ability (AUC=0.96). While the models consistently distinguished classic IBC (cT4d) from early-stage disease, classification of IBC from locally advanced non-IBC with extensive skin involvement and/or edema (cT4b) remains a challenge, reflecting known clinical diagnostic difficulties. Future work will incorporate combined-sequence analysis, probabilistic deep learning for uncertainty quantification, and an expanded cT4b cohort to improve distinction between IBC and non-inflammatory locally advanced breast cancers. These results demonstrate the potential of a deep learning-based clinical decision support system to improve diagnostic accuracy and help guide personalized treatment decisions for patients with IBC.
Presentation numberPS3-04-18
Ai-enhanced imaging of quantum dot-encoded fluorescent magnetic nanoparticles enables sensitive and multiplexed detection of circulating tumor cells in breast cancer
Aihua Fu, NVIGEN, Inc, Santa Clara, CA
A. Fu, K. Wang, H. Jin, W. Gu; R&D, NVIGEN, Inc, Santa Clara, CA.
Background: Circulating tumor cells (CTCs) are valuable biomarkers for early detection, prognosis, and therapeutic monitoring in breast cancer. However, their rarity and the need for multiplexed marker detection present significant technical challenges. We introduce a novel platform combining quantum dot-based fluorescent magnetic nanoparticles (MyQuVigen) with AI-enhanced imaging to achieve sensitive, multiplexed CTC isolation and detection using standard fluorescence microscopy. Methods: NVIGEN’s MyQuVigen fluorescent magnetic nanoparticles are conjugated with antibodies targeting epithelial tumor markers (e.g., EpCAM, HER2, PD-L1, VEGF, EGFR), enabling efficient magnetic capture of CTCs from whole blood. The nanoparticles incorporate semiconductor quantum dots as fluorescent reporters, offering key advantages: broad excitation with narrow, tunable emissions for true multiplexing, high photostability, low autofluorescence background, and enhanced signal-to-noise ratio. An AI-powered image correction algorithm was developed to process images captured from standard color CCD cameras, improving contrast and signal clarity without requiring specialized multispectral equipment. Results: Using spiked breast cancer cell lines, the platform demonstrated >90% CTC recovery efficiency and successful multiplexed visualization of 3-5 protein markers per cell. AI image correction improved signal-to-noise ratio by 2-10×, enabling clear detection of low-abundance markers with simplified imaging systems. The workflow is compatible with widely available fluorescence microscopes, making it suitable for both research and translational applications. Conclusions: This integrated approach leveraging quantum dot fluorescence, magnetic nanoparticle-based CTC capture, and AI-enhanced imaging offers a powerful, scalable solution for sensitive and multiplexed CTC analysis in breast cancer. The technology’s simplicity, sensitivity, and adaptability make it well-positioned for broader use in biomarker-driven oncology research and diagnostics.
Presentation numberPS3-04-19
Health Economic Evaluation of an Artificial Intelligence assisted Breast Cancer Multi Disciplinary Team Meeting/Tumour Board: Preliminary results from a UK Single Centre Simulation Trial
Olubukola Ayodele, University of Leicester, Leicester, United Kingdom
J. Tan1, P. Garodia2, R. Williams3, L. Cook3, M. Hasanova4, A. Kirby2, B. Lamb5, G. Hunnings2, G. Langton6, R. Pearson7, S. Adomah8, O. Ayodele9, A. Ghose4, A. Maniam3; 1University of Manchester, London, United Kingdom, 2University College London, London, United Kingdom, 3Isle of Wight NHS Trust, Newport, United Kingdom, 4OncoFlow AI, London, United Kingdom, 5North East London Cancer Alliance, London, United Kingdom, 6The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, United Kingdom, 7Lord Lister Health Centre, London, United Kingdom, 8The Royal Marsden NHS Foundation Trust, London, United Kingdom, 9University of Leicester, Leicester, United Kingdom
Background Cancer Multi-Disciplinary Team Meetings (MDTMs) in the UK ensure comprehensive, patient-centred decision-making. They operate at an industrial scale across the National Health Service England (NHSE), with an estimated annual cost of approximately £154.3. Each patient may be discussed upto an average of four times, with a base cost of £428 per patient, increasing to £485 when accounting for opportunity costs. While MDTMs aim to improve outcomes, the substantial financial burden raises questions about efficiency and value. Standard NHS Consultant Programmed Activity (PA) is 4 hours, worth almost £100. The Isle of Wight (IOW) Breast Cancer MDT comprises 2 Oncologists/Equivalent, 3 Surgeons, 1 each of Radiologist, Pathologist, Radiographer, Clinical Nurse Specialist (CNS) and MDT Coordinator, with weekly MDTMs involving 1560 to 2080 cases annually. The OncoflowTM Artificial Intelligence (AI) powered Cancer MDTM CoPilot platform is a class 1 UKCA (UK Conformity Assessed) MHRA (Medicines and Healthcare products Regulatory Agency) registered medical device (RN 32434) that uses proprietary Large Language Model (LLM) based Data Extraction and Treatment Matching. A simulation trial of this intervention vs standard manual practice was conducted to inform a cost benefit and effective analysis. Methods The Simulation study involved 2 prospective Breast Cancer MDTMs with 10 cases each, matched to similar disease stage (early or metastatic) and complexity (simple/edge/complex). Phase 1 was standard practice and Phase 2 was the OncoFlow intervention. Manual preparation for this 10 case set involved 120 minutes or 1.5 hours of time from 10 IOW MDT members as aforementioned, with role-specific hourly rates applied. OncoFlow was assumed to eliminate labour input, incurring only an annual licence fee of £25,000. A cost-analysis model compared the resource use and associated costs of standard versus OncoFlow AI assisted breast cancer MDTM preparation. Results Standard manual preparation cost per 10 breast cancer cases taking a total 1.5 hours time was £378.00, equating to £37.80 per case. This included £262.50 for 7 consultants’ time (£25/hour) and £115.50 for 3 other MDT staff (£25 for radiographer, £22 for MDTM coordinator, £30 for CNS). In contrast, OncoFlow’s cost per case was calculated for two scenarios based on MDTM volume, i.e., £16.03 for 1,560 cases/year and £12.02 for 2,080 cases/year, based solely on the annual licence fee. This resulted in savings of £21.77 and £25.78 per case, respectively. Annualised savings were projected from £33,961.20 (1,560 cases) to £53,622.40 (2,080 cases) for 52 weekly breast cancer MDTMs. OncoFlow also reduced preparation time from 13.5 hours per case to negligible levels (~0.01 hours), yielding an Incremental Cost Effectiveness Ratio (ICER) of £1.91 per hour saved. This implies that for every hour of clinician time saved through automation, the cost difference was £1.91, favouring OncoFlow as a cost-saving intervention. Conclusion The OncoflowTM AI Cancer MDTM CoPilot significantly reduces the cost burden associated with manual MDTM preparation in breast cancer pathways. The system delivers cost savings of up to £25.78 per case. With an ICER of £1.91 per hour saved, OncoFlow offers a highly cost-effective solution for modernising cancer MDTM workflows, operating at scale. Broader implementation across tumour types and MDTMs could amplify these cost benefits, contributing to more sustainable, efficient NICE (National Institute for Health and Care Excellence) commissioned cancer care delivery across the NHS.
Presentation numberPS3-04-21
Bridging the Reality Gap: A Multimodal Evaluation of Large Language Models for Real-World Breast Cancer Decision Support
Zheng Qu, University of Pennsylvania, PHILADELPHIA, PA
Z. Qu1, X. Wang2, S. Pei3, Y. Fang3; 1Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, PHILADELPHIA, PA, 2College of Information Sciences and Technology, The Pennsylvania State University, State College, PA, 3Department 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.
Background: Large Language Models (LLMs) have shown promising accuracy in generating guideline-based cancer treatment recommendations. However, their alignment with real-world breast cancer management decisions remains poorly characterized. This study presents a novel benchmark evaluating LLM-generated recommendations against both clinical guidelines and actual treatments recorded in electronic health records (EHR), while also assessing the models’ capacity for patient-centered communication. Methods: We extracted structured and unstructured EHR data from 500 newly diagnosed non-metastatic breast cancer patients (2023-2025) at Penn Medicine, including imaging data. Each case was processed through a multi-turn, agent-based LLM prompting framework using a range of models, including general-purpose LLMs (GPT-4o, Claude 3.7, DeepSeek R1, Grok), as well as medically tuned models (MedGemma, HealthBench). Each model received basic case data and dynamically queried for additional clinical information before recommending a treatment plan. Recommendations were compared to (1) NCCN/ESMO guidelines and (2) the actual treatment received (ATR). Concordance rates, over-/under-treatment frequencies, and the “Reality Gap” (cases where recommendations matched guidelines but diverged from real-world practice) were analyzed. Propensity score-based causal inference was used to identify demographic and socioeconomic drivers of discordance. Additionally, a multidisciplinary panel rated LLM responses to common patient questions in terms of clarity, empathy, and shared decision-making support. Results: Across all models, guideline concordance was high (GPT-4o: 90.2%, Claude 3.7: 89.4%, Deepseek: 88.6%, MedGemma: 87.9%). However, concordance with real-world treatment was notably lower (GPT-4o: 67.8%, Claude 3.7: 65.1%, MedGemma: 62.3%), with LLMs generally recommending more intensive treatment than was actually administered. In 22.3% of cases, the LLM recommendation aligned with guidelines but diverged from the real-world decision, reflecting a significant “Reality Gap.” Multivariable analysis identified Medicaid insurance (OR 2.14, 95% CI 1.31-3.49), age ≥70 (OR 1.72, 95% CI 1.08-2.75), and documented treatment refusal as independent predictors of discordance. In the communication task (N=100 dialogues), GPT-4o showed relatively strong performance in accuracy (4.61/5) and decision support (4.55/5), though differences between models were modest. Conclusions: This study directly addresses a central question in AI-enabled oncology: how far are large language models (LLMs) from actual clinical decision-making? While LLMs are capable of generating medically sound and guideline-concordant recommendations, real-world factors—such as patient preferences, socioeconomic barriers, and health literacy—are often overlooked, despite playing a critical role in shaping care delivery. By jointly evaluating clinical validity and contextual fidelity, our framework quantifies the “reality gap” between LLM recommendations and actual treatment. Importantly, we do not advocate for LLMs to conform to all real-world deviations, nor should models be encouraged to offer suboptimal plans based on non-clinical attributes. Instead, our findings highlight the importance of using social context to enhance interpretation and patient-centered communication—without compromising evidence-based care. Our work provides a concrete model for evaluating and improving AI alignment with real-world clinical decisions in breast cancer care. Further analyses are underway to expand the scope and robustness of these findings.
Presentation numberPS3-04-22
A mathematical framework to optimize combination therapy for triple-negative breast cancer in obese mice
Krithik Vishwanath, The University of Texas at Austin, Austin, TX
K. Vishwanath1, C. I. Crawford2, A. G. Sorace2, T. E. Yankeelov3, E. A. Lima1; 1Oden Institute for Computational Engineering & Sciences, The University of Texas at Austin, Austin, TX, 2Radiology, The University of Alabama at Birmingham, Birmingham, AL, 3Biomedical Engineering, The University of Texas at Austin, Austin, TX.
Introduction. Triple-negative breast cancer (TNBC) disproportionately affects obese individuals, with emerging evidence suggesting that obesity may decrease or paradoxically enhance the efficacy of certain immunotherapies. However, obesity-driven intratumoral hypoxia presents additional challenges for treatment, motivating the use of evofosfamide (a hypoxia-activated prodrug) as a therapeutic adjunct. With this dual challenge in mind, we set out to build a clinically oriented ‘virtual trial’ platform that can forecast tumor response and pinpoint dosing schedules that maximize immunotherapy + evofosfamide benefit while minimizing toxicity in obesity-linked TNBC. Methods. We have developed a mathematical framework that allows us to conduct ‘virtual trials’ on the computer, screening thousands of possible dosing protocols before any animal or patient is treated. The framework is built on a nonlinear system of ordinary differential equations that simultaneously describe intrinsic tumor growth, immune response, angiogenesis and vessel regression, and the effects of evosfamide and immunotherapy—whose interaction is modulated by the well-vascularized fraction of the tumor. Model parameters are inferred with Markov Chain Monte Carlo, an iterative random-sampling algorithm that scans the parameter space and converges on the combinations that most closely replicate the observed data. Model calibration relies on 34 days of serial data from C57BL/6J mice (N=24). Prior to data acquisition, the obese model of TNBC was established with 13 weeks of a high-fat diet (HFD) and then orthotopically implanted into the mammary fat pad with E0771 TNBC cells 12 days before baseline (day 0). For each mouse, we tracked tumor volume by caliper and the well-vascularized fraction by [18F]-fluoromisonidazole (FMISO) PET across four study arms—control, evofosfamide alone, immunotherapy on normoxic mice, and the combination on hypoxic mice. Mice were grouped seven days before baseline. By varying dosing and scheduling, we optimize treatment for three objectives: 1) reducing final tumor volume, 2) minimizing total tumor burden (tumor volume integrated over the experimental interval), and 3) limiting therapy toxicity. Results. Our mathematical framework accurately reproduces (i.e., calibrate) tumor growth dynamics under control and individual treatment conditions, achieving an average concordance correlation coefficient (CCC) of 0.97 ± 0.005. The model is also able to accurately predict the response of tumors to combination therapy, yielding a CCC of 0.93. Through differential evolution optimization—a mathematical technique that mimics natural selection by iteratively combining and selecting the best treatment parameter sets—we identify candidate treatment protocols that we hypothesize will yield a 67.5% reduction in average final tumor volume, a 68.2% reduction in tumor burden, and an 18.7% reduction in total therapy dosage compared to the standard of care. Conclusion. The findings underscore the efficacy of reducing hypoxia to enhance immunotherapy in HFD mice, which can be significantly strengthened through optimized scheduling of evofosfamide and immunotherapy combination therapies. The proposed mathematical framework demonstrates a robust ability to predict tumor response and optimize treatment strategies, highlighting the potential for translating mathematical systems to guide combination therapies and develop digital twins for personalized medicine. The optimized schedules point to the possibility of achieving significantly better tumor control with less total drug dose, potentially translating into reduced toxicity while preserving benefit. These optimized regimens should be regarded as hypothesis-generating; their safety and therapeutic benefit must be experimentally confirmed.
Presentation numberPS3-04-23
Artificial intelligence (AI)-generated electronic medical record summarization in breast oncology
Ko Un Park, Dana-Faber Cancer Institute/ Brigham and Women’s Hospital, Boston, MA
K. Park, C. A. Minami, L. N. Butler, J. Gittzus, K. McLean, A. Dunn, E. A. Mittendorf, T. A. King; Surgery, Dana-Faber Cancer Institute/ Brigham and Women’s Hospital, Boston, MA.
Introduction: Pre-charting for oncology patients (pts) requires manual review of documentation, often with varying formats. AI trained to summarize records for pre-charting may enhance efficiency in providing consistent information to clinicians, thereby enhancing the quality of care. We developed and clinically validated an AI agent to summarize records of pts with breast cancer. Methods: A generative AI agent utilizing GPT-4o and Retrieval-Augmented Generation (RAG) for processing radiology, pathology, and consultation reports was developed on a secure platform. Prompt engineering was employed to guide GPT-4o in creating oncology-specific summaries. For 50 consecutive breast cancer pts, history of present illness summaries (HPIs) generated by 4 breast surgery physician assistants (PAs) were compared to AI HPIs of identical records. Two breast surgeons evaluated the AI HPIs by rating the organization (clarity’) and contextual relevance (relevance’) of the information on a 5-point Likert scale (1=strongly disagree, 5=strongly agree). They clinically validated the AI HPIs by comparing to the PA HPIs using a structured evaluation framework focused on accuracy (factually correct/incorrect) and completeness of information (‘complete’ if AI HPIs aligned with the key information that oncologists would anticipate in a PA HPI) across predefined domains: patient age, imaging findings (mammogram, ultrasound, MRI, biopsy modality), pathology findings (histology, receptor status), medical history, and pending workup. Results: The overall clarity and relevance of the AI HPIs were high (average clarity 4.01, average relevance 3.87). AI accurately stated pt age in 94% of HPIs, with discrepancies attributed to birthday timing since initial workup. All pts had a mammogram, 86% had an US, and 36% MRI. Accuracy of imaging report summaries varied by modality, with MRI showing lowest accuracy (83.3%) and completeness rates (44.4%) (Table). Among the 85 biopsies, the biopsy modality was missing in 39.8%; AI HPI stated ‘core biopsy’ rather than ‘stereotactic guided’, ‘US guided’ or ‘MRI guided’ core biopsy. Pathology reporting demonstrated high accuracy among the 85 biopsies: histology 97.6%, hormone receptors and HER2 receptor 89.8%, and tumor grade 95%. Inaccurate AI responses resulted when grade or receptor stain percentage were reported in ranges. Two HPIs misrepresented past medical history. Among 12 patients requiring additional workup, 41.7% of AI HPIs appropriately identified pending studies. Conclusion: RAG-enabled GPT-4o demonstrated a high level of accuracy in breast cancer record summarization for pre-encounter preparation. Human review is necessary, but AI shows promise for clinical integration. Iterative prompt engineering, integration into clinical workflow, and evaluating the effectiveness of AI-assisted breast oncology medical record summarization is ongoing.
| Accurate Information | Complete information | |
| Mammogram (n=50) | 47 (94%) | 46 (92%) |
| US (n=43) | 37 (86%) | 39 (90.7%) |
| MRI (n=18) | 15 (83.3%) | 8 (44.4%) |
Presentation numberPS3-04-24
Common and rare germline variants together with somatic mutations alter the integrity of cancer hallmark regulatory networks
Jiawei Dai, Yale School of Medicine, New Haven, CT
J. Dai1, M. Posta2, M. Marczyk3, T. Qing1, B. Győrffy2, L. Pusztai1; 1Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Department of Bioinformatics, Semmelweis University, Budapest, HUNGARY, 3Department of Data Science and Engineering, Silesian University of Technology, Gliwice, POLAND.
Background:How germline and somatic alterations together contribute to alterations in cancer relevant pathways in individual cancers remains poorly understood.Methods:We mapped all germline and somatic alterations of individual cancer samples from The Cancer Genome Atlas (TCGA) and the Breast Cancer Genome Guided Therapy Study (BEAUTY) onto Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomics (KEGG) pathways. To quantify pathway-level disturbances from germline and somatic origins, we calculated gene-level impact scores by integrating the functional importance of a gene in sustaining cell survival with the predicted impact of its variants. These scores were then used to compute a pathway disturbance score (CanSys score), that quantified the combined effect of gene-level alterations across all pathway members. Results:We found that almost all cancers have common and rare protein function altering germline alterations in cell cycle, telomere maintenance, and DNA repair hallmark pathways. At somatic mutation level, 12 pathways were altered in >75% of cancers corresponding to core cancer hallmarks, however different genes within the same pathway are affected in different individuals, illustrating phenotypic convergence. In addition, 404 pathways were affected in >25% but <50% of cancers which may contribute to the unique clinical course of each cancer. A freely available web tool (https://cansysplot.com) developed for this study enables visualization of GO/KEGG pathway disturbances from germline and somatic origins at the individual sample level.Conclusion:Our findings implicate that the individuals with cancer harbor protein function altering germline alterations in genes involved with cell cycle regulation, telomere maintenance, and DNA repair, whereas somatic mutations primarily affect pathways involved in cell adhesion, cell motility, metabolism, and a broad range of signal transduction processes. The unique combination of pathway alterations might explain the unique behavior of each cancer.
Presentation numberPS3-04-25
Artificial intelligence (AI) based image analysis of PD-L1, TIL, immune signature and ctDNA for prediction of response to neoadjuvant chemotherapy in breast cancer
Eun Young Kim, Kangbuk Samsung Hospital, Seoul, Korea, Republic of
E. Kim1, I. Do2, S. Chae2; 1General Surgery, Kangbuk Samsung Hospital, Seoul, KOREA, REPUBLIC OF, 2Pathology, Kangbuk Samsung Hospital, Seoul, KOREA, REPUBLIC OF.
Background Tumor-infiltrating lymphocytes (TILs) have been recognized as prognostic and predictive biomarkers in breast cancer. However, challenges such as interobserver variability and lack of standardized cut-off values have limited their clinical implementation. Recent advancements in artificial intelligence (AI)-based pathology have shown potential to enhance reproducibility and objectivity in TIL quantification. Concurrently, circulating tumor DNA (ctDNA) has emerged as a minimally invasive biomarker capable of detecting treatment response after neoadjuvant chemotherapy (NAC). Objectives This study aims to: (1) evaluate the predictive value of AI-quantified stromal TILs from H&E-stained slides; and (2) validate the clinical utility of ctDNA-based biomarkers for detection of responses in breast cancer patients after NAC. Methods TILs were assessed from pre-treatment tumor H&E slides using Lunit SCOPE IO, a validated AI-powered WSI analysis tool. Immune phenotypes were classified as inflamed, immune-excluded, or desert. PD-L1 expression was evaluated using PD-L1 IHC 22C3 pharmDx. TIL cut-off was set at 30%, Formalin-fixed paraffin-embedded samples (FFPEs) were obtained from biopsy tissue, and plasma samples were collected at baseline, pre-surgery (after NAC). Forty-seven breast cancer-related genes were analyzed using next-generation sequencing. The diagnostic performance of ctDNA was evaluated, and logistic regression analyses were conducted to assess the impact of clinical and molecular factors on ctDNA status. Parallel plasma samples were collected at predefined intervals for ctDNA extraction and sequencing using a customized Breast NGS panel targeting 157 hotspot mutations across 12 genes. Mutation validation was performed using ddPCR. Preliminary Results A total of 101 patients with early breast cancer were enrolled. The most frequently identified gene was TP53 (FFPE, 66.7%; ctDNA, 46.4%), followed by PIK3CA (FFPE, 36.2%;ctDNA, 17.4%). The triple-negative breast cancer (TNBC) subtype exhibited the strongest association with ctDNA detection (odds ratio [OR] 209.50, p = 0.005) in multivariate analysis. Patients with inflamed TIL phenotype at diagnosis and ctDNA clearance after NAC had higher pathological complete response (pCR) rate (38.5% vs. 11.1%, p = 0.238). Conclusion Our integrated approach using AI-based TIL quantification and ctDNA analysis using NGS and ddPCR provides a feasible and sensitive approach for NAC response in breast cancer patients. These findings support the clinical utility of combining digital pathology and liquid biopsy for treatment monitoring and risk stratification in conjunction with precision oncology.
Presentation numberPS3-04-26
Reproducibility and Robustness of an AI-based Stromal Tumor-Infiltrating Lymphocyte Pipeline in Triple-Negative Breast Cancer
Pingjun Chen, The University of Texas MD Anderson Cancer Center, Houston, TX
S. Ranjbar1, A. San Lucas2, P. Chen1, K. Shah2, M. Suchko2, B. Guerrouahen2, C. Ercan3, E. Barrientos Toro3, K. Sweeney4, X. Pan1, J. DeLa Cruz2, B. Rodriguez1, N. Reddy2, C. Yam5, G. Mantha2, M. Riben4, L. Huo4, Y. Yuan1; 1Institute for Data Science In Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Division of Pathology and Laboratory Medicine Clinical Trial Operations, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 5Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
Introduction – Triple-negative breast cancer (TNBC) has limited treatment options and significant treatment-related toxicity. Recent studies have shown that quantifying stromal tumor-infiltrating lymphocytes (sTILs) can aid in identifying early-stage TNBC patients likely to achieve a pathological complete response to neoadjuvant chemotherapy (Abuhadra et al., 2023), suggesting its utility in treatment stratification. However, manual assessment of sTILs is prone to interobserver variability (Van Bockstal et al., 2021). Artificial intelligence (AI) promises a more consistent alternative, but its reproducibility and robustness to input variability remains insufficiently evaluated, limiting its clinical adoption (Park et al., 2020). Methods – We aimed to validate the reproducibility and robustness of an in-house AI-sTIL pipeline (AbdulJabbar et al., 2020; Pan et al., 2025) developed using H&E-stained slides from the ARTEMIS trial (NCT02276443), scanned between 2016 and 2024 on a Leica Aperio AT2 scanner at 20x magnification. We first compared AI-generated sTIL scores with two independent sets of manual sTILs (n=201) provided by pathologists according to the International TIL Working Group guidelines. Moreover, to establish deployment readiness, we examined the effects of input variability, including scanner type, scanning magnification, operator, and slide aging by rescanning up to 150 slides and conducting controlled, pairwise comparisons. Rescanned images were generated at 20x and 40x magnifications using Aperio AT2 and Hamamatsu NanoZoomer scanners, and at 40x using Aperio GT450. To assess operator viability, the scanning was run independently by different operators with AT2 (20x and 40x) and GT450 (40x). Results – Manual sTILs showed strong concordance between pathologists (ρ=0.884, R²=0.855). Comparing this agreement to the inter-operator reproducibility of the pipeline (ρ=0.980-0.994, R²=0.978-0.984) suggests AI-sTIL is robust to typical user handling variations in clinical workflows and offers a more consistent assessment. The pipeline correlated well with individual manual sTILs (ρ=0.768, R²=0.502 for pathologist1, and ρ=0.765, R²=0.579 for pathologist2) and even more strongly with the average of the two sets of manual sTILs (ρ=0.793, R²=0.583), indicating the pipeline may approximate a consensus interpretation while minimizing individual variability. Scores remained highly consistent across scanner models (AT2 vs. GT450: ρ=0.972, R²= 0.941; AT2 vs. Hamamatsu: ρ=0.987, R²=0.975), though a systematic bias toward higher scores for AT2 (regression slopes of 0.853 and 0.885, when compared with GT450 and Hamamatsu scores) suggests calibration may be necessary to enable accurate cross-platform comparisons. Similarly, scores were consistent across scanning magnification (20x vs. 40x: AT2: ρ=0.984, R²=0.964; Hamamatsu: ρ=0.992, R²=0.979). Interestingly, we observed lower scores for rescanned images of older slides (median slide age: 6.3 years), likely due to stain fading. However, this is unlikely to impact clinical deployment, as such delays between tissue sectioning and scanning are rare in routine clinical practice. The pipeline demonstrated excellent inter-run consistency with 100% concordance (ρ=0.991, R²=0.988), confirming the repeatability and stability of the pipeline. Conclusion – This study highlights the reproducibility and robustness of our in-house developed AI-sTIL pipeline across common variations in pathology workflows. Through controlled data acquisition experiments, we identified the appropriate data acquisition setup to pair with the pipeline. Our approach provides a practical framework for institutions seeking to assess deployment readiness of pathology AI tools before formal validation and clinical integration.
Presentation numberPS3-04-27
Mutational burden and clonal expansion in lymph nodes correlate with IBC postoperative relapse
Qing Ye, MD Anderson Cancer Center, Houston, TX
Q. Ye1, K. Chen1, A. Alexander2, R. Upadhyay3, S. Krishnamurthy4, C. Yam2, The MDACC Inflammatory Breast Cancer Team, A. Lucci5, W. Woodward6, B. Lim2; 1Bioinformatics & Comp Biology, MD Anderson Cancer Center, Houston, TX, 2Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 3Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, 4Anatomical Pathology, MD Anderson Cancer Center, Houston, TX, 5Department of Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, 6Breast Radiation Oncology, MD Anderson Cancer Center, Houston, TX.
Background Inflammatory breast cancer (IBC) is a rare and aggressive subtype of breast cancer characterized by early metastasis and poor prognosis. Analyzing mutational patterns between primary tumors and lymph node metastases is critical for understanding postoperative relapse yet remains challenging due to marked intratumoral heterogeneity (ITH). Methods Whole-exome sequencing (WES) was performed on six paired samples of primary tumors and matched lymph node metastases from patients treated with endocrine therapy and pembrolizumab recruited through a trial (NCT02971748), with two patients remaining disease-free and four experiencing postoperative relapse that ultimately led to death. Somatic variants from WES data were identified using Mutect2, annotated with OpenCRAVAT, and filtered to retain only high-confidence, protein-altering, non-synonymous mutations with sufficient sequencing depth and gene mapping. Clonal architecture and cellular prevalence were analyzed using PyClone. Copy number variations (CNVs) were identified using FACETS, which performs joint segmentation across all samples and classifies CNV segments into structural variant types such as deletions, duplications, and loss of heterozygosity. Results To explore mutation patterns linked to postoperative relapse, we compared mutated genes between primary breast tumors and matched lymph node metastases (LNM), classifying them as either shared or unique to each tissue. Initial analysis focused on cancer-related genes from the Cancer Gene Census. Genes mutated non-recurrently between the primary tumors and the autologous LNM samples showed low variant allele frequencies, with median VAFs of 7.3% and 11%, respectively, suggesting limited clonal evolution. In contrast, genes mutated in both tissues, such as NF1, PIK3CA, and TP53, tended to show higher VAFs, particularly in the LNMs (median VAF: 22%). Within individual patients, few annotated mutations were shared between the primary tumor and lymph node, with most being site-specific, indicating substantial intra-patient mutational heterogeneity. To extend beyond annotated cancer genes, we analyzed all mutated genes to explore the relationship between mutation patterns and clinical outcomes. In patients with favorable outcomes, the VAF in the LNMs showed a maximum decrease of ~33% from the primary tumor, while in patients with poor outcomes, VAFs increased by ~70% in lymph nodes relative to the primary site. This pattern suggests a potential association between higher mutational burden in lymph nodes and postoperative relapse. Clonal structure analysis further revealed outcome-associated differences: in patients with favorable outcomes, one exhibited very low clonal prevalence in the lymph nodes (~2%), and another showed a ~40% decrease compared to the primary tumor. In contrast, patients with postoperative relapse displayed mixed patterns, but notably included mutations with clonal prevalence increasing by over 20% in lymph nodes, suggesting potential clonal expansion linked to metastasis. Copy number variation analysis revealed distinct structural variant patterns across patients, with deletion counts increasing from primary tumors to lymph nodes in favorable outcome cases, but decreasing in those with poor outcomes. The increase in deletions was most prominent on chromosome 19 in patients with disease-free, where three overlapping genes, HCN2, CD177P1, and TEX101, were deleted. Conclusion These findings suggest that disease-free HR+ IBC were associated with lower mutation burden and limited clonal expansion in lymph node metastases, while postoperative relapse are linked to increased clonal dominance and structural alterations, particularly in the metastatic site. Future studies can validate such finding as potential biomarker and therapeutic targets.
Presentation numberPS3-04-28
Espwa: a deep learning-enabled computational pathology tool that facilitates precision oncology for haitian breast cancer patients
Albert E Kim, Massachusetts General Hospital, Boston, MA
D. Pulido-Arias1, R. Henderson2, J. Lormil3, C. Millien4, M. Djenane Jose5, G. Flambert6, M. Mathelier6, M. Corrielus6, T. Goncalves1, J. Kalpathy-Cramer7, E. Gerstner8, K. Landgraf9, A. Brown9, P. Castle10, J. Jeronimo10, S. Sirintrapun11, S. Wander8, D. Milner9, J. Brock9, C. Bridge1, A. E. Kim8; 1Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, 2OBGYN, University of Alabama-Birmingham, Birmingham, AL, 3Cancer Center, Partners-in-Health/Zanmi Lasante, Port-au-Prince, HAITI, 4Mirebalais University Hospital, Partners-in-Health, Mirebalais, HAITI, 5Pathology, Partners-in-Health/Zanmi Lasante, Port-au-Prince, HAITI, 6Shands Hospital, University of Florida College of Medicine, Gainesville, FL, 7Division of Artificial Medical Intelligence, University of Colorado, Aurora, CO, 8Cancer Center, Massachusetts General Hospital, Boston, MA, 9Center for Global Health, American Society for Clinical Pathology, Chicago, IL, 10Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, 11Pathology, Massachusetts General Hospital, Boston, MA.
ESPWA: A deep learning-enabled computational pathology tool that facilitates precision oncology for Haitian breast cancer patients Background: Global disparities in breast cancer outcomes are a public health crisis. In Haiti, one of the most resource-limited health care settings in the world, the challenge is especially dire. With only five practicing pathologists serving the entire country, timely and complete pathologic assessment of tumor tissue is nearly impossible. As estrogen receptor (ER) assessment with immunohistochemistry (IHC) cannot be performed, all Haitian breast cancer patients are empirically treated with endocrine therapy (ET). This “one-size-fits-all” strategy results in increased morbidity and mortality. To address this unmet need, we present ESPWA: ER Status assessment using deep learning-enabled histoPathology Whole slide imaging (WSI) Analysis, a low-cost tool that informs precision-based use of ET for Haitian patients. Methods: To develop ESPWA, we curated a dataset of H&E WSIs and tissue-matched ER status for 3448 breast cancer patients from Zanmi Lasante (ZL), the largest health care provider in Haiti. This dataset encompasses primary tumors (n = 2933) and lymph node metastases (n = 515). Using the ZL and The Cancer Genome Atlas (TCGA)-breast cancer datasets (n = 1349), we used weakly-supervised attention-based multiple instance learning to train models, using H&E as input, that predict ER status for each patient. Binary classification (output: ER-positive vs negative) and regression (output: quantitative expression of ER– 0-100%) models were trained. Label-preserving image augmentation methods (e.g., color augmentation, rotations, flips) were applied to increase diversity of training data. To assess performance, both the TCGA and ZL datasets were split into training (70%), validation (15%), and testing (15%) sets, and the results were compared across both patient populations. Results: First, we trained an ER classification model using the TCGA dataset. Performance of this TCGA-trained model was sensitive to the domain shift between the Black and Caucasian populations, with a performance of an area under receiver operating characteristic (AUROC) of 0.846 [95% confidence interval: 0.831-0.876] on the “held-out” TCGA test set, and 0.671 [0.655-0.683] on the ZL test set. Next, using 10-fold cross validation and bootstrapping analyses, we trained ER classification and regression models with the ZL dataset. These models demonstrated an AUROC of 0.790 [0.774-0.805] and an intraclass correlation coefficient of 0.524 [0.471, 0.551] on the ZL test sets. Subgroup analyses revealed that ESPWA performed less well for Haitian patients harboring ER-weak positive tumors (compared to ER-negative and strong positive tumors) and poorly differentiated tumors (compared to well-differentiated tumors). A head-to-head comparison between ESPWA and a breast pathologist with 22 years of experience (J.E.B.) illustrated ESPWA’s potential to outperform human experts. ESPWA (accuracy: 0.726 [0.711, 0.740]) displayed superior performance in determining ER status from an H&E compared to the pathologist (accuracy: 0.639 [0.627, 0.651]) for the entire dataset and across nearly all subgroups. Conclusions: In collaboration with the American Society of Clinical Pathology, we are planning clinical trials in ZL and other African countries to deploy ESPWA (“Hope” in Haitian Creole) to inform precision-based use of endocrine therapy for prospective patients.
Presentation numberPS3-04-29
Machine Learning predicts early tumor progression in Hormone Receptor-positive, Human Epidermal growth factor Receptor 2-negative (HR+/HER2-) advanced breast cancer (aBC) patients treated with first-line endocrine therapy (ET) plus Cyclin Dependent Kinase 4/6 inhibitors (CDK4/6i)
Leonardo Provenzano, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
L. Provenzano1, R. Caputo2, M. Dieci3, P. Vigneri4, M. Giuliano5, G. Curigliano6, A. Toss7, A. Botticelli8, R. Pedersini9, S. Cinieri10, M. Lambertini11, G. Rizzo12, B. Tagliaferri13, M. Sirico14, M. Giordano15, L. Gerratana16, I. Meattini17, M. Piras18, A. Fabi19, A. Zambelli20, F. Pantano21, A. Gennari22, N. La Verde23, D. Sartori24, O. Garrone25, D. Generali26, F. Ligorio1, G. Pruneri27, F. De Braud1, C. Vernieri1, PALMARES-2 study group; 1Medical Oncology 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 2Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Napoli, ITALY, 3Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padua, ITALY, 4Department Clinical and Experimental Medicine, University of Catania, Misterbianco, ITALY, 5Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II, Naples, Naples, ITALY, 6Division of Early Drug Development for Innovative Therapy, IRCCS European Institute of Oncology, Milan, ITALY, 7Division of Medical Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, ITALY, 8Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, ITALY, 9Medical Oncology Department, ASST Spedali Civili of Brescia, Brescia, ITALY, 10U.O.C. Oncologia medica, Presidio Ospedaliero “Antonio Perrino”, Brindisi, ITALY, 11Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Milan, ITALY, 12Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, ITALY, 13Medical Oncology Unit, ICS Maugeri-IRCCS, Pavia, ITALY, 14Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, Meldola, ITALY, 15SC Oncologia, ASST Lariana, Como, ITALY, 16Breast Medical Oncology, CRO Aviano National Cancer Institute, Aviano, ITALY, 17Unità di Radioterapia Oncologica, Azienda Ospedaliero Universitaria Careggi, Firenze, ITALY, 18Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, ITALY, 19UOSD Medicina di Precisione in Senologia, IRCCS Fondazione Policlinico Universitario Agostino Gemelli, Milan, ITALY, 20Oncology Department, Papa Giovanni XXIII Hospital, Milan, ITALY, 21Medical Oncology, Università Campus Bio-Medico di Roma, Rome, ITALY, 22Department Translational Medicine, University of Piemonte Orientale, Novara, ITALY, 23Department of Oncology, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, ITALY, 24Oncology Unit, AULSS3 Veneziana, Mirano, ITALY, 25SC Oncologia, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milan, ITALY, 26SC Oncologia Multidisciplinare, ASST Cremona, Cremona, ITALY, 27Medical Oncology 1, Department of Diagnostic Innovation, Milan, ITALY.
Introduction: CDK4/6i combined with ET reshaped the first-line treatment paradigm for HR+/HER2-advanced Breast Cancer (aBC). However, early progressors, as defined as patients (pts) undergoing tumor progression within 6 months from treatment initiation, remain a major clinical challenge. Indeed, identifying early progressors may lead to the use of alternative, potentially effective, treatments. Machine learning (ML) has demonstrated significant capability in integrating data extracted from electronic health records to successfully predict clinical outcomes in oncology. Methods: PALMARES-2 (NCT06805812) is an observational, retrospective-prospective, multicenter Italian study that is investigating the effectiveness of first-line ET plus CDK4/6i in HR+/HER2- aBC pts. We used the log-rank test to evaluate the impact of early progression on real-world (rw) time to chemotherapy (rwTTC) and overall survival (rwOS). Four ML architectures, namely linear regression with elastic net penalisation (glmnet), random forest (RF), XGBoost (XGB) and neural network (NN), were trained to predict the risk of early progression (rw Progression-Free Survival <6 months). As model inputs, we used 73 variables, including clinical, pathological and laboratory ones. Missing data were imputed through the missForest method, while undersampling was applied to address class imbalance. The best hyperparameters were selected based on 10-fold cross-validation (CV) results. After model development in the training set, model performance was confirmed on a test set (20% of the overall population) and on an independent cohort of pts from Istituto Nazionale dei Tumori, Milan. Results: Among 4104 evaluable pts, 521 (12.7%) experienced early tumor progression. Early progressors had significantly worse rwTTC (median 5.3 vs 43.4 months, P <0.001) and rwOS (median 21.4 vs 73.0 months, P <0.001) when compared to pts who did not experience early progression. Early progressors were more likely to have lower tumor estrogen receptor and progesterone receptor expression, higher Ki67 expression, endocrine-resistant disease at diagnosis, worse ECOG PS and liver metastases (P <0.001). All models demonstrated consistent accuracy, with acceptable generalizability in the independent test cohort (Table). Conclusions: Early tumor progression during first-line ET+CDK4/6i is associated with remarkably worse outcomes in HR+/HER2- aBC pts. We developed the first ML model to predict the risk of early progression in this setting. If prospectively validated, this tool may assist clinicians in selecting pts who could benefit from alternative first-line treatment approaches, thus potentially improving long-term outcomes, such as TTC and OS.
| RF training CV | RF test | RF independent cohort | Glmnet training CV | Glmnet test | Glmnet independent cohort | XGB training CV | XGB test | XGB independent cohort | NN training CV | NN test | NN independent cohort | ||||||||||||||
| Accuracy | 0,71 | 0,75 | 0,6 | 0,69 | 0,71 | 0,59 | 0,73 | 0,75 | 0,57 | 0,67 | 0,72 | 0,57 | |||||||||||||
| Sensitivity | 0,76 | 0,65 | 0,66 | 0,65 | 0,63 | 0,75 | 0,79 | 0,58 | 0,62 | 0,67 | 0,62 | 0,74 | |||||||||||||
| Specificity | 0,65 | 0,76 | 0,59 | 0,72 | 0,73 | 0,56 | 0,65 | 0,78 | 0,57 | 0,72 | 0,74 | 0,55 |
Presentation numberPS3-04-30
Developing virtual tumors using spherical harmonics and quantitative MRI data
Gauri Patel, University of Texas at Austin, Austin, TX
G. Patel1, D. A. Hormuth II2, R. J. Patel1, C. E. Stowers2, A. Chaudhuri2, E. A. Lima3, J. Kileel4, T. E. Yankeelov5; 1Biomedical Engineering, University of Texas at Austin, Austin, TX, 2Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, 3Oden Institute for Computational Engineering and Sciences, Texas Advanced Computing Center, University of Texas at Austin, Austin, TX, 4Department of Mathematics, Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, 5Biomedical Engineering, Diagnostic Medicine, Oden Institute, Livestrong Institute, Imaging Physics, University of Texas at Austin, Austin, TX.
Introduction. It is well-recognized that response to a particular treatment can vary greatly from patient to patient. However, clinical trials are limited in the range of treatment regimens they can evaluate due to constraints on patient numbers. To address this issue in a time- and cost-effective manner, our group has developed biology-based mathematical models that quantify the spatiotemporal changes in tumor cell number in response to treatment, using patient-specific MRI data1. These models enable in silico clinical trials, allowing us to evaluate responses to many regimens in virtual patient populations. Such trials require virtual populations that are realistic but more heterogeneous than those observed in traditional trials. Current methods of generating virtual patients are limited in capturing intra- and inter-tumor heterogeneity beyond that observed in narrow, historical trial populations. This study introduces a method of generating large, realistic, and heterogeneous virtual patient populations by applying spherical harmonics-based expansion to tumor cellularity maps of triple negative breast cancer (TNBC) patients. Methods. Pretreatment diffusion weighted MRI (DW-MRI) and dynamic contrast enhanced MRI (DCE-MRI) data from 27 TNBC patients were acquired from the I-SPY 2 trial2. Tumor regions were identified from DCE-MRI scans. DW-MRI scans were used to calculate apparent diffusion coefficient maps, ADC(x), from which tumor cell number maps, N(x), were calculated for each patient3. We applied spherical harmonics-based expansion to each N(x) to compute an approximation of tumor cellularity, N*(x), as a weighted sum of spherical harmonic functions multiplied by suitable radial functions4. We used the concordance correlation coefficient (CCC) to determine a minimum number, n, of functions needed to obtain 90% agreement between N(x) and N*(x). To generate realistic, heterogeneous tumors, probabilistic distributions were fit to coefficients corresponding to pairs of consecutive basis functions. As the basis functions are ordered in decreasing importance for reconstructing N(x), the distributions aimed to preserve statistical relationships between pairs with similar importance. Specifically, n/2 multivariate normal distributions (MVNs) were fit to describe the set of all coefficients. Sampling all MVNs, we generated n coefficients that parameterized a virtual tumor. The two-sample Kolmogorov-Smirnov (KS) test applied to tumor volume and cellularity distributions quantified agreement between virtual and historical populations. Results. Spherical harmonics-based expansion yielded 252,279 basis functions that reconstruct N(x) with high agreement (mean ± standard deviation CCC = 0.99±.004); importantly, 500 basis functions were sufficient for accurate reconstruction (CCC = 0.92±.024). 250 MVNs were sampled 100 times to generate 100 virtual tumors. KS test results indicated that tumor volume distributions of virtual and historical populations are not significantly different (p = 0.002) and that tumor cellularity distributions are significantly different (p = 0.79). Conclusion. Using our spherical harmonics-based framework, we generated a virtual population that preserved tumor sizes of the historical population but allowed for variation in tumor cell distribution over the volume; this heterogeneous virtual population can be used to systematically quantify the efficacy of many treatment regimens and identify interventions to test experimentally. References.1. Wu C et al., MRI-Based Models Forecast Patient-Specific Treatment Cancer Res. 20222. Barker A et al. I-SPY 2: An Adaptive Breast Cancer Trial Design. Clin Pharm Ther. 2009.3. Jarrett AM et al. Quantitative MRI and Tumor Forecasting of Breast Cancer. Nat Protoc. 2021.4. Kileel J et al. Fast expansion into harmonics. SIAM J Sci Comput. 2025.
Presentation numberPS3-06-02
Prospective Evaluation of “PreciseBreast” AI Tool in Early-Stage Invasive Breast Cancer Risk Stratification
Talar Telvizian, Main Line Health, Wynnewood, PA
T. Telvizian1, A. P. Maity1, M. Weiss2, O. Agyei1, R. Ciocca3, C. Carruthers4, J. Sabol3, S. Kjelstrom5, A. Ghaneie1; 1Hematology/Oncology, Main Line Health, Wynnewood, PA, 2Radiation Oncology, Main Line Health, Wynnewood, PA, 3Breast Surgery, Main Line Health, Wynnewood, PA, 4Breast Surgery, Main Line Health, Paoli, PA, 5Lankenau Institute for Medical Research, Main Line Health, Wynnewood, PA.
Background: Hormone receptor-positive breast cancer is the most common type of breast cancer among women in the United States. Multigene assays such as Oncotype DX (ODX) assess recurrence risk and the potential benefit of chemotherapy, guiding treatment decisions. These assays have limitations, including high cost and lengthy processing time. This study evaluates the novel AI-powered PreciseBreast (PDxBR) risk score, developed by PreciseDx, which uses digital pathology and machine learning to provide a rapid, phenotype-based assessment of recurrence risk. The primary objective is to evaluate PDxBR as a reliable tool for guiding treatment based on recurrence risk. Methods: This prospective correlational study was conducted within a community health system. Eligible participants included female patients with newly diagnosed early-stage, hormone receptor-positive invasive ductal carcinoma who qualified for ODX testing. Pathology slides were analyzed using the PDxBR assay, which incorporates morphologic features such as nuclear pleomorphism, mitotic activity, tubule formation, and tumor-infiltrating lymphocytes. ODX defines low risk of distance recurrence as a score of 0-25, and high risk as ≥26. The RSClin tool’s low risk is defined as ≤ 3%, intermediate risk as 3-5%, and high risk as ≥5% in validation studies. PDxBR and ODX scores were compared using McNemar’s test, and PDxBR and RSClin scores were compared using a symmetry test. Turnaround time was compared using a paired t-test. The results of this study were for research purposes and did not affect patient management. Results: A total of 32 patients were enrolled. The mean age was 63 years, ranging from 40 to 82 years. Twenty-six patients (81.3%) had stage IA disease, 4 patients (12.5%) had stage IIA, and 2 patients (6.3%) had stage IIB disease. Three patients (9.4%) had carcinoma with mixed invasive ductal and lobular features, while the remaining 29 patients (90.6%) had pure invasive ductal carcinoma. Among the seven premenopausal patients, ODX scores ranged from 6 to 16, with one patient scoring 18. Among patients classified as ODX low risk (N=6), 100% were deemed high risk by PDxBR. Only one patient had a high-risk ODX score, and that patient was classified as low risk by PDxBR (p = 0.059). There was a significant association between RSClin and PDxBR classifications: 66.7% of PDxBR high risk patients were also high risk by RSClin, while most RSClin intermediate and low risk patients were classified as low risk by PDxBR (p < 0.0001). The mean turnaround time for ODX was 6.3 days (SD 3.4) versus 2.0 days (SD 1.5) for PDxBR (p < 0.0001). The list price for PDxBR was $1,500 compared to $4,620 for ODX. Conclusion: The PDxBR assay offers significantly faster turnaround time and lower cost compared to ODX. The PDxBR risk score demonstrates strong concordance with the RSClin tool, and while there is discordance with ODX, the small sample size limits definitive conclusions. A larger study is needed to further evaluate concordance, non-inferiority, and clinical impact. Additional investigation is warranted to explore the basis of discordant results. Ultimately, a combined approach incorporating gene expression profiling, clinical features, and tumor morphology may provide the most accurate recurrence risk assessment. AI-powered digital pathology tools like PDxBR may complement or, in certain settings, serve as cost-effective alternatives to multigene assays, especially in resource-limited settings. This study is ongoing, and continued enrollment will allow for a more robust evaluation.
Presentation numberPS3-06-03
Refining adjuvant CDK4/6 inhibitor eligibility in early HR+ HER2- breast cancer with artificial intelligence
Nicholas P McAndrew, University of California Los Angeles, Los Angeles, CA
N. P. McAndrew1, E. Chiru2, E. Senkus-Konefka3, A. Bardia4, J. Cappadona5, K. G. Zeng5, K. J. Geras5, J. Witowski5; 1Department of Medicine, University of California Los Angeles, Los Angeles, CA, 2Medical Oncology, Basel University Hospital, Basel, SWITZERLAND, 3Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, POLAND, 4UCLA Health Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, 5AI, Ataraxis AI, New York, NY.
Background: CDK4/6 inhibitors have demonstrated efficacy in reducing disease recurrence among HR+ HER2- breast cancer patients meeting high risk of recurrence (RoR) criteria. These criteria, defined in trials such as NATALEE, consist of factors including Ki67>20%, G3, or Oncotype DX>26. However, these criteria are suboptimal predictors of recurrence risk and leave room for improvement with respect to only providing CDK4/6i to truly high RoR patients. We evaluated Ataraxis Breast (ATX), an artificial intelligence tool which integrates morphological features extracted from H&E-stained slides with clinical information, to (1) establish the RoR by ATX broadly, and (2) assess ATX-based RoR performance within risk groups defined by NATALEE criteria. Methods: We generated ATX scores in data from 2,228 HR+ HER2- patients from five institutions with available H&E slides and complete clinical information. Results were pooled for analysis. Patients received standard of care treatment which did not include CDK4/6i as they were not available at the time of collection. Clinical High-Risk and Low-Risk groups were defined according to NATALEE trial CDK4/6i eligibility criteria. T2N0 patients without available Ki-67 or Oncotype scores were excluded. The ATX model generated 5-year RoR scores, with a primary cut-off at 10% to classify patients into ATX-High (greater or equal than 10%) and ATX-Low (less than 10%) risk groups. The primary endpoint was disease-free interval, evaluated using hazard ratios (HR per 0.1 increase in ATX) and C-index. ATX was not trained with any data used in this analysis. Results: Out of 2,228 HR+ HER2- patients, 918 (41%) were Clinical High-Risk, i.e., eligible for adjuvant CDK4/6i according to NATALEE criteria, and 1,310 were Clinical Low-Risk, i.e., not eligible for adjuvant CDK4/6i by NATALEE. In the Clinical High-Risk group, ATX was prognostic, with a HR of 1.81 (1.58-2.06) and C-index of 0.71 (0.68-0.74). In the Clinical Low-Risk group, ATX was also prognostic with a HR of 2.53 (1.57-4.06) and C-index of 0.72 (0.69-0.75). Among all Clinical Low-Risk patients, the ATX-High subgroup (N=90) had a 5-year recurrence rate of 16%, comparable to 17% recurrence rate in the Clinical High-Risk cohort, suggesting that ATX might identify high-risk CDK4/6i-eligible patients not captured by the current clinical criteria. Conversely, among Clinical High-Risk patients with ATX-Low scores (N=376), the 5-year recurrence rate was 10%, similar to the 6% rate seen across Clinical Low-Risk patients with primary T1N0 disease, suggesting that ATX might identify Clinical High-Risk patients who might not be appropriate candidates for CDK4/6i based on clinical factors alone. Conclusions: ATX score enhances risk stratification in early-stage HR+/HER2− breast cancer patients and could potentially redefine which patients are eligible for adjuvant CDK4/6i relative to the current criteria.
| High-risk group definition (Potential CDK4/6i selection criteria) | Patients in high-risk group | Share of all 5-year recurrences captured in the high-risk group | 5-y recurrence rate in the high-risk group | Patients in low-risk group | 5-y recurrence rate in the low-risk group | ||||||
| Clinical High-Risk (NATALEE eligible) | 918 (41%) | 92/135 (68%) | 14% | 1,310 | 4.3% | ||||||
| ATX ≥6% | 1,165 (52%) | 110 (81%) | 13% | 1,063 | 3.1% | ||||||
| ATX ≥8% | 825 (37%) | 93 (69%) | 16% | 1,403 | 3.8% | ||||||
| ATX ≥10% | 632 (28%) | 79 (59%) | 18% | 1,596 | 4.6% | ||||||
| ATX ≥15% | 358 (16%) | 55 (41%) | 24% | 1,870 | 5.5% |
Presentation numberPS3-06-04
Spatial representation of deep-learning markers show additional prognostic value in breast cancer patients
Constance Boissin, Karolinska Institutet, Stockholm, Sweden
C. Boissin1, J. Hartman2, M. Rantalainen1; 1Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SWEDEN, 2Department of Oncology-Pathology, Karolinska Institutet, Stockholm, SWEDEN.
Introduction: Intra-tumour heterogeneity has been hypothesised to increase risk of recurrence in breast cancer patients but has not been studied systematically. Deep-learning enables systematic extraction of prognostic information from H&E histopathology whole slide images (WSI), however, local tile level features are typically aggregated without spatial context. Research objective: To investigate the spatial distribution of an AI-based prognostic marker (DeepGrade) within tumour areas to identify patterns of spatial heterogeneity . Materials and Methods: 3325 WSIs from resected breast tumours of patients diagnosed in two hospitals in Sweden were used to predict DeepGrade status on a tile-level. Tumour regions were segmented and divided spatially into a tumour front and a tumour centre. For each tumour region, the total number of tiles, and the proportion of high-risk tiles were calculated. Clusters of high-risk tiles (regions of 25 neighboring tiles) were defined in the tumour front as a representation for tumour aggressiveness. Analyses were performed by tumour subtypes (Luminal A, Luminal B, Her2-enriched and Basal-like). Cox Proportional Hazards analyses was used to evaluate prognostic performance with progression free survial as primary endpoint. Results: The proportion of DeepGrade-high tiles in the centre area were higher in Her2+ and basal-like patients than luminal patients, (49.6% vs 12.8% had > 80% of centre tiles DeepGrade-high). In the subgroup of luminal patients (2268 patients), those who had a cluster of DeepGrade-high tiles in the tumour front area had higher recurrence propability with a multivariate hazard ratio of 1.97 (CI: 1.19-3.25; p-value=0.008); and within the subgroup of luminal patients with DeepGrade-low status (1356 patients), having a cluster in the tumour front had a univariate hazard ratio of 3.20, and a multivariate hazard ratio of 1.98 (CI: 1.13-3.49, p-value=0.017) when controlling for age, tumour size, lymph node status, and grade. Conclusions: The presence of at least one cluster of high-risk tiles within the tumour front was found to be an independent prognostic factor. More generally, the spatial distribution of high-risk tumour areas in a histopathology slides can have prognostic implications and should be characterised with greater detail in the future.
Presentation numberPS3-06-05
A Single-Cell Atlas of the Breast Cancer Microenvironment Reveals Subtype-Specific Immune States and Therapeutic Vulnerabilities
Liron Pola Zisman Schachter, Technion, Rappaport Faculty of Medicine, Haifa, Israel
L. P. Zisman Schachter, A. Pinhasi, K. Yizhak; Department of Cell Biology and Cancer Science, Technion, Rappaport Faculty of Medicine, Haifa, ISRAEL.
BackgroundBreast cancer (BC) is the most commonly diagnosed cancer and a leading cause of cancer-related mortality among women worldwide. Despite advancements in detection and treatment, challenges persist due to the disease’s molecular heterogeneity and racial disparities in outcomes.MethodsWe performed a meta-analysis of single-cell RNA-seq (scRNA-seq) data from 376 samples with various stages of BC. The dataset comprises 1.2 million tumor, immune, and stromal cells across the major subtypes: Triple-Negative (TN), Luminal A (LA), Luminal B, and HER2-enriched.ResultsComparative analysis across subtypes revealed distinct immune phenotypes consistent with the “hot” versus “cold” tumor paradigm. In localized disease, TNBC displayed the highest immune infiltration, while LA tumors exhibited immune exclusion. This difference diminished in metastatic lesions. Ligand–receptor analysis highlighted that LA tumors had a higher frequency of anti-inflammatory chemokine signaling than TNBC.In localized LA tumors, we observed enrichment for anti-inflammatory macrophages (MC1), whereas metastases contained more pro-inflammatory macrophages (MC0). The MC0/MC1 ratio correlated with a gene program enriched in SPP1 expression—previously linked to bone metastases. Additionally, we found that estrogen receptor (ER) expression declined in LA metastases, suggesting a loss of ER signaling during disease progression. However, HER2 expression remained high in ER-positive LA cells, comparable to clinically defined HER2-enriched tumors, indicating a subset of HER2+ LA subset.To quantify ER-dependent transcriptional programs, we constructed an ER score based on genes enriched in either ER+ or ER− tumor cells. A high ER+ score was associated with improved overall survival, greater response to endocrine therapy (ET), and reduced immune infiltration. This ER+ program also correlated with fibrotic fibroblasts and MC1 macrophages, pointing to an immune-excluded TME. Notably, part of the ER+ score genes showed chromosomal amplification in ER+ population. Conversely, genes enriched in ER− tumors including MYC were linked to poor prognosis, ET resistance, and an inflamed TME.In TNBC, immune checkpoint expression diverged by disease stage. Primary TNBCs exhibited high PD1 and LAG3 expression, corresponding with improved response to immune checkpoint inhibitor (ICI) therapy in combination with chemotherapy. In contrast, metastatic TNBC showed predominant CTLA4 signaling, suggesting stage-specific immune escape mechanisms. In parallel, we identified EMT-associated tumor programs associated with metastatic progression and poor survival, partly driven by chromosomal amplifications observed in metastatic samples. These programs showed elevated TROP2 expression, nominating them as potential candidates for TROP2-targeted therapies. ConclusionThis study presents the first in-depth single-cell analysis of transcriptional heterogeneity within LA and TNBC tumors. In LA we identified two major tumor cell profiles: ER+ and ER-. These were strongly associated with differences in patient survival and response to ET. MYC was identified as a potential driver of ER- cells and may contribute to therapeutic resistance. The distinct profiles also corresponded with specific TME characteristics: ER+ tumors were linked to immune exclusion and fibrotic stroma enriched in anti-inflammatory macrophages, whereas ER- tumors showed increased immune infiltration and a pro-inflammatory stromal profile. In TNBC, we mapped the IC landscape across disease stages—findings that may guide future ICI trial designs. Finally, EMT-programs with high TROP2 expression nominate a subset of TNBC tumors as potential candidates for targeted therapies such as Sacituzumab govitecan.
Presentation numberPS3-06-06
A comparison of prognostic predictors in node-positive hr+/her2- breast cancer
Marcus Vetter, Cantonal Hospital Baselland, Liestal, Switzerland
E. Chiru1, S. Muenst2, C. Kurzeder3, S. Ebner4, F. Schwab3, B. Seifer1, M. Vetter1; 1University Center for Medical Oncology and Hematology, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 2Institute of Medical Genetics and Pathology, Basel University Hospital, Basel, SWITZERLAND, 3Breast Center, Basel University Hospital, Basel, SWITZERLAND, 4Frauenklinik, Cantonal Hospital Baselland, Liestal, SWITZERLAND.
Background: RxPONDER showed that Oncotype DX (ODX) can assist with adjuvant chemotherapy selection in postmenopausal women, but in premenopausal patients, treatment benefit prediction is unresolved. RSClinN+, an online calculator that adds clinical factors to ODX, was introduced to close this gap. However, RSClin+ lacks independent validation, and requires prior ODX testing, which risks tissue exhaustion, financial burdens, and significant turnaround times. Ataraxis Breast RISK (ATX) is a multimodal artificial intelligence prognostic test that natively combines H&E‑derived morphology with clinicopathologic data and predicts risk of recurrence (RoR). We compared its performance with ATX in a real‑world cohort. Methods: We retrospectively analyzed 112 HR+/HER2- patients with 1-3 positive lymph nodes from Basel University Hospital who previously underwent ODX testing. 12 patients (11%) experienced breast cancer recurrence. RSClinN+ scores were computed with the online tool. ATX scores were generated using a locked model using baseline diagnostic H&E slide and clinical data. Prognostic accuracy of ODX, RSClin+ and ATX was assessed using Harrell’s C-index (concordance index). Results: ATX was significantly associated with overall recurrence (p<0.01) with a hazard ratio of 1.67 (95% CI: 1.44-2.44). In a head-to-head comparison, ATX continuous risk score was more accurate with a C-index of 0.74 (0.61-0.86), compared to 0.67 (0.50-0.84) for RSClinN+ and 0.53 (0.35-0.71) for ODX. In multivariable Cox analysis including both ATX and RSClin, ATX added independent prognostic information (LR χ² = 4.81, p=0.028). Using an exploratory threshold of 10% risk of 5-year recurrence, the high-risk group (N=64) had a recurrence rate of 18.8%, while there were no recurrences in the low-risk group (N=48). Conclusions: ATX and RSClinN+, two multi-modal prognostic tools, were more accurate than ODX alone. ATX had the greatest prognostic accuracy, outperforming both RSClin+ and ODX in a real-world cohort of N+ patients.
Presentation numberPS3-06-07
Aligning AI Recurrence Predictions with Real-world Recurrence Rates Through Survival Calibration
Krzysztof J Geras, Ataraxis AI, New York, NY
K. G. Zeng1, J. Witowski1, J. Cappadona1, B. Machura1, C. Fernandez-Granada2, K. J. Geras1; 1AI, Ataraxis AI, New York, NY, 2Data Science, Center for Data Science, New York, NY.
Background: Prognostic tools such as genomic assays are typically evaluated in whether they can distinguish between high-risk and low-risk patients. However, these scores are uncalibrated, meaning they do not directly correspond to the probability of recurrence, therefore, they lack a straightforward interpretation. As a result, while such tools may be prognostic, translating their outputs into clinical decisions can be challenging. Calibration offers a solution by aligning predicted risks with observed event rates to produce absolute recurrence risk estimates. However, calibration within survival analysis remains challenging due to censoring. In this study, we demonstrate the critical role of calibration in prognostic model development which ensures that scores are aligned with observed event rates. Methods: First, we calibrated a novel AI-based recurrence prediction model using data from 5,351 early-stage breast cancer (BC) patients across 13 cohorts. We evaluated the AI model’s calibration on a hold-out set of 3,457 patients from 5 independent cohorts (separate from the 13 training cohorts). Additionally, we evaluated Oncotype DX recurrence score (ODX RS) calibration according to data based on the SWOG 8814 trial. To calibrate the AI model, we used spline-based methods. During evaluation, we stratified evaluated patients into quartiles based on predicted AI risk. Within each quartile, we compared the median AI score with the observed recurrence rate estimated via Kaplan-Meier estimator. To evaluate ODX RS, patients were divided into quartiles based on RS in our cohorts, and the median score within each quartile was mapped to the corresponding 5-year Disease-Free Survival (DFS) estimate from the SWOG trial. The analysis was performed separately for different treatment regimens (CAF+Tamoxifen vs. Tamoxifen alone) and nodal status (1-3 vs. ≥4 positive nodes). We use Lin’s concordance correlation coefficient (CCC) to quantify calibration for all analyses. Results:Within the 3,457 evaluated patients, while both calibrated and uncalibrated scores showed strong monotonic associations with recurrence (Pearson R-squared = 0.98), the uncalibrated AI model is poorly calibrated, with a CCC of 0.29 (95% CI: [-0.14, 0.63], p=0.19). Model calibration substantially enhanced calibration to a CCC of 0.92 (95% CI: [0.53, 0.99], p=0.001). Among patients with ODX RS, the AI model remained calibrated, with a CCC of 0.90 (95% CI: [0.19, 0.99], p=0.02). In comparison, ODX RS was poorly calibrated, with CCC ranging from 0.13 (for patients with ≥4 nodes, CMF+Tam) to 0.47 (1-3 nodes, Tam only). Conclusion: Calibration enables the model to produce risk estimates that more accurately reflect true recurrence probabilities, enhancing clinical interpretability and utility for treatment decision-making.
| Percentile | Observed recurrence rate | Uncalibrated | Calibrated | ||||||||||||||||
| 0-25th | 2.5% [1.5%-4.1%] | 17.2% [15.1%-18.4%] | 3.7% [2.1%-4.8%] | ||||||||||||||||
| 25-50th | 4.9% [3.5%-6.8%] | 19.9% [18.4%-22.0%] | 6.3% [4.8%-8.6%] | ||||||||||||||||
| 50-75th | 11.5% [9.3%-14.3%] | 24.7% [22.0%-29.0%] | 11.9% [8.6%-17.4%] | ||||||||||||||||
| 75-100th | 19.5% [16.2%-23.4%] | 34.5% [29.0%-77.9%] | 25.5% [17.4%-81.2%] |
Presentation numberPS3-06-08
Somatic variant detection using long read RNA sequencing reveals HLA-restricted targets for immunotherapy
Felipe Batalini, Mayo Clinic, Phoenix, AZ
F. Batalini1, S. Henry2, Y. Zhang3, R. Foster4, J. M. Gayton5, A. Hostal6, E. A. Wilson7, A. Singharoy3, M. Gustafson2, B. Pockaj5, J. Park3, K. S. Anderson3; 1Hematology and Oncology, Mayo Clinic, Phoenix, AZ, 2Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, 3Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 4Hematology and Oncology, Phoenix Children’s Hospital, Phoenix, AZ, 5Surgery, Mayo Clinic, Phoenix, AZ, 6Health Sciences Campus, Creighton University, Phoenix, AZ, 7Icahn School of Medicine, Mount Sinai, New York, NY.
Introduction: Accurate detection and prioritization of tumor-specific somatic variants is foundational to the development of personalized immunotherapies. Traditional pipelines for neoepitope identification rely heavily on short-read next-generation sequencing (NGS) and DNA-based variant calling, followed by RNA expression filtering and HLA-binding prediction. However, this approach is limited by time-intensive workflows, potential false positives from DNA-only calls, and poor capture of complex isoforms or RNA-level editing. Moreover, the accuracy of neoepitope prediction is constrained by conventional sequence-based HLA binding algorithms that do not account for structural and biophysical features of peptide-MHC interactions, particularly across underrepresented HLA alleles. Methods: To overcome these limitations, we developed a streamlined, sequencing-to-vaccine framework that leverages long-read RNA sequencing for direct detection of expressed somatic variants, including fusions, indels, and single nucleotide variants. Using Oxford Nanopore Technologies’ cDNA-PCR approach, we enable comprehensive, full-length transcript characterization from both fresh and frozen breast tumor samples. Our bioinformatic pipeline, NeoTarget, integrates alignment, variant calling, and neoepitope inference from tumor RNA in a single, modular workflow. A core innovation of this pipeline is HLA-Inception, a deep convolutional neural network trained to predict peptide-MHC class I binding using features derived from molecular dynamics-based modeling of electrostatic potential at the peptide-HLA interface, enhancing generalizability across diverse HLA subtypes. Results: Expanding on previous results, the current NeoTarget pipeline identifies, evaluates, and prioritizes single nucleotide variants, indels, and fusions. To validate this system, we used a set of 9 human breast cancer tumors benchmarked with short read sequencing. Identified neoantigens included most variants shown by clinical short read sequencing and also novel structural variants (i.e., fusions) only revealed with long read sequencing with high confidence. Conclusion: Here, we describe the design and performance of the NeoTarget pipeline, and show proof-of-concept feasibility of applying this fast and cost-effective workflow for HLA-restricted epitope discovery in breast tumors using long-read RNA sequencing. Overall, these methods can be applied to rapidly identify and prioritize neoantigens for personalized neoantigen-based immune therapies.
Presentation numberPS3-06-09
Her2-ladder: a spatially interpretable artificial intelligence-driven model for tailored dual-targeted therapy in HER2-positive breast cancer
Xiang-Rong Wu, Fudan University Shanghai Cancer Center, Shanghai, China
X. Wu1, D. Ma1, H. Lv2, S. Zhao1, W. Yang2, Y. Jiang1, G. Di1, Z. Shao1; 1Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 2Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Background Neoadjuvant dual HER2 blockade with trastuzumab and pertuzumab plus chemotherapy represents the current standard of care for HER2-positive breast cancer. However, marked heterogeneity in patient response has intensified discussions on treatment de-escalation for low-risk individuals and alternative therapies for resistant cases. Presently, clinically robust precision oncology tools facilitating such individualized therapeutic strategies remain unavailable. Methods We developed HER2-LADDER (Layered AI-based Dual-targeteD anti-HER2 Recommendation), an AI-driven decision-support framework that integrates clinicopathological information with spatial features derived from routine hematoxylin and eosin (H&E) and HER2 immunohistochemistry (IHC) whole-slide images. The predictive model was trained and tested using patients from the PCbHP arm of the phase II FASCINATE-N trial (NCT05582499) and subsequently validated externally in multiple real-world cohorts (total N = 381). Spatial in situ profiling techniques were employed to elucidate the biological mechanisms underlying the model’s predictive capacity. Results HER2-LADDER achieved outstanding predictive performance, demonstrated by an area under the curve (AUC) of 0.986 in the training cohort and 0.939 in the testing cohort, with consistent external validation results. Based on individualized HER2-LADDER scores, patients were stratified into Low-, Medium-, and High-score groups. These classifications effectively identified suitable candidates for treatment de-escalation (THP or PCbH/TCbH), standard dual-targeted therapy (PCbHP/TCbHP), or alternative regimens, including next-generation anti-HER2 antibody-drug conjugates (ADCs) and tyrosine kinase inhibitors (TKIs), respectively. Spatial analyses revealed key biological determinants underpinning HER2-LADDER predictions, specifically highlighting homogeneous HER2-enriched tumor cell patterns and neutrophil-helper T cell interactions. Conclusions HER2-LADDER offers clinicians a powerful, clinically actionable tool to personalize neoadjuvant HER2-targeted treatments. By accurately predicting individual responses and delineating optimal therapeutic pathways, it substantially advances precision medicine for patients with HER2-positive breast cancer.
Presentation numberPS3-06-10
Pan-cancer ai foundation models yield accurate biomarker and survival predictions in breast cancer
Krzysztof Geras, Ataraxis AI, New York, NY
J. Cappadona, J. Witowski, K. Zeng, J. Park, B. Machura, K. Geras; AI, Ataraxis AI, New York, NY.
Background: The application of machine learning methods to oncology has historically been challenging due to the high resolution of medical imaging modalities and scarcity of downstream supervised data. Over the past years, however, foundation models have enabled the field to bypass these constraints by leveraging self-supervised learning on large quantities of unsupervised imaging data. These foundation models learn effective latent representations for the various morphologies seen throughout the training data. These representations can then be used downstream for supervised machine learning tasks with minimal additional training. Methods: A digital pathology foundation model was trained using self-supervised learning method DINOv2 on 250M patches extracted from 260k whole slide images (WSIs). We evaluated the model on biomarker classification and survival analysis tasks across a total of 17 cohorts covering 12 cancer types. For all evaluations, we kept the foundation model frozen and trained regressors or classifiers on top of mean-pooled embeddings obtained by passing hematoxylin and eosin (H&E)-stained slide patches through the model. No clinical data was used for any evaluation. For all tasks, we perform 5-fold cross-validation and report AUROC for classification tasks and C-index for survival tasks. For classification tasks, we used k-NN (k=20), and for survival tasks, we used the Cox Proportional Hazards model. Results: Downstream models performed particularly well in breast cohorts, with mutation classification AUROCs of 0.70 ± 0.07 (TP53) and 0.71 ± 0.18 (CDH1) in CPTAC-BRCA, and a breast cancer-specific survival (BCSS) C-index of 0.67 ± 0.06 in PLCO-Breast. The models also predicted disease-specific survival (DSS) in other cancer types, including bladder (PLCO) with a C-index of 0.77 ± 0.09 despite only 39 events among 285 unique patients. AI models also predicted PIK3CA mutation status in the CPTAC-GBM cohort (AUROC of 0.73±0.17), TP53 in CPTAC-LUAD (0.63±0.10), and homologous recombination deficiency (HRD) in pan-cancer TCGA cohorts (SARC: 0.80±0.06; STAD: 0.68±0.06; UCEC: 0.67±0.07), and microsatellite instability (MSI) in TCGA-STAD (0.69±0.06). See Table 1 for results for all tasks and cohorts. Conclusions: Pathology foundation models are applicable to a wide variety of tasks across a range of cancer subtypes, even in spite of sparse data and naive model architectures. With the right data, similar methodology could be used to train predictors of recurrence risk, metastasis risk, treatment benefit, and more.
| Cohort | # Patients | # Slides | Task | C-index / AUROC | |||||||||||||||
| PLCO-Bladder | 285 | 483 | DSS | 0.771 ± 0.091 | |||||||||||||||
| PLCO-Breast | 1012 | 2908 | DSS | 0.673 ± 0.057 | |||||||||||||||
| PLCO-Colorectal | 749 | 2762 | DSS | 0.615 ± 0.026 | |||||||||||||||
| PLCO-Lung | 492 | 1521 | DSS | 0.587 ± 0.027 | |||||||||||||||
| PLCO-Ovarian | 227 | 873 | DSS | 0.632 ± 0.012 | |||||||||||||||
| PLCO-Prostate | 1094 | 2966 | DSS | 0.645 ± 0.050 | |||||||||||||||
| TCGA-BLCA | 372 | 452 | DSS | 0.622 ± 0.067 | |||||||||||||||
| TCGA-LUAD | 460 | 558 | DSS | 0.616 ± 0.060 | |||||||||||||||
| CPTAC-BRCA | 120 | 395 | TP53-mut | 0.699 ± 0.074 | |||||||||||||||
| CPTAC-GBM | 99 | 246 | TP53-mut | 0.652 ± 0.108 | |||||||||||||||
| CPTAC-LUAD | 106 | 498 | TP53-mut | 0.629 ± 0.102 | |||||||||||||||
| CPTAC-BRCA | 120 | 395 | PIK3CA-mut | 0.609 ± 0.089 | |||||||||||||||
| CPTAC-GBM | 99 | 246 | PIK3CA-mut | 0.730 ± 0.165 | |||||||||||||||
| CPTAC-BRCA | 120 | 395 | CDH1-mut | 0.711 ± 0.176 | |||||||||||||||
| CPTAC-BRCA | 120 | 395 | AKT1-mut | 0.607 ± 0.101 | |||||||||||||||
| CPTAC-LUAD | 105 | 492 | HRD | 0.693 ± 0.095 | |||||||||||||||
| TCGA-HNSC | 431 | 457 | HRD | 0.614 ± 0.064 | |||||||||||||||
| TCGA-LUAD | 437 | 522 | HRD | 0.580 ± 0.071 | |||||||||||||||
| TCGA-SARC | 237 | 577 | HRD | 0.801 ± 0.056 | |||||||||||||||
| TCGA-SKCM | 400 | 454 | HRD | 0.621 ± 0.105 | |||||||||||||||
| TCGA-STAD | 402 | 428 | HRD | 0.681 ± 0.057 | |||||||||||||||
| TCGA-UCEC | 467 | 543 | HRD | 0.672 ± 0.065 | |||||||||||||||
| TCGA-COAD | 330 | 345 | MSI | 0.597 ± 0.076 | |||||||||||||||
| TCGA-STAD | 358 | 383 | MSI | 0.685 ± 0.064 |
Presentation numberPS3-06-11
A Novel Agent-Based AI System to Unlock Unstructured Data in Synoptic Reports for Advanced Population Health Analysis in Breast Cancer
Steven N Hart, Mayo Clinic, Rochester, MN
S. N. Hart; Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
Background: While synoptic reports have standardized breast cancer data, a wealth of critical information remains locked in unstructured free-text, including nuanced biomarker details and procedural notes. This data is not easily computable, creating a significant barrier to understanding comprehensive population-level statistics and subtle clinical trends. Traditional data extraction methods are often insufficient to capture this complexity. We developed and validated a novel, flexible AI-powered system using an agent-based architecture to extract, normalize, and discretize this valuable unstructured data, enabling previously infeasible population-scale analytics. Methods: Our system employs a modular, agent-based framework where each “agent” is a specialized large language model (LLM) tasked with a specific function in a sequential workflow. The process begins with an initiator agent that parses the synoptic report. Subsequent agents then perform targeted tasks, such as identifying and normalizing tumor characteristics, procedural details, and biomarker information from free-text fields. A final agent aggregates this information into a structured, computable format. To rigorously test our approach, we first developed a synthetic data generator to create realistic, anonymized report snippets with known ground-truth values. We then evaluated the performance of several leading LLMs of various sizes (e.g., gemini, llama3.3, phi4, deepseek-r1, gemma3) on this synthetic dataset. Results: Our early analysis on simulated synoptic reports demonstrated high accuracy in data extraction and normalization. The gemini-1.5-pro-002 model achieved an overall accuracy of 96.9%, with a sensitivity of 99.3%, specificity of 94.3%, and an F1-score of 97.0%. The llama3.3:70b model also showed strong performance with a 98.7% accuracy, (F1: 98.8%, sensitivity: 99.8%, specifically: 97.6%). Gemma3:27b was the least performant model tested (F1: 66.2%), substantially worse than much smaller models (deepseek-r1:1.5b and phi4:14b). A detailed error analysis of false positives and negatives has allowed us to iteratively refine the agent logic. By the time of the symposium, we will have analyzed a large cohort of real, de-identified synoptic reports and will present comprehensive performance metrics and initial findings on population-level trends. Conclusion: This AI-powered, agent-based system offers a powerful and scalable solution to overcome the limitations of structured data in synoptic reports. By accurately extracting and normalizing previously inaccessible free-text information, our approach has the potential to provide clinicians and researchers with near real-time insights into population-level statistics and trends in breast cancer. This will facilitate a more profound understanding of disease patterns and ultimately support evidence-based advancements in patient care and clinical research.
Presentation numberPS3-06-13
Early Prediction of Pathologic Complete Response in Breast Cancer via Graph Network Modeling of Intratumoral and Peritumoral Dynamics on DCE-MRI
Yuyi Tan, Institute of Automation Chinese Academy of Sciences, Beijing, China
Y. Tan1, S. Du2, L. Zhang2, Z. Liu1, J. Tian1; 1CAS Key Laboratory of Molecular Imaging, Institute of Automation Chinese Academy of Sciences, Beijing, CHINA, 2Department of Radiology, The First Hospital of China Medical University, Shenyang, CHINA.
Background: Pathological complete response (pCR) is an important marker associated with outcomes in breast cancer patients receiving neoadjuvant chemotherapy (NAC). However, pCR is confirmed only after surgery through pathological examination, making it difficult to assess treatment efficacy early and adjust therapy in time. Therefore, it is necessary to modelling intratumoral and peritumoral dynamics to predict pCR in early stage. Methods: We collected pre-treatment and early-treatment (one-cycle) DCE-MRI scans from Center A (n=259) and ACRIN 6698 trial (n=227). Center A was divided into training and internal testing sets in 8:2 ratio, and ACRIN 6698 trial served as an external test sets. Tumor regions were segmented and expanded by 5 mm to define the peritumoral regions. To capture intratumoral information, each tumor was partitioned into 100 superpixels, and then were categorize into three habitats using k-means clustering. Then we constructed graph with ten nodes—tumor, peritumoral region, and three habitats at both pre-treatment and one-cycle timepoints. A three-layer Graph Convolutional Network (TriGCN) was employed to model interactions between nodes. Finally, the output was concatenated with clinicopathological characteristics to build an integrated model (TriGCN-C). Results: The results presented the area under the curve (AUC) of the receiver operating characteristic curve. The integrated model (TriGCN-C) demonstrated superior performance, achieving AUCs of 0.85 in the training set, 0.81 in the internal testing set, and 0.79 in the external testing set. These results outperformed those of the clinical model (AUC: 0.69-0.75), as well as the single-timepoint models (AUC: 0.66-0.76) in the testing sets. Although the single-timepoint models outperformed the integrated model in the training set, they suffered from overfitting in the testing sets. In contrast, the integrated model captured the dynamic changes of the tumor during treatment, enabling better generalization and demonstrating robust prediction. Conclusion: Our study shows that DCE-MRI from pre-NAC and early-NAC can predict pCR, assisting in early adjustments of therapeutic strategies. Graph-based models have proven effective in capturing the interactions among different tumor nodes. These findings emphasize the model’s promise for predicting treatment response in clinical practice.
Presentation numberPS3-06-16
Mapping neutrophil architecture with integrative spatial analysis to reveal clinically relevant functional heterogeneity
Yu-Zheng Xu, Fudan University Shanghai Cancer Center, Shanghai, China
Y. Xu, D. Ma, X. Wu, Z. Qiu, L. Dai, H. Zhang, S. Zhao, Y. Xiao, Y. Jiang, Z. Shao; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Mapping neutrophil architecture with integrative spatial analysis to reveal clinically relevant functional heterogeneity Background: Neutrophils are increasingly recognized as a key component of the tumor microenvironment. However, their spatial organization and interactions with other cells remain poorly characterized. The clinical and biological significance of these spatial patterns requires further exploration. Our study aims to identify clinically relevant neutrophil spatial architectures in breast cancer and elucidate their biological underpinnings. Methods: We established a large-scale breast cancer cohort covering all standard therapeutic regimens (adjuvant setting, n = 629; neoadjuvant setting, n = 381), with matched hematoxylin and eosin (H&E)-stained whole slide images (WSIs). We refined an artificial intelligence-based computational pathology framework we had proposed1 to identify neutrophils in WSIs and characterize their spatial patterns (i.e., their spatial relationships with neighboring cell types). Then we studied the correlation between these patterns and patient outcomes. To investigate the biology basis of distinct neutrophil spatial patterns, we performed single-cell spatial transcriptomics profiling (n = 104) and integrative analysis, with functional validation using patient-derived organoids (PDOs). Results: We found neutrophil spatial architectures demonstrated significant prognostic and predictive value, particularly in triple-negative breast cancer (TNBC). Specifically, we identified two clinically relevant patterns in TNBC: (1) Neutrophils in closer proximity to tumor cells predicted improved prognosis, which was validated in independent cohorts. Mechanistically, neutrophils upregulated MHC class I on tumor cells via secreted factors (e.g., TNFα), thereby enhancing CD8+ T cell-mediated recognition and cytotoxicity. This process was further corroborated by evidence from a neutrophil-PDO-CD8+ T cell co-culture system. (2) Neutrophils neighboring immune cells correlated with superior response to neoadjuvant immunotherapy. These neutrophils developed potent antigen-presenting capabilities through concurrent upregulation of MHC class I and II molecules. They primed CD4+ naive T cells and expanded CD8+ effector/memory T cells, thus fostering an immunotherapy-responsive microenvironment. Conclusion: Our study delineates the spatial heterogeneity of neutrophils, mapping their functional plasticity to spatial patterns with clinical relevance. These findings may inform the development of clinically actionable biomarkers for prognosis stratification and treatment response prediction in breast cancer patients. Key Words: Neutrophil; spatial architecture; breast cancer; computational pathology; spatial omics References 1Zhao, S. et al. Single-cell morphological and topological atlas reveals the ecosystem diversity of human breast cancer. Nat Commun 14, 6796, doi:10.1038/s41467-023-42504-y (2023).
Presentation numberPS3-06-17
Prognostic performance analysis of artificial intelligence test and 21-gene assay in premenopausal node-positive HR+ HER2- breast cancer patients
Jailan Elayoubi, Karmanos Cancer Institute, Wayne State University, Detroit, MI
J. Elayoubi1, K. Choucair1, K. Kalinsky2, K. Pogoda3, F. J. Esteva4, K. Zeng5, J. Cappadona5, B. Machura5, K. J. Geras5, J. Witowski5; 1Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 2Division of Hematology & Oncology, Department of Hematology and Medical Oncology, Glenn Family Breast Center, Winship Cancer Institute, Emory University, Atlanta, GA, 3Department of Breast Cancer and Reconstructive Surgery, Maria Skłodowska-Curie National Research Institute of Oncology, Warszawa, POLAND, 4Division of Hematology/Oncology, Northwell Health Cancer Institute, Lenox Hill Hospital, New York, NY, 5AI, Ataraxis AI, New York, NY.
Background: The RxPONDER trial was a landmark study demonstrating that the 21-gene Oncotype DX (ODX) assay can help avoid unnecessary adjuvant chemotherapy in postmenopausal women with 1-3 positive nodes (N+) and an ODX recurrence score (RS) ≤25. However, N+ premenopausal women with RS≤25 showed a significant but variable benefit to adjuvant chemotherapy despite low or intermediate ODX RS. Current NCCN guidelines therefore recommend ODX only as a consideration in this subgroup, leaving risk-adapted decision-making unresolved. Ataraxis Breast (ATX) is a multi-modal artificial intelligence test that integrates morphological features extracted from standard H&E-stained slides with baseline clinical data. Here we validate the prognostic accuracy of ATX in four premenopausal N+ cohorts. Methods: 150 premenopausal HR+/HER2- node-positive cases were extracted from Providence Health (n=48), TCGA (n=59), Basel (n=32) and Karmanos (n=11) cohorts. ATX recurrence scores were generated using a locked model. 43 patients (Basel and Karmanos cohorts) also had ODX RS available. Prognostic performance of ATX and ODX RS was assessed for disease-free interval (DFI) endpoint using Harrell’s C-index and hazard ratio (HR per 0.1 increase in ATX). Reported HR and C-index are pooled using random-effects meta-analysis. Results: In a univariate model, ATX was a significant predictor of DFI in premenopausal N+ women with a hazard ratio of 2.77 (1.53-5.01, p=0.02) and C-index of 0.78 (95% CI: 0.64-0.92). ODX did not reach statistical significance with a HR of 1.05 (1.00-1.10, p=0.05) and C-index of 0.46 (0.32-0.59). In multivariate analysis adjusting for age, tumor size and grade, ATX was the only variable significantly associated with DFI (p<0.01). The estimated 5-year recurrence rates for ATX bottom and top quartiles were 9.1% and 25.7%. When divided into exploratory low/high ATX risk groups with threshold at 10% 5-year risk of recurrence (RoR), the ATX <10% RoR group included 39 patients with an observed recurrence rate of 5%, while ATX ≥ 10% RoR included 111 patients with an observed recurrence rate of 16%. Conclusions: ATX demonstrates promising independent prognostic performance in premenopausal node‑positive breast cancer across diverse datasets. Larger studies are needed to confirm its efficacy and utility.
| Dataset | N total patients in the cohort | N HR+HER2- premenopausal node-positive | N of recurrences in HR+HER- prem. N+ group | C-index (95% CI) | HR (95% CI) |
| Providence | 1687 | 48 | 6 | 0.67 (0.34-0.95) | 3.41 (0.94-12.93) |
| TCGA | 930 | 59 | 5 | 0.90 (0.77-1.0) | 2.91 (1.22-6.93) |
| Basel | 267 | 32 | 6 | 0.67 (0.48-0.86) | 1.96 (0.50-7.64) |
| Karmanos | 162 | 11 | 3 | 0.75 (0.37-1.0) | NA |
Presentation numberPS3-06-18
Deep Learning-guided Spatial Transcriptomics Identifies KRAS signaling as the Dominant Mechanism of Resistance to CDK4/6 Inhibitors
Maki Tanioka, Okayama University, Okayama, Japan
M. Tanioka1, T. Nakayama2, T. Marutani3, Y. Niki3, M. Tohda3, Y. Hirose3, L. Zehao4, T. Daisuke5, K. Morooka4, A. Nakaya6, T. Shien7; 1Clinical Oncology Center, Okayama University, Okayama, JAPAN, 2Medical School, Okayama University, Okayama, JAPAN, 3School of Engineering, Okayama University, Okayama, JAPAN, 4Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, JAPAN, 5Breast Surgery, Shikoku Cancer Center, Matsuyama, JAPAN, 6Graduate School of Frontier Sciences, Tokyo University, Chiba, JAPAN, 7Breast and Endocrine Surgery, Okayama University, Okayama, JAPAN.
Background: Resistance to CDK4/6 inhibitors remains poorly understood in many patients. While ESR1 mutations and PI3K/AKT pathway alterations have been linked to resistance, these explain only a subset of cases and drugs targeting these mutations show short-lived clinical benefit. We hypothesized that both intratumoral heterogeneity and the tumor microenvironment contribute to therapeutic response, and that AI-based pathology and spatial transcriptomics could uncover dominant resistance pathways. Methods: We developed a fine-tuned GigaPath foundation model (Xu et al., Nature, 2024) to predict progression-free survival (PFS), using 511 H&E-stained slides from 178 metastatic breast cancer patients treated with first-line CDK4/6 inhibitors (Palbociclib,108; Abemaciclib,70) plus endocrine therapy. Grad-CAM visualizations identified spatial regions associated with PFS. Spatial transcriptomics (Visium v2, 10x Genomics) was applied to 12 pre-treatment samples to compare gene expression between AI-identified PFS-long (sensitive) and PFS-short (resistant) regions. Additionally, 7 matched pre- and post-treatment tumor pairs underwent spatial transcriptomic profiling using Visium HD. GSEA was performed to analyze enriched pathways. Results: The model achieved an AUC of 0.84 in distinguishing cases with PFS <6 months versus ≥15 months. Grad-CAM identified intratumoral regions associated with sensitivity and resistance in 9 samples. In these regions, GSEA revealed that resistant areas were enriched for KRAS signaling (e.g., KRAS.600.LUNG.BREAST_UP.V1_UP and HALLMARK_KRAS_SIGNALING_UP), whereas sensitive areas were enriched for gene sets associated with suppressed KRAS activity (e.g., KRAS.600_UP.V1_DN) (Table). In the 7 pre- and post-treatment tumor pairs, spatial transcriptomics within tumor regions detected 29 pathways in at least 5 out of 7 tumor pairs. Among them, eight were related to KRAS signaling—including KRAS.LUNG.BREAST_UP.V1_UP and KRAS.BREAST_UP.V1_UP—five were associated with polycomb group proteins (BMI1, MEL18), and five involved tumor suppressor pathways (p53, BRCA1, PTEN, ATM). These results indicate that KRAS signaling was the most frequently enriched pathway following treatment. Conclusion: Two independent spatial transcriptomic analyses-comparing AI-identified regions and treatment timepoints—consistently identified KRAS signaling as the dominant mechanism of resistance to CDK4/6 inhibitors in breast cancer. These findings provide a compelling rationale to explore KRAS pathway inhibition to overcome resistance to CDK4/6 inhibitors in breast cancer.
| Terms in sensitive regions | Count (N=9) |
|
HALLMARK_EPITHELIAL_MESENCHYMAL_TRANSITION
|
7 |
|
GAO_LARGE_INTESTINE_24W_C1_DCLK1POS_PROGENITOR
|
7 |
|
HAY_BONE_MARROW_STROMAL
|
6 |
|
KRAS.600_UP.V1_DN
|
6 |
|
HE_LIM_SUN_FETAL_LUNG_C1_SMG_BASAL_CELL
|
6 |
|
LEF1_UP.V1_UP
|
6 |
|
GAO_LARGE_INTESTINE_ADULT_CJ_IMMUNE_CELLS
|
6 |
|
TRAVAGLINI_LUNG_ALVEOLAR_FIBROBLAST_CELL
|
6 |
| Terms in resistant regions | Count (N=9) |
|
CUI_DEVELOPING_HEART_VALVAR_ENDOTHELIAL_CELL
|
9 |
|
HE_LIM_SUN_FETAL_LUNG_C1_PROXIMAL_BASAL_CELL
|
8 |
|
HE_LIM_SUN_FETAL_LUNG_C1_PROXIMAL_SECRETORY_PROGENITORS
|
8 |
|
KRAS.600.LUNG.BREAST_UP.V1_UP
|
8 |
|
AIZARANI_LIVER_C21_STELLATE_CELLS_1
|
7 |
|
HALLMARK_KRAS_SIGNALING_UP
|
7 |
|
HE_LIM_SUN_FETAL_LUNG_C3_OMD_POS_ENDOTHELIAL_CELL
|
7 |
Presentation numberPS3-06-19
Phylogenetic and Transcriptomic Analyses Reveal Specific Adaptations for Progression of Breast Cancer
Mariano Russo, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA
M. Russo1, P. Raj-Kumar1, J. Liu1, A. Praveen-Kumar1, A. Kovatich2, A. Tan3, H. Hu1, C. Shriver4, X. Lin1; 1Bioinformatics, Chan Soon-Shiong Institute of Molecular Medicine at Windber, Windber, PA, 2Murtha Cancer Center Research Program, The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 3Oncological Sciences & Biomedical Informatics, Huntsman Cancer Institute, Salt Lake City, UT, 4Murtha Cancer Center Research Program, Walter Reed National Military Medical Center, Bethesda, MD.
Introduction: Metastatic spread and cancer evolution in response to treatment represent major obstacles to positive patient outcomes. Preliminary evidence on cancer metastatic progression supports evolutionary selection from primary tumors as a proposed mechanism for metastasis. PanCancer and focused studies have demonstrated an overall pattern of increased genomic alterations concurrent with increased clonality in metastasis compared to primary tumors. This circumstance is likely to arise due to ongoing mutational processes during cancer progression that result in selection for a specific subclone seeding. This selection process will manifest in genomic adaptations such as small mutations, copy number alterations, or epigenetic modifications regulating gene expression and directed towards enhancing tumor survival and proliferation. The genomic composition of seeding clones across tumors may give us unique insight into metastatic progression. Methods: Our study aims at defining networks of progression in metastasis of breast cancer with a total of 2077 patients participating from the ORIEN consortium. The current analysis focused on 112 paired cases of primary and metastasis (regional lymph node or distal). Subclonal deconvolution was used to define seeding clone clusters and reconstruct tumor phylogenies based on PyClone and CONIPHER modeling. Transcriptomics analysis of a subset of cases with successful RNASeq (N=62) was performed to identify differential expression between primary and metastasis using limma. Copy number alterations were identified with ASCAT or Sequenza and analyzed using GISTIC2. dNdS mutation rates were used to model positive selection. Gene Set Enrichment Analysis was used to identify differentially expressed pathways. Results: Our analysis of 112 cases of paired breast tumors has identified specific oncogenes and tumor suppressors, including transcription factors, kinases, and chromatin remodelers, under positive selection for mutation at different stages of progression. These genes include GATA3, TP53, and 4 potentially novel breast cancer driver genes. Copy number analysis identified a unique profile for amplifications and deletions in selection for metastatic progression, including the early acquisition of broad chromosome arm events and focal enrichment. Transcriptomics analysis revealed 5 specific pathways enriched in progression programs such as EMT and immune regulation. Tumor microenvironment and mutation signatures are being analyzed to elucidate the progression networks of breast cancer. Hierarchical clustering and machine learning approaches will be used to model clinical, molecular, and seeding clone features for additional insights into the evolutionary networks that drive metastasis. Conclusion: Our study uses innovations in cancer genomics to determine the subclonal progression into metastasis with a comprehensive genomic and transcriptomic profile. This approach can yield valuable insights into cancer evolution and therapeutic markers. Disclaimer: The contents of this publication are the sole responsibility of the author(s) and do not necessarily reflect the views, opinions, or policies of Uniformed Services University of the Health Sciences (USUHS), the Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., the Department of Defense (DoD) or the Departments of the Army, Navy, or Air Force. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. government.
Presentation numberPS3-06-20
A rapid and accurate diagnostic system based on label-free multimodal multiphoton microscopy and deep learning
Yulei Wang, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
G. Zhang1, Y. Wang1, Y. Zhang2, X. Zhu3, B. Xu3, N. Liao1; 1Department of Breast Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, CHINA, 2Department of Breast Surgery, Guangdong Provincial People’s Hospital’s Nanhai Hospital, Foshan, CHINA, 3/, Femtosecond Research Center (Guangzhou), Guangzhou, CHINA.
Despite the significant growth of computational pathology (CPATH) as a promising solution to improve cancer diagnosis in the past years, limited generalizability due to the technical variabilities introduced during slide preparation and digitization processes is still the single most important obstacle for its implementation into the clinic. To overcome this issue, we develop the first highly generalizable auxiliary diagnostic system integrating slide-free, stain-free multimodal microscopy imaging with specially designed deep learning algorithm. By virtue of standardized information-rich images as input data, a small training set of 130 specimens is sufficient to achieve an area under the curve (AUC) of 0.9934 for breast cancer detection, when tested on 500 independent patient specimens. More importantly, such performance is well maintained in various external tests with less than 1% drop in AUCs. We also demonstrate our system’s great potential for molecular subtyping that traditionally requires immunohistochemical evaluation. High generalizability and small training set size needed warrant this system to significantly improve the applicability of deep learning-based diagnostic systems into the clinic, and help enable precision oncology which relies heavily on rapid, accurate and robust diagnosis.
Presentation numberPS3-06-21
Differential Network Analysis Reveals Pathways Associated with APOBEC3 Mutational Signature in Breast Cancer
Mohadeseh Soleimanpour, texas biomedical research institute, San Antonio, TX 78227, TX
M. Soleimanpour; HPI, texas biomedical research institute, San Antonio, TX 78227, TX.
Differential Network Analysis Reveals Pathways Associated with APOBEC3 Mutational Signature in Breast CancerMohadeseh Soleimanpour1, Diako Ebrahimi1,2,31 Texas Biomedical Research Institute, San Antonio, Texas, USA2 University of Texas Health San Antonio, San Antonio, Texas, USA3 The University of Texas San Antonio, San Antonio, Texas, USAThe human genome encodes seven APOBEC3 enzymes (A3A/B/C/D/F/G/H) known for their ability to mutate DNA. These enzymes primarily function to restrict viral infections by introducing C-to-U mutations in viral genomes. However, certain A3 family members, particularly A3A and A3B, can also mutate cellular DNA, contributing to tumor initiation and progression. Several cancers including breast, bladder, cervical, head and neck, and skin exhibit characteristic C-to-T mutations (SBS2 and SBS13) associated with aberrant A3 enzyme expression or activity. While SBS2 and SBS13 mutational signatures are generally more prevalent in cancers with elevated A3A and A3B transcript levels, this correlation does not consistently hold at the individual TCGA donor level. Many donors exhibit high SBS2/13 levels despite little or no A3A/B mRNA expression, and vice versa. This discrepancy is thought to arise from varying levels of DNA repair among donors and/or the episodic nature of A3 dysregulation. Here, we propose and test a third hypothesis: that changes in the interaction networks of A3 enzymes, rather than their expression levels alone, underlie SBS2/13 mutational signatures. Specifically, we hypothesize that the A3 interactome determines whether tumors undergo A3-induced tumorigenesis in a donor-specific manner. To investigate this, we analyzed 1,082 TCGA breast tumors, grouping them into high and low SBS2/13 categories. Using Parsimonious Gene Correlation Network Analysis (PGCNA), we constructed gene expression networks for each group and performed a differential network analysis. Preliminary findings reveal significant differences between the networks, particularly involving genes such as GATA3, CDH1, and PIK3CA. Our ongoing work aims to extend this analysis across all TCGA tumors to fully explore gene network changes associated with A3 mutational signatures.
Presentation numberPS3-06-22
Artificial Intelligence Predicts OncotypeDX Recurrence Scores Directly from H&E-Stained Whole Slide Images of ER+/HER2- Node-Negative Breast Cancer Surgical Sections
Savitri Krishnamurthy, The University of Texas MD Anderson Cancer Center, Houston, TX
C. Feng1, H. Muhammad2, S. S. Chavan2, D. K. Almaraz3, H. Basu2, W. Huang4, R. Roy2, G. Wilding5, S. Kummar6, S. Krishnamurthy3; 1Computational science, Pathomiq Inc, Cupertino, CA, 2Computational Science, Pathomiq Inc., Cupertino, CA, 3Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 4Pathology, UW School of Medicine and Public Health, Madison, WI, 5Computational Science, Pathomiq Inc., Houston, CA, 6Medical Oncology, OHSU Knight Cancer Institute, Portland, OR.
Title: Artificial Intelligence Predicts OncotypeDX Recurrence Scores Directly from H&E-Stained Whole Slide Images of ER+/HER2- Node-Negative Breast Cancer Surgical Sections Background:Oncotype DX (ODX) is a widely used genomic assay for risk stratification and guiding adjuvant treatment decision making in early stage ER+/HER2- node-negative breast cancer. However, its cost and turnaround time can significantly limit its use, particularly in low-resource settings. We developed a deep learning model to predict the ODX recurrence score directly from H&E-stained whole-slide images (WSIs) of surgically resected ER+/HER2- node-negative invasive breast tumors and externally validated its performance using retrospective real-world clinical cases. Methods: We developed our model using publicly available datasets consisting of WSIs with associated RNA-Seq data. The ODX score was estimated from the expression levels of individual genes included in the ODX assay. A two-step deep learning framework was utilized to predict the estimated recurrence score from WSIs of H&E stained tissue sections of surgically resected breast tumor. First, our internal Phenotype Atlas, a library of pan-cancer morphological features, was used to identify distinct morphological phenotypes across slides. Second, these phenotypes were mapped to the estimated ODX score using weakly supervised learning to associate ODX scores to morphological features, thus generating a slide-level prediction. For external validation, we used WSIs of an independent cohort of 300 early stage ER+/HER2- node-negative breast cancers with available ODX scores and over 10 years of follow-up. Model performance was assessed using the area under the receiver operating characteristic curve (AUC) for high and low ODX categories as defined by thresholds set in the TAILORx clinical trial (High: ODX>25, Low: ODX≤25 when age is greater than 50). In addition, Kaplan-Meier analysis was used to compare patient outcomes based on treatment decisions guided by either ODX or our model.Results: The median age at diagnosis of the 300 patients in our external validation cohort was 53 years (range: 26–82). The ODX recurrence scores ranged from 0 to 71, with 150 patients having an ODX score below 26. Ninety-five patients (32%) were premenopausal. Our AI model predicted ODX scores directly from H&E WSI alone with an AUC of 0.82. When the AI-predicted ODX score was combined with clinical variables including age, tumor size, tumor grade in a linear regression model, performance improved to an AUC of 0.86. Furthermore, we assessed the performance of the model on premenopausal patients and achieved an AUC of 0.91. No significant difference was found in prediction of patient risk of distant metastasis using Oncotype score in comparison to our AI model (log-rank test p=0.9). Conclusions:Our AI model can estimate ODX recurrence scores directly from H&E WSIs with high accuracy. This model has the potential to serve as a low-cost substitute for ODX with a rapid turnaround time that can be utilized for guiding adjuvant therapy decisions in early stage ER+/HER2- node-negative breast cancer patients.
Presentation numberPS3-06-23
Abstracting Lines of Therapy in Breast Cancer using Large Language Models
James C Dickerson, Stanford University School of Medicine, Stanford, CA
J. C. Dickerson1, M. Shaw1, N. Dalal1, M. McClure2, N. Smith1, J. Caswell-Jin1; 1Medicine (Oncology), Stanford University School of Medicine, Stanford, CA, 2NIH Oncology Fellowship, National Institute of Health, Rockville, MD.
Background: Breast cancer research relies on accurate data from the electronic medical record (EMR). Manual chart review is time-consuming and error prone. To address this, we sought to use large language models (LLMs) to extract treatment data for complex patients with breast cancer. If successful, this approach will increase the data available for breast cancer research. Methods: We randomly selected 100 patients who had ≥ 2 lines of treatment and were diagnosed between 2010 – 2022 from an institutional database. To establish a ground truth, the medical oncologists on the team chart reviewed patients using all available data in the EMR. Research coordinators separately abstracted the data for comparison. We extracted the drugs used, the number of therapy lines, treatment intent (curative/palliative), start/stop dates, and the reason for discontinuation. We then developed an open-source pipeline using commercial HIPAA-compliant LLMs. We created text-delimited files containing either (1) the most recent oncology note, (2) Beacon chemotherapy plans, (3) medication lists, or (4) all these materials. We did not provide instructions on source prioritization in the prompt. We evaluated the performance of three LLMs: OpenAI’s ChatGPT-4.1, an older ChatGPT, and Google Gemini 2.5, using cold temperature and default settings with sufficient token windows for a single API call. Outcomes examined were (1) identification of the exact anti-cancer drug(s), (2) the reason for stopping palliative intent therapy, and (3) palliative treatment duration ± 60 days. We report these outcomes as percentages correct, with the oncologist’s chart review as the gold standard. Results: Among the 100 patients, 85% received curative therapy, and 89% received palliative therapy (median lines: 5; IQR, 3-7). The GPT-4.1 model, using a single oncology note, accurately identified all curative lines in 88% of patients (n = 85) and correctly identified 84% (n = 390 lines) of palliative intent lines. These results were similar for the Gemini 2.5 model (92% and 82%), and both outperformed an older version of ChatGPT (82% and 77%) and human research coordinators (78% and 73%). After excluding the final line of treatment, which may have been ongoing at the time of data censoring, GPT-4.1 and Gemini 2.5 identified the reason for stopping palliative intent therapy in 71% and 73% of lines where the model had already correctly identified the drugs. In comparison, the old ChatGPT was correct at 69%, while the research coordinators were correct at 56%. Adding unprocessed medication lists and Beacon plans to the note had a minimal impact on accuracy for all tasks (± 2%). Using Beacon or the med list alone led to poor accuracy in identifying lines of therapy, ranging from 0% to 39%. We reviewed the 5% of cases where all LLMs struggled. These were all patients who had some of their treatment with a different oncologist than the note author (e.g., second opinions). GPT-4.1 average process time per case was 12 seconds; Gemini 2.5 was 63 seconds. The oncologists required 15 – 75 minutes per case. Conclusions: LLMs can rapidly collect data from clinical notes with moderate accuracy, even without hyperparameter tuning, input standardization, or a foundational model. GPT-4.1 was more accurate than the research coordinators for all tasks. Our findings emphasize the importance of carefully considering inputs, prompts, model settings, and pipelines to ensure accurate variable abstraction. Further work is needed before researchers can use unsupervised abstraction at scale.
Presentation numberPS3-06-24
Transcriptomics-guided AI outperforms and expands on OncotypeDX to predict Hormonal therapy response and Chemotherapy benefit from H&E-stained Whole Slide images in ER+/HER2- Breast Cancer
Savitri Krishnamurthy, The University of Texas MD Anderson Cancer Center, Houston, TX
H. Muhammad1, S. S. Chavan1, C. Feng1, D. K. Almaraz2, H. Basu1, W. Huang3, R. Roy1, G. Wilding1, G. Mills4, S. Kummar5, S. Krishnamurthy2; 1Computational Science, Pathomiq Inc., Cupertino, CA, 2Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Pathology, UW School of Medicine and Public Health, Madison, WI, 4Oncology, OHSU Knight Cancer Institute, Portland, OR, 5Medical Oncology, OHSU Knight Cancer Institute, Portland, OR.
Title: Transcriptomics-Guided AI Outperforms and Expands on OncotypeDX to Predict Hormonal Therapy Response and Chemotherapy Benefit from H&E-Stained Whole Slide Images in ER+/HER2- Breast CancerPurposeOncotype DX (ODX) is one of the most widely used genomic tests for risk stratification and prediction of response to hormonal therapy (HT) of early stage ER+/HER2- node-negative breast cancer. However, this test does not provide guidance on the extent of chemotherapy (CT) benefit. We developed an AI-based model that predicts response to HT and failure of HT+CT directly from H&E-stained whole slide images (WSI) of the breast tumor in surgical resections through inferring underlying genomic and signaling pathway alterations directly from morphology. Our model was developed to improve upon the performance of ODX in risk stratification and prediction of distant metastasis in HT treated patients but also extend its clinical utility by predicting HT+CT failure to enable consideration of other therapy options.MethodsOur AI technology leverages cross-modality transfer learning to infer transcriptomic signals from morphological features. We first developed a biomarker for HT-response and combined HT+CT-failure using only genomic data by mapping RNA-Seq to metastasis outcomes. These biomarkers integrated thousands of gene expressions and signaling pathway activation. After validating the predictive accuracy of these genomic biomarkers, we trained a deep learning model to infer the biomarkers directly from H&E WSIs of the invasive breast tumor. For external validation, we used our AI model to predict on H&E-stained WSIs from 300 early stage ER+/HER2- node-negative breast cancer patients including 150 treated with HT and 150 with HT+CT, based on ODX recommendations. For each WSI, we predicted the presence or absence of our HT-response and HT+CT-failure biomarkers, enabling further stratification of patients within their respective groups. Kaplan-Meier survival estimation and log-rank statistical testing were used to measure the magnitude and significance of stratification in the HT and HT+CT treated groups.ResultsThe 150 ER+/HER2-node negative breast cancer patients who were treated with HT alone based on ODX scores ranging from 0 to 25 were stratified with our HT-response biomarker with a hazard ratio (HR) of 0.28 (p < 0.002). Fifteen patients with ODX<20 treated with HT alone metastasized and our AI model identified 11/15 (73%) of these patients as HT-failures. The 150 patients treated with HT+CT based on ODX scores ranging from 26 to 71 were stratified using our CT-failure biomarker with an HR of 2.42 (p < 0.02), identifying 22/35 (65%) patients who metastasized after being treated with HT+CT. In a multivariate analysis, ODX score added prognostic information to the AI- based HT-response biomarker (p=0.04) and age added prognostic information to the AI- based HT+CT-failure biomarker (p=0.03). By integrating signaling pathway alterations together with changes in gene expressions, we were able to utilize these enriched genomic changes using our AI model to identify responders and non-responders to HT and HT+CT. ConclusionTo our knowledge, this is the first AI model that can predict response to HT and HT+CT in early stage ER+/HER2- node-negative breast cancer. Our AI model by itself identified 73% patients with low ODX scores who developed metastasis following HT alone and 63% patients with high ODX who did not benefit from HT+CT. Combining ODX recurrence scores with the output of our AI model improved the prediction of response in HT treated patients.
Presentation numberPS3-06-25
Simulating Personalized Patient Journeys for Early Cancer Detection Using Artificial Intelligence Synthetic Data
Oge Marques, Florida Atlantic University, Boca Raton, FL
N. H. Borges1, L. E. Silva e Oliveira1, I. C. Cazagranda1, C. B. de Albuquerque1, O. Marques2, C. d. Costa3; 1Department of Research, Spesia, Curitiba, BRAZIL, 2College of Engineering Computer Science, Florida Atlantic University, Boca Raton, FL, 3Department of Research, Instituto de Oncologia do Paraná – IOP, Curitiba, BRAZIL.
BACKGROUND: Developing and validating AI/ML models for early cancer detection is significantly hampered by the scarcity, sensitivity, and ethical complexities associated with real patient data. Synthetic data offers a compelling solution, providing a controlled environment for rigorous model development and testing without compromising privacy. Furthermore, Modifiable Risk Factors (MRFs) such as diet, physical activity, alcohol consumption, sleep patterns, and smoking, are critical determinants of cancer risk, including breast cancer. The ability to simulate the dynamic interplay of these factors and their impact on health outcomes is crucial for designing effective personalized prevention strategies. This study aims to establish a robust, privacy-preserving framework for simulating personalized patient journeys to advance early cancer detection, with a particular focus on breast cancer. Our core objectives are: 1) To develop a rich synthetic dataset of patient personas augmented with detailed MRFs and longitudinal health parameters. 2) To enable “what-if” scenario analysis, allowing for the simulation of the impact of behavioral changes and clinical interventions on individual cancer risk trajectories. METHOD: This is a synthetic cohort dataset generated with artificial intelligence, designed to serve as a foundational resource for building persona simulation engines. We initiate persona generation by adapting NVIDIA’s Nemotron-Personas dataset, leveraging its inherent demographic diversity (e.g., age, gender, occupation, geographic distribution) as a robust base. These generic personas are then augmented using Synthea, an open-source synthetic electronic health record (EHR) generator. We specifically utilize Synthea’s capabilities to model realistic, longitudinal patient histories, incorporating a dedicated breast cancer module to simulate disease progression and relevant clinical events over time. Each persona can be customized with a comprehensive set of parameters, including nuanced dietary patterns, specific lifestyle behaviors (e.g., physical activity levels, sleep patterns), social factors, and simulated environmental risks, with a strong emphasis on quantifiable MRFs. This rich data will eventually make it possible to simulate dynamic patient journeys. It will allow researchers to explore complex “what-if” questions by adjusting modifiable risk factors (MRFs) and observing their effects on cancer risk progression. For example, we could model scenarios such as “What if this patient had annual mammogram screenings for the past three years?” or “What if that patient stops smoking today?” RESULTS: A high-fidelity, privacy-preserving synthetic dataset of diverse patient personas with rich, longitudinal health trajectories relevant to breast cancer risk. This dataset serves as a useful testbed for developing and evaluating AI/ML models for early cancer detection. Future work will add “what-if” simulation capabilities that are expected to provide valuable insights into the personalized impact of MRF modifications and early interventions, laying the groundwork for future precision prevention strategies in oncology.
Presentation numberPS3-06-26
Target Proteasome for Immunogenic Synthetic Kill in Triple Negative Breast Cancer
Jinyu Lu, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY
X. Zheng1, D. Zheng1, J. Lu2; 1Genetics, Albert Einstein College of Medicine, Bronx, NY, 2Oncology, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of Medicine, Bronx, NY.
Triple negative breast cancer (TNBC) is the most aggressive subtype in breast cancer and metastatic TNBC (mTNBC) particularly carries a poor prognosis. While immunotherapy has brought exciting progress in treating mTNBC, only 30~40% of mTNBC patients with high PD-L1 expression are eligible and treatment resistance is common. Proteasome inhibitors (such as Bortezomib) have been approved by FDA and revolutionized the treatment of multiple myeloma. Proteasome inhibitors can induce immunogenic cell death and anti-tumor immune response in solid tumors, but their application in unselected breast cancer patients has been limited. Previous studies suggest that proteasome addiction may be a therapeutic vulnerabilities in TNBC. Using a computational approach, we analyzed expression of proteasome subunits in TCGA dataset. Our analysis showed that constitutive proteasome subunit PSMB5 was highly expressed in breast cancer tumor tissues but not in adjacent non-tumor tissues. Importantly, a higher level expression of PSMB5 significantly correlated with worse disease free survival in breast cancer patients in TCGA database (p < 0.001). Examination of breast cancer molecular subtypes showed that patients with high PSMB5 expression carry a worse disease free survival than PSMB5 low expression group, particularly in basal like breast cancers. We examined DepMap drug sensitivity database and found that TNBC cell lines are more sensitive to proteasome inhibitors than ER-positive and HER2-positive breast cancers. Basal-like breast cancer (overlapping with TNBC) is significantly more sensitive to proteasome inhibitor Bortezomib than other molecular subtypes. Analysis of transcriptomic data from TCGA and CCLE across different immune subtypes predicts significant greater sensitivity to Bortezomib in fully inflamed (FI) and stroma-restricted (SR) groups than margin-restricted (MR) and immune desert (ID) group. Furthermore, inhibition of proteasome causes synthetic lethality with oncogenic Ras signaling. Our data suggest that targeting protein homeostasis network by proteasome inhibitors such as Bortezomib may represent a novel approach to treating triple negative breast cancers that are already under heightened endoplasmic reticulum stress.
Presentation numberPS3-06-27
Deep Learning to Predict Pathological Complete Response in Patients Receiving NAC using Pre-treatment Clinical and Imaging Features
Ryan Gifford, The Ohio State University, Columbus, OH
R. Gifford1, J. Hawley2, C. Taylor2, B. Griffith2, A. Cubbison2, S. Beyer3, T. Andraos3, R. Young3, J. Eckstein3, S. Jhawar3, S. Krening1; 1Integrated Systems Engineering, The Ohio State University, Columbus, OH, 2Radiology, The Ohio State Wexner Medical Center, Columbus, OH, 3Radiation Oncology, The Ohio State Wexner Medical Center, Columbus, OH.
Background: Pathological complete response (pCR) after Neoadjuvant Chemotherapy (NAC) is a strong predictor of survival, making its early prediction clinically valuable. Current research is developing machine learning (ML) models that use pre- and post-treatment MRIs to predict pCR non-invasively. However, there is a critical need for models that rely solely on pre-treatment data to help assess if NAC is appropriate. While some ML models have been developed for this purpose, they have substantially lower accuracy compared to models that add post-treatment data. We address this gap by presenting a novel ML architecture that uses only pre-treatment MRIs while nearing the accuracy of methods that use post-treatment MRIs. Our method uses the pre-treatment MRI to predict radiomic features of the post-treatment MRI, which is then substituted for actual post-treatment imaging. We use the I-SPY2 dataset to train and validate our model. Methods: Our method is 3 steps. Step 1. A Convolutional Neural Network (CNN) is trained using both pre- and post-treatment MRIs. This CNN learns the important radiomic features in the MRI to predict pCR.Step 2. A second CNN is trained using the pre-treatment MRI to predict the radiomic features of the post-treatment MRI, essentially predicting the patient’s post-treatment MRI. The CNN predicts the changes in the radiomic features from the pre- to post-treatment MRI, as learned by the first CNN. Step 3. The predicted post-treatment radiomic features from Step 2 are substituted into Step 1, resulting in a model that does not need any post-treatment data. Results: Our method increases accuracy over a CNN that was trained using only pre-treatment MRIs while maintaining 87% of the accuracy obtained by the CNN that uses both treatment time-points. On the ISPY2 dataset, the Area Under the Curve (AUC) for the pre-treatment only CNN is 0.53, the pre- and post-treatment CNN is 0.71, and our proposed method is 0.62. If clinical data (molecular subtype and age) are added as inputs to the model, the AUC for the pre-treatment only CNN is 0.70, the pre- and post-treatment CNN is 0.78, and our proposed method is 0.73, a 3% increase over the model using only pre-treatment MRI. Conclusion: Our study presents a novel deep learning architecture that predicts pCR using only pre-treatment data with an accuracy of 3-9% higher than other pre-treatment methods. This method raises the AUC from 0.53 to 0.62, and up to 0.73 with clinical data, approaching the accuracy of models that use post-treatment data. By eliminating the need for post-treatment scans, our approach could help identify patients who might benefit from alternative or non-operative strategies early in their treatment. Leveraging the robust I-SPY2 dataset, our findings support the potential of radiomics-driven ML models to guide more personalized, less invasive care in breast cancer.
| Pre-Treatment MRI | Pre- and Post-Treatment MRI | Pre- and Estimated Post-Treatment MRI (Our Method) | |||||
| MRI Only | 0.53 | 0.71 | 0.62 | ||||
| MRI, Molecular Subtype, and Age | 0.70 | 0.78 | 0.73 |
Presentation numberPS3-06-28
Identification of APOBEC3 Protein Sectors in Cancer Through Evolutionary Conservation and Co-variation Analysis
Niloofar Haghjoo, Texas Biomedical Research Institute, San Antonio, TX
N. Haghjoo; Host-Pathogen Interaction, Texas Biomedical Research Institute, San Antonio, TX.
The functionality of proteins is intrinsically linked to their structural composition. The primary structure refers to the linear sequence of amino acids. Secondary structures, such as α-helices and β-sheets, arise from hydrogen bonding within the polypeptide backbone. These elements fold into the tertiary structure, defining the protein’s three-dimensional shape. Beyond these traditional classifications, the concept of protein sectors offers a dynamic view of protein functionality. Protein sectors consist of co-evolving amino acid groups, highlighting functional interdependence. Human DNA codes for 11 Cytidine deaminases including 7 APOBEC3 enzymes known for their roles in DNA mutagenesis. These enzymes play a critical role as antiviral proteins but their aberrant activity has been shown to lead to mutations in cellular DNA leading to cancer. APOBEC3 enzymes are multifunctional proteins, however, currently, it is unclear how amino acid residues of these enzymes have co-evolved and whether they form distinct sectors, each responsible for a specific function. The mechanisms underlying functional specificity of members of this enzyme family are also mostly unknown. For example, APOBEC3A and APOBEC3B are localized in the nucleus, and are involved in inhibiting the replication of endogenous elements and exogenous DNA viruses. In contrast, A3D/F/G/H are found in the cytoplasm, primarily targeting RNA viruses, especially retroviruses. It’s plausible that the unique localization of APOBEC3A and APOBEC3B is influenced by a specific sector that enables nuclear localization, possibly through interaction with unique cellular proteins. Here, we introduce a novel application of statistical coupling analysis (SCA) to identify APOBEC3 protein sectors and elucidate their functional roles. SCA identifies pairs of amino acid positions that co-vary across a set of homologous proteins, indicating functional or structural importance. SCA applies spectral analysis to a matrix combining correlation information with sequence conservation, revealing patterns of correlated mutations. The significant co-evolved sections are distinguished by eigenvector decomposition, helping to understand the evolutionary dependencies and functional interdependencies within proteins, providing insights into protein structure and function. Using this methos, we recently analyzed 5000 unique sequences (APOBEC1, APOBEC2, APOBEC3A/B/C/D/F/G/H, APOBEC4, AID) from a diverse range of species. Our analysis identified five distinct protein sectors. Mapping co-evolving sectors onto the APOBEC3A reference structure (PDB ID: 4XXO) revealed distinct functional compartmentalization among key catalytic and DNA-binding residues. Specifically, sector 2 exclusively encompassed the zinc-coordinating triad—His70, Cys101, and Cys103—highlighting its likely role in enzymatic catalysis. In contrast, sector 4 clustered residues Asn57, Trp98, and Tyr130, which are known to mediate substrate DNA recognition and binding. Ongoing analyses aim to characterize additional sectors that may modulate substrate specificity, mutagenic intensity, and/or regulatory interactions.In conclusion, given the multifunctional nature of APOBEC enzymes, it is crucial to discern, using a systematic evolutionary approach, how functions like DNA binding, RNA binding, deaminase activity, cellular localization, and other unknown roles have evolved distinctly in APOBEC proteins, and which amino acids play a part in each functional domain.
Presentation numberPS3-06-29
Retrieval-augmented GPT-4 improves NCCN-concordant breast cancer treatment recommendations
Bradley Callas, Creighton University School of Medicine- Phoenix, Phoenix, AZ
C. Yost1, A. Aseem2, B. Callas1, S. Monick3, L. Bonilla4, R. Nguyen5, R. Galamaga6, S. Osborn7, Y. Kumar8, N. Ertz-Archambault9; 1Internal Medicine, Creighton University School of Medicine- Phoenix, Phoenix, AZ, 2Medicine, University of Illinois Chicago Department: Medicine, Chicago, IL, 3Oncology Hematology, Mayo Clinic Alix School of Medicine- Arizona, Phoenix, AZ, 4Hematology and Oncology, St. Luke’s University Health Network, Easton, PA, 5Internal Medicine, University of Illinois School of medicine, Chicago, IL, 6Oncology Hematology, Barrow Neurological Institute, Phoenix, AZ, 7School of Medicine, St. George’s University School of Medicine, Phoenix, NY, 8Endocrine and diabetes., Liaquat national medical college hospital, karachi, PAKISTAN, 9Internal Medicine, Creighton university School of Medicine- Phoenix, Phoenix, AZ.
Guideline-concordant breast cancer treatment selection is increasingly complex due to rapid evolution in biomarker-driven therapies. Large language models (LLMs), such as GPT-4, offer a potential tool to support clinical decision-making. However, hallucinations and deviation from evidence-based guidelines remain significant concerns. This study evaluated whether retrieval-augmented generation (RAG) using National Comprehensive Cancer Network (NCCN) guidelines improves GPT-4 performance in treatment recommendation tasks. We developed a retrieval-augmented GPT-4 agent (RAG-GPT) by indexing 2025 NCCN Breast Cancer Guidelines into a vector database using sentence embeddings. Forty clinical vignettes were created from published case reports to reflect diverse scenarios, including HER2-low, triple-negative, and BRCA-mutated disease across all stages. Two models—a baseline GPT-4 (without augmentation) and the RAG-GPT—were prompted with structured templates requesting treatment plans, rationale, clinical trial options, and citations.Outputs were independently scored by two blinded oncology-trained reviewers using the modified Generative AI Performance Score (mG-PS). The mG-PS scored outputs for 2 categories, guideline concordance (Gold standard = 1.0, Acceptable = 0.5, Non-concordant = 0.0) and hallucination errors (Severe = -1.0, Moderate = -0.5, Mild = -0.25, None = 0.0). Guideline Concordance was deemed based on NCCN guidelines and what an oncologist would due based on using the guidelines for each case. Hallucinations are a term used for falsehoods in Artificial Intelligence, like made-up facts or studies, incorrect information, improper doses, or inappropriate or wrong treatments. To ensure consistency and minimize inter-rater variability, reviewers were provided with comprehensive scoring guidelines and detailed instructions for each evaluation metric. Scores were normalized between -1.0 and 1.0 for improved evaluation of tools. Readability and rationale clarity were rated on a 5-point Likert scale. Inter-rater reliability was assessed using Cohen’s kappa. The RAG-GPT demonstrated a significantly higher mean mG-PS score (0.599 ± 0.102, 95% CI: 0.498-0.702) compared to the baseline GPT-4, which not only scored lower but had a negative mean (-0.21 ± 0.143, 95% CI: -0.353 to -0.067). For the mG-PS Cohen’s Kappa test resulted in a score of .69 with substantial aggreament. An independent sample t-test showed a difference (t(78) = 29.1, p < 0.0001). Severe hallucinations were also absent in RAG-GPT outputs (0/40) but occurred in 8/40 baseline GPT-4 outputs. The evaluation of readability and rationality also showed notable differences, with the augmented LLM scoring significantly higher (M = 4.35, SD = 0.66) compared to the naïve LLM (M = 3.05, SD = 1.06). An independent samples t-test confirmed this difference t(65.3) = 6.58, p < 0.0001, further supporting the benefit of augmented Large Language models. Cohen’s Kappa test resulted in a score of .76 for a substantial agreement for readability and rational clarity scale. Limitations include small sample size; further fine-tuning and larger real-world validation studies remain necessary. Linking GPT-4 to NCCN guidelines via retrieval-augmented generation significantly improves treatment accuracy and reduces hallucination severity in breast cancer care scenarios. While further refinement is needed, the reduced incidence of severe hallucinations, increase in guideline-concordant recommendations, and enhanced readability findings support further development and prospective validation of guideline-anchored LLMs for clinical use.
Presentation numberPS3-06-30
Mammoscope: a clinically-informed foundation model for high-resolution mammography interpretation
Christoph Sadée, Stanford University, Palo Alto, CA
C. Sadée1, C. Lin1, D. Raymond1, P. Shah2, K. Sangani1, Q. Xu1, N. Hundal3, A. Chun1, T. Onyemeh4, Z. Onah5, A. Ilo5, K. Hartmann6, B. Dashevsky2, I. Okoye5, E. McDonald6, O. Gevaert1; 1BMIR, Stanford University, Palo Alto, CA, 2Department of Radiology, Stanford University, Stanford, CA, 3Department of Computer Science, UC Davis, Davis, CA, 4Medicine, University of Nigeria, Enugu, NIGERIA, 5Department of Radiology, University of Nigeria Teaching Hospital, Enugu, NIGERIA, 6Department of Radiology, University of Pennsylvania, Philadelphia, PA.
Artificial intelligence (AI) models in breast imaging have shown promise in automating classification tasks but often lack the nuanced decision-making radiologists apply in clinical practice. We developed a high-resolution vision transformer-based foundation model trained on over 650,000 mammograms from 14 public datasets and validated on internal cohorts. The model, termed MammoScope, incorporates radiologist-informed approaches such as bilateral comparison, priors, and lesion-context reasoning using image registration and multiview analysis. Self-supervised pretraining was performed using the DINO framework for improved performance, followed by fine-tuning on downstream tasks including benign versus malignant classification, BI-RADS prediction, and lesion subtype stratification.
Presentation numberPS3-07-01
Ki67 change between diagnostic biopsy and surgery: impact of hormonal replacement therapy or contraception (HRT-HC) withdrawal at breast cancer diagnosis
Khalil Zaman, Lausanne University Hospital CHUV, Lausanne, Switzerland
L. Fontannaz1, J. Sauser2, B. Bisig3, N. Dris1, A. Kakourou1, B. Giacomuzzi-Moore1, A. Stravodimou1, A. Liapi1, C. Perrinjaquet1, A. Dolcan1, B. Wolf1, E. Dubruc3, D. Hastir3, K. Zaman1; 1Department of Medical Oncology, Lausanne University Hospital CHUV, Lausanne, SWITZERLAND, 2Clinical Trials Unit, Lausanne University Hospital CHUV, Lausanne, SWITZERLAND, 3Institute of Pathology, Lausanne University Hospital CHUV, Lausanne, SWITZERLAND.
Background: Change in Ki67 expression between diagnostic biopsy and surgery of a breast cancer (BC) – the so-called window of opportunity – is commonly used as a surrogate marker of treatment response or to assess endocrine sensitivity as in the WSG-ADAPT HR+/HER2- trial. HRT-HC were shown to increase BC risk and are typically discontinued at BC diagnosis. Whether this withdrawal alone influences Ki67 dynamics remains unclear and may represent a potential confounding factor in the window of opportunity studies. Methods: This retrospective study included 382 female patients (pts) who underwent primary surgery at our breast center over a 4-year period (2020-2023). Clinical and tumor characteristics, including Ki67 levels at biopsy and surgery, were extracted from our institutional database. The primary objective was to compare the change in log Ki67 levels between the initial biopsy and the surgical specimen in pts with vs. without HRT-HC in HR+/HER2- BC (n=308). For this comparison, a linear regression adjusting for log Ki67 level at biopsy, HRT-HC use, age, menopausal status, year, stage, grade, expression levels of progesterone (PgR) and estrogen receptors, and histology was considered using the ratio of geometric means as an estimate of the effect of hormonal treatment. Results: Among patients with HR+/HER2- BC, 38% were premenopausal and 62% postmenopausal. At diagnosis, 27.9% of patients were receiving HRT-HC (39.3% of premenopausal and 20.9% of postmenopausal women). The distribution of Ki67 levels remained stable across the four years of assessment. Ki67 level was significantly correlated with the BC grade (1 vs. 2 vs. 3) and stage (I vs. II-III), at biopsy and at surgery. The correlation with menopausal status was significant only at surgery. No correlation was observed with PgR < vs. > 20%. Median time from biopsy to surgery was 39 days. Ki67 level was 9.2% lower at surgery than at biopsy (p=0.025). There was no significant difference in terms of Ki67 level at biopsy in pts with HRT-HC vs. without. However, the geometric mean of Ki67 at surgery for pts with HRT-HC was 29% lower than for pts without HRT-HC, adjusted for Ki67 level at biopsy (p=0.007). They also had a higher likelihood to present a Ki67 level <10% at surgery (estimate 0.52, p=0.015). This difference stayed significant in multivariable analysis (p<0.001). Conclusion: HR+/HER2- BC pts who interrupt HRT-HC upon BC diagnosis present a statistically significant drop in Ki67 level between diagnostic biopsy and surgery; a change which is not observed in patients HRT-HC naive. A significantly higher proportion of pts with HRT-HC also achieved a Ki67 level below 10% at surgery, a commonly used cutoff for decision. Thus, HRT-HC may act as a confounder factor when administrating a window of opportunity treatment to guide adjuvant systemic treatment or to predict a treatment efficacy based on Ki67 levels. These findings and their potential clinical relevance warrant validation in a larger multicenter cohort.
Presentation numberPS3-07-02
An Exploratory Clinical Trial of CDK4/6 Inhibitor Dalpiciclib Combined with Aromatase Inhibitors as Neoadjuvant Therapy for Stage II-III HR-positive HER2-negative Breast Cancer
Qingmo Yang, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
Q. Yang1, W. Chen1, S. Chen1, M. Meng2, Q. Yang1, G. Lin3, L. Lin4, Z. Zheng5, R. Zeng6, B. Chen7, W. Li2, Q. Zhao5; 1Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, CHINA, 2Department of Breast Surgery, The Affiliated Zhongshan Hospital of Xiamen University, Xiamen, CHINA, 3Department of the Thyroid and Breast Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, CHINA, 4Department of Breast Surgery, Affiliated Hospital of Putian University, Putian, CHINA, 5Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Xiamen, CHINA, 6Department of Medical Oncology, The First Affiliated Hospital of Xiamen University, Xiamen, CHINA, 7Department of Thyroid and Breast Surgery, Hongai Hospital, Xiamen, CHINA.
An Exploratory Clinical Trial of CDK4/6 Inhibitor Dalpiciclib Combined with Aromatase Inhibitors as Neoadjuvant Therapy for Stage II-III HR-positive HER2-negative Breast Cancer BackgroundHormone receptor positive (HR+) / human epidermal growth factor receptor 2 negative (HER2-) breast cancer accounts for ~60% of all breast cancer cases. Neoadjuvant endocrine therapy (NET) offers comparable objective response rate (ORR) and breast-conserving rate to chemotherapy but with lower toxicity. Dalpiciclib, a cyclin-dependent kinase 4/6 inhibitor (CDK4/6i), arrests the cell cycle to suppress tumor proliferation. This trial evaluates its combination with aromatase inhibitors (AIs) in stage II-III HR+/HER2- breast cancer, seeking an optimized, low-toxicity neoadjuvant strategy. MethodsThis single-arm, multicenter, open-label phase II trial enrolled 45 treatment-naive women with stage II-III HR+ (estrogen receptor >10%) and HER2- breast cancer. Participants receive dalpiciclib 150 mg/d (d1-21 every 28 days) + AIs (letrozole 2.5 mg/d, anastrozole 1 mg/d, or exemestane 25 mg/d); premenopausal women add ovarian suppression (goserelin/leuprolide). The primary endpoint was ORR. Secondary endpoints encompassed pathological complete response (pCR, ypT0/is ypN0), Ki-67 reduction from baseline, preoperative endocrine prognostic index (PEPI), Miller/Payne (MP), and safety. ResultsFrom March 2023 to June 2025, 45 patients were enrolled. Median age was 57 years (28-79). Among the subjects, 48.8% (21/45) were pre/perimenopausal women. Most patients had T2 (55.6%), N1 (53.3%), Stage III (53.3%) tumors. 24 completed the neoadjuvant treatment and underwent surgery (8 patients are still receiving treatment, 12 patients withdrew from the study, and 1 patient showed progression). ORR was 46.9% (95% CI: 29.6%-64.2%), with 32 patients having completed at least one cycle of treatment and undergone efficacy evaluation, including 3.2% (1/32) complete response (CR) and 45.2% (14/32) partial response (PR). No patient achieved pCR. Ki-67 decreased from a baseline of 20% (IQR: 10%-40%) to 5% (IQR: 3%-26.25%) before surgery, indicating suppressed tumor proliferation; the interquartile range may suggest heterogeneous patient responses. The median MP grade was 3 (IQR: 2.75-3), with the majority (70.8%) showing grade 3, indicating moderate pathological response to therapy. Median PEPI was 4 (IQR: 3-5), and 33.3% (8/24) patients were classified as medium-risk (1≤PEPI≤3). Grade ≥3 adverse event was neutropenia (35.2%), with no treatment-related deaths. Conclusion Dalpiciclib combined with AIs demonstrates promising efficacy and manageable toxicity as neoadjuvant therapy for stage II-III HR+/HER2- breast cancer, offering a new chemotherapy-free option. Future research should focus on biomarker-guided patient selection, long-term outcomes assessment, and comparative trials with other neoadjuvant therapies, aiming to improve the treatment paradigm for HR+/HER2- breast cancer.
Presentation numberPS3-07-03
Evaluating Nodal Burden in T1a-T2 HR+HER2- Breast Cancer: Implications for SLNB Omission in Patients ≥50 Years
Yerin R Lee, University of Toronto, Toronto, ON, Canada
Y. R. Lee1, D. W. Lim2; 1Faculty of Medicine, University of Toronto, Toronto, ON, CANADA, 2Department of Surgery, Women’s College Hospital, Toronto, ON, CANADA.
Purpose: The recent INSEMA and SOUND randomized trials support sentinel lymph node biopsy (SLNB) omission in postmenopausal patients ≥50 years with clinically node-negative T1-T2 hormone receptor-positive (HR+) HER2- breast cancer and negative axillary ultrasound. We used real-world data to evaluate pathologic nodal positivity rates by tumor size for HR+HER2- breast cancer to assess the appropriateness of SLNB omission. Methods: We performed a population-based retrospective cohort study using ICES administrative databases. Patients diagnosed with T1-T2 HR+HER2- invasive breast cancer from 2000-2019 in Ontario, Canada were identified through the Ontario Cancer Registry. Demographic, tumor, and treatment data were linked. Nodal positivity rates across T1a-T2 were calculated for the full cohort and stratified by age group (<50, 50-69, ≥70). Multivariable logistic regression adjusting for age, T stage, and hormone receptor subtype was performed using SAS®. Results: There were 30,969 HR+HER2- tumors, including 4895 (15.8%) patients <50 years; 16,987 (54.9%) between 50-69 years; and 9087 (29.3%) ≥70 years. Among patients aged <50, 1944 (39.7%) were node-positive, most of whom (1590, 81.8%) had N1 disease, followed by 278 (14.3%) N2 and 76 (3.9%) N3. By T stage, there were 21 (1.1%) T1a, 75 (3.9%) T1b, 582 (29.9%) T1c, and 1266 (65.1%) T2 tumors with nodal involvement. The majority of N2 and N3 disease occurred in T2 tumors [212 (76.3%) N2; 60 (78.9%) N3], followed by T1c tumors [60 (21.6%) N2; 15 (19.7%) N3]. Among patients aged 50-69, 4756 (28.0%) were node-positive, of whom there were 3905 (82.1%) N1, 634 (13.3%) N2, and 217 (4.6%) N3 disease. By T stage, there were 45 (0.9%) T1a, 306 (6.4%) T1b, 1775 (37.3%) T1c, and 2630 (55.3%) T2 tumors with nodal involvement. The majority of N2 and N3 disease occurred in T2 tumors [463 (73.0%) N2; 168 (77.4%) N3], followed by T1c tumors [151 (23.8%) N2; 39 (18.0%) N3]. Among patients aged ≥70, 2295 (25.3%) were node-positive, of whom there were 1890 (82.4%) N1, 298 (13.0%) N2, and 107 (4.7%) N3 disease. By T stage, there were 10 (0.4%) T1a, 100 (4.4%) T1b, 755 (32.9%) T1c, and 1430 (62.3%) T2 tumors with nodal involvement. The majority of N2 and N3 disease occurred in T2 tumors [216 (72.5%) N2; 92 (86.0%) N3], followed by T1c tumors [75 (25.2%) N2; 13 (12.1%) N3]. In our cohort, SLNB was omitted in 288 (3.2%) patients over age 70. On multivariable logistic regression, tumor size was the strongest predictor of nodal positivity. Compared to T1a tumors, the odds of nodal involvement were significantly higher for T1b (OR 1.9, 95% CI 1.5-2.5, P<0.01), T1c (OR 6.0, 95% CI 4.8-7.6, P<0.01), and T2 tumors (OR 17.0, 95% CI 13.4-21.5, P<0.01). Age was also inversely associated with nodal positivity. Compared to patients <50, the odds of nodal involvement were significantly lower for those ≥50 years (OR 0.7, 95% CI 0.7-0.8, P<0.01) and those ≥70 years (OR 0.6, 95% CI 0.5-0.6, P<0.01). Hormone receptor subtype was also associated with nodal positivity. Compared to ER+PR+ tumors, the odds of nodal involvement were significantly lower for ER+PR- (OR 0.9, 95% CI 0.8-1.0, P=0.02) and ER-PR+ (OR 0.7, 95% CI 0.5-0.9, P=0.01). Conclusion: Rates of nodal positivity are not insignificant for early-stage breast cancer. In women over the age of 50 where SLNB omission may be considered, rates of nodal positivity increased from 5% for pT1a lesions to 25% for pT1c lesions. Most of the nodal burden was N1 disease. This suggests that SLNB omission would not have impacted systemic therapies, as most of these patients would receive genomic assay testing. However, radiotherapy decisions would be impacted as the identification of nodal disease is an indication for locoregional radiation. These findings need to be considered in the era of surgical and radiotherapy de-escalation.
Presentation numberPS3-07-04
Neoadjuvant chemotherapy combined with Toripalimab for HR+/HER2- breast cancer : a prospective, single-arm, multi-center clinical study (NEOTORCH-BREAST01)
Zhijun Dai, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
Y. Chen1, G. Zhong1, L. Chen1, J. Ma1, H. Cui1, B. Wei1, W. Shao1, Y. Wang2, P. Fu1, Z. Dai1; 1Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, CHINA, 2Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, CHINA.
Background:Hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) exhibits low pathologic complete response (pCR) to neoadjuvant therapy, urgently requiring more effective alternatives. The phase III trials KEYNOTE-756 and CheckMate-7FL demonstrate the potential of neoadjuvant immunotherapy combined with chemotherapy in HR+/HER2- breast cancer. Here we aim to evaluate the efficacy and safety of toripalimab combined with neoadjuvant chemotherapy plus adjuvant toripalimab in HR+/HER2- BC, with the goal of providing more treatment options and potential theoretical support for precision therapy in breast cancer.Method:This is a prospective, single-arm, phase II clinical trial. Eligible patients are women aged 18-75 years, treatment-naïve, with histologically confirmed invasive breast cancer at stage cT1c-3/N0-3M0 and HR+/HER2- status. Following enrollment, patients will receive neoadjuvant treatment consisting of toripalimab (240 mg, Q3W) combined with epirubicin (100 mg/m²) or liposomal doxorubicin (35 mg/m²), each administered with cyclophosphamide (600 mg/m²) for 4 cycles, followed by nab-paclitaxel (260 mg/m², Q3W) for 4 cycles.Surgical treatment will be performed 2-4 weeks after completion of neoadjuvant therapy. After definitive surgery, patients will receive adjuvant treatment with toripalimab (240 mg, Q3W) for 8 administrations, combined with endocrine therapy (ET).The primary endpoints are pathologic complete response (pCR) and residual cancer burden (RCB) 0-1. Secondary endpoints include safety, event-free survival (EFS), progression-free survival (PFS), and disease-free survival (DFS).Results:As of July 6, 2025, 37 patients had been enrolled in the study. The median age was 52 years. Of all patients, 40.5% (15/37) were premenopausal, 73% (27/37) had node-positive disease, 81.1% (30/37) had clinical stage II, 18.9% (7/37) had clinical stage III. 73.0% (27/37) had ER expression≥50%, 37.8% (14/37) had PR expression≥50%, and 89.2% (33/37) had Ki67 expression≥15%, 37) .Among the 29 patients completed neoadjuvant therapy and underwent surgery, the current pCR was 44.9% (13/29), and the postoperative RCB0/1 was 65.5% (19/29). Subgroup analyses among the 29 patients who and underwent surgery further showed: 31.3% (5/16) patients in ER≥90 subgroup reach pCR. For CPS (Combined Positive Score) within this operated cohort, 1 patient with CPS < 1 did not achieve pCR but reached RCB0/1; among 7 patients with 1 ≤ CPS < 10, none achieved pCR and only 1 reached RCB0/1; in 15 patients with CPS ≥10, 11 achieved pCR and all 15 attained RCB0/1. For TILs (Tumor-Infiltrating Lymphocytes) in the same operated group, among 11 patients with TILs < 10, 2 patients achieved pCR and 4 patients reached RCB0/1; in 8 patients with 10 ≤ TILs < 30, 3 achieved pCR and 5 reached RCB0/1; in 6 patients with TILs ≥30, 5 achieved pCR and all 6 attained RCB0/1. 65.5% (19/29) of patients experienced at least one treatment-related adverse event (TRAE). Grade 3 or higher TRAEs occurred in 37.9% (11/37) of patients.Conclusion:Preliminary results indicate that toripalimab-based neoadjuvant chemotherapy achieves favorable efficacy signals in HR+/HER2- breast cancer, with a safety profile consistent with prior immunotherapy studies. These data support the potential of this regimen to expand therapeutic options for patients with HR+/HER2- BC, and long-term follow-up will further clarify its impact on survival endpoints including EFS, PFS, and DFS.Clinical trial information: NCT06611813.
Presentation numberPS3-07-05
Clinical and Demographic Factors Associated with Discordant Oncotype (RS) and MammaPrint (MP) Scores in Patients with Hormone-Positive (HR+) Breast Cancer (BC). An Analysis of the National Cancer Database (NCDB)
Devashish Desai, SUNY Upstate Medical University, Syracuse, NY
D. Desai1, C. Widholm2, P. Ashok Kumar1, E. Hill2, S. Sammons3, A. Sivapiragasam4; 1Department of Hematology and Medical Oncology, SUNY Upstate Medical University, Syracuse, NY, 2Department of Biostatistics Shared Resources, Medical University of South Carolina, Charleston, SC, 3Department of Hematology and Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 4Department of Hematology and Medical Oncology, Medical University of South Carolina, Charleston, SD.
Background: Patients with early-stage ER-positive breast cancer frequently undergo genomic assays like RS or MP to guide chemotherapy decisions. A discordance between these tests is observed, yet no prospective head-to-head trials exist to determine which assay is more predictive, posing challenges for clinicians. This study evaluates the clinical and pathological features of patients with discordant results and explores associated factors. Methods: We utilized the 2022 NCDB to include non-metastatic female breast cancer patients over the age of 18 who are hormone receptor-positive (HR+), HER2-negative, N0-1 with tumor stage T1-4, and who underwent both RS and MP. From this cohort, we compared the characteristics of patients with concordant results to those with discordant results. Within the discordant group, we identified two subgroups: High MP/Low RS, and High RS/Low MP. Tumor and patient characteristics were assessed using univariate analysis. Results: Out of 4,231,162 breast cancer patients, 2,014 patients underwent both MP and RS, of whom 35.25% (N=710) exhibited discordant results. Cases with discordant results were more likely to be high grade (grade 3: 21% vs. 17%; grade 2: 63% vs. 60%; p<0.001), have ductal histology (85% vs. 79%, p<0.001), and N0 disease (70% vs. 64%, p=0.005), and were more frequently Black (10% vs. 7.1%, p=0.011) compared to cases with concordant results. More patients with discordant results received chemotherapy compared to those with concordant results (41% vs. 24%; p<0.001). Of the 710 discordant cases, 92.1% (n=654) had high MP/low RS, while 7.9% (n=56) had high RS/low MP. Compared to the high RS/low MP group, the high MP/low RS cases were more likely to be grade 3 (22% vs. 9.1%, p=0.016), have ductal histology (87% vs. 65%, p<0.001), and include a higher proportion of premenopausal patients (34% vs. 16%, p=0.005). Chemotherapy utilization was equal at 41% in both discordant groups (p=0.96). Factors predicting chemotherapy utilization in the high MP/low RS group included median oncotype score (20 vs. 17), premenopausal status (49% vs. 23%), ductal histology (91% vs. 84%), T2 stage (41% vs. 26%), N1 stage (46% vs. 19%), and grade 3 disease (33% vs. 15%). Conclusion: This analysis revealed that the discordance rate between RS and MP is approximately 35%, with 92.1% of discordant cases exhibiting high MP and low RS. Chemotherapy utilization in both discordant groups is equal, indicating that providers are not making decisions based on one assay over the other. However, clinical factors such as premenopausal status, higher T stage, N1 disease, and grade 3 tumors influence chemotherapy decisions in discordant cases. Our study lacks survival data analyses due to a limited number of events.
| Variable | Overall (N=2014) | Discordant (N=710) | Concordant (N=1304) | p-value | |||||
| Race | 0.011 | ||||||||
| Black | 162 (8.1) | 71 (10) | 91 (7.1) | ||||||
| White | 1691 (85) | 577 (82) | 1114 (87) | ||||||
| Other | 135 (6.8) | 57 (8.1) | 78 (6.1) | ||||||
| Median (Q1, Q3) Oncotype Score | 17.0 (11.0, 23.0) | 19.0 (14.0, 23.0) | 16.0 (10.0, 25.0) | <0.001 | |||||
| Histology | <0.001 | ||||||||
| Ductal | 1591 (81) | 583 (85) | 1008 (79) | ||||||
| Ductal and Lobular | 95 (4.8) | 20 (2.9) | 75 (5.9) | ||||||
| Lobular | 278 (14) | 79 (12) | 199 (16) | ||||||
| Chemotherapy | <0.001 | ||||||||
| Did not Receive | 1409 (70) | 416 (59) | 993 (76) | ||||||
| Received | 605 (30) | 294 (41) | 311 (24) | ||||||
| T stage | 0.27 | ||||||||
| T1 | 1305 (65) | 460 (65) | 845 (65) | ||||||
| T2 | 637 (32) | 232 (33) | 405 (31) | ||||||
| T3 | 68 (3.4) | 17 (2.4) | 51 (3.9) | ||||||
| T4 | 3 (0.1) | 1 (0.1) | 2 (0.2) | ||||||
| N stage | 0.005 | ||||||||
| N0 | 1305 (66) | 489 (70) | 816 (64) | ||||||
| N1 | 678 (34) | 211 (30) | 467 (36) | ||||||
| Grade | <0.001 | ||||||||
| 1 | 405 (20) | 108 (15) | 297 (23) | ||||||
| 2 | 1213 (61) | 445 (63) | 798 (60) | ||||||
| 3 | 376 (19) | 151 (21) | 225 (17) |
Presentation numberPS3-07-06
Real-world Prognostic Evaluation of Adjuvant Endocrine Therapy, Ovarian Function Suppression, and Chemotherapy in Premenopausal HR-positive/HER2-negative Early Breast Cancer in Japan: The PEACE Study
Tatsunori Shimoi, National Cancer Center Hospital, Tokyo, Japan
T. Shimoi1, K. Narui2, K. Kataoka3, S. Orihara 4, K. Kida 5, R. Nakamura 6, K. Adachi7, Y. Horimoto8, M. Oshi9, A. Yamada9, K. Matsumoto10, J. Tsurutani11, Y. Kajiura5, H. Nogi12, S. Akashi-Tanaka13, Y. Hasegawa14, K. Wakita15, T. Kubota16, M. Taguri4, T. Ishikawa7; 1Department of Medical Oncology, National Cancer Center Hospital, Tokyo, JAPAN, 2Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Kanagawa , JAPAN, 3Center for Translational Research, The Institute of Medical Science Hospital, The University of Tokyo, Tokyo, JAPAN, 4Department of Health Data Science, Tokyo Medical University, Tokyo, JAPAN, 5Department of Breast Surgical Oncology, St. Luke’s International Hospital, Tokyo, JAPAN, 6Department of Breast Surgery, Chiba Cancer Center Hospital, Chiba, JAPAN, 7Department of Breast Surgical Oncology, Tokyo Medical University Hospital, Tokyo, JAPAN, 8Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, JAPAN, 9Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Kanagawa, JAPAN, 10Division of Medical Oncology, Hyogo Cancer Center, Hyogo, JAPAN, 11Advanced Cancer Translational Research Institute, Showa Medical University, Tokyo, JAPAN, 12Department of Breast and Endocrine Surgery, The Jikei University School of Medicine, Tokyo, JAPAN, 13Department of Breast Surgery, Tokyo Women’s Medical University, Tokyo, JAPAN, 14Department of Breast Surgery, Hachinohe City Hospital, Aomori, JAPAN, 15Department of Breast Surgery, Yodogawa Christian Hospital, Osaka, JAPAN, 16Department of Breast Surgery, Kamiiida Daiichi General Hospital, Aichi, JAPAN.
Background: Premenopausal women with hormone receptor-positive, HER2-negative early breast cancer (HR+HER2- EBC) exhibit higher incidence rates in Asia than in Western populations. Chemotherapy (CT) is routinely recommended for premenopausal node-positive patients, while it remains unclear whether its apparent benefit derives primarily from ovarian function suppression (OFS). We therefore evaluated long-term outcomes of adjuvant endocrine therapy (ET) alone, ET plus OFS, and ET plus chemotherapy (±OFS) in a nationwide Japanese cohort, stratified by nodal status. Methods: We retrospectively analyzed 7,396 premenopausal HR+HER2- T1-4N0-1 patients who underwent surgery at 25 Japanese centers between January 2008 and December 2017. Patients were divided into node-positive (N1; n=2,041) and node-negative (N0; n=5,355) cohorts. Three treatment groups were defined: ET alone, ET+OFS, and ET +chemotherapy (±OFS). Survival probabilities of eight-year disease-free survival (DFS) and overall survival (OS) were estimated by Kaplan-Meier methods; hazard ratios (HRs) for treatment groups were estimated using Cox models with inverse probability of treatment weighting (IPTW) adjusted for age, tumor size, grade, receptor status, lymphovascular invasion, systemic treatment pattern, surgery type, and radiotherapy. Results: Median follow-up was 8.4 years. In the overall population (n=7,396), 8-year DFS and OS were 95.2% and 98.0%, respectively. Among N0 cohort (n = 5,355), 68.4% ≦T1 and 31.6% ≧T2. Histological grade 3 comprised 12.7%. ER positive in 82.3%; PgR positive in 75.1%. N0 patients’ 8-year DFS was 97.1% (95% CI 96.6-97.6), and 8-year OS 98.8% (95% CI 98.4-99.2), with no significant DFS benefit for ET+OFS or CT versus ET alone. Among N1 cohort, 42.1% ≦T1 and 58.8% ≧T2. Histological grade 3 comprised 28.5%. N1 patients’ 8-year DFS was 90.4% (95% CI 88.7-92.1), and 8-year OS 96.0% (95% CI 94.9-97.1). Compared with ET alone, ET+OFS conferred a significant DFS reduction, whereas CT did not reach significance. OS differences were non-significant in both subgroups. In a Cox model for DFS in N1 patients, the following factors were independently associated with statistically favorable DFS; ET+OFS treatment pattern (vs ET alone), PgR positive (vs PgR null). ET+Chemotherapy was favorable DFS trend with ET alone. Conclusions: Japanese premenopausal HR+HER2- EBC patients demonstrate excellent long-term survival, comparable to Western clinical-trial populations. In node-positive disease, OFS plus ET provided numerically greater DFS benefit than chemotherapy, suggesting that OFS may allow omission of chemotherapy in selected N1 patients. Prospective validation, is warranted to optimize adjuvant treatment in this population.
| Cohort | Treatment | 8-year DFS (%) [95% CI] | HR (vs ET alone) [95% CI] |
| All (N=7,396) | Observation±ET±OFS±Chemo | 95.2 [94.6 – 95.8] | Not applicable |
| Node-negative (N0, N=5,355) | ET alone or Observation | 95.6 [94.6 – 96.6] | 1.00 (ref) |
| ET + OFS | 97.0 [95.9 – 98.1] | 0.92 [0.59 – 1.44] (n.s.) | |
| ET ± OFS + Chemo | 95.8 [93.0 – 98.7] | 0.91 [0.51 – 1.64] (n.s.) | |
| Node-positive (N1, N=2,041) | ET alone or Observation | 88.3 [83.2 – 93.3] | 1.00 (ref) |
| ET + OFS | 92.2 [87.6 – 96.8] | 0.46 [0.22 – 0.97]* | |
| ET ± OFS + Chemo | 89.4 [87.6 – 91.1] | 0.83 [0.49 – 1.40] (n.s.) |
Presentation numberPS3-07-07
Tulip4n+ study: total tumor load as a predictor of ≥4 axillary lymph node metastases in HR+/HER2− breast cancer patients
Vicente Peg, Vall d’Hebron University Hospital, Barcelona, Spain
V. Peg1, E. Zamora2, I. Sperduti3, A. Piñero4, J. Fougo5; 1Pathology, Vall d’Hebron University Hospital, Barcelona, SPAIN, 2Oncology, Vall d’Hebron University Hospital; Vall d’Hebron Institute of Oncology, Barcelona, SPAIN, 3U.O. di Biostatistica e Bioinformatica, IRCCS Istituto Nazionale Tumori Regina Elena, Rome, ITALY, 4Surgery, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, SPAIN, 5Surgery, Centro Hospitalar Universitário de São João, Porto, PORTUGAL.
Background In early-stage hormone receptor-positive, HER2-negative (HR+/HER2−) breast cancer, the presence of ≥4 metastatic axillary lymph nodes has emerged as a key criterion for selecting candidates for adjuvant abemaciclib, based on data from the MonarchE trial. However, the increasing omission of axillary lymph node dissection (ALND) in clinical practice limits the ability to accurately determine nodal burden. Alternative methods for identifying patients with extensive nodal involvement are therefore needed. The One-Step Nucleic Acid Amplification (OSNA) assay enables intraoperative, quantitative detection of CK19 mRNA in sentinel lymph nodes (SLNs), providing a measure known as total tumor load (TTL). Preliminary studies have suggested a correlation between TTL and nodal involvement, but its utility in predicting ≥4 positive nodes remains to be validated in large cohorts. Objective To evaluate the predictive performance of TTL for identifying ≥4 metastatic axillary lymph nodes in HR+/HER2− early breast cancer and to establish an optimal TTL cut-off to support clinical decision-making, particularly to identify candidates for adjuvant abemaciclib treatment. Methods A retrospective, multicenter analysis was conducted using data from 5,032 patients with HR+/HER2− invasive breast cancer included in six previously published cohorts (2009-2016). All patients underwent SLN biopsy analyzed by OSNA, followed by conventional ALND. Patients with carcinoma in situ, de novo metastatic disease, or who received neoadjuvant therapy were excluded. The association between TTL and the presence of ≥4 positive axillary lymph nodes was analyzed using univariate and multivariate logistic regression models. Additional analyses were performed in histological subgroups, including lobular carcinoma. Results A TTL threshold of ≥1.0 × 10⁵ CK19 mRNA copies demonstrated a positive predictive value (PPV) of 32.5% for identifying patients with ≥4 positive lymph nodes. In the subgroup with lobular carcinoma, the predictive performance improved when TTL was combined with tumor grade and lymphovascular invasion, reaching a PPV of 50%. This multivariate model will be made available as an online calculator to support clinical decision-making. Conclusions The TULIP4N+ study supports the clinical utility of OSNA-based TTL for identifying patients with ≥4 metastatic lymph nodes in HR+/HER2− early breast cancer. In the current era of axillary surgery de-escalation, sentinel lymph node biopsy assessed by OSNA —and especially the TTL value— emerges as a reliable, quantitative tool to guide the selection of patients eligible for adjuvant therapies such as abemaciclib.
Presentation numberPS3-07-08
Sustained Ovarian Function Suppression with Triptorelin in Premenopausal HR+ HER2− Breast Cancer: Results from the Second Interim Analysis of the ROSE Study
Alessandra Fabi, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
A. Fabi1, G. V. Bianchi2, F. Riccardi3, A. Palazzo4, L. Carbognin1, A. Rossi1, C. Mocerino3, R. Ruocco3, G. Mazzoli2, G. Capri2, E. Di Monte4, V. Frescura4, M. Fournier5, G. Mauri6, N. Bianco7, M. Milano7, E. Munzone7; 1UOC Ginecologia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, ITALY, 2Medical Oncology Department, Fondazione IRCCS – Istituto Nazionale dei Tumori, Milan, ITALY, 3Oncology Unit, AORN Antonio Cardarelli, Naples, ITALY, 4Medical Oncology Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, ITALY, 5Global Medical Affairs, Ipsen, Paris, FRANCE, 6Medical & Regulatory Affairs, Ipsen, Milan, ITALY, 7Division of Medical Senology, IEO, European Institute of Oncology IRCCS, Milan, ITALY.
Background: Ovarian function suppression (OFS) is a cornerstone in the adjuvant treatment of premenopausal women with hormone receptor-positive (HR+), HER2-negative early breast cancer (EBC). Triptorelin is commonly used in this setting, but real-world evidence on its effectiveness across various treatment combinations remains limited. Methods: The ROSE study (NCT05377684) is a multicenter, observational trial designed to assess quality of life (QoL) as its primary endpoint in premenopausal women with HR+ HER2− EBC treated with triptorelin as OFS plus oral endocrine therapy. Chemotherapy was administered based on relapse risk factors and multidisciplinary discussion. Enrollment of total population (n=451) was completed as of December 2024. This second interim analysis, which included the first 103 patients who completed the follow-up as per protocol, focuses on a secondary endpoint: the evaluation of hormonal suppression over time. Premenopausal patients ≥18 years with stage I-IIIA HR+ HER2− EBC were eligible. Patients received adjuvant endocrine therapy (tamoxifen or aromatase inhibitors [AI]) plus triptorelin (1- or 3-month formulation), started either concurrently or sequentially with chemotherapy. Results: This analysis includes 103 participants who completed ≥2 hormone assessments (estradiol [E2], estrone [E1], FSH) and had 18 months of follow-up. OFS was defined as E2 <30 pg/mL, according to average values collected in Italian centers as per protocol. Results were stratified by treatment combinations and chemotherapy status. The cohort had a mean age of 46.2 years (SD: 4.9); 17.5% received chemotherapy with almost half of them received concomitant triptorelin, and 9.7% received abemaciclib in addition to endocrine therapy. Most patients received AIs (82.5%) and 1-month triptorelin (88.3%). At 6, 12, and 18 months, OFS (E2 <30 pg/mL) was achieved in 84.2%, 91.5% and 93.0%, respectively. Hormone levels showed a sustained suppression over time across all treatment subgroups. Median E2 levels dropped markedly from the first triptorelin injection to 6 months [median: 12.50 (Q1:6.70, Q3: 23.00)] and remained consistently low at 12 [median: 12.16 (Q1: 5.00, Q3: 23.00)] and 18 months [median: 13.95 (Q1: 5.00, Q3: 23.00)] FSH levels rose accordingly, confirming suppression. Among patients (9.7%) receiving triptorelin + AI + abemaciclib, hormonal suppression seemed comparable to that observed in other AI-based combinations (71.8%). The average duration of triptorelin exposure was similar across all subgroups, with a mean of 79.59 (SD: 6.11) weeks in the overall population. No significant variation in OFS achievement was observed by chemotherapy status or triptorelin formulation (1-month vs. 3-month). This uniformity supports comparability of endocrine effect regardless of treatment combinations, such as chemotherapy status or type of endocrine therapy. Outliers in E2 levels were rare and will be described for each time point after the ongoing post-hoc analysis. Conclusion: In this second interim analysis of the ROSE study, triptorelin was shown to effectively and durably suppress ovarian function over time in premenopausal women with HR+ HER2− EBC, across multiple treatment settings. These findings support the role of long-term hormonal monitoring and reinforce the use of OFS as a key component of endocrine-based strategies in this population. Final analyses from the full cohort will help refine patient selection and personalization of OFS regimens.
Presentation numberPS3-07-09
Extended Endocrine Therapy (eET) Following Five Years of Adjuvant LHRH-agonist (LHRHa) in Premenopausal Patients with Node-Positive, Hormone Receptor (HR)-Positive early Breast Cancer (eBC): a subanalysis according to surrogate subtypes
Carmine Valenza, European Institute of Oncology, Milan, Italy
C. Valenza1, Y. Zheng2, M. Milano3, D. Trapani1, E. Giordano1, L. Guidi1, P. Berton Giachetti1, L. Boldrini1, G. Castellano1, J. Katrini1, B. Malagutti1, G. Antonarelli1, F. Conforti4, G. J. Kirkner5, C. Sangalli6, D. E. Kate5, M. Colleoni3, M. M. Regan2, E. Munzone3, G. Curigliano1, A. H. Partridge5; 1Division of New Drugs and Early Drug Development, European Institute of Oncology, Milan, ITALY, 2Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 3Division of Breast Oncology, European Institute of Oncology, Milan, ITALY, 4Division of Medical Oncology, Humanitas Gavazzeni, Bergamo, ITALY, 5Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 6Clinical Trial Office, European Institute of Oncology, Milan, ITALY.
Background: In a previous cohort study analysis on 503 patients with node-positive early breast cancer (eBC) who remained premenopausal after 5 years of LHRHa treatment, we showed that the extension of endocrine therapy (ET) either with tamoxifen alone or by continuing previous LHRHa was associated with reduced invasive and distant breast cancer recurrences (Valenza, ASCO 2025). Due to the relative higher incidence of long-term recurrences in patients with luminal A-like eBC, the benefit of eET may differ according to surrogate breast cancer subtypes. Methods: We conducted a cohort study on two ongoing prospective datasets (Young Women’s Study and European Institute of Oncology’s Breast Cancer Dataset) to evaluate the clinical benefit of eET in women who had completed 5 years of adjuvant LHRHa, remained premenopausal and had no evidence of distant or locoregional recurrence. This study included women <40y at diagnosis (from 2005-2016) with node-positive HR+ eBC, with ductal, lobular, or ductolobular histotype, receiving or not eET (tamoxifen monotherapy or LHRHa+tamoxifen/aromatase inhibitor [AI]). Endpoints included invasive breast cancer-free survival (IBCFS) and distant recurrence-free survival (DRFS) calculated from the 6th year of ET (study baseline) and assessed with the propensity score (PS) weighted analysis. We performed a subgroup analysis according to the following surrogate breast cancer subtypes: luminal A-like (i.e., Ki67<20%, and progesterone receptor (PgR)≥20%, and Grade 1/2, and HER2-negative), luminal B-like/HER2-negative (i.e., Ki67≥20%, or PgR<20%, or Grade 3; and HER2-negative), and HER2-positive (i.e., HER2 positive per ASCO/CAP). Results: 487 patients were included (see Table): 276 received eET for a median duration of 3.7 years (IQR: 2.2-5.0). Overall, 89 (18.3%), 298 (61.2%) and 100 (20.5%) had a luminal A-like, luminal B-like and HER2-positive disease. At a median follow-up of 7.3 years (from the study baseline), the PS weighted Hazard Ratio (HR) for IBCFS comparing the eET to the non-eET group was 0.64 (95% CI, 0.45-0.91) in all patients, and 0.68 (95% CI, 0.34-1.40) in luminal A-like, 0.63 (95% CI, 0.41-0.98) in luminal B-like, and 0.62 (95% CI, 0.22-1.78) in HER2-positive subgroup. The PS weighted cause-specific HR for DRFS was 0.50 (95% CI, 0.32-0.79) in all patients, and 0.34 (95% CI, 0.12-0.92) in luminal A-like, 0.53 (95% CI, 0.31-0.92) in luminal B-like, and 0.77 (95% CI, 0.18-3.22) in HER2-positive subgroup. Conclusion: In this subanalysis, the benefit of eET was observed across all surrogate breast cancer subtypes. Notably, the magnitude of clinical benefit in terms of DRFS appeared greater in patients with a luminal A-like eBC, a finding that warrants confirmation in larger, prospective cohorts.
| Characteristic | eET (N=276) | No eET (N=211) |
| Age at diagnosis, median (IQR) | 37 (35-39) | 37 (33-39) |
| Dataset: IEO|YWS, % | 99|1 | 99|1 |
| Histology: Ductal|Lobular|Ductolobular, % | 92|4|4 | 95|3|2 |
| pT1|pT2|pT3-4, % | 37|50|13 | 41|52|7 |
| pN1|pN2|pN3, % | 63|22|15 | 73|17|10 |
| Luminal A-like|Luminal B-like (HER2-)|HER2+, % | 17|63|20 | 20|58|22 |
| ET during the firs 5y: LHRHa+Tam|LHRHa+AI|LHRHa, % | 65|34|1 | 77|21|1 |
| Previous chemotherapy, % | 77 | 70 |
Presentation numberPS3-07-10
Low dose (Z)-endoxifen in the I-SPY2 Endocrine Optimization Pilot
Karthik V Giridhar, Mayo Clinic Comprehensive Cancer Center, Rochester, MN
K. V. Giridhar1, M. P. Goetz1, C. Yau2, A. M. Wallace3, C. Falkson4, J. Boughey5, M. Arora6, E. Stringer-Reasor7, C. Nangia8, C. Gallagher9, A. Elias10, A. Zimmer11, M. S. Trivedi12, C. Omene13, L. van ‘t Veer14, L. Brown Swigart14, G. L. Hirst2, W. Symmans15, A. D. Borowsky16, N. Onishi17, N. Hylton17, C. Vakalvas18, M. Wei18, J. M. Reid1, S. Alhafaji19, N. Chen20, R. M. Mukhtar2, L. Huppert21, O. Opopade20, L. J. Esserman2, A. Chein21; 1Department of Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 2Department of Surgery, University of California San Francisco, San Francisco, CA, 3Department of Surgery, University of California San Diego, La Jolla, CA, 4Department of Medicine, University of Rochester – Pluta Cancer Center, Rochester, NY, 5Department of Sugery, Mayo Clinic Comprehensive Cancer Center, Rochester, MN, 6Department of Internal Medicine, University of California – Davis Comprehensive Cancer Center, Sacramento, CA, 7Division of Hematology Oncology, UAB O’Neal Comprehensive Cancer Center, Birmingham, AL, 8Department of Medical Oncology, HOAG Cancer Center, Irvine, CA, 9Department of Hematology Oncology, MedStar Georgetown Cancer Institute at MedStar Washington Hospital Center, Washington, DC, 10Department of Medicine, Medical Onocolgy, University of Colorado, Aurora, CO, 11Department of Medicine, Oregon Health and Science University, Portland, OR, 12Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, 13Division of Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 14Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 15Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 16Department of Pathology and Laboratory Medicine – Center for Immunology and Infectious Diseases, University of California Davis, Sacramento, CA, 17Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, 18Department of Medicine, University of Utah Huntsman Cancer Institute, Salt Lake City, UT, 19Department of Labaratory Medicine, University of California San Francisco, San Francisco, CA, 20Department of Medicine, University of Chicago, Chicago, IL, 21Department of Medicine, University of California San Francisco, San Francisco, CA.
Background: (Z)-endoxifen (ENDX) is an oral SERM that dually targets ERα (> 5 ng/ml) and PKCβ1 (>500 ng/ml). It has been tested in the endocrine resistant setting at doses of 20-360 mg/day; however, the optimal dose in an endocrine naïve population is not defined. We explored 10 mg/day dose of neoadjuvant ENDX in the Endocrine Optimization Pilot (EOP) of I-SPY2. Methods: EOP eligibility included pts with Stage II/III HR+ HER2- breast cancer, MammaPrint (MP) low. Pts with MP High1 scores were eligible if clinically node-negative. Pts received ENDX 10 mg orally once daily for 6 cycles (each cycle = 28 days). Premenopausal pts started ovarian function suppression on cycle 2, day 1. ENDX was continued until the day prior to surgery. The primary study endpoint was feasibility, defined as >75% of pts completing >75% of ENDX. Baseline (T0), 3-wk (T1) biopsies, and the surgical specimen (T3) were assessed centrally for Ki-67. Rates of modified preoperative endocrine prognostic index (mPEPI) 0 were reported, defined as pT1-2 pN0, Ki67< 2.7%, or pathologic complete response. A genomic index for sensitivity to endocrine therapy (SET) index was done at T0. Breast MRI measurement of functional tumor volume (FTV) was performed at T0, T1, 12 weeks (T2), and pre-operatively (T3). Blood was collected for tumor informed ctDNA at T0, T1, T2, T3 and for pharmacokinetic (PK) steady state concentrations (Css) at T1. Safety data using CTCAE V5 is reported. Results: Between March 2023 and May 2024, 20 pts were enrolled in this arm. The median age was 52.5 years (range 32 -73), 13 (65%) were premenopausal, and 55% had clinically positive lymph nodes. Tumors were MPHigh1 in 3 pts (15%) and SET2,3 index high in 16 pts (80%). The primary endpoint was met with 95% of study participants completing ≥75% of therapy. The T0 median Ki-67 was 10.5% (range 2-60%). At T1 (n=19) and T3 (n=18), median Ki-67 was 5% (range 0-35%) and 5% (range 0-45%), respectively. The 3-week rate of Ki-67 ≤10% at T1 was 61.5% in premenopausal and 71.4% in postmenopausal women. The rate of Ki-67 ≤10% at surgery was 81.8% in premenopausal and 57.1% in postmenopausal women. An mPEPI score of 0 was observed in 1 (5%) of participants. The median T0 FTV was 8.9 ccs (range 0.9 to 251.1). The median T3 FTV was 1.3 ccs (range 0 to 11.2) and the median change in FTV from T0 to T3 was -72.0% (range -88.5% to -61.6%). ctDNA analysis was performed in 17 pts longitudinally across 64 blood samples.10 pts had positive ctDNA at T0, with 7 converting to ctDNA undetectable during treatment with ENDX. 1/7 pts who was ctDNA negative at T0 became positive at T1 and then reverted to undetectable ctDNA. The other 6 pts remained ctDNA negative at all timepoints. The median ENDX T1 Css was 79.3 ng/mL (range 73.6 – 906.4 ng /mL). The most common side effects reported were Grade 1 hot flush (70%), nausea (40%), fatigue (40%), and insomnia (35%). Grade 2 side effects included fatigue (15%) and night sweats (5%). No dose reductions or discontinuations due to toxicity occurred. Conclusions: ENDX at 10 mg was well tolerated and demonstrated reductions in Ki-67, MRI FTV, and ctDNA. The ISPY2 EOP is currently enrolling patients to ENDX 40 mg/day (intended to dually target ERα and PKCβ1) in combination with abemaciclib (with and without OFS).
Presentation numberPS3-07-11
Defining postpartum breast cancer based on 21-gene recurrence score
Shiliang Zhang, University of California, Los Angeles, CA
S. Zhang1, K. J. Queen1, N. Duggirala2, M. Lipsyc-Sharf1, K. Hong3, Y. Suresh3, C. Hamilton3, M. Pitta3, N. Chien3, E. La3, P. Spellman1, A. Bardia1, N. S. Kapoor4; 1Hematology and Oncology, University of California, Los Angeles, CA, 2David Geffen School of Medicine, University of California, Los Angeles, CA, 3College of Letters and Science, University of California, Los Angeles, CA, 4Surgery, University of California, Los Angeles, CA.
Background: Postpartum breast cancer (PPBC), which arises during breast involution after childbirth and lactation, is linked to worse outcomes in young women with breast cancer (YWBC). Definitions of PPBC are not standardized, with some sources defining PPBC as 0-5 years postpartum and others defining it as up to 10 years postpartum. Prior studies suggest PPBC is associated with higher 21-gene recurrence scores (RS) and other high-risk molecular features, but detailed molecular profiles of breast cancer risk based on different PPBC definitions are limited. We aimed to identify a subset of PPBC that may be biologically distinct by comparing RS, individual gene expression levels, and pathologic tumor grade in different groups of PPBC stratified by timing of postpartum breast cancer diagnosis. Methods: We identified women aged ≤45 years with hormone receptor (HR)-positive, HER2-negative breast cancer who underwent RS testing at a single academic cancer center between 2011 and 2024. Reproductive and clinicopathologic data, including parity and timing of most recent childbirth relative to tumor testing were collected through electronic medical record review. Patients were stratified into four groups: nulliparous, PPBC1 (diagnosis 10-years postpartum. Patients who were diagnosed with breast cancer during pregnancy and patients with incomplete reproductive data were excluded. Linear regression was used to evaluate for differences in mean RS and gene expression scores for estrogen receptor (ER), progesterone receptor (PR), and HER2. Ordinal logistic regression was used to evaluate for differences in odds of increased RS categories (0-15, 16-25, >25) and increased pathologic tumor grade. All models were adjusted for age at diagnosis and nodal status. Results: A cohort of 381 HR+HER2- breast cancer patients were identified and classified as: nulliparous (n=149), PPBC1 (n=71), PPBC2 (n=92), and >10-years postpartum (n=69). In our study, the average RS for women in the PPBC1 group was 5.13 points higher than for nulliparous women (95% CI: (2.38, 7.88), p 10-years postpartum group (p = 0.065). Similarly, after adjusting for age and nodal status, the odds of having a higher RS category for the PPBC1 group were 2.44 times that of nulliparous women (95% CI: (1.41, 4.22), p 10-years postpartum group (p = 0.055). Regarding PR gene expression, the average score decreased by approximately half a point for all postpartum women compared to nulliparous women (p = 0.002 for PPBC1, 0.047 for PPBC2, 0.006 for >10-years postpartum). There was no statistically significant difference in ER or HER2 gene score between nulliparous and postpartum women. In terms of histological grade, the odds of increasing by one pathology grade unit for women with PPBC1 were 2.00 times that of women who are nulliparous (95% CI: (1.14, 3.52), p = 0.015). The odds were no different for women who are PPBC2 or >10-years postpartum compared to nulliparous women (p = 0.90, 0.76, respectively). Conclusions: In this cohort of patients, breast cancer diagnosed within five years postpartum is associated with significantly higher RS and histological grade compared to nulliparous young women, independent of nodal involvement. On the other hand, breast cancer diagnosed 5-10 years postpartum does not show this association. These findings support refining the definition of PPBC in ER+HER2- breast cancer as disease diagnosed within five years postpartum, based on its distinct molecular risk profile.
Presentation numberPS3-07-12
The CONCISE Study : A Pan-UK Real-World Audit of Adjuvant Abemaciclib Use in High-Risk Early Breast Cancer
Olubukola Ayodele, University College London Hospital Cancer Collaborative, Princess Alexandra Hospital NHS Trust, Harlow, and 25.Leicester Cancer Research Centre, University of Leicester, Leicester, UK UK, Leicester, United Kingdom
B. Baraka1, A. Ghose2, H. Abdallah3, A. Maniam4, Y. Owoseni5, A. Konstantis6, S. Horne7, A. Nazir7, R. Kussaibati7, L. Mooney8, F. Nazeer9, N. Atsumi10, E. Daniels11, L. McAvan12, M. Abraham13, D. Morgan14, G. Langford15, E. Bean16, E. Morris17, R. Muhammad18, S. Khan19, R. Pravinkumar20, S. Mohammed21, S. A. Raza22, C. Blair23, R. Khan24, M. Tsalic7, R. Douglas25, K. Panagiotis26, S. Waters14, J. Smith27, E. Papadimitraki28, C. Michie29, C. Wilson24, O. Ayodele30; 1Oncology, Nottingham University hospitals, Nottingham, UNITED KINGDOM, 2Oncology Department, Barts Cancer Centre, Barts Health NHS Trust, London, UNITED KINGDOM, 3Oncology Department, Cambridge Clinical Research Centre, Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UNITED KINGDOM, 4Oncology Department, Cancer Centre, Portsmouth Hospitals University NHS Trust, Portsmouth, and 5.Cancer Services, Isle of Wight NHS Trust, Newport, UK, Portsmouth, UNITED KINGDOM, 5Oncology Department, University Hospitals of Leicester NHS Trust, Leicester, UK, Leicester, UNITED KINGDOM, 6Oncology Department, University College London Hospital Cancer Collaborative, Princess Alexandra Hospital NHS Trust, Harlow, UK, Essex, UNITED KINGDOM, 7Oncology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK, Birmingham, UNITED KINGDOM, 8Oncology Department, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, UNITED KINGDOM, 9Oncology Department, Royal Surrey Cancer Centre, Royal Surrey NHS Foundation Trust, Guildford, and Surrey and Sussex Healthcare NHS Trust, Redhill, UK, Surrey, UNITED KINGDOM, 10Oncology Department, Portsmouth Hospitals University NHS Trust, and Cancer Services, Isle of Wight NHS Trust, Newport, UKK, PO30 5TG, UNITED KINGDOM, 11Oncology Department, Dyson Cancer Centre, Royal United Hospitals Bath NHS Foundation Trust, Bath, UK, Bath, UNITED KINGDOM, 12Oncology Department, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK, Coventry and Warwickshire, UNITED KINGDOM, 13Oncology Department, Dorset Cancer Centre, University Hospitals Dorset NHS Foundation Trust, Poole, UK, Poole, UNITED KINGDOM, 14Oncology Department, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UK, Cardiff, UNITED KINGDOM, 15Oncology, Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UK, Cardiff, UNITED KINGDOM, 16Oncology Department, Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK, Newcastle, UNITED KINGDOM, 17Oncology, University College Hospital Macmillan Cancer Centre, University College London Hospitals NHS Foundation Trust, London, UK, London, UNITED KINGDOM, 18Oncology Department, University College London Hospital Cancer Collaborative, Princess Alexandra Hospital NHS Trust, Harlow, UK, London, UNITED KINGDOM, 19Oncology Department, Nottingham University Hospital, Nottingham, UNITED KINGDOM, 20Oncology, Edinburgh Cancer Centre, NHS Lothian, Edinburgh, UK, Edinburgh, UNITED KINGDOM, 21Oncology Department, Great Western Hospitals NHS Foundation Trust, Swindon, UK, Nottingham, UNITED KINGDOM, 22Oncology Department, Queen’s Centre for Oncology and Haematology, Hull University Teaching Hospitals NHS Trust, Cottingham, UK, Hull, UNITED KINGDOM, 23Oncology Department, Bristol Haematology and Oncology Centre, University Hospitals of Bristol and Weston NHS Trust, Bristol, UK, Bristol, UNITED KINGDOM, 24Oncology, The Christie NHS Foundation Trust, Manchester, UK, Manchester, UNITED KINGDOM, 25Oncology, Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, UK, Belfast, UNITED KINGDOM, 26Oncology Department, Royal Surrey Cancer Centre, Royal Surrey NHS Foundation Trust, Guildford,UK, Surrey, UNITED KINGDOM, 27Oncology, Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK, Newcastle upon Tyne, UNITED KINGDOM, 28Oncology Department, University College Hospital Macmillan Cancer Centre, University College London Hospitals NHS Foundation Trust, London, UK, London, UNITED KINGDOM, 29Oncology Department, Edinburgh Cancer Centre, NHS Lothian, Edinburgh, UK, Edinburgh, UNITED KINGDOM, 30Oncology Department, University College London Hospital Cancer Collaborative, Princess Alexandra Hospital NHS Trust, Harlow, and 25.Leicester Cancer Research Centre, University of Leicester, Leicester, UK UK, Leicester, UNITED KINGDOM.
Background Adjuvant Abemaciclib (AA) combined with endocrine therapy (ET) is an established treatment for hormone receptor-positive (HR+), HER2-negative (HER2-), node-positive early breast cancer (EBC) at high risk of recurrence, as demonstrated in the monarchE trial. However, data on its tolerability and use in routine UK clinical practice are limited. This study presents multicentre real-world data on the use of adjuvant AA across the UK, focusing on treatment duration, dose modifications, discontinuations and toxicity. Methods CONCISE is a retrospective, multicentre observational study across 21 NHS Trusts including patients aged ≥18 years with HR+/HER2-, node-positive, high-risk EBC treated with AA between July 2022 and April 2025. High-risk was defined as ≥4 positive axillary lymph nodes or 1-3 nodes with tumour size ≥5 cm or histological grade 3. Clinical data were obtained from patients electronic records and a data registry was established on Ledidi. Primary endpoint: real-world treatment duration of AA. Secondary endpoints: rates of dose reduction, discontinuation, and grade ≥2 toxicities; correlation with age, endocrine therapy type and prior chemotherapy. Results A total of 1,028 patients were included; 98.5% were female and 82.8% Caucasian, with a median age of 54 years. Majority were post-menopausal (56.5%). Most patients had stage II (57.5%) and grade 2 (52.6%) tumours. Lymph node involvement was present in 95% (median = 5 nodes). Chemotherapy was administered in 88.4% (neoadjuvant 16.3%, adjuvant 72.1%). Endocrine backbone (AI, 86.2% vs Tamoxifen 13.8%).The median duration of AA was 15.5 months. At the time of analysis, 25.3% had completed the full 2-year course with 74.7% remaining on treatment. AA was discontinued in 15.9% due to toxicity and 3.8% due to disease progression (median discontinuation time: 10.3 months). Single dose level reduction occurred in 64.6%, with further reduction in 25.4%.Common toxicities included diarrhoea (76.4%, ≥Grade 2 in 40%), neutropenia (43.4%, ≥Grade 2 in 61.1%), anaemia (33.2%, ≥Grade 2 in 11.7%), thrombocytopenia (10%, ≥Grade 2 in 6.8%) and transaminitis (17.2%, ≥Grade 2 in 25%). There were 21 deaths (2.0%) of which 16 were cancer-related and one attributed to treatment-related renal failure. Conclusion This large UK real-world cohort confirms the feasibility of AA use in routine practice, though dose modifications and early discontinuations were more frequent than reported in clinical trials. Toxicities remain consistent with known safety profiles, but adherence may be impacted by age, comorbidities, and supportive care access. Optimising management of side effects is critical to maintaining long-term adherence and maximising clinical benefit in real-world settings.
Presentation numberPS3-07-13
High genomic risk by multigene assay in germline BRCA1/2 mutated tumors among patients with ER-positive/HER2-negative early breast cancer
Ee Jin Kim, Asan medical center, Seoul, Korea, Republic of
E. Kim1, S. Baek2, S. Bae3, J. Jeong3, S. Ahn3, T. Yoo1, S. Lee1, I. Chung1, H. Kim1, B. Ko1, J. Lee1, B. Son1, J. Kim1; 1Department of Surgery, Asan medical center, Seoul, KOREA, REPUBLIC OF, 2Department of Surgery, Yongin Severance Hospital, Gyeonggi-do, KOREA, REPUBLIC OF, 3Department of Surgery, Gangnam Severance Hospital, Seoul, KOREA, REPUBLIC OF.
High genomic risk by multigene assay in germline BRCA1/2 mutated tumors among patients with ER-positive/HER2- negative early breast cancer Introduction Indication of BRCA testing is expanding with growing evidence on optimal surveillance and treatments for BRCA-associated breast cancer (BC). As BRCA-associated hormone receptor(HR) positive, HER2 negative metastatic BC has shown to display worse outcome from previous studies, we aim to investigate the genomic risk profile and subsequent outcome in early HRpos/HER2neg BRCA-associate BC compared to BRCA non-associated BC.Methods A retrospective review of 5,081 early HRposHER2neg BC cases between Jan 2011 ~ Dec 2023 in three institutes was done. Among the 1,401 BRCA-tested patients, 102 (7.3%) was found to have germline BRCA pathogenic variant (PV) and 1,299 (92.1%) with BRCA wild type (WT)/VUS. To minimize the effect of potential BRCA-carrier effect we excluded cases with BRCA testing indicated yet not tested (n=339). Total 3,518 cases were analyzed including 3,320 patients not- indicated for BRCA testing as control. We compared genomic risk using Oncotype recurrence score (RS) (cut-off 26) and Mammaprint (MMP) high vs low risk. Disease free survival and cumulative incidence of contralateral breast cancer was analyzed.Results Among the 3,518 cases analyzed, BRCA-pathogenic variant (PV) displayed high genomic risk compared to control group (high vs low, 50.0% vs 26.7%, p<0.001). Mean Oncotype RS was higher in BRCA-PV group (18 ± 9 vs 25 ± 12, p<0.001) and 89.5% (17/19) of BRCA-PV group was MMP-high while 43.2% (286/680) in control group. BRCA-PV group showed younger age at diagnosis (46y±11 vs 51y±6, p<0.001), and BRCA-PV BC was independently associated with high genomic risk, high grade and high Ki67 compared to control group (all p<0.005).Median f/u was 49.0 months (IQR 30.9-63.6). The BRCA-PV group displayed worse disease-free survival (89.3% vs 96.0% at 5years, 70.8% vs 90.0% at 10years, p<0.01). While BRCA mutation did not affect overall survival, 5-yr and 10yr-cumulative CBC incidence was higher in BRCA-PV group than in control group (OS: 98.1% vs 99.3% at 5years, p=0.78; CBC: 3.1% (3/102) vs 0.9% (26/4,617) at 5years, 11% (5/102) vs 2.7% (35/4,617) at 10years, p<0.05). BRCA-mutation and multigene-assay genomic high risk was both independently associated with high risk of recurrence (HR 2.13, 95%CI 1.05-4.33 and HR 1.51, 95%CI 1.00-2.28, respectively).In subgroup analysis, BRCA-PV patients in both high and low genomic risk group showed worse outcome than control group. Further analysis incorporating actual treatment data is planned. Conclusion BRCA-associated BC displayed high genomic risk and subsequent worse outcome. Worse outcome was observed in both high and low genomic risk group, suggesting the necessity in identifying optimal treatment strategy for this BRCA-associated BC population. Also, risk reducing mastectomy should be considered for younger age BRCA-PV BC patients.
Presentation numberPS3-07-14
Predictors of early discontinuation of adjuvant palbociclib in early HR+/ HER2- breast cancer: final analysis of the PALLAS trial integrating patient-reported outcomes
Marcus Vetter, Cantonal Hospital Baselland, Liestal, Switzerland
M. Vetter1, A. Wiencierz2, M. Gnant3, D. Hlauschek4, C. Kurzeder5, C. Grasic Kuhar6, N. Zdenkowski7, E. Shinn8, J. Suga9, D. Egle10, J. Meisel11, S. Antolin-Novoa12, E. Munzone13, T. Haddad14, S. Loibl15, A. Holynskyj16, C. Fesl3, A. Dueck17, A. DeMichele18, E. Mayer19, on behalf of the PALLAS groups and investigators (ABCSG, AFT, BIG,PReCOG, GBG, NSABP); 1Cancer Center Baselland, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 2CTU, University Hospital Basel, Basel, SWITZERLAND, 3ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer C, ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria, Wien, AUSTRIA, 4ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer C, ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria, Wien, AUSTRALIA, 5Breast Clinic, University Hospital Basel, Basel, SWITZERLAND, 6Institute of Oncology Ljubljana, Institute of Oncology Ljubljana, Ljubljana, SLOVENIA, 7The University of Newcastle, The University of Newcastle, East Maitland, AUSTRALIA, 8MD Anderson Cancer Center, The University of Texas, Houston, TX, 9Medical Oncology, Kaiser Permanente Vallejo Medical Center, Vallejo, CA, 10ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer C, ABCSG, Austrian Breast and Colorectal Cancer Study Group, Vienna, Austria AND Comprehensive Cancer Center, Medical University of Vienna Vienna, Austria AND Medical University of Innsbruck, Wien, AUSTRIA, 11Winship Cancer Institute, Emory University Atlanta, Atlanta, GA, 12Medical Oncology Department, Breast Unit, A Coruña University Hospital, A Coruña, A Coruña, SPAIN, 13European Institute of Oncology IRCCS, European Institute of Oncology IRCCS, Milan, ITALY, 14Medical Oncology, Mayo Clinic, Rochester, MN, USA, Rochester, MN, MN, 15German Breast Group, German Breast Group, Germany AND Goethe University Frankfurt/M, AND Clinical Consultant Centre for Haematology and Oncology/Bethanien Frankfurt/M, Neu-Isenburg, GERMANY, 16Pfizer, Pfizer, New York, NY, 17Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, MN, 18Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, 19Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA.
Background: The PALLAS trial (ABCSG-42, AFT-05, BIG-14-03) investigates adjuvant palbociclib plus endocrine therapy (ET) versus ET alone in patients (pts) with HR+/HER2− early breast cancer. While palbociclib improves outcomes in metastatic settings, final analysis from PALLAS did not demonstrate benefit in survival outcomes from the addition of palbociclib to ET. A number of pts discontinued palbociclib before the planned 2 year duration though degree of palbociclib exposure was not associated with clinical outcomes. This analysis explores factors predictive of palbociclib early discontinuation and dose reductions, including baseline characteristics, toxicities, and patient-reported outcomes (PROs). Methods: Using the safety population of 5736 pts (2840 receiving palbociclib + ET), we assessed early palbociclib discontinuation within a competing-risks framework (competing events: progression, death). Cumulative incidence curves (Aalen-Johansen estimates) and cause-specific hazard models were used to explore associations with clinical and demographic factors (age, menopausal status, stage, race, endocrine therapy type), key toxicities (neutropenia, anemia, nausea), and baseline PROs (patient assessed quality of life [QoL], anxiety, fatigue, pain, frailty/ dependency, depression, and low income. financial difficulties). An automated model selection based on the Akaike information criterion (AIC) starting from the cause-specific hazard model incorporating all considered predictors including PROs. Results: Early discontinuation of palbociclib not due to progression or death occurred in 1171 pts (41.3%); an additional 343 pts (12.1%) stopped due to results of a futility analysis. In 108 (3.8%) and 2 (0.1%) pts treatment was discontinued due to progression or death. Median time to early discontinuation was 7.7 months in pts that discontinued treatment (median interquartile range 3.45-13.60). Very early discontinuation (≤3 months) occurred in 261 pts (9.2%). Dose reductions to 100mg and 75mg occurred in 1566 pts (55.2%) and 951 pts (33.6%) of 2840 pts, respectively. In univariable analyses, higher age, lower N-stage, and specific baseline PROs (baseline QoL, fatigue, anxiety, depression, frailty/ dependency and financial difficulties) were linked with increased risk of discontinuation. The best-fitting multivariable model identified age, nodal stage (N-stage), low quality of life (QoL), and low income as key predictors of early treatment discontinuation. Conclusions: In the PALLAS trial, our findings highlight the importance of integrating PROs into risk prediction of early treatment discontinuation. Additionally, when starting pts on oral targeted therapies in the adjuvant setting, symptom control, social work and financial counseling resources may support pts at higher risk of non-adherence. This is in line with findings from the ESMO Breast QoL analysis (Bjelic-Radisic et al.), which emphasized the added value of subjective patient-reported outcomes alongside objective clinical parameters in understanding treatment burden and adherence. Support: AFT, ABCSG, Pfizer, ECOG-ACRIN, NSABP Foundation, GBG, BIG; https://acknowledgments.alliancefound.org; ClinicalTrials.gov Identifier: NCT02513394.
Presentation numberPS3-07-15
Efficacy of a quarterly formulation of human of luteinizing hormone-releasing hormone agonists in inducing ovarian suppression during adjuvant treatment of hormone receptor-positive localized breast cancer in premenopausal women – a retrospective study.
Deborah Cathalá Ribeiro Dias, AC Camargo Cancer Center, São Paulo, Brazil
D. C. Dias, L. Leite, S. Sanches, V. Cordeiro; Oncology, AC Camargo Cancer Center, São Paulo, BRAZIL.
Introduction Premenopausal women with hormone receptor positive (HR+) early breast cancer (BC) who receive chemotherapy have significant reduced risk of distant relapse when treated with ovarian function suppression (OFS) and aromatase inhibitors (AI) in the adjuvant setting. Clinical trials supporting this practice used predominantly monthly luteinizing hormone-releasing hormone agonists (LHRHa) formulations. However, due to the convenience of dosing, other formulations are frequently used in clinical practice. The aim of this study is to compare the efficacy of quarterly LHRHa dosing with monthly LHRHa dosing in inducing OFS in premenopausal HR+ patients undergoing adjuvant AI plus LHRHa. Methodology This was a retrospective cohort of all HR+ (HER2-positive or negative) BC patients treated with AI plus LHRHa between 2015 and 2021 in a single center. Patients were evaluated for ovarian suppression failure (OSF), a composite outcome defined by one of the following criteria: E2 levels above the postmenopausal range (according to local laboratories references); occurrence of menstrual bleeding during treatment; need to switch AI to tamoxifen due medical judgement of castration failure and need for salpingo-oophorectomy due to OSF. In patients receiving the quarterly LHRHa, the need to switch to the monthly formulation as per medical judgement due to castration failure was also considered OSF. As secondary outcomes, we compared the impact of both LHRHa on relapse free survival (RFS) and overall survival (OS), as well as the impact of OSF on survival outcomes. Statistical comparisons included Fisher’s Exact test, Chi-square, Mann- Whitney, Kaplan-Meier, and regression analyses. The significance level was set at 5%. Results Among 332 patients, 238 received monthly LHRHa and 94 patients received quarterly LHRHa. Baseline clinical characteristic were balanced between the two groups, except for the duration of treatment, which was longer in the quarterly versus the monthly LHRHa group (55 months x 38 months, p < 0.01). This imbalance in the duration of treatment occurred because 152 patients in the monthly LHRHa group changed to quarterly LHRHa administration after some time, due to patient convenience. OSF occurred in 27 patients, 20/94 (21.3%) in quarterly LHRHa vs. 7/238 (2.9%) in monthly LHRHa (p < 0.01). A sensitivity analysis was performed to adjust for the duration of treatment, excluding patients in the monthly group who changed to quarterly administration. In this analysis 180 patients were included, 86 in the monthly LHRHa and 94 in the quarterly LHRHa group. OSF still was higher in the quarterly vs. monthly group (21.3% x 2.3%). Considering the entire population the median follow-up of 59 months, 18 patients (5.4%) had disease recurrence (4 local/14 distant) and 7 patients died, with no diference between initial choice of LHRHa formulation (HR 0.65 95%CI 0.23 – 1.82), On uni and multivariate analysis of RFS age, cT, cN+, HER2 status and histologic grade were not significant factors, and only OSF remained significant in a model including clinical stage and type of analogue (HR 5.11 95%CI 1.54 – 17, p= 0.008). OS was also not impacted by initial type of LHRHa used (HR 0.20 95%CI 0.02 – 1.71, p=0.14), but further analysis were limited by the number of events. Conclusion The use of the quarterly dosage of the LHRH analogue was linked to higher ovarian suppression failure, biased by the longer treatment duration of this formulation in our sample. OSF was an important prognostic factor for RFS and estradiol levels should be monitored. Prospective studies are needed to clarify doubts regarding the efficacy of quarterly LHRH analogues.
Presentation numberPS3-07-16
Validation of Late Distant Recurrence Prediction Model in Premenopausal Women with ER-Positive/HER2-Negative Breast Cancer
Jong-Ho Cheun, SMG-SNU Boramae Medical Center, Seoul, Korea, Republic of
J. Cheun1, D. Shin2, D. Noh3, J. Ahn4, Y. Lee5, E. Kang2, J. Lee6, J. Lee7, S. Kwon3, H. Lee2, J. Ryu8, S. Ahn9; 1Surgery, SMG-SNU Boramae Medical Center, Seoul, KOREA, REPUBLIC OF, 2Surgery, Seoul National University Hospital, Seoul, KOREA, REPUBLIC OF, 3Information and Biomedical Engineering, Pusan National University, Pusan, KOREA, REPUBLIC OF, 4Surgery, Severance Hospital, Seoul, KOREA, REPUBLIC OF, 5Surgery, Asan medical center, Seoul, KOREA, REPUBLIC OF, 6Surgery, Ewha Womans University Mokdong Hospital, Seoul, KOREA, REPUBLIC OF, 7Surgery, Soonchunhyang University Hospital Seoul, Seoul, KOREA, REPUBLIC OF, 8Surgery, Samsung Medical Center, Seoul, KOREA, REPUBLIC OF, 9Surgery, Gangnam Severance Hospital, Seoul, KOREA, REPUBLIC OF.
Background: The Clinical Treatment Score post-5 years (CTS5) model was developed to predict late recurrence in estrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) patients who have completed five years of endocrine therapy (ET). However, CTS5 has shown lower predictive accuracy and risk underestimation in premenopausal women, and the model may not fully reflect the currently improved prognosis as it was developed based on trials conducted decades ago. To address these limitations, we previously developed a machine learning-based model to predict the probability of late distant metastasis (DM) using a multi-institutional cohort of premenopausal women. In this study, we aimed to externally validate our model and compare its performance with that of CTS5. Methods: We retrospective reviewed the patients who underwent primary breast cancer surgery between 2000 and 2013 at Asan Medical Center, Severance Hospital, Boramae Medical Center, and those enrolled in the ASTRRA trial. Eligible patients were premenopausal women aged ≤45 years with ER-positive/HER2-negative BC who received ET with or without ovarian function suppression (OFS). Patients who discontinued ET before 4.5 years, received neoadjuvant chemotherapy, and had distant metastasis within five years were excluded. Our previously developed machine learning-based prediction model utilized eight clinicopathologic features: age, tumor size, number of positive lymph nodes, nuclear grade, histologic grade, progesterone receptor status, use of OFS, and chemotherapy. Patients were stratified into high- or low-risk groups based on a model output probability of 0.480. The primary endpoint was distant metastasis-free survival (DMFS) between five and ten years after surgery. Results: A total of 1,465 patients were included. The median age at the time of operation was 41.0 years (IQR, 38.0-43.0 years). T1 stage and N0 stage tumors were present in 920 (62.8%) and 897 (61.2%) patients, respectively. ET duration was extended in 283 patients (19.3%), and OFS was administered in 537 (36.7%). Among 1,182 patients who did not undergo ET extension, the 10-year DMFS rate was 95.3% during a median follow-up of 157.7 months (IQR 137.6-185.6). The high-risk group had significantly worse DMFS than the low-risk group (p<0.001, HR 2.71; 95% CI, 1.60-4.58). When including those who extended ET, high-risk patients appeared to benefit from ET extension (p=0.008, HR 4.36; 95% CI, 1.33-14.29), whereas low-risk patients did not (p=0.126). In comparison with CTS5, our model achieved a slightly better AUC (0.701 vs. 0.654). According to the CTS5 classification, high-risk group demonstrated significantly worse DMFS (p<0.001, HR, 3.39; 95% CI, 1.91-6.00), but the intermediate-risk group did not show a different survival outcome (p=0.885) compared to the low-risk group. Notably, 45 among 698 patients classified as low-risk by CTS5 were identified as high-risk by our model, and had significantly worse DMFS (p=0.023, HR, 3.26; 95% CI, 1.11-9.57). Finally, we developed our model using the combined development and validation datasets (n=2,395). The updated model demonstrated robust performance, with an AUC of 0.744, accuracy of 0.718, sensitivity of 0.615, specificity of 0.725, positive predictive value of 0.128, and negative predictive value of 0.966. Conclusions: Our machine learning-based model provides valuable prognostic information on the risk of late distant recurrence between five and ten years after surgery in premenopausal, ER-positive/HER2-negative BC patients. This model would assist clinicians in personalizing decisions regarding the duration of ET and the need for OFS in patients who remain recurrence-free at five years postoperatively.
Presentation numberPS3-07-17
Circulating tumor DNA predicts relapse in early ER positive breast cancer
Mark Basik, Lady Davis Institute, Montreal, QC, Canada
M. Basik, A. Klemantovich, R. Talia, L. Josiane, L. Cathy, A. Aguilar-Mahecha; Oncology, Lady Davis Institute, Montreal, QC, CANADA.
Introduction: Early hormone receptor positive breast cancer is treated with surgery followed by adjuvant systemic therapy, including chemotherapy and endocrine therapy. Recent studies have reported the added benefit of CDK4/6 inhibitors in the adjuvant setting in high-risk patients. Despite the high risk of relapse in patients for whom these drugs are currently indicated, the relapse free benefits are very small. There is a need for a better method to determine more precisely the prognosis of these high risk ER+ breast cancer patients. Circulating tumor DNA (ctDNA) offers promise for real-time, personalized monitoring of cancer. Tumor bespoke ctDNA assays have been shown to accurately predict tumor relapse. In our Q-CROC-03 and TRICIA trials we validated the prognostic value of ctDNA in early triple negative breast cancer. We now present our first results of serial ctDNA testing in patients with early ER+ breast cancer in the adjuvant setting. Our aim was to detect relapse in early breast cancer patients at least 9-12 months prior to clinical relapse. Methods: Plasma samples were collected post-operatively in 35 patients with mostly stage II/III ER+ breast cancer every 3-12 months up to 5 years after surgery. We performed a case-control study in which 14 patients with relapse were compared with 21 patients without relapse. Serial plasma samples were analysed in the 2 years prior to relapse in relapsed patients and up to 5 years post-operatively in non-relapsed patients. Patient-specific digital PCR assays (5/patient) were designed in-house using somatic tumor mutations identified by whole exome sequencing. Fractional abundance (cFA) was corrected by subtracting the fractional abundance of each variant in normal control plasma. Negative cFA were labeled zero (negative). Results: We determined an optimal threshold of cFA of 0.015% detected in at least 2 of the 5 variants to consider the sample ctDNA positive (ctDNA+). The median time to relapse was 35 months. 12 of 14 (86%) relapsed patients were ctDNA+ compared to 6 of 21 non-relapsed patients in at least one time point. Patients with negative ctDNA (ctDNA-) had a relapse-free survival (RFS) of 88% compared to 19% in ctDNA+ patients (p = 0.003, HR = 0.16 (95% CI = 0.05 to 0.4)), a negative predictive value (NPV) of 88%, sensitivity of 86% and specificity of 71%. When examining only samples collected between 6-18 months after surgery, ctDNA- patients reached an RFS of 90% compared to 21% in ctDNA+ patients (p<0.0001, HR = 0.08 (95% CI = 0.02 to 0.3)), reaching a NPV of 90%, sensitivity of 83% and specificity of 86%. Moreover, the mean cFA of tested variants in relapsed patients was over 10-fold higher than that of non-relapsed patients. The cFA of tested variants in relapsed patients increased with each time point, whereas in non-relapsed patients the fractional abundance in tested variants decreased with each time point. We were able to detect relapses in ER+ breast cancer patients for a median of 21.6 months (range 4.9 -53.3) prior to clinical relapse.Conclusion: Circulating tumor DNA (ctDNA) is a strong predictive biomarker for relapse in ER+ breast cancer patients following surgery. Our results show that detectable ctDNA predicts relapse with a lead-time of nearly 2 years offering a substantial lead time for potential intervention.
Presentation numberPS3-07-18
Barriers to Enrollment in NRG-BR009 in West Virginia: Patient Treatment Preference as a Key Challenge in a Prospective Randomized Trial of Premenopausal Women with Intermediate Oncotype Score
Gary Monroe, West Virginia University Cancer Institute, Morgantown, WV
M. Hartzell, J. Hill, G. Monroe, D. Safi, S. Kurian, S. Nethagani, B. Jiang; Hematology and Medical Oncology, West Virginia University Cancer Institute, Morgantown, WV.
Background: NRG-BR009 is a phase III randomized trial evaluating whether chemotherapy can be safely omitted in premenopausal women with early-stage, estrogen receptor (ER)–positive, HER2-negative breast cancer and an intermediate Oncotype DX Recurrence Score® (16–25). The trial compares standard chemotherapy plus endocrine therapy to ovarian function suppression (OFS) plus endocrine therapy alone. Despite the clinical promise of de-escalation, national accrual has been limited. This study explores enrollment barriers within the West Virginia University Cancer Institute (WVUCI), a rural-academic network spanning both university and satellite cancer centers. Methods: A real-time Epic-based dashboard was developed to track all patients for whom Oncotype DX testing was ordered from 5/22/2024 to 6/30/2025. Structured interviews with clinical research teams and treating physicians were conducted to assess reasons for enrollment or non-enrollment, focusing on patient decision-making, provider communication, and operational barriers. Results: Among the 177 patients who underwent Oncotype DX® testing during the study period, 166 (93.8%) were female, 20 (11.3%) were under the age of 50, and 21 (11.9%) had a Recurrence Score (RS) greater than 25. Nodal status revealed that 26 patients (14.7%) were node-positive, 125 (70.6%) were node-negative, and 25 (14.1%) had unknown nodal status.Five patients (2.8%) met eligibility criteria for the NRG-BR009 trial. Of these, 4 were treated at the university hospital and 1 at a community site. All 5 patients were node-negative; 1 had an RS between 16–21 and 4 had RS values between 22–25. The clinical trial was offered at the initial oncology consultation. One patient with an RS of 23 enrolled and was randomized to the chemotherapy arm. 3 patients declined participation and subsequently received endocrine therapy with ovarian function suppression (OFS) outside of the trial. One additional patient, with an RS of 16, was recommended OFS by the treating oncologist post-test and was not randomized.Physicians consistently cited both the limited number of eligible patients and strong pre-existing patient treatment preferences as key barriers to enrollment. The trial option was typically introduced during the initial multidisciplinary oncology visit. However, initial interest in NRG-BR009 was low. Many premenopausal patients expressed a preference for chemotherapy prior to surgery but later shifted toward OFS plus endocrine therapy after receiving an intermediate Recurrence Score postoperatively. In most cases, patients had already made a treatment decision by the time of trial discussion and expressed reluctance to accept randomization. Preferences were primarily driven by concerns regarding side effects, fertility preservation, and perceptions of limited absolute benefit from chemotherapy. Conclusions: In a rural-academic cancer network, despite a high enrollment rate 20%, enrollment in NRG-BR009 was limited by both a small pool of eligible patients and strong, pre-existing treatment preferences among premenopausal women with intermediate Oncotype DX scores. Despite timely trial discussions and centralized eligibility tracking, most patients declined participation due to discomfort with randomization and a preference for personalized treatment decisions based on fertility concerns, perceived side effects, and anticipated chemotherapy benefit. These findings highlight the need for earlier engagement in shared decision-making, tailored decision-support tools, and strategies to maintain equipoise when offering clinical trials in preference-sensitive populations.
Presentation numberPS3-07-19
Impact of Endocrine Therapy in Estrogen Receptor-Low Breast Cancer: A Systematic Review and Meta-Analysis
Davide Massa, Università degli Studi di Padova, Padova, Italy
D. Massa1, E. Chiappe2, V. Delucchi3, F. Girardi4, E. Blondeaux5, M. La Commare1, V. Guarneri1, M. Lambertini2, M. Dieci1; 1Dipartimento di Scienze Chirurgiche, Oncologiche e Gastroenterologiche (DiSCOG), Università degli Studi di Padova, Padova, ITALY, 2Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genova, Genova, ITALY, 3U. O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 4Oncology 2, Istituto Oncologico Veneto – IRCCS, Padova, ITALY, 5Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, ITALY.
Background: The current threshold for estrogen receptor (ER) positivity in breast cancer (BC) is ≥1% of positively stained cancer cells by immunohistochemistry. However, tumors with ER-low expression (ER 1-9%) share biological features with triple-negative breast cancer (TNBC), including gene-expression profiles, immune features, and response rates to neoadjuvant chemotherapy and immunotherapy. The clinical benefit of endocrine treatment (ET) in this subgroup of patients remains uncertain. Methods: This systematic review and meta-analysis was conducted in accordance with PRISMA guidelines. We searched Ovid MEDLINE and Embase (last updated February 2, 2025) for randomized trials and observational studies including patients with non-metastatic ER-positive (≥1%)/HER2-negative BC, presenting estimates of ET efficacy or effectiveness within the ER-low subgroup, or enabling outcome comparisons between ER-low and ER≥10% tumors. Manual citation screening was also performed to identify additional eligible studies. Two reviewers independently screened studies and extracted data. Random-effects meta-analyses were performed to derive pooled hazard ratios (HR) with 95% confidence intervals (95% CI) for overall (OS), disease-free survival (DFS) in the two ER strata, and to assess the association between ET use and outcomes within the ER-low group. Heterogeneity was assessed with the I² statistic. Egger’s test was used to evaluate publication bias. Sensitivity analyses included leave-one-out and exclusion of reconstructed data or surrogate endpoints. Results: Of 8,982 records screened, 17 studies were included, comprising 9,711 patients with ER-low tumors and 58,660 with ER≥10% tumors. In pooled analyses, patients with ER-low tumors had worse DFS (HR 2.18 [95% CI: 1.58-3.00]; I² = 78%) and OS (HR 2.08 [95% CI: 1.30-3.31]; I² = 96%) compared to those with ER≥10% tumors. Among patients with ER-low tumors, receipt of endocrine therapy was associated with improved DFS (HR 0.74 [95% CI: 0.55-1.00]; I² = 25%) and OS (HR 0.80 [95% CI: 0.70-0.92]; I² = 21%). No significant publication bias was detected by Egger’s test. Sensitivity analyses confirmed the robustness of the findings.Conclusions: Our systematic review and metanalysis showed that patients with ER-low breast cancer experience a significantly worse prognosis compared to those with ER>10%, and derive a survival benefit from ET. These results suggest that, in addition to therapeutic strategies appropriate for such high-risk profile – such as chemotherapy – the potential benefit of adjuvant endocrine therapy should not be overlooked and ought to be part of the clinical discussion for patients with early-stage ER-low tumors.
Presentation numberPS3-07-20
Prognostic implication of tumor grade in patients with node-negative breast cancer aged ≤ 50 years with 21-gene recurrence scores of 11-25
Soong June Bae, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, Korea, Republic of
S. Bae1, S. Lee2, Y. Kook1, A. Kim1, J. Jeong1, S. Lee3, S. Ahn1; 1Surgery, Gangnam Severacne Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Surgery, Inha University Hospital, Inha University College of Medicine, Incheon, KOREA, REPUBLIC OF, 3Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, KOREA, REPUBLIC OF.
Background The TAILOR-X trial demonstrated that clinical risk scores incorporating histologic grade and tumor size provided additional prognostic stratification in premenopausal, node-negative breast cancer patients with 21-gene recurrence scores (RS) of 16-25. We evaluated the prognostic significance of tumor grade in patients with node-negative, hormone receptor-positive (HR+), and human epidermal growth factor receptor 2 (HER2)-negative breast cancers treated according to RS-guided strategies. Patients and Methods In this multicenter retrospective cohort study, we analyzed 1,944 patients with node-negative, HR+/HER2− breast cancer treated between 2011 and 2020 at two academic centers. The recurrence-free interval (RFI), defined as the time from diagnosis to the first invasive local, regional, or distant recurrence, was analyzed by tumor grade, stratified by age (≤ 50 vs. > 50 years), and RS category. Results Among all patients, a high tumor grade was associated with a shorter RFI (P < 0.001). This association was evident in patients aged ≤ 50 years (P 50 years. In 802 women aged ≤ 50 with intermediate RS (11-25), high-grade tumors were associated with adverse features, including lymphovascular invasion, high Ki-67 expression, and chemotherapy receipt, and demonstrated reduced RFI (P < 0.001). In this subgroup, tumor grade remained an independent prognostic factor in multivariable analysis (hazard ratio, 6.96; 95% confidence interval, 2.69-17.99), particularly in patients who did not receive chemotherapy. Conclusion A high tumor grade was associated with increased tumor recurrence in younger node-negative patients with an intermediate RS. These findings support the incorporation of histological grade into prognostic assessment and treatment decision-making in this group.
Presentation numberPS3-07-21
Validation of the CTS5 as a predictor of late distant recurrence (DR) for HR+ HER2-negative Invasive Lobular Carcinoma (ILC)
Jason A Mouabbi, MD Anderson Cancer Center, Houston, TX
J. A. Mouabbi1, A. S. Raghavendra1, S. Pasyar2, B. L. Roland2, R. A. Mukhtar3, M. Giancarlo4, M. D. Wright4, A. K. Bisen4, A. N. Iheme4, C. H. Barcenas1, V. Valero1, A. Nasrazadani1, B. Lim1, D. L. Ramirez1, A. Hassan4, H. M. Kuerer5, T. Adesoye5, P. R. Paula1, F. Meric-Bernstam6, S. H. Giordano1, J. K. Litton1, R. M. Layman1; 1Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX, 2Biostatistics, MD Anderson Cancer Center, Houston, TX, 3Surgical Oncology, Univeristy of California San Francisco, San Francisco, CA, 4General Oncology, MD Anderson Cancer Center, Houston, TX, 5Breast Surgical Oncology, MD Anderson Cancer Center, Houston, TX, 6Invest. Cancer Therapeutics, MD Anderson Cancer Center, Houston, TX.
Background: ILC is a distinct subtype of breast cancer characterized by unique histopathological features and a higher risk of late distant recurrence (DR) compared to invasive ductal carcinoma (IDC). Accurate risk stratification is critical for optimizing adjuvant management. While CTS5 is a widely used and readily available tool for predicting late DR (after 5 years) in HR+/HER2- breast cancer, it has not been evaluated and validated in ILC. We report the first validation of CTS5 in ILC and compare its performance to the recently developed ILC-specific prognostic model (MDA-iLobulaRX).Methods: We retrospectively analyzed data from patients with stage I-III HR+/HER2- ILC treated at MD Anderson Cancer Center, between 1980-2020, using a prospectively curated database. Two models were evaluated for predicting late DR: (1) MDA-iLobulaRX, incorporating age, ER%, tumor grade, presence of LCIS, ILC histology, lymph node status, tumor size, and adjuvant endocrine therapy (tamoxifen or aromatase inhibitor [AI]); and (2) CTS5, including age, tumor size and grade, and nodal status. Patients with less than five years of follow-up were excluded to ensure adequate time at risk for late DR. Additionally, patients who did not receive endocrine therapy were excluded. Model performance was assessed using: (1) Akaike Information Criterion (AIC) to compare model fit; (2) Harrell’s concordance index (c-index) to assess discrimination; and (3) Receiver operating characteristic (ROC) curves with area under the curve (AUC), based on a binary classification of recurrence (yes/no) after 5 years for clinical interpretability.Results: The cohort included 2,253 women with a median follow up time of 10.3 years with a range of 5 to 40 years. The median age was 57 years, median tumor size was 23 mm, 52% were node-positive and 67% were postmenopausal. Classical ILC was the predominant subtype (90%) compared to 10% non-classical subtypes. Endocrine therapy included tamoxifen (43%), non-steroidal AIs (55%) and steroidal AI (2%). Model 1 (MDA-iLobulaRX) had a slightly better fit (AIC: 9115 vs 9157) while both models demonstrated comparable discrimination (c-index 0.698 for model 1 vs 0.704 for model 2) and AUC (0.74 for model 1 and 0.75 for model 2).Conclusion: CTS5 is a valid, readily accessible tool for predicting late DR in HR+/HER2- ILC, with performance comparable to ILC-specific models. While CTS5 does not guide treatment or surveillance decisions, it may support risk stratification discussions in clinical practice and offers a pragmatic tool for estimating late recurrence risk in this population.
Presentation numberPS3-07-22
Correlation between MRI background parenchymal enhancement and serum estradiol in premenopausal patients on neoadjuvant endocrine therapy for breast cancer
Sarah Choi, University of California, San Francisco, San Francisco, CA
S. Choi1, N. Onishi2, P. Metanat2, R. Mukhtar1, K. Giridhar3, M. P. Goetz3, L. Huppert4, A. Elias5, O. Olopade6, I-SPY2 Imaging Working Group, I-SPY2 Investigator Network, L. Esserman1, J. Chien4, N. Hylton2; 1Department of Surgery, University of California, San Francisco, San Francisco, CA, 2Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 3Department of Oncology, Mayo Clinic, Rochester, MN, 4Department of Medicine, University of California, San Francisco, San Francisco, CA, 5Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 6Department of Medicine, University of Chicago, Chicago, IL.
Purpose Background parenchymal enhancement (BPE), the contrast enhancement of normal fibroglandular tissue on dynamic contrast-enhanced breast MRI, is hormonally dependent and fluctuates with the menstrual cycle, lactation, and pregnancy. In patients with breast cancer, both endocrine therapy and chemotherapy can decrease BPE levels during treatment. Notably, a lack of BPE suppression was associated with inferior response after neoadjuvant chemotherapy in hormone receptor-positive (HR+) cancer. In HR+ breast cancer, BPE may be particularly relevant as estrogen fuels the tumor’s growth. However, there is limited clinical research directly comparing BPE and serum estradiol levels. This study aimed to investigate 1) the correlation between BPE and serum estradiol levels in treatment-naïve premenopausal patients with HR+ breast cancer and 2) the association between BPE suppression and serum estradiol suppression during neoadjuvant endocrine therapy (NET). Methods We retrospectively reviewed data from premenopausal patients enrolled in the Endocrine Optimization Pilot (EOP), a sub-study of the I-SPY 2 TRIAL, which tests new endocrine strategies in patients with stage 2/3, MammaPrint (MP) low-risk (index 0 to 1), HR+/HER2-negative (HER2-) breast cancer. Patients with MP High1 (index -0.57 to 0) tumors were eligible if clinically node negative. This study included 58 EOP patients who had both serial serum estradiol levels (drawn monthly) and BPE data available. These patients started ovarian function suppression (OFS) at various times based on their assigned treatment regimen: before NET start, n=28; at cycle 1, n=16; at cycle 2, n=14. The average estradiol levels over six months on NET were categorized into two groups: suppressed to the postmenopausal range (<10 pg/mL) or non-suppressed (≥10 pg/mL). MRI scans at baseline (before NET) and pre-surgery (after six months of NET) were analyzed. BPE was calculated on the contralateral breast as the mean early (~150s post-contrast injection) percent enhancement of the central 50% of the axial slices. The Spearman’s correlation coefficient and the Wilcoxon rank-sum test were used to examine the relationship between BPE and estradiol. Results Treatment-naïve estradiol levels were available for 25 of the 58 patients, with the median [1st, 3rd quartile] of 82.7 [32.4, 250] pg/mL. A positive correlation was observed between treatment-naïve estradiol and baseline BPE, with a correlation coefficient of ρ = 0.74 (p < 10-5). There was no significant difference in the treatment-naïve estradiol levels between patients who later showed estradiol suppression to the menopausal range during NET (n=14 [56 %]) and those who did not (n=11 [44 %]) (81.3 pg/mL versus 86.5 pg/mL, respectively (p=0.39)). During NET, estradiol levels were suppressed to the postmenopausal range in 26 of the 58 patients (45%). Estradiol suppression was significantly associated with greater reduction in post-treatment BPE: -42.7% [-54.3, -21.8] BPE reduction for the suppressed estradiol group versus -15.0% [-40.6, 6.85] for the non-suppressed estradiol group (p=0.002). Conclusion This study demonstrates the estrogen dependency of BPE in premenopausal patients with HR+/HER2- breast cancer. Baseline estradiol levels positively correlated with baseline BPE, and the suppression of on-treatment estradiol was associated with greater BPE suppression following NET. Our findings further support exploration of the use of BPE as a reflection of estradiol level under NET and OFS in addition to other MRI biomarkers such as functional tumor volume. These complementary imaging biomarkers could independently reflect both non‑malignant and tumor-specific biological processes to improve predicted performance.
Presentation numberPS3-07-24
Breast Cancer Index re-stratifies 21-gene assay risk groups for risk of recurrence and extended endocrine therapy benefit: Final Analysis of the BCI Registry Study
Valerie Chuy, University of Illinois Hospital, Chicago, IL
V. Chuy1, N. Siuliukina2, B. O’Neal3, A. K. Anderson3, K. Koskins1, Y. K. Zhang3, K. Treuner2, V. K. Gadi1; 1Department of Medicine, University of Illinois Hospital, Chicago, IL, 2R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 3R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA.
Background Patients with hormone receptor-positive (HR+) early-stage breast cancer face a prolonged risk of recurrence even after adjuvant endocrine therapy. The Breast Cancer Index (BCI) is a genomic classifier that provides the risk of overall (0-10y) and late (post 5 years) distant recurrence (DR) and predicts the likelihood of benefit from extended endocrine therapy (EET) based on the H/I (HOXB13/IL-17BR) ratio. The 21-gene assay (Recurrence Score; OncotypeDX) provides the 9-year risk of DR and the benefit from adjuvant chemotherapy (CT) in HR+/HER2− breast cancer. Here, we investigated the concordance between BCI score and H/I ratio versus RS in HR+/HER2- patients from the full cohort of the BCI Registry study. Methods The BCI Registry study is a prospective, multi-institutional study that enrolled over 3400 patients to determine the long-term clinical outcome, decision impact and medication adherence in HR+ early-stage breast cancer patients receiving BCI testing as part of routine clinical care. Based on TAILORx and RxPONDER results, RS stratified patients into four risk groups: Age ≤ 50, No CT benefit (RS ≤ 15); Age ≤ 50, CT benefit (RS >15); Age > 50, No CT benefit (RS≤25); and Age > 50, CT benefit (RS >25). Kendall’s correlation coefficient (R) was used to estimate the correlation between BCI score, H/I ratio and RS as continuous variables. Cohen’s kappa was used to measure the agreement between H/I and RS categories. Results BCI and RS results were reported for 1238 patients (76% T1; 58% grade II; 84% N0). The H/I ratio classified 834 patients (67%) as H/I-Low and 404 (33%) as H/I-High, respectively, while BCI stratified 649 patients (52%) as low-risk and 589 (48%) as high-risk for late DR. RS classified 45 patients (4%) as Age ≤ 50, No CT benefit, 54 (4%) as Age ≤ 50, CT benefit, 976 (79%) as Age > 50, No CT benefit and 163 (13%) as Age > 50, CT benefit. When analyzed as continuous variables, the BCI score and H/I ratio showed a weak correlation with RS with R=0.2 and R=0.18, respectively. Based on categorical groups, H/I had low concordance with RS for younger women (Age ≤ 50, Cohen’s kappa=0.08, p=0.29) but showed some correlation with RS for older women (Age > 50, Cohen’s kappa=0.21, p<0.001). Among those older than Age 50, the H/I ratio identified 29% of those with no predicted CT benefit as likely to benefit from EET while 39% of those with predicted CT benefit as not likely to benefit from EET. Table 1. Cross-tabulation of RS and H/I groups
| RS risk categories | H/I-Low | H/I-High | |||
| Age ≤ 50, No CT benefit | 38 (84%) | 7 (16%) | |||
| Age ≤ 50, CT benefit | 41 (76%) | 13 (24%) | |||
| Age > 50, No CT benefit | 692 (71%) | 284 (29%) | |||
| Age > 50, CT benefit | 63 (39%) | 100 (61%) |
Conclusion This comparative analysis between BCI and the 21-gene assay revealed a low concordance between the BCI score and H/I ratio with RS risk categories. The H/I ratio consistently re-stratified RS risk categories into distinct H/I-Low and -High groups, confirming that prediction of CT benefit does not equate to prediction of EET benefit.
Presentation numberPS3-07-25
Clinical Characteristics of Chinese Patients with Early Invasive Breast Cancer Tested with 70-gene signature: Comparison with the MINDACT Cohort
Hong Liu, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
H. Liu1, G. Liu2, W. Zhou3, N. Li4, Y. Wang5, H. Hu6, Y. Sheng7, D. Zhou8, C. Wang9, G. Liu10; 1Department of Breast Surgery, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA, 2Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA, 3Department of Breast Surgery, Jiangsu Province Hospital, Nanjing, CHINA, 4Department of Thyroid, Breast and Vascular Surgery, The First Affiliated Hospital of Air Force Medical University, Xi’an, CHINA, 5Department of Breast Surgery, Shandong Cancer Hospital, Jinan, CHINA, 6Department of Breast Surgery, Shenzhen People’s Hospital, Shenzhen, CHINA, 7Department of Thyroid and Breast Surgery, Changhai Hospital, Shanghai, CHINA, 8Department of Breast Surgery, Foshan First People’s Hospital, Foshan, CHINA, 9Department of Breast Surgery, Shanghai Second People’s Hospital, Shanghai, CHINA, 10Department of Surgery, Anyang Tumour Hospital, Anyang, CHINA.
Background: The 70-gene signature (MammaPrint®) predicts distant metastasis risk in early invasive breast cancer, especially HR+/HER2- patients with 0-3 nodes. Despite guideline inclusion, its performance in Chinese populations needs validation. This study compares clinicopathological features between a Chinese cohort and MINDACT.Methods: Retrospective analysis of 1900 Chinese early breast cancer patients tested with MammaPrint®. Clinical risk (C-High [C-H] or C-Low [C-L]) used modified Adjuvant! Online. Genomic risk (MammaPrint®): High Risk (G-H) or Low Risk (G-L), subcategorized by quartile (Ultra-Low [G-UL], Low but Not Ultra-Low [G-LNUL], High but Not Ultra-High [G-H1], Ultra-High [G-H2]). Compared G-H vs G-L within Chinese cohort, and key characteristics (age, T-stage, grade, nodal status) across overall population and four risk groups (C-H/G-H, C-H/G-L, C-L/G-H, C-L/G-L) with MINDACT (n=6693). Categorical variables were analyzed using Fisher’s exact test, with statistical significance defined as a two-sided P < 0.05.Results:Within the Chinese Cohort (G-H vs. G-L): Of 1,900 patients, 1,120 (58.9%) were G-L and 780 (41.1%) were G-H (distribution: G-UL 11.0%, G-LNUL 48.0%, G-H1 38.0%, G-H2 3.0%). Significant differences (p 14%: 51.2% (G-L) vs. 83.1% (G-H).Chinese Cohort vs. MINDACT (Overall & Risk Groups):Overall, Chinese patients were younger (<50 years: 40.3% vs. 33.3%, p < 0.001), had higher T2 stage (41.6% vs. 27.2%, p < 0.001), higher Grade 2 frequency (75.9% vs. 49.1%, p < 0.001), and slightly higher node positivity (23.7% vs. 21.0%, p = 0.013). Group-specific differences (p < 0.001) included:C-H/G-L: Higher Grade 2 (86.8% vs. 64.2%), higher T2 (69.1% vs. 54.4%), lower T1 (27.2% vs. 41.8%)C-H/G-H: Higher T2 (67.7% vs. 46.7%), predominant Grade 2 (60.1% vs. 23.3%), lower Grade 3 (37.5% vs. 75.6%)C-L/G-H: Younger age (<50 years: 49.9% vs. 30.1%), minimal advanced disease (T3: 0%; G3: 2.5%)C-L/G-L: Younger age (<50 years: 45.9% vs. 29.1%); predominantly T1, Grade 2, node-negative tumorsClinical-genomic discordance (C-H/G-L) was 24.0% in China, consistent with MINDACT (23.0%); overall concordance was 57.1%.Conclusion: Significant differences exist between Chinese and MINDACT cohorts in age (younger), T-stage (higher T2), and grade (higher Grade 2), persisting across MammaPrint® risk subgroups. These findings underscore the necessity of validating the 70-gene signature’s predictive value for chemotherapy decisions using China-specific data. Survival analysis of this large cohort and prospective studies will further evaluate its risk stratification performance and clinical utility in Chinese early breast cancer patients.
Presentation numberPS3-07-26
Precision neoadjuvant treatment with Artificial Intelligence assisted subtyping in HR+/HER2- breast cancer: a randomized, open-label, phase 2 trial FASCINATE-N
Jun-Jie Li, Fudan University Shanghai Cancer Center, Shanghai, China
J. Li, L. Ma, P. Ji, L. Chen, J. Wu, G. Liu, Y. Hou, G. Di, Z. Hu, Y. Jiang, K. Yu, L. Fan, Z. wang, Z. Shao; Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, CHINA.
Identification of effective neoadjuvant personalized treatment in hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2−) breast cancer remains a topic of ongoing research. We previously performed next-generation sequencing combined with metabolomics and proteomics on HR+/HER2- breast cancer sample. Using large-scale multi-omics data and similarity network fusion (SNF), we classified HR+/HER2- breast cancer into four novel subtypes: SNF1 (canonical luminal), SNF2 (immunogenic), 126 SNF3 (proliferative), and SNF4 (RTK-driven). Then we developed a digital pathology based on artificial intelligence (AI) assisted- classification approach employing convolutional neural networks derived from pathology whole-slide images (WSIs) to classify molecular subtypes. In this phase 2 trial, patients with stage II-III HR+/HER2- breast cancer were categorized into endocrine-based and targeted-based group based on the proposed similarity network fusion (SNF) subtyping by AI assisted digital pathology classification and randomly assigned (1:1) to receive precision or control treatment. Precision treatments included six cycles of dalpicilib (CDK4/6 inhibitor) plus endocrine therapy every 4 weeks for endocrine-based group and six cycles of targeted therapy (SHR-1316 [PD-L1 inhibitor] for SNF2, fuzuloparib [PARP1 inhibitor] for SNF3 and apatinib [VEGFR inhibitor] for SNF4) plus nab-paclitaxel and carboplatin every 4 weeks for targeted-based group. Control treatment was nab-paclitaxel plus carboplatin every 4 weeks for six cycles. The primary endpoint was pathological complete response (pCR). Among 251 randomized patients, SNF subtyping classified 49 patients (19.5%) into the endocrine-based group and 202 (80.5%) into the targeted-based group. Patients receiving precision treatment had significantly higher pCR rate (11.1% [14/126; 90% CI, 6.8-16.8] vs. 4.0% [5/125; 90% CI, 1.6-8.2]; P = 0.033). In the endocrine-based group, no pCR events were observed with endocrine treatment, compared with one pCR event in the control group (P = 0.490). In the targeted-based group, the pCR rate was significantly higher with precision treatment than with control (13.9% vs. 4.0%; P = 0.026). Grade 3 or higher treatment-related adverse events occurred similar in both treatment groups. No treatment-related deaths occurred. Our findings showed precision therapy could improve the benefit of neoadjuvant therapy for HR+/HER2- breast cancer patients. NET plus CDK4/6 inhibitor was verified to replace chemotherapy with better tolerance in the endocrine-base group while precision therapy was proved promising clinical activity and manageable safety profile in the targeted-based group. (ClinicalTrials.gov: NCT05582499).
| EB-study(n=25) | EB-control(n=24) | p | TB-study(n=101) | TB-control(n=101) | p | |
| CR+PR | 18 | 17 | 0.928 | 91 | 78 | 0.013 |
| MP4&5 | 2 | 3 | 0.667 | 40 | 16 | 0.001 |
| RCB0&1 | 1 | 2 | 0.609 | 28 | 11 | 0.002 |
| change in Ki67 | -12 | -10 | 0.854 | -20 | -14 | 0.002 |
Presentation numberPS3-07-27
Identifying Premenopausal Patients with Early-Stage Hormone Receptor-Positive Node-Negative Breast Cancer at Minimal Risk of Distant Recurrence by Breast Cancer Index
Ruth O’Regan, University of Rochester, Rochester, NY
R. O’Regan1, Y. Ren2, Y. Zhang3, G. F. Fleming4, P. A. Francis5, O. Pagani6, B. A. Walley7, R. Kammler8, P. Dell’Orto9, G. Viale10, S. Loi5, M. Colleoni10, K. Treuner3, M. Regan2; 1Medicine, University of Rochester, Rochester, NY, 2IBCSG Statistical Center, Dana-Farber Cancer Institute, Boston, MA, 3R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 4Hematology/Oncology, The University of Chicago Medical Center, Chicago, IL, 5Oncology, Peter MacCallum Cancer Center, Melbourne, AUSTRALIA, 6Medicine, Interdisciplinary Cancer Service Hospital Riviera-Chablais Rennaz, Geneva, SWITZERLAND, 7Oncology, University of Calgary, Calgary, AB, CANADA, 8Translational Research, ETOP IBCSG Partners Foundation, Bern, SWITZERLAND, 9Pathology and Laboratory Medicine, Instituto Europeo Di Oncologia IRCCS, Milan, ITALY, 10Medicine, Instituto Europeo Di Oncologia IRCCS, Milan, ITALY.
Background: Adjuvant endocrine therapy for 5 years with ovarian function suppression (OFS) is recommended for premenopausal breast cancer patients with endocrine-sensitive tumors at high enough risk of recurrence, e.g. younger age, high grade tumor or nodal involvement. Genomic risk assessment may help to personalize OFS-based treatment selection. The Breast Cancer Index (BCI) test was previously confirmed as prognostic in premenopausal patients in the SOFT and TEXT trials [1]. For hormone receptor-positive (HR+), node-negative (N0) breast cancers, an adjusted BCI model with an additional cutpoint was recently developed that re-stratified postmenopausal patients with BCI low risk breast cancer into BCI minimal or low risk; where the new minimal risk group had a 10-year risk of distant recurrence of <5% [2]. Here, we evaluated the prognostic performance of the adjusted BCI model in premenopausal patients with HR+ N0 breast cancer in the SOFT and TEXT trials; the median follow-up was 12 and 13 years, respectively.Methods: The analysis included 1110 N0 patients from SOFT (N = 383 assigned tamoxifen; N = 727 assigned exemestane+OFS or tamoxifen+OFS), of whom 70 experienced DR; and 915 N0 patients from TEXT (assigned exemestane+OFS or tamoxifen+OFS), of whom 64 had DR. The new BCI cutpoint, along with the current cutpoints, was used to re-stratify N0 patients as minimal or low risk, with those in intermediate or high risk groups remaining as previously classified. The primary endpoint was distant recurrence-free interval (DRFI). 10-year DR was reported as one minus Kaplan-Meier estimate of DRFI with pointwise 95% confidence intervals (CI).Results: The adjusted BCI model re-classified 18% and 20% of N0 patients in SOFT and TEXT into the BCI minimal risk group; 43% and 38% remained classified in the low-risk group, respectively. Of those with minimal-risk BCI tumors in SOFT, 86% were ≤2cm and 51% were grade 1 tumors, 85% did not receive chemotherapy, 62% were age ≥45y. Of those with minimal-risk BCI tumors in TEXT, 80% were ≤2cm and 35% were grade 1 tumors, 73% did not receive chemotherapy, 51% were age ≥45y. 10-yr DR estimates are tabulated.Conclusions: This study confirms the prognostic ability of the adjusted BCI model to identify a subset of premenopausal patients whose estimated 10-year risk of DR was <5%. These findings provide clinicians and premenopausal patients with additional information for discussions about potential benefits and side effects of adjuvant endocrine therapy regimens. References: [1] (O’Regan et al; PMID 39145953). [2] (Jilderda et al; PMID 40145943)
| BCI Risk Group, Estimated 10-yr DR (95% CI) | |||||||||
| N0 Population (N. pts) | Minimal | Low | Intermediate | High | |||||
| SOFT (1110) | 2.3% (0.9-6.0) | 4.1% (2.6-6.5) | 8.4% (5.4-13.0) | 10.4% (6.8-15.9) | |||||
| SOFT assigned tam (383) | 3.5% (0.9-13.3) | 7.2% (4.0-13.1) | 9.3% (4.7-17.8) | 11.4% (5.6-22.6) | |||||
| SOFT assigned exe+OFS or tam+OFS (727) | 1.7% (0.4-6.6) | 2.7% (1.4-5.3) | 7.9% (4.3-14.2) | 9.9% (5.7-16.8) | |||||
| TEXT assigned exe+OFS or tam+OFS (915) | 2.0% (0.7-6.2) | 4.6% (2.8-7.7) | 9.0% (5.7-13.9) | 13.1% (8.5-19.7) |
Presentation numberPS3-07-28
Prognostic performance of Breast Cancer Index in patients with early-stage HR+ HER2+ breast cancer (BC) treated with adjuvant trastuzumab: NCCTG N9831 (Alliance)
Saranya Chumsri, Mayo Clinic, Jacksonville, FL
S. Chumsri1, E. Liu2, K. Edwards3, Y. Wen4, Y. Zhang5, A. K. Anderson6, A. H. Partridge7, L. Carey8, K. Ballman3, E. Perez9, K. Treuner5, M. Goetz2, E. Thompson10; 1Oncology, Mayo Clinic, Jacksonville, FL, 2Oncology, Mayo Clinic, Rochester, MN, 3Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, 4Alliance, University of Chicago, Chicago, IL, 5R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 6Oncology, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 7Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 8Oncology, The University of North Carolina, Chapel Hill, NC, 9Hematology/Oncology, Mayo Clinic, Jacksonville, FL, 10Department of Cancer Biology, Mayo Clinic, Jacksonville, FL.
Background: The Breast Cancer Index (BCI) is clinically validated to stratify patients with hormone receptor-positive (HR+), early-stage BC based on the risk of overall (0-10 years [y]) and late (5-10 y) distant recurrence (DR) with N0/N1 lymph node involvement. Validation of BCI in patients with HR+ HER2+ BC has been limited, especially in patients treated with adjuvant trastuzumab-based chemotherapy. The HOXB13 and IL17BR genes constitute the predictive component of BCI and are known to exhibit an ER-dependent correlation pattern with HER2. We evaluated the prognostic performance of BCI in patients with HR+ HER2+ disease enrolled in the NCCTG N9831 (Alliance) trial treated with the combination of chemotherapy and trastuzumab. Methods: All available tumor specimens from patients with HR+ HER2+ N0/N1 BC in arms B (sequential trastuzumab) and C (concurrent trastuzumab) of the trial were included. BCI testing was performed blinded to clinical outcomes. The primary endpoint was distant recurrence-free interval (DRFI). BCI (N0) and BCIN+ (N1) risk groups were calculated based on prespecified cut points. Kaplan-Meier analysis and log-rank testing were used to assess the prognostic significance of BCI/BCIN+ risk groups based on DR. Cox proportional hazard models with and without clinical covariates were used to estimate hazard ratios (HRs), and the associated 95% confidence intervals (CIs). Results: A total of 454 HR+ patients (234 arm B, 220 arm C), including 69 N0 and 385 N1 patients, were analyzed (median age was 47 y, 85% N1, 55% T2, 63% G3). BCIN+ scores were available for 378 of the 385 N1 patients due to missing tumor grade for 7 patients. The median follow-up was 12.7 years. In the overall cohort, BCI/BCIN+ stratified 51 (11%), 24 (5%) and 372 (83%) patients as low-, intermediate- and high-risk with 10-y DRFI rates of 96% (95% CI: 84-99%), 100% (100-100%) and 89% (86-92%), respectively. Since the low and intermediate risk groups had few patients and a similar risk of 10-y DRFI, they are combined into a single low-risk group hereafter. Thus, for overall 10-y DR, 75 (17%) and 372 (83%) of patients were classified by BCI/BCIN+ as low and high risk with 10-y DRFI rates of 97% (95% CI: 89-99%) and 89% (95% CI: 86-92%), respectively (HR=4.50, 95% CI: 1.09-18.61; p = 0.038). BCI/BCIN+ remained statistically significant in the multivariable analysis, adjusting for age, treatment, and nodal status (HR=7.54, 95% CI: 1.34-42.64; p = 0.022). For 69 N0 patients, BCI stratified 54 (78%) and 15 (22%) patients as low and high risk with 10-y DRFI rates of 100% (95% CI, 100-100%) and 77% (95% CI, 44-92%), respectively (p = 0.012). For 378 N1 patients, BCIN+ stratified 21 (6%) and 357 (94%) of patients as low and high risk with 10-y DRFI of 90% (95% CI, 66-97%) and 90% (95% CI, 86-93%), respectively (p = 0.07). Of the 21 patients classified as BCIN+ low-risk, two had BCIN+ scores of 6.75 and 6.73 in close proximity to the cut point (6.93) and experienced DR after 5 years, resulting in the same 10-y DRFI for both BCIN+ low- and high-risk patients. Conclusions: BCI is significantly prognostic for the risk of overall DR (0-10 y) in patients with HR+ HER2+ disease treated with adjuvant trastuzumab-based chemotherapy. Optimization of BCI cut points may further improve prognostic performance in HR+ HER2+. Additional validation in other HR+ HER2+ cohorts treated in the modern setting with anti-HER2-based therapy is warranted, given the small sample size of N0 patients and relatively low DR rate in the late recurrence cohort. In summary, these results provide further evidence of BCI’s utility to support clinical decision-making for adjuvant endocrine therapies for HR+ HER2+ BC. Support: U10CA180821, U10CA180882, U24CA196171, Genentech; Clinicaltrials.gov Id: NCT00005970; https://acknowledgments.alliancefound.org.
Presentation numberPS3-07-29
Real-world outcomes by Estrogen Receptor Expression Subgroups in Patients with HER2-negative Breast Cancer: A Population-based Cohort Study
qiao yang, Karolinska Institutet, Stockholm, Sweden
q. yang, X. Chen, J. Hartman, B. Acs; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SWEDEN.
Background:In Sweden, patients with breast cancer (BC) expressing 10-100% estrogen receptor (ER) are treated as ER-positive, whereas those with ER-low (1-9%) BC are managed as triple-negative disease (TNBC). The distinct clinicopathological and prognostic significance of ER-subgroups, especially low and intermediate ER-expressing BC, remains incompletely characterized. We aimed to describe real-world patient and tumor characteristics, treatment patterns and overall survival across ER subgroups in a Swedish population-based cohort of patients with HER2-negative BC. Methods:In this nationwide cohort study, all Human Epidermal growth factor Receptor 2 (HER2)-negative BC patients diagnosed in Sweden (2007-2023) were stratified by ER status: ER-zero (0%), ER-low (1-9%), ER-sublow (10-29%), ER-medium (30-59%; ER-med), and ER-high positive (60-100%; ER-high). Cohorts were analyzed separately for adjuvant and neoadjuvant settings. Overall survival (OS) was evaluated using Kaplan-Meier analysis and multivariable Cox proportional hazards models. Models were adjusted for age, year of diagnosis, tumor stage, grade, and treatment modality. Results:We analyzed 75,473 eligible patients, stratified into adjuvant (n = 70,159: ER-zero = 4,559; ER-low = 536; ER-sublow = 504; ER-med = 1,425; ER-high = 63,135) and neo-adjuvant cohorts (n = 5,314; ER-zero = 1,766; ER-low = 205; ER-sublow = 132; ER-med = 145; ER-high = 3,066). ER-sublow demonstrated significantly worse OS than higher ER expression subgroups in the adjuvant cohort. In univariable analysis, ER-sublow patients had 1.27-fold increased mortality risk versus ER-med (hazard ratio (HR) = 1.27, 95% confidence interval (CI) = 1.04-1.55, p = 0.017) and 1.63-fold increased risk versus ER-high (HR = 1.63, 95% CI = 1.37-1.93, p < 0.001). ER-sublow showed no significant difference in OS compared to ER-zero (HR = 0.86, 95% CI = 0.72-1.03, p = 0.106) or ER-low (HR = 1.12, 95% CI = 0.88-1.43, p = 0.347). Multivariable analysis confirmed significantly poorer survival for ER-sublow versus ER-high (adjusted HR = 1.58, 95% CI = 1.33-1.88, p < 0.001). Notably, ER-med demonstrated worse OS compared to ER-high in both univariable analysis (HR = 1.19, 95% CI = 1.08-1.32, p < 0.001) and multivariable analysis (adjusted HR = 1.26, 95% CI = 1.13-1.39, p 0.05). Conclusion:ER-sublow (10-29%) patients have characteristics and prognosis similar to patients with ER-low (1-9%) and ER-zero BC, while patients with ER-med (30-59%) BC have inferior outcomes than patients with ER-high (60-100%) BC. ER expression should be considered as a continuum rather than a binary (positive/negative) biomarker in clinical decision-making.
Presentation numberPS3-07-30
Genomic Drivers of Racial Disparity in HR-Positive Early Breast Cancer
Jennifer Vu, University of Illinois Chicago, Chicago, IL
J. Vu1, J. Gor1, Y. Simons2, G. E. Chlipala3, M. W. Geise4, A. Ibraheem1, O. C. Danciu1, V. K. Gadi1, M. Singh5, K. F. Hoskins1, G. Mohapatra5; 1Division of Hematology and Oncology, University of Illinois Chicago, Chicago, IL, 2Department of Medicine, NYU Grossman School of Medicine, New York, NY, 3Research Informatics Core, University of Illinois Chicago, Chicago, IL, 4Data Integration Shared Resource, University of Illinois Cancer Center, Chicago, IL, 5Department of Pathology, University of Illinois Chicago, Chicago, IL.
Background: Nearly all breast cancer disparities research has focused on the higher incidence of triple-negative breast cancer (TNBC) among Black women despite the fact that hormone receptor-positive (HR+) breast cancer is three times more common than TNBC, and Black women with HR+, early-stage breast cancer (EBC) are 80% more likely to die and 30% more likely to have biologically aggressive HR+ tumors compared with Non-Hispanic White (NHW) women (Hoskins et al, JAMA Oncol 2021). Genomic subtyping studies of HR+ EBC reveal that compared with NHW women, Black women have a higher prevalence of the aggressive luminal B and HR+ basal subtypes, but the molecular mechanisms underlying this racial difference are unknown. We analyzed genomic profiles of HR+/HER2- primary tumors in a diverse cohort of EBC patients in order to identify molecular drivers of the aggressive HR+ tumor phenotype that disproportionately affects Black women. Methods: A series of cases of HR+/HER2- EBC treated at the University of Illinois Chicago (UIC) between 2018-2021 underwent NGS profiling of the primary tumor in the Laboratory of Genomic Medicine at UIC for DNA and RNA somatic driver alterations with the Oncomine Comprehensive Assay v3 (OCAv3), which includes 161 driver genes and detects SNVs, CNVs, indels, and gene fusions. Enrichment of alterations according to self-identified race was tested with Fisher’s exact tests and odds ratios with 95% confidence intervals (CI). Publicly available TCGA data was downloaded from the Genomic Data Commons portal (https://portal.gdc.cancer.gov) to validate findings. Indicators of neighborhood disadvantage were obtained by geocoding residential addresses and linking to census tract measures of the social and environmental context to examine associations between tumor genomic profile and neighborhood contextual factors. Results: In total, 128 primary tumors were evaluated (Black, n=72; non-Black, n=56). Results for the most prevalent alterations are presented in the table. Inactivating SNVs and indels in ARID1A and ATM were more common in tumors from Black patients. Results of analyses for survival and for associations between neighborhood- and individual-level indicators of disadvantage and tumor genomic profile, and validation with TCGA data will be presented. Conclusion: Inactivation of ARID1A (a key component of the SWI/SNF chromatin remodeling complex) in HR+ breast cancer cells leads to loss of luminal identity, switch to an estrogen-independent basal-like phenotype, and resistance to endocrine therapy (Xu et al. Nature Genetics 2020). Enrichment of ARID1A somatic driver mutations in HR+/HER2- tumors from Black patients with EBC provides a mechanistic explanation for the higher rate of HR+ basal tumors in that population. This finding along with the higher rate of ATM alterations can inform development of targeted therapeutic strategies designed to mitigate racial disparity in EBC survival.
| Gene | Variant type | Black patients (n=72) | Non-Black patients (n=56) | Odds ratio (95% CI) | p-value |
| ARID1A | SNV/Indel | 16.70% | 3.60% | 5.4(1.24-24.9) | 0.02 |
| ATM | SNV/Indel | 12.50% | 1.80% | 7.9 (1.19-87.6) | 0.04 |
| CCND1 | CNV | 15.30% | 16.70% | 0.94 (0.36-2.3) | >.99 |
| ERBB2 | SNV | 12.50% | 5.40% | 2.5 (0.7-8.9) | 0.22 |
| PIK3CA | SNV | 51.00% | 71.00% | 0.42 (0.20-0.90) | 0.03 |
Presentation numberPS3-08-01
Validation of the Modified Preoperative Endocrine Prognostic Index (mPEPI) in a single institution cohort of patients with invasive lobular carcinoma
Anna Vertido, University of California San Francisco, San Francisco, CA
A. Vertido1, A. Quirarte1, J. A. Mouabbi2, J. Chien3, R. A. Mukhtar1; 1Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, CA, 2Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 3Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA.
Validation of the Modified Preoperative Endocrine Prognostic Index (mPEPI) in a single institution cohort of patients with invasive lobular carcinoma Background: Neoadjuvant endocrine therapy (NET) is increasingly used in the management of early-stage estrogen receptor-positive (ER+), HER2-negative (HER2-) breast cancer to downstage tumors and evaluate treatment response. The Preoperative Endocrine Prognostic Index (PEPI) score is a validated tool for estimating recurrence risk and informing adjuvant chemotherapy decisions following NET. Prior trials, including IMPACT and Z1031, have shown that patients with a low PEPI score (PEPI=0) have a significantly lower risk of recurrence compared to those with a higher score (PEPI >0). The modified PEPI (mPEPI) score, which omits the ER Allred score, was also developed for patients treated with selective estrogen receptor degraders (SERDs) and is currently being evaluated in the ALTERNATE trial. However, neither the PEPI nor mPEPI score has been studied specifically in patients with invasive lobular carcinoma (ILC), a histologic subtype accounting for 10–15% of all breast cancers and known for its unique clinical features. We evaluated the ability of the mPEPI score to predict recurrence-free survival (RFS) in patients with ILC. Methods: We retrospectively analyzed a prospectively maintained institutional ILC database to identify patients with stage I-III ER+, HER2- ILC treated with NET. An mPEPI score for each patient was calculated as 0-3, with 1 point given for each of the following factors: pathologic tumor size >5 cm, positive pathologic nodal status, and post-treatment Ki-67 >2.7%. RFS was analyzed using the Kaplan-Meier method and log-rank tests, with patients grouped by mPEPI score (0 versus 1+; 0–1 versus 2–3) and stratified by menopausal status. Statistical analyses were conducted using R software. Survival analyses were right-censored at 100 months. Results: We identified 124 patients with ER+, HER2- ILC who received NET and had available post-treatment Ki-67 data. Of these, 81 (65.3%) patients were postmenopausal and 43 (34.7%) premenopausal. Eleven (8.9%) patients had a prior history of ipsilateral breast cancer, and 15 (12.1%) had prior exposure to endocrine therapy. At surgery, the mean tumor size was 4.5 cm (range: 0.11-13.7 cm), and most patients were node-negative (n=78, 62.9%). Tumor grade was predominantly grade 1 (29.0%) or grade 2 (67.7%). Most patients received an aromatase inhibitor (n=88, 71.0%), including five (4.0%) with a CDK4/6 inhibitor and six (4.8%) with ovarian suppression. Thirteen (10.5%) patients received a SERD-based regimen, while 22 (17.7%) received tamoxifen alone. mPEPI scores were distributed as follows: score 0 in 19 (15.3%) patients, score 1 in 51 (41.1%), score 2 in 39 (31.5%), and score 3 in 15 (12.1%). Fourteen recurrence events occurred during follow-up (median 41.0 months). When comparing patients with mPEPI 0–1 versus 2–3, RFS at 100 months was significantly higher in the 0–1 group (82.2% versus 73.6%, p=0.04). No significant differences were observed when comparing mPEPI 0 versus 1+, or when stratified by menopausal status regardless of grouping. Conclusions: In this retrospective cohort of ILC patients treated with NET, the mPEPI score demonstrated a significant association with RFS when grouped as 0–1 versus 2–3. These findings suggest that the mPEPI score may help stratify recurrence risk in patients with ILC. Larger studies are warranted to validate the prognostic utility of the mPEPI score in ILC.
Presentation numberPS3-08-02
Impact of Hormone Receptor-Positive Breast Cancer Subtypes on Oral Endocrine Therapy Adherence and Persistence
Wilmene S Hercule, Rush University Medical Center, Chicago, IL
W. S. Hercule, J. O. Dorcelien, M. A. Allen, L. Anoruo, C. Ikedi, T. Pham, M. L. Barbosa, T. N. Christ, R. Rao; Internal Medicine, Division of Hematology, Oncology, and Cell Therapy, Rush University Medical Center, Chicago, IL.
Introduction: Hormone receptor-positive (HR+) breast cancer accounts for approximately 78% of cases in the U.S. and is primarily treated with adjuvant oral endocrine therapy (AOET) to reduce recurrence and improve survival. HR+ tumors are further classified into molecular subtypes—luminal A and luminal B—based on hormone receptor expression, Ki-67 proliferation index, and HER2 status, which together guide treatment intensity and strategy. Despite the established benefits of AOET, adherence and persistence remain suboptimal, with up to 50% of patients discontinuing therapy prematurely, increasing the risk of recurrence and mortality. Although side effects and comorbidities are known barriers, limited research has examined how these factors vary by subtype. Understanding these relationships is essential to developing targeted adherence interventions for HR+ breast cancer survivors. Objective: To assess the association between HR+ breast cancer subtypes (luminal A, B, and HER2) and AOET adherence and persistence, and examine the impact of sociodemographic and clinical factors. Methods: This retrospective chart review included patients diagnosed with stage I-III hormone receptor-positive (HR+) breast cancer in 2014 at Rush University Medical Center who were prescribed adjuvant oral endocrine therapy (AOET). Of 391 patients identified through the Rush Cancer Registry, 133 met eligibility criteria after exclusions for stage 0, IV, or unknown; insufficient documentation; or transfer of care. Molecular subtypes were classified as luminal A, luminal B, luminal HER2, or mixed, based on hormone receptor, HER2, and Ki-67 status. Oncotype recurrence scores were available for a subset. Clinical variables, comorbidities, and sociodemographic factors—including age and race/ethnicity—were recorded. Adherence and persistence were assessed by AOET duration, completion of ≥5 years of therapy, and documentation of adherence behaviors, side effects, and reasons for discontinuation. Statistical analyses included descriptive and inferential tests to evaluate associations between subtype, sociodemographic and clinical factors, and adherence outcomes. Results: Among 133 patients, 35% had luminal A, 19% luminal B, 5% luminal HER2, and 29% mixed-subtype tumors. Although AOET duration appeared longer in luminal A compared to other subtypes, these differences were not statistically significant. Receipt of chemotherapy was significantly associated with shorter AOET duration (p = 0.042), suggesting treatment intensity may influence persistence. Consistent with prior studies, patients who reported endocrine-related side effects were significantly more likely to discontinue therapy early (p = 0.026) and were less likely to take AOET as prescribed (p = 0.005 for multi-level adherence; p = 0.003 for binary adherence). Oncotype recurrence scores also differed significantly by subtype (p < 0.001), though scores were not routinely calculated in HER2+ or mixed-subtype patients. Conclusion: Molecular subtype was not a statistically significant predictor of AOET adherence or persistence in this cohort. However, treatment-related side effects and chemotherapy exposure were significantly associated with reduced persistence and nonadherence. While the link between side effects and endocrine nonadherence is well established, our findings confirm that this relationship persists across diverse subtypes and real-world practice settings. These results underscore the need for survivorship strategies that address symptom burden and treatment intensity, especially for patients receiving multimodal therapy. Future work should explore how subtype-specific treatment planning may inform adherence interventions in HR+ breast cancer.
Presentation numberPS3-08-03
The mystery of ER-negative/PR-positive breast cancer: revisiting the endocrine therapy controversy using national data
Shawn Michael Doss, Medical College of Georgia, Augusta, GA
S. M. Doss, P. Raval; Department of Medicine, Medical College of Georgia, Augusta, GA.
Background: The existence of estrogen receptor-negative (ER-), progesterone receptor-positive (PR+) breast cancer is rare, biologically confusing, and often attributed to technical artifact. Previous national analyses found no benefit from endocrine therapy (ET) in ER-/PR+ tumors, but more recent data suggest a survival benefit. To explore this inconsistency, we analyzed the association between receiving ET and survival in ER-/PR+ early-stage breast cancer treated with chemotherapy over time. Methods: We used the National Cancer Database (NCDB) to identify women age 40 and older diagnosed with stage I-III, ER-/PR+ breast cancer from 2010 to 2020 who received chemotherapy. Multivariate logistic regression evaluated factors associated with ET omission. Multivariate Cox regression analyzed the association between ET receipt and three-year overall survival (OS), stratified by diagnosis period (2010-2017 vs. 2018-2020) based on 2018 changes for prognostic staging and NCDB site-specific factors. Analyses adjusted for stage at diagnosis, age, comorbidity, HER2 status, race/ethnicity, grade, facility type, and zip code income. Robustness was tested using propensity score matching (PSM), landmark analysis, and HER2-negative subgroup analysis. Results: We identified 24,198 (0.94%) cases of ER-/PR+ breast cancer, of which 18,788 (77.6%) were stage I-III at diagnosis. Of these, 13,957 (74.3%) received chemotherapy, and 11,558 with complete covariate data were analyzed. Most cases were HER2-negative (60.2%) and high grade (75.1%). Stage distribution was 38.5% stage I, 43.5% stage II, and 18.0% stage III. Two-year overall survival was 93.3% for patients diagnosed from 2010-2017 and 92.9% for 2018-2020. ET was omitted in 33.8% of patients (median age 58 vs. 57 among ET recipients). In multivariate logistic regression, factors associated with omission of ET were diagnosis from 2018-2020 (adjusted odds ratio [aOR] 1.95, 95% CI 1.75-2.17, p<0.001), increasing age (age 70-80 vs. 40-50: aOR 1.34, 95% CI 1.16-1.55, p<0.001), and high grade (aOR 1.15, 95% CI 1.03-1.28, p=0.02). In multivariate Cox regression, receiving ET was associated with improved three-year OS for patients diagnosed from 2010-2017 (n=7,517; adjusted hazard ratio [aHR] 0.54, 95% CI 0.47-0.62, p<0.001) and 2018-2020 (n=4,041; aHR 0.60, 95% CI 0.48-0.77, p<0.001). Interaction testing for diagnosis period and ET benefit was not significant (p=0.59). After PSM by all covariates, OS benefit remained for both 2010-2017 (n=5,696; aHR 0.55, 95% CI 0.47-0.65, p<0.001) and 2018-2020 (n=2,344; aHR 0.64, 95% CI 0.48-0.86, p=0.003). Conclusions: Omission of ET in ER-/PR+ early-stage breast cancer treated with chemotherapy was associated with worse overall survival, both before and after 2018. The biological and technical uncertainty of this subtype remains unresolved, but these findings warrant further investigation.
| Analysis | 2010–2017 (aHR, 95% CI) | 2010–2017 p-value | 2018–2020 (aHR, 95% CI) | 2018–2020 p-value |
| Multivariable Cox | 0.54 (0.47–0.62) | <0.001 | 0.60 (0.48–0.77) | <0.001 |
| HER2-negative only | 0.59 (0.50–0.69) | <0.001 | 0.63 (0.48–0.81) | <0.001 |
| Landmark (patients surviving ≥6 months) | 0.60 (0.52–0.70) | <0.001 | 0.70 (0.55–0.89) | 0.004 |
| Propensity score matched | 0.57 (0.49–0.66) | <0.001 | 0.66 (0.50–0.87) | 0.003 |
Presentation numberPS3-08-04
Dalpiciclib Combined with Letrozole as Neoadjuvant Therapy for Stage II-III HR-Positive, HER2-Negative Breast Cancer: A Phase II Study
Yan Mao, The affiliated hospital of Qingdao University, Qingdao, China
Y. Mao, H. Wang, W. Li, T. Ma, X. Gao, W. Cao, X. Wang, Z. Niu, J. Cui, H. Hu, C. Liu, D. Jiang, G. Nie, C. Liu, X. Sun; Breast Disease Center, The affiliated hospital of Qingdao University, Qingdao, CHINA.
Background: Hormone receptor -positive (HR+)/ human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC) typically shows lower rates of pathologic complete response (pCR) and objective response rates (ORR) to neoadjuvant therapy, underscoring a critical need for more effective strategies. Phase III DAWNA‑1/2 trials have demonstrated that adding dalpiciclib, a cyclin-dependent kinase 4/6 (CDK4/6) inhibitor, to endocrine therapy significantly prolongs progression-free survival in patients with HR+/HER2- advanced BC. However, its efficacy and safety in the neoadjuvant setting remain underexplored. This study aims to explore the efficacy and safety of dalpiciclib plus letrozole in patients with stage II-III HR+/HER2- BC. Methods: In this single-arm, open-label study (NCT05512416), early or locally advanced HR+/HER2- BC patients received six 28-day cycles of dalpiciclib (150 mg orally, once daily on days 1-21) plus letrozole (2.5 mg orally, once daily), with or without goserelin (3.6 mg, intramuscularly). Tumor response was evaluated every two cycles by MRI according to RECIST v1.1 criteria until surgery. Surgical resection was scheduled 2-4 weeks after completion of neoadjuvant therapy. The primary endpoint was complete cell cycle arrest (CCCA), defined as Ki-67 ≤ 2.7% on Day 15 of Cycle 1 (C1D15). Secondary endpoints included ORR, residual cancer burden score of 0 or I (RCB 0/I), total pCR (tpCR; ypT0/is and ypN0), and safety. Results: From August 2022 to December 2024, a total of 37 patients were enrolled. All patients with estrogen receptor (ER) expression ≥ 50%, 43.2% (16/37) had Ki-67 expression ≥ 20%. Among the 35 patients with biopsy at C1D15, 74.3% (26/35) achieved CCCA. Of 34 patients who completed neoadjuvant therapy and underwent surgery, the RCB 0/I was 3.0% (1/34), breast pCR rate was 11.8% (4/34), and ORR was 60.0% (21/35). The most common adverse events (AEs) were neutropenia (100%) and leukopenia (94.6%), with grade ≥ 3 AEs reported in 59.5% and 16.2% of patients, respectively. Most other AEs were grade 1-2. Translational biomarkers analyses are ongoing. Conclusion: Our results showed that dalpiciclib plus letrozole demonstrates a promising safety profile and antitumor activity in HR+/HER2− early/locally advanced BC, with significant suppression of proliferation (Ki‑67). These results support its potential as a fully oral, chemotherapy-free neoadjuvant regimen. Further studies are warranted to confirm these preliminary results.
Presentation numberPS3-08-06
Elegant: elacestrant versus standard endocrine therapy (ET) in women and men with node-positive, estrogen receptor-positive (ER+), HER2-negative (HER2-), early breast cancer (eBC) with high risk of recurrence in a global, multicenter, randomized, open-label phase 3 study
Aditya Bardia, University of California Los Angeles (UCLA) Health Jonsson Comprehensive Cancer Center, Santa Monica, CA
A. Bardia1, V. Kaklamani2, J. O’Shaughnessy3, P. Schmid4, T. Beck5, M. De Laurentiis6, G. Curigliano7, H. S. Rugo8, C. H. Barcenas9, W. J. Gradishar10, M. Ignatiadis11, D. A. Cameron12, G. Tonini13, S. Scartoni13, J. Crozier14, T. Wasserman15, S. Tolaney16; 1Hematology and Oncology, University of California Los Angeles (UCLA) Health Jonsson Comprehensive Cancer Center, Santa Monica, CA, 2Breast Medical Oncology, University of Texas Health Sciences Center, San Antonio, TX, 3Breast Medical Oncology, Baylor University Medical Center, Texas Oncology, US Oncology, Dallas, TX, 4Oncology, Barts Cancer Institute, Queen Mary University of London, London, UNITED KINGDOM, 5Oncology, Highlands Oncology, Springdale, AR, 6Oncology, Istituto Nazionale Tumori Napoli IRCCS “Fondazione Pascale”, Napoli, ITALY, 7Oncology, Istituto Europeo di Oncologia, IRCCS, and University of Milano, Milano, ITALY, 8Breast Medical Oncology, City of Hope, Duarte, CA, 9Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 10Oncology, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, 11Breast Medical Oncology, Breast Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), Brussels, BELGIUM, 12Oncology, Institute of Genetics and Cancer and Usher Institute, University of Edinburgh, Edinburgh, UNITED KINGDOM, 13Biostatistics, Menarini Group, Florence, ITALY, 14Clinical Development, Menarini Group, New York, NY, 15Medical Affairs, Menarini Group, New York, NY, 16Breast Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Background: Adjuvant ET is the standard of care (SOC) for treating ER+/HER2- eBC. Despite advances to optimize adjuvant treatment in high-risk ER+/HER2- eBC, there continues to be a risk of local and metastatic (incurable) recurrence that persists, and new therapies with desirable safety profiles are warranted. Elacestrant is a next-generation oral SERD that provides a novel mechanism of action that has shown both SERD (degradative) and SERM (partial agonist) activity that differs from currently available adjuvant ET (Wardell, ERC 2015). In the EMERALD trial, elacestrant significantly prolonged PFS vs SOC ET in the overall population (HR 0.70; 95% CI 0.55-0.88; P=0.0018) and in patients with ESR1-mut tumors (HR 0.55; 95% CI 0.39-0.77; P=0.0005) (Bidard, JCO 2022). In patients with ESR1-mut tumors who received prior ET+CDK4/6i ≥12 mo, mPFS with elacestrant was 8.6 vs 1.9 mo with SOC ET (Bardia, CCR 2024). In a preoperative, window of opportunity ER+/HER2- eBC trial (SOLTI-1905-ELIPSE), elacestrant was associated with complete cell cycle arrest (defined as Ki67<2.7%) rate of 27% and a statistically significant mean change from baseline, shifting tumor biology toward a more endocrine-sensitive and less proliferative tumor phenotype (Vidal, CCR 2025). Given that elacestrant demonstrated efficacy in mBC regardless of ESR1-mut status relative to SOC ET and has shown biologic activity in eBC, it is hypothesized that elacestrant can prolong invasive breast cancer-free survival (IBCFS) in patients with high-risk eBC who received prior adjuvant ET±CDK4/6i. Design and Methods: ELEGANT (NCT06492616) is a global, multicenter, open-label phase 3 study designed to evaluate elacestrant vs SOC ET (AI or tamoxifen) in patients with eBC and a high risk of recurrence. Patients will be randomized 1:1 to continue SOC ET or to elacestrant for a duration of 5 yrs. Eligible patients are women or men with ER+/HER2− node-positive eBC who have completed 24-60 mo of adjuvant ET±CDK4/6i and have ECOG PS ≤1. Patients who received a prior CDK4/6i or a PARP inhibitor must have already completed or discontinued these treatments. Pre/perimenopausal women and men will be administered a LHRH agonist. Exclusion criteria include inflammatory breast cancer, history of prior invasive breast cancer, and >6 mo continuous interruption of prior SOC adjuvant ET or discontinuation of adjuvant ET >6 mo prior to randomization. The primary endpoint is IBCFS. Key secondary endpoints include distant relapse-free survival, overall survival, invasive disease-free survival, safety, patient-reported outcomes-quality of life, and pharmacokinetics. Status: Planned enrollment is 4,220 patients; recruitment is ongoing.
Presentation numberPS3-08-07
Comparative Analysis of Breast Cancer Index Testing in Hispanic and Non-Hispanic Populations
Ana C Sandoval-Leon, Baptist Health South Florida. Miami Cancer Institute, Miami, FL
A. C. Sandoval-Leon1, N. Siuliukina2, Y. Liu2, Y. Chamorro1, N. Dempsey3, M. M. Zerey4, L. Dumeny5, D. Tolman3, P. Bhatt3, L. Carcas3, Y. Zhang2, A. Anderson2, K. Treuner2, R. Mahtani3; 1Medical Oncology, Baptist Health South Florida. Miami Cancer Institute, Miami, FL, 2R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 3Medical Oncology, Baptist Health South Florida. Miami Cancer Institute, Plantation, FL, 4Radiation Oncology, Baptist Health South Florida. Miami Cancer Institute, Miami, FL, 5Family Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL.
Background: Hispanic and non-Hispanic patients with breast cancer (BC) exhibit distinct clinicopathologic profiles that may impact treatment outcomes. The Breast Cancer Index (BCI) is a validated gene expression assay that assesses overall (0-10 years) and late (5-10 years) distant recurrence (DR) risk and predicts the likelihood of benefit from extended endocrine therapy (EET). This analysis compares BCI results and clinicopathologic features between Hispanic patients with hormone receptor positive (HR+) BC from Miami Cancer Institute (MCI) and non-Hispanic patients enrolled in the BCI Registry study. Methods: After IRB approval, we performed a retrospective review of patients treated at MCI with HR+ early-stage BC who underwent BCI testing as part of routine clinical care. (n=776). The BCI Registry is a prospective, multi-institutional study that enrolled over 3400 patients with early-stage HR+ BC to evaluate long-term clinical outcomes, decision impact, and medication adherence. Statistical analyses included Fisher’s exact test and the Cochran-Mantel-Haenszel (CMH) test to assess associations between different variables (clinicopathologic vs. ethnicity; clinicopathologic vs. BCI results), and the Breslow-Day (BD) test to evaluate whether ethnicity influences the association between clinicopathologic factors and BCI results (BCI Predictive and Prognostic groups). Results: The BCI Registry study included 2,406 non-Hispanic patients, while the MCI dataset comprised 408 self-identified Hispanic patients. Compared to non-Hispanic patients, Hispanic patients were more likely to identify as white (98% vs. 89%, p < 0.001), were younger (<50 years: 23% vs. 7%, p < 0.001), and were less likely to be postmenopausal (80% vs. 90%, p < 0.001). They were also less likely to start adjuvant endocrine therapy with an aromatase inhibitor (AI) (65% vs. 74%, p < 0.001) and more likely to switch therapy (20% vs. 13%). Clinicopathologic features were largely similar between groups, including tumor size, tumor grade, nodal status, and HER2 status. BCI Predictive results showed fewer Hispanic patients with a high likelihood of EET benefit (33% vs. 39%, p = 0.046), and BCI Prognostic results indicated a lower proportion categorized as High Risk for late distant recurrence (49% vs. 53%, p = 0.087). In BCI Predictive groups, stratified BD analysis revealed significant ethnic differences in associations between EET benefit and clinical variables, including age, surgery type (BD p = 0.01), menopausal status, tumor size, tumor grade, nodal status, and HER2 status (BD p < 0.001), with moderate effects for BMI (BD p = 0.042). In BCI Prognostic groups, strong ethnic stratification was observed for tumor size, tumor grade, nodal status, HER2 status, and surgery type (BD p < 0.001), with moderate effects for age (BD p = 0.03) and race (BD p = 0.013), suggesting ethnicity may influence how these factors relate to late distant recurrence. Conclusion: Hispanic patients with BC showed distinct clinical and molecular profiles compared to non-Hispanic patients, including younger age at diagnosis, differing endocrine therapy patterns, lower predicted benefit from EET, and more favorable tumor biology. These findings highlight the need to account for ethnic variability in personalized treatment planning and call for further research into individualized risk assessment in early-stage HR+ BC. However, these findings should be interpreted with caution given the retrospective, single institution nature of the Hispanic cohort. Long-term clinical outcomes from the MCI cohort will be reported in future analyses to better understand how BCI results correlate with real-world treatment decisions and patient outcomes.
Presentation numberPS3-08-08
Anthracycline vs. Non-Anthracycline Chemotherapy Regimens for ER+/HER2− Early-Stage Breast Cancer Based on Oncotype DX Recurrence Score: A Retrospective Study
Mei Wei, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
M. Wei1, C. Carroll2, S. Kimani3, H. N. Lynn4, F. Bingjian5; 1Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 2Dermatology, University of Utah, Salt Lake City, UT, 3Internal Medicine, University of North Carolina, Chapel Hill, NC, 4Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 5Dermatology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT.
Introduction: Choosing between anthracycline (A)-based versus non-A-based chemotherapy (chemo) regimens among patients with estrogen receptor-positive (ER+), HER2-negative (HER2-) early-stage breast cancer (BC) remains challenging due to the lack of data to guide this decision. In this study, we utilized the Flatiron Health Research Database to examine demographic, clinical, and pathologic factors associated with administration of A versus non-A-based chemo regimens for patients with stage I-II, ER+/HER2- BC. Methods: Patients age 18+, who were diagnosed with stage I-II, N0-1, ER+/HER2- BC, who initiated chemo within 180 days of BC diagnosis, and who had an available Oncotype DX recurrence score (RS) were included in this cohort. Patients who received at least one dose of doxorubicin were designated as having received A-based chemo. Logistic regression was performed to determine associations between the A-based chemo decision and each clinical factor. Cox regression was performed to compare survival probabilities between patients who received A- versus non-A-based chemo, using propensity score matching to balance baseline clinical characteristics and reduce confounding due to treatment selection. Results: Of the 512 patients included in the analysis, 406 (79.3%) received non-A-based chemo. Factors significantly associated with receiving A-based chemo included higher stage, larger tumor size, lymph node involvement, younger age, and higher RS (Table 1). Tumor grade was not significantly associated. In a multivariable analysis, factors that remained significant included stage, lymph node, age, and RS. We matched patients 1:1 on these factors, resulting in 190 participants for survival analysis. Patients who received A-based regimens had numerically better, though not statistically significant, recurrence-free survival (RFS) compared to those who received non-A-based regimens (hazard ratio [HR]=0.64 [0.24-1.70], p=0.37). A-based regimen had a lower HR in the subset of patients with RS>30 (HR=0.57 [0.12-2.64], p=0.90) than those with RS<=30 (HR=0.71 [0.20-2.52], p=0.60), although the difference was not significant. Conclusion: Our study suggests that oncologists use treatment with anthracycline-containing chemotherapy regimens for patients who are younger and those with higher-risk BC. In this relatively small, retrospective cohort, there was a suggestion of improved RFS with anthracycline, but confidence intervals were wide. Larger randomized studies or meta-analyses are needed to determine whether anthracyclines truly improve outcomes in these patients.
| A based N (%) | non-A-based N (%) | A- vs. non-A-based regimen * | |
| Stage | |||
| I | 43 (40.6%) | 249 (61.3%) | |
| II | 63 (59.4%) | 157 (38.7%) |
OR = 2.32 [1.50,3.59] p = 0.00015 |
| Tumor size | |||
| T1 | 48 (45.3%) | 247 (60.8%) | |
| T2/3 | 58(54.7%) | 158(38.9%) |
OR = 1.71 [1.15,2.56] p = 0.0084 |
| Lymph nodes | |||
| N0 | 57 (53.8%) | 297 (73.2%) | |
| N1 | 48 (45.3%) | 105 (25.9%) |
OR = 2.38 [1.53,3.71] p = 0.00013 |
| Grade | |||
| 1 | 7 (6.6%) | 50 (12.3%) | |
| 2 | 48 (45.3%) | 178 (43.8%) | |
| 3 | 50 (47.2%) | 178 (43.8%) |
OR = 1.24 [0.89,1.73] p = 0.20 |
| Age | |||
| <40 | 7 (6.6%) | 13 (3.2%) | |
| 40-54 | 41 (38.7%) | 127 (31.3%) | |
| 55-69 | 50 (47.2%) | 190 (46.8%) | |
| ≥70 | 8 (7.5%) | 76 (18.7%) |
OR = 0.65 [0.49,0.86] p = 0.0027 |
| OncotypeRS | |||
| ≤15 | 14 (13.2%) | 48 (11.8%) | |
| 16-25 | 23 (21.7%) | 130 (32.0%) | |
| 25-30 | 15 (14.2%) | 86 (21.2%) | |
| 30-40 | 26 (24.5%) | 97 (23.9%) | |
| >40 | 28 (26.4%) | 45 (11.1%) |
OR = 1.28 [1.08,1.52] p = 0.0051 |
Presentation numberPS3-08-09
Postpartum breast cancer as a potential driver of increased 21-gene recurrence score (RS) and RS category for node-positive HR+/HER2- breast cancer
Shiliang Zhang, University of California, Los Angeles, CA
S. Zhang1, K. J. Queen1, M. Lipsyc-Sharf1, N. Duggirala2, P. Spellman1, A. Bardia1, N. S. Kapoor3; 1Hematology and Oncology, University of California, Los Angeles, CA, 2David Geffen School of Medicine, University of California, Los Angeles, CA, 3Surgery, University of California, Los Angeles, CA.
Background: The 21-gene recurrence score (RS) is widely used to guide adjuvant chemotherapy decisions in hormone receptor-positive (HR+) HER2-negative (HER2-) early breast cancer (BC). However, in premenopausal women with node-positive disease, RS has limited utility in identifying low-risk individuals who may forgo chemotherapy. One understudied subgroup is postpartum BC (PPBC). PPBC is associated with higher rates of lymph node metastases, increased risk of distant recurrence, and worse overall prognosis, even after adjusting for traditional clinicopathologic factors. As such, PPBC, particularly in women with nodal involvement, may confound RS interpretation. We aimed to evaluate whether PPBC is associated with higher RS in premenopausal women with node-positive HR+HER2- BC compared to non-PPBC and nulliparous women, and if this impacted outcomes. Methods: We identified women aged ≤45 years with HR+ HER2- BC with node positive disease who underwent RS testing at a single academic institution between 2011 and 2024. Reproductive and clinicopathologic data were collected through medical record review. Patients were stratified into three groups: nulliparous, PPBC (diagnosis 25) between the groups. Time-to-event outcomes were analyzed using Kaplan-Meier survival estimates, and invasive disease-free survival (IDFS) was compared across groups using Cox proportional hazards regression models. Results: A cohort of 78 patients were included: nulliparous (n=27), PPBC (n=20), and non-PPBC (n=31). Compared to nulliparous women, patients with PPBC had a higher mean RS, with an average increase of 8.78 points (p=0.002). No difference in RS was observed between the non-PPBC group and nulliparous women (p = 0.63). Women in the PPBC group had 4.78 times the odds of being classified in a higher RS category compared to nulliparous women (p = 0.007), and 4.71 times the odds compared to nulliparous and non-PPBC groups combined (p = 0.003). No significant difference in odds of higher RS category was observed in the non-PPBC group compared to nulliparous women. Among all 78 patients, 46 (59.0%) received chemotherapy and this increased with RS category: 38.0% (RS 25) (p=0.00022). Median follow-up time was 2.75 years (range: 0.115, 8.977, IQR=3.02), and there were 6 recurrences (7.7%). Five-year IDFS was 81.7% (0.608-1.00) in PPBC patients and 78.6% (0.544-1.00) in all others, with a non-significant trend toward lower recurrence risk in non-PPBC and nulliparous patients compared to PPBC (HR 0.47; 95% CI: 0.07-2.61; p=0.36). Among the subgroup of patients who did not receive chemotherapy (n=24), five-year IDFS was 66.7% (0.300-1.00) in the PPBC group versus 46.9% (0.117-1.00) in all other patients. Conclusions: In this cohort of young women with HR+HER2- node-positive BC, PPBC was associated with higher RS and RS category than BC in nulliparous women. Larger studies with longer follow-up will be needed to evaluate whether distinguishing PPBC from nulliparous and non-PPBC patients can refine RS interpretation in the node-positive setting and enhance its utility in identifying premenopausal patients with biologically low-risk disease who may safely forgo chemotherapy.
Presentation numberPS3-08-10
Efficacy of LHRHa treatment in luminal type breast cancer and in vivo estrogen-independent breast cancer cell lines
Muhan Yu, Chiba University, Chiba, Japan
M. Yu1, M. Takada1, H. Yamada2, T. Nagashima1, H. Fujimoto1, J. Sakakibara1, M. Takaku3, M. Otsuka1; 1General Surgery, Chiba University, Chiba, JAPAN, 2General Surgery, Kimitsu Chuo Hospital, Kisarazu, JAPAN, 3Biomedical Sciences, University of North Dakota, Grand Forks, ND.
Hormone receptor-positive (ER+/PR+), HER2-negative (luminal type) breast cancer accounts for 75.6% of all cases. For premenopausal patients, luteinizing hormone-releasing hormone agonists (LHRHa) suppress ovarian estrogen and progesterone production and are often combined with antiestrogen therapies like tamoxifen or aromatase inhibitors. Clinical trials, such as SOFT and TEXT, demonstrate that LHRHa improves outcomes. However, challenges include side effects (e.g., menopausal symptoms, bone density loss) and identifying which patients benefit most. It is important to personalize treatment strategies to optimize LHRHa use.We performed orthotopic injection of MCF7 cells into bilaterally ovariectomized mice (estrogen depleted). After 2-4 months of observation in the estrogen-depleted state, multiple in vivo-estrogen-independent breast cancer cell lines (iEIBCCs) were established. This may be a model that approximates luminal-type recurrent breast cancer under anti-estrogen therapy. Here, we investigated the effects of LHRHa combination therapy in luminal type breast cancer patients and iEIBCC cell lines. The clinicopathological factors and prognosis of patients with Stage I-III Luminal breast cancer from 2016 to 2021 who were treated at Chiba University hospital were analyzed. We selected 152 analytic cohort patients and split them into two groups by treatment with or without LHRHa. Two kinds of iEIBCCs, 1LR and E10, and parental MCF-7 cells were transplanted into ovariectomized SCID mice, LHRHa-treated mice, and wild-type mice. Furthermore, we conducted RNA sequencing to define the differential gene expression between 1LR, E10, and parental MCF-7. The 5-year RFS of the patients treated with LHRHa (N=48) was statistically higher than without LHRHa (N=104) (p = 0.0150). In the parental MCF-7 group, compared to wild-type mice (N=4), the tumor growth was inhibited by both ovariectomize (N=5) and LHRHa-treated (N=7). In 1LR group, the tumor growth was inhibited by LHRHa-treated (N=7) but was not inhibited by ovariectomized (N=4). On the contrary, in E10 group, the tumor growth was inhibited by ovariectomized (N=4) but was not inhibited by LHRHa-treated (N=6). According to RNA sequencing, there are some differential gene expressions between the 1LR, E10, and parental MCF-7 cells. The main differences in GO pathways enrichment are axonogenesis, small GTPase-mediated signal transduction, protein localization, regulation of neuron projection development, and amide metabolic process. Our results suggest that LHRHa treatment is effective in patients with a high risk of recurrence. The differential gene expression between the E10 and 1R, and the resistance and sensitivity in E10, 1LR provide crucial context for interpreting how LHRHa impacts cancer cells. These insights underscore the complexity of cancer cell responses to hormone depletion therapies like LHRHa. Understanding the specific adaptations that confer resistance or sensitivity in these cell lines can guide the development of combination therapies.
Presentation numberPS3-08-11
Correlation between clinical-pathological features and recurrence score from a multigene predictive assay in hormone receptor-positive/human epidermal growth factor receptor 2-negative early breast cancer: a real-world, retrospective, monocentric analysis of over 700 patients.
Giacomo Mazzoli, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
G. Mazzoli1, G. Fotia1, M. De Santis2, A. Fumagalli3, G. Armani1, L. Lozza2, D. Presti1, R. Agresti4, M. Gennaro4, C. Volpi5, C. Vernieri1, S. Folli4, G. Pruneri5, G. Scaperrotta6, F. De Braud1, G. Capri1, G. Bianchi1; 1Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 2Radiation Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 3Department of Oncology and Hematology-Oncology, University of Milan, Milan, ITALY, 4Breast Surgery Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 5Department of Diagnostic Innovation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY, 6Breast Radiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, ITALY.
Background: Adjuvant cytotoxic chemotherapy followed by endocrine therapy, with or without cyclin dependent Kinase 4/6 (CDK4/6) inhibitors, significantly reduced the risk of tumor recurrence in patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) early breast cancer (eBC) and high-risk clinical-pathological features. Oncotype DX is a validated genomic assay that provides valuable prognostic information to identify patients with eBC who benefit from adjuvant chemotherapy. However, the association between Oncotype DX recurrence score (RS) and clinical-pathological features has not been fully clarified. Here we present a large real-world retrospective analysis of patients with HR+/HER2- eBC undergoing Oncotype DX test at IRCCS Istituto Nazionale dei Tumori. Methods: We conducted a monocentric, retrospective study to investigate the association between Oncotype DX and clinical-pathological features in HR+/HER2− eBC between patients who underwent breast surgery between June 2009 and December 2024. Patients were divided into three risk groups according to RS: low (≤10), intermediate (11-25), and high (≥26). The association between clinical-pathological features and categorized RS was reported as odds ratio (OR). A multivariable cumulative link model was fitted to adjust the associations for ten clinically relevant covariates in both the overall population and the subgroups of patients with (N+) or without (N0) lymph node involvement. Results: Among 732 patients included in the study, Oncotype DX identified 149 (20.3%) patients as low genomic risk, 443 (60.5%) as intermediate risk, and 140 (19.2%) as high risk. After multivariable adjustment, lower progesterone receptor (PgR) expression and higher Ki67 levels were independently associated with a higher RS, whereas lobular histology and older age were associated with lower RS (Table). PgR and Ki67 expression were also significantly associated with RS in both N0 and N+ subgroups (Table). Lobular histology and older age were significantly associated with lower RS only in N0 and N+ patients, respectively (Table). Conclusions: Among clinical-pathological features, Ki67 and PgR showed the most consistent association with genomic risk. In case of limited access to genomic testing, PgR and Ki67 may represent the most reliable markers for prognostic and predictive stratification, alongside lobular histology in N0 tumors and age in N+ cases.
| Ki67, aOR (95%CI), p val | PgR, aOR (95%CI), p val | Lobular histology, aOR (95%CI), p val | Age, aOR (95%CI), p val | ||||||||||||||||
| Overall | 1.86 (1.54-2.25), <0.001 | 0.73 (0.69-0.77), <0.001 | 0.51 (0.32-0.83), 0.006 | 0.86 (0.76-0.96), 0.006 | |||||||||||||||
| N0 | 1.91 (1.51-2.43), <0.001 | 0.72 (0.67-0.77), < 0.001 | 0.42 (0.22-0.81), 0.009 | 0.92 (0.79-1.06), 0.256 | |||||||||||||||
| N+ | 2.11 (1.47-3.09), <0.001 | 0.75 (0.68-0.83), <0.001 | 0.53 (0.25-1.20), 0.113 | 0.75 (0.62-0.91), 0.004 |
aOR: adjusted odds ratio; 95%CI: 95% confidence interval; p val: p-value; RS: recurrence score.
Presentation numberPS3-08-12
Machine Learning Prediction of High Oncotype DX® RS based on Clinicopathologic features: Results from the GBECAM 0520 Multicenter Retrospective Study
Julio araujo, ICESP – Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
J. araujo1, D. araujo2, L. Oliveira3, P. de Souza2, D. Rosa4, A. Katz5, D. Suzuki6, D. Argolo3, S. Sanches7, L. Testa1, J. Bines8, R. Kaliks9, R. Sousa10, T. Corrêa11, A. Shimada5, C. dos Anjos5, R. Linck5, T. Megid12, D. Batista8, D. Gomes8, M. Cesca7, D. Gaudêncio3, L. Moura3, R. Bonadio8, Z. Souza11, J. Beal9, M. Lopes13, L. Sales12, J. Marlière12, M. Mano12, D. Gagliato14; 1Oncologia, ICESP – Instituto do Câncer do Estado de São Paulo, São Paulo, BRAZIL, 2Oncologia, Huna, São Paulo, BRAZIL, 3Oncologia, Oncoclínicas&CO – Medica Scientia Innovation Research (MedSir), São Paulo, BRAZIL, 4Oncologia, Hospital Moinhos de Vento, Porto Alegre, BRAZIL, 5Oncologia, Hospital Sírio Libanes, São Paulo, BRAZIL, 6Oncologia, Hospital Sírio Libanês, Brasília, Brasília, BRAZIL, 7Oncologia, AC Camargo Cancer Center, São Paulo, BRAZIL, 8Oncologia, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 9Oncologia, Hospital Israelita Albert Einstein, São Paulo, BRAZIL, 10Oncologia, Grupo Américas, São Paulo, BRAZIL, 11Oncologia, Hospital Sírio Libanês, Brasília, São Paulo, BRAZIL, 12Oncologia, Instituto de Ensino e Pesquisa Sírio-Libanês, São Paulo, BRAZIL, 13Oncologia, Hospital Moinhos de Vento, São Paulo, BRAZIL, 14Oncologia, BP – A Beneficência Portuguesa de São Paulo, São Paulo, BRAZIL.
Machine Learning Prediction of High Oncotype DX® RS based on Clinicopathologic features: Results from the GBECAM 0520 Multicenter Retrospective Stud Background: The 21-gene Oncotype DX® assay is a validated prognostic and predictive tool for assessing chemotherapy (CT) benefit in patients with estrogen receptor–positive (ER+), HER2-negative (HER2–) early-stage breast cancer. It has demonstrated clinical utility in node-negative (N0) patients and postmenopausal patients with N1 disease. However, financial cost associated with the test may significantly limit its accessibility to patients. The present study evaluates whether machine learning models based on routinely available clinicopathologic and immunohistochemical data could accurately predict high likelihood of a Recurrence Scores greater than 25, possibly providing support for clinical decision-making. Methods: We retrospectively assembled a cohort of 897 patients diagnosed with ER+ breast cancer (BC) diagnosed between 2005–2024 at seven cancer centers. All patients had Oncotype DX® results and a complete pathology report. Of these, 158 (18%) had RS > 25. Statistical analysis identified Ki-67 (%), progesterone receptor (PR) status, and histologic grade as the most predictive variables. These were included in a ridge regression model to estimate continuous RS values (Table 1). Discriminative performance for RS > 25 was measured by AUC. To test robustness, we performed leave-one-center-out cross-validation: training on six centers and testing on the seventh. Performance was averaged across folds. Results: Among the 897 patients included, 708 (78.9%) had N0 disease, 62 (6.9%) N1mic, and 118 (13.2%) N1 disease. Of note, nodal information was unavailable for 9 patients (1.0%). The median age was 54 years (range, 24–83). Oncotype DX® testing indicated low (25) genomic risk in 165 (18.4%), 574 (64.0%), and 158 (17.6%) of patients, respectively. The model achieved a mean Pearson correlation of 0.50 ± 0.03 between predicted and actual RS, and an AUC of 0.78 ± 0.03 for detecting high-risk (RS ≥ 25) tumors. AUC stratified by menopausal status showed no significant difference (p = 0.858), indicating similar performance across groups. After min–max scaling, Ki-67 was the strongest positive predictor, followed by histologic grade conversely, PR status was inversely correlated with RS. These findings are consistent with established tumor biology and previous studies, in which a high proliferative index, poor histologic differentiation, and low immunohistochemistry PR expression are features associated with more aggressive tumor characteristics. Conclusion: A simple machine learning model using only Ki-67, histologic grade, and PR status moderately predicts the Oncotype DX® score and accurately identifies high-risk cases. In resource-limited settings, this approach could support clinical decision-making. Prospective validations are needed to confirm its clinical utility.Table 1: Significant variables according to Recurrence Score (RS) categories (RS ≤ 25 and RS > 25). Values are expressed as mean ± standard deviation for the continuous variable (Ki-67) and as number (percentage) for all categorical variables.
Presentation numberPS3-08-13
Exploratory analysis of palbociclib benefit in the PALLAS trial by SET<sub>ERPR</sub> index and prior chemotherapy regimens (ABCSG-42/AFT-05/BIG-14-13)
Otto Metzger, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
O. Metzger1, P. O’Brien2, K. Ballman2, M. Gnant3, M. Watson4, E. Chen5, K. Tran5, D. Hlauschek6, M. Martin7, J. Balko8, Z. Nowecki9, O. Hahn10, C. Denkert11, C. Curtis12, F. Liu13, A. Dueck14, C. Fesl15, E. Mayer1, A. De Michele16, W. Symmans17; 1Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 2Statistics and Data Center, Mayo Clinic, Rochester, MN, 3Medical Oncology, University of Vienna, Vienna, AUSTRIA, 4Siteman Cancer Center Biorepository Programs, Washington University School of Medicine, St. Louis, MO, 5Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, 6Department of Internal Medicine I, ABCSG and Medical University of Vienna, Vienna, AUSTRIA, 7Department of Medical Oncology, GEICAM and Hospital General Universitario Gregorio Marañón, Madrid, SPAIN, 8Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 9Breast Cancer & Reconstructive Surgery, The Maria Sklodowska Curie Centre, Warsaw, POLAND, 10Department of Medicine, University of Chicago, Chicago, IL, 11Institute of Pathology, Philipps‑Universität Marburg, Marburg, GERMANY, 12Medical Oncology, Stanford University, Stanford, CA, 13Translational Oncology, Pfizer, San Diego, CA, 14Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, 15Department of Statistics, Austrian Breast & Colorectal Cancer Study Group (ABCSG), Vienna, AUSTRIA, 16Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 17Department of Pathology, University of Texas MD Anderson Cancer Center,, Houston, TX.
Background PALLAS trial compared palbociclib plus endocrine therapy (ET) vs. ET alone in stage II-III HR+/HER2- breast cancer. SETER/PR index quantifies transcriptional activity related to estrogen and progesterone receptors and serves as a measure of endocrine sensitivity. In CALGB 9741, low SETER/PR (< 0.75) – reflecting endocrine resistance – predicted survival benefit from adjuvant anthracycline/taxane-based treatment. In PALLAS low SETER/PR was associated with worse prognosis and early recurrence. Early recurrences driven by endocrine-resistant tumors may obscure the benefit of palbociclib by disproportionately contributing events from a more resistant population. We hypothesized that palbociclib efficacy might vary by endocrine sensitivity and prior chemotherapy regimen, with greatest benefit in patients with intermediate SETER/PR index. Methods Invasive disease-free survival (iDFS) was summarized with Kaplan-Meier plots and the treatment arms were compared with a log-rank test. Cox models estimated the hazard ratio (HR) with 95% confidence interval (95%CI) (significance level used was a two-sided 0.05). SETER/PR index was successfully obtained for 3,090 PALLAS patients (TransPALLAS ITT cohort, Metzger et al. ASCO 2024). SETER/PR index cut points (0.75 and 1.50) were proposed from previous studies comparing different adjuvant chemotherapy regimens. Results Table 1 summarizes the efficacy of adjuvant palbociclib by prior (neo)adjuvant chemotherapy regimen. Palbociclib was associated with longer iDFS in the group who received prior anthracycline-paclitaxel chemotherapy (AC-P), though the chemo regimen x palbociclib interaction term was not significant (Table 1). In the 1,132 patients from the TransPALLAS cohort who received AC-P chemotherapy, the 518 (46%) patients with intermediate SETER/PR (0.75≤ SETER/PR <1.50) had longer iDFS with palbociclib (HR 0.58; 95% CI: 0.40-0.84; p=0.004; 7-yr iDFS 80.2% vs. 68.4%), but this subset did not predict palbociclib benefit relative to all others, i.e., 206 (18%) patients with low SETER/PR (<0.75) and 408 (36%) high SETER/PR (≥1.50); P interaction=0.11. Exploratory analysis of SETER/PR index as a continuous variable in patients who received prior AC-P chemotherapy revealed a non-linear relationship with the hazard ratio favoring palbociclib observed in the subset with SETER/PR values between 1.1 and 1.8, compared to all others (P interaction=0.01). Conclusions In PALLAS, we observed a numerical benefit favoring palbociclib among pts treated with adjuvant AC-P; in this subset a moderate level of endocrine transcriptional activity (SETER/PR index between 1.1 and 1.8) predicted palbociclib benefit. These results are hypothesis-generating. Table 1. Efficacy of adjuvant palbociclib by prior (neo)adjuvant chemotherapy regimen.
| N = | IDFs, TransPALLAS HR (95% CI) | P-value | N = | IDFs, PALLAS Trial (ITT) HR (95% CI) | P-value | ||||||||
| AC-P | 1,132 | 0.73 (0.55 – 0.95) | 0.019 | 2,131 | 0.78 (0.64 – 0.94) | 0.008 | |||||||
| AC-D | 737 | 0.95 (0.68 – 1.33) | 0.76 | 1,460 | 0.99 (0.79 – 1.25) | 0.95 | |||||||
| 1st gen. | 360 | 1.17 (0.67 – 2.04) | 0.58 | 622 | 0.96 (0.64 – 1.44) | 0.83 | |||||||
| Other | 225 | 0.72 (0.41 – 1.29) | 0.27 | 482 | 0.92 (0.63 – 1.33) | 0.64 | |||||||
| None | 636 | 0.90 (0.60 – 1.36) | 0.63 | 1,053 | 1.03 (0.75 – 1.41) | 0.86 | |||||||
| All Groups | Interaction Term | 0.50 | Interaction Term | 0.45 | |||||||||
| Footnote: A – anthracycline; C – cyclophosphamide; P – paclitaxel; D – docetaxel; 1st gen – first generation regimens such as TC or AC; Other – multiple or not well characterized; None – no prior chemotherapy. HR and P-values are based on Cox model according to each patient’s randomized treatment assignment. (Reference: Arm B – ET Alone) | |||||||||||||
Presentation numberPS3-08-14
Typer—A randomized controlled trial evaluating the effect of typology-based coaching on therapy management for patients with hr+/her2- early breast cancer under adjuvant ribociclib treatment
Manfred Welslau, Klinikum Aschaffenburg, Hösbach, Germany
M. Welslau1, C. Zeder-Goess2, R. Haidinger3, B. Welter4, B. Aktas5, P. Wimberger6, P. A. Fasching2, H. Kolberg7, J. C. Radosa8, C. Rudlowski9, A. Hein10, S. Seitz11, D. Fischer12, C. Iuliano13, C. Mann13, M. Braun14, V. Mueller15, E. Stickeler16, M. Thill17, N. Ditsch18; 1Oncology Department, Klinikum Aschaffenburg, Hösbach, GERMANY, 2Department of Gynecology and Obstetrics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, GERMANY, 3Breast Cancer, Brustkrebs Deutschland e.V., Hohenbrunn, GERMANY, 4Breast Cancer, Mammazone e.V., Augsburg, GERMANY, 5Department of Gynecology, University of Leipzig Medical Center, Leipzig, GERMANY, 6National Center for Tumor Diseases Dresden and Department of Gynecology and Obstetrics, University Hospital Dresden, TU Dresden, Dresden, GERMANY, 7Department of Gynecology and Obstetrics, Marienhospital Bottrop, Bottrop, GERMANY, 8Department of Gynecology and Obstetrics, Saarland University Hospital, Homburg, GERMANY, 9Department of Gynecology, Evangelical Hospital Bergisch Gladbach, Bergisch Gladbach, GERMANY, 10Department of Gynecology and Obstetrics, Hospital Esslingen, Esslingen am Neckar, GERMANY, 11Department of Gynecology and Obstetrics, St. Josef Hospital, Regensburg, GERMANY, 12Department of Gynecology, Hospital Ernst von Bergmann, Potsdam, GERMANY, 13Oncology, Novartis Pharma GmbH, Nürnberg, GERMANY, 14Department of Gynecology, Rotkreuzklinikum München, München, GERMANY, 15Department of Gynecology, Hamburg-Eppendorf University Medical Center, Hamburg, GERMANY, 16Department of Obstetrics and Gynecology, Center for Integrated Oncology (CIO Aachen, Bonn, Cologne, Düsseldorf), University Hospital of RWTH Aachen, Aachen, GERMANY, 17Department of Gynecology and Gynecologic Oncology, Agaplesion Markus Hospital, Frankfurt am Main, GERMANY, 18Gynecology, Obstetrics and Senology, Faculty of Medicine, University of Augsburg, Breast Center, University Hospital Augsburg, CCC Augsburg, Germany, Bavarian Cancer Research Center (BZKF), Augsburg, GERMANY.
Endocrine therapy is the backbone of hormone receptor-positive, HER2-negative (HRpos/HER2neg) early breast cancer (eBC) treatment, yet early discontinuation rates are high, compromising long-term outcomes. With ribociclib plus a non-steroidal aromatase inhibitor (NSAI) now approved by the FDA and EMA for eBC at risk of recurrence, it is expected to be widely used in the adjuvant setting. Adjuvant CDK4/6 inhibitor trials show discontinuation in 30-35% of patients. Thus, adherence and persistence are crucial for an effective management of adjuvant ribociclib therapy. Recent studies have highlighted the positive impact of supportive care programs provided by qualified nurses for patients with cancer. Patient-focused motivation techniques and education performed by nurses could strengthen patients’ understanding of the risks and benefits of anticancer treatments. Personalized coaching and structured tools like the “Multinational Association of Supportive Care in Cancer (MASCC) Oral Agent Teaching Tool” (MOATT) may strengthen patient engagement and optimize therapy management. TYPER (EU Clinical Trials Number: 2024-520290-12-00) is a prospective, multicenter, randomized, controlled, phase IV study with the objective of investigating the effect of personalized patient coaching on therapy persistence in patients with HRpos/HER2neg eBC receiving adjuvant ribociclib plus NSAI according to the summary of product characteristics (SmPC). In total, 548 patients will be enrolled across 50 study sites in Germany and randomized 1:1 to receive either local routine (control arm) or continuous personalized coaching utilizing the MOATT tool (study arm) during the first 12 months of adjuvant ribociclib therapy. A patient classification model organizes individuals into four distinct groups based on responses to a typology questionnaire. It is based on the idea that personal acceptance of and perceived control over the illness determine the type of support that is most helpful for each individual. The different patient groups therefore call for varied approaches to patient support. Personalized patient coaching will be tailored to individual patient profiles based on patient typology and use the MOATT tool and must be performed by trained (TYPER) coaches. As part of the study, healthcare professionals will be trained to become TYPER coaches. The primary objective is to evaluate the effect of patient education and patient typology-based coaching on persistence, with the primary endpoint being time to permanent ribociclib discontinuation. Secondary endpoints include persistence rates at month 6, 12 and 36, side effect management, and unplanned therapy interruptions. A biomarker program will explore correlations with clinical outcomes. Participants will remain in the study until completion or death, with a maximum individual participation period of 60 months, comprising a 24-month enrollment phase and 36-month follow-up phase. Patient recruitment is planned between Q2/2025 and Q2/2027, with overall study completion expected by Q2/2030.
Presentation numberPS3-08-15
Prognostic Significance of Oncotype DX (RS) and MammaPrint (MP) in Post-Menopausal Estrogen Receptor-Positive (ER+), Node-Positive (N1) Breast Cancer (BC) patients: A Comparative Analysis by Race Using the National Cancer Database (NCDB)
Devashish Desai, SUNY Upstate Medical University, Syracuse, NY
D. Desai1, C. Widholm2, P. Ashok Kumar1, E. Hill2, S. Sammons3, A. Sivapiragasam4; 1Department of Hematology and Medical Oncology, SUNY Upstate Medical University, Syracuse, NY, 2Department of Biostatistics Shared Resources, Medical University of South Carolina, Charleston, SC, 3Department of Hematology and Medical Oncology, Dana Farber Cancer Institute, Boston, MA, 4Department of Hematology and Medical Oncology, Medical University of South Carolina, Charleston, SC.
Background: Postmenopausal ER+ patients often undergo genomic testing with RS or MP to assess chemotherapy benefits. However, the prognostic value of RS, particularly among ethnic minorities, remains unclear.Methods: We analyzed postmenopausal breast cancer patients (age > 50) from the 2022 NCDB with non-metastatic, ER+, HER2-negative, N1, T1-4 tumors who had undergone either RS or MP. They were categorized as African American (AA) or Caucasian (W). Associations of tumor and patient characteristics with race were assessed using Wilcoxon rank sum test and chi-square/Fisher’s exact tests. Kaplan-Meier estimates of 5-yr overall survival (OS) were determined for AA and W with respect to the results of MP and RS. Unadjusted and covariate adjusted hazard ratios (HR) were estimated with Cox proportional hazard regression to assess the association between race and OS. Subgroup analyses were conducted to explore disparities and trends.Results: 37,951 postmenopausal females were identified who met the above criteria, of whom 30,398 had RS<26 [W:27,889 (91.75%), AA: 2,509 (8.25%)], 1,982 had low risk MP [W: 1,802 (90.92%), AA: 180 (9.08%)], 4,459 had RS ≥26 [W: 3,970 (89.03%), AA: 489 (10.97%)] and 1,112 had high MP [W: 942 (84.71%), AA: 170 (15.29%)]. AA compared to W had higher rates of grade 3 disease in patients with RS<26 (13% vs 9.9%, p<0.001), RS ≥26 (53% vs 45%, p=0.014), while no difference in high MP (40% vs 37%, p=0.73) and had lower rates in low MP (6.9% vs 7.5%, p=0.011), though grade 2 rates were higher (73% vs 62%, p=0.011). Receipt of chemotherapy and endocrine therapy was similar in both races in all groups, except receipt of endocrine therapy in AA was less than W in high MP (89% vs 95%, p=0.006). Adjusted HRs showed no significant difference by race for high MP, RS ≥26, and low MP, while AA had almosta 15% increased risk of death compared to W if RS<26 [HR 1.149 (1.02-1.294), p=0.022]. Subgroup analysis of RS<26 showed that AA had increased risk of dying if Age ≤ 65 [HR 1.28(1.08-1.52)], grade 3 disease [HR 1.38 (1.05-1.82)], or had RS 11-25 [HR 1.23 (1.08-1.41)].Conclusion: In patients with low-risk RS, AA had increased risk of death compared to W, while there was no difference for high-risk RS, low or high MP. Equal endocrine therapy and chemotherapy in low RS suggest no apparent gap in care, hence these findings point towards racial difference seen in prognostication between RS and MP. Further studies are needed to determine the validation of such prognostication tests across races.
| Setting | Hazard Ratio | 95% CL | p-value |
| Low Risk Oncotype | 1.149 | 1.02-1.294 | 0.022 |
| Low Risk Mammaprint | 0.925 | 0.469-1.824 | 0.824 |
| High Risk Oncotype | 0.998 | 0.798-1.248 | 0.986 |
| High Risk Mammaprint | 1.109 | 0.798-1.248 | 0.671 |
| Other Covariates used in Adjusted Model: Age, T Stage, Grade, ER/PR status, Type of
Surgery, Radiation Therapy, Chemotherapy, Hormonal Therapy, Charlson-Deyo Score, Type of Facility, Income, Insurance, Geography |
Presentation numberPS3-08-16
Real-world patterns of chemotherapy use in premenopausal women with node-negative early-stage HR+/HER2− breast cancer and intermediate genomic risk
Ismail Ajjawi, Yale School of Medicine, New Haven, CT
I. Ajjawi1, M. Wong1, W. Wei1, T. Park2, J. Du1, M. Rozenblit1, S. Schellhorn1, A. Kahn1, N. Casasanta1, E. Winer1, M. Lustberg1; 1Yale Cancer Center, Yale School of Medicine, New Haven, CT, 2Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY.
Background:The optimal use of adjuvant chemotherapy in premenopausal women with early-stage hormone receptor-positive (HR+), HER2-negative (HER2−), node-negative breast cancer and intermediate Oncotype DX recurrence scores remains an area of clinical uncertainty. While prospective trials have provided clearer guidance for postmenopausal women, real-world treatment patterns and decision-making processes among premenopausal patients remain less well defined.Methods:We performed a retrospective analysis using the National Cancer Database (NCDB) to identify premenopausal women (<50 years) diagnosed from 2010-2021 with stage I-II, HR+/HER2−, node-negative breast cancer and recurrence scores of 11-25. We examined trends in chemotherapy omission, identified sociodemographic and clinical predictors of omission using multivariate logistic regression, and evaluated survival outcomes using Kaplan-Meier analysis and Cox regression.Results:Among 27,459 patients, 77.9% had stage I disease (n=21,408) and 22.1% had stage II (n=6,051). Chemotherapy was administered to 28.3% (n=7,773), while 71.7% (n=19,686) omitted it. Median age was 42 years among those who received chemotherapy and 44 years among those who did not. Among stage I patients, 24.2% received chemotherapy; in stage II, 42.6% received it. Chemotherapy use declined significantly from 35.4% in 2010 to 23.6% in 2021 (p < 0.001).Factors associated with chemotherapy omission included age 40-50 (OR 1.82, p < 0.001), diagnosis in 2014-2017 (OR 1.45, p < 0.001) or 2018-2021 (OR 1.58, p < 0.001) compared to 2010-2013, treatment at comprehensive (OR 1.09, p = 0.023), academic (OR 1.13, p = 0.002), or network centers (OR 1.11, p = 0.006) versus community centers, Medicare insurance (OR 1.81, p < 0.001), higher median income ≥$46,000 (OR 1.14, p = 0.002), and higher Charlson-Deyo scores (score 2: OR 1.23, p = 0.001; score ≥3: OR 1.32, p = 0.012).Conversely, chemotherapy omission was less likely among Black (OR 0.85, p < 0.001), South Asian (OR 0.68, p < 0.001), Hispanic (OR 0.83, p < 0.001), and rural patients (OR 0.88, p = 0.005) compared to non-Hispanic White and metropolitan patients, respectively.Five-year overall survival was similar between chemotherapy and no chemotherapy groups (98.1% vs. 97.6%, p = 0.244), with no significant survival benefit in Cox regression (HR 0.98, 95% CI 0.89-1.07, p = 0.51).Conclusions: Chemotherapy omission has increased over time among node negative premenopausal women with intermediate recurrence scores, yet practice variability remains. In this real-world population where treatment decisions were made by patients and physicians, there did not appear to be a survival benefit associated with chemotherapy.Ongoing prospective studies are further evaluating the safety of chemotherapy omission in premenopausal women with low genomic and higher anatomic risk, including the OFSET Trial (NRG-BR009, NCT05879926).
Presentation numberPS3-08-17
HR+ HER2- Early Breast Cancer (EBC) Patients ≥70 with High Risk 70-gene signature benefit from (Neo)adjuvant chemotherapy (CT): Real World FLEX Study
Reshma Mahtani, Miami Cancer Institute, Plantation, FL, FL
R. Mahtani1, H. Wildiers2, S. Lin1, S. Ana1, A. Schreiber3, L. Carcas1, N. Dempsey1, V. Poillucci4, M. Landon4, C. Dreezen5, H. Ramaswamy5, W. Audeh6, J. O’Shaughnessy7; 1Medical Oncology, Miami Cancer Institute, Plantation, FL, FL, 2Medical Oncology, University Hospital Leuven, Leuven, BELGIUM, 3Medical Oncology, University of Colorado Hospital, Aurora, CO, 4Medical Affairs, Agendia, Inc., Irvine, CA, 5Clinical Science, Agendia, Inc., Irvine, CA, 6CMO, Agendia, Inc., Irvine, CA, 7Medical Oncology, Baylor University Medical Center, Texas Oncology, Dallas, TX and Sarah Cannon Research Institute, Dallas, TX.
BACKGROUND: Women ≥70 are less likely to receive CT due to quality-of-life concerns and comorbidities. These patients (pts) are underrepresented in studies assessing the utility of genomic profiling to guide CT decisions. To identify the utility of a 70-gene recurrence risk assay MammaPrint (MP) we examined the relationship of age (≥70 vs <70), comorbidities, and treatment outcomes stratified by MP result in pts with HR+HER2- EBC. METHODS: The FLEX Study (NCT03053193) includes stage I-III pts with EBC who had MP performed and consented to full transcriptome and clinical data collection. Clinical characteristic differences between age groups were assessed by Chi-squared, Fisher’s exact, or Wilcoxon-Mann-Whitney tests. 4-year (yr) DRFS was assessed using Kaplan-Meier survival analysis. Log-rank tests assessed differences in endpoints between treatment groups. Comorbidity scoring assigned conditions into 7 categories. Pts were assigned 1 point for each category with a reported condition; multiple conditions in a single category counted as 1 point (Table). A multivariate Cox regression model evaluated predictors of DRFS in pts ≥70, adjusting for treatment-genomic risk interactions. RESULTS: A total of 6237 HR+HER2- EBC pts were included. 1145 were ≥70 (18%) and 4792 <70 (82%). MP results showed higher rates of low-risk tumors in the ≥70 vs <70 group (Ultralow 15.2% vs 14.6%, Low 41.5% vs 39.5%, High 1 (H1) 37.4% vs 36.7%, and High 2 (H2) 6.0% vs 9.2%, p = 0.013). Pts ≥70 vs <70 were less likely to receive (neo)adjuvant CT (26.0% vs 42.5%, p<0.001). 16.7% of pts ≥70 had no comorbidities, vs 42.0% of those <70 (p<0.001). A larger proportion of ≥70 pts vs <70 had a comorbidity score of 2 (25.2% vs 13.1%, p<0.001). Scores of 3+ were rare at 2.2% in both groups. Pts aged ≥70 with High -Risk cancers were less likely to receive CT than those <70 (H1: 49.4% vs 77.1%, H2: 72.1% vs 90.9%, p<0.001). The 4-year DRFS for pts ≥70 with High -Risk cancers trended toward superiority for those treated with CT vs endocrine thx alone, especially in H2 cancers (H1 88.5% vs 84.8%, p=.031, H2 88.1% vs 78.3%, p=.064). In multivariate analysis, CT was associated with improved DRFS in H1 (HR=0.51, p=0.015) and H2 (HR=0.35, p=0.009) pts ≥70. CONCLUSION: This Real-World Evidence study demonstrates that pts ≥70 with HR+HER2− EBC are less likely to receive CT than younger pts, despite a substantial number being classified as MP High. Among pts ≥70 with MP High, CT use was associated with improved DRFS, even in the presence of comorbidities. While the unadjusted difference in 4-year DRFS for the H2 subgroup did not reach significance, multivariate analysis showed a significant association between CT and reduced recurrence risk. These findings suggest that MP may provide additional risk information supporting individualized treatment decisions in pts ≥70 with HR+HER2- EBC even with underlying comorbidities.
Presentation numberPS3-08-18
Are we Choosing Wisely in the Omission of Radiation Therapy in Patients with Ductal Carcinoma In situ: A Review of Compliance Using the National Cancer Database
Olutayo Sogunro, Johns Hopkins, Baltimore, MD
O. Sogunro, F. Shojaeian, R. Marmor; Surgery, Johns Hopkins, Baltimore, MD.
Olutayo Sogunro1, Fatemeh Shojaeian1, Rebecca Marmor11 Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
Introduction: The Choosing Wisely (CW) campaign, initiated by the American Board of Internal Medicine, promotes evidence-based care by reducing unnecessary procedures and minimizing redundant clinical practices. In 2016, CW guidelines specific to breast oncology recommended that women over 70 years of age with small (≤2 cm), strongly hormone receptor-positive tumors and clinically negative axillary lymph nodes could safely omit both sentinel lymph node biopsy and whole breast radiation. In this study, we evaluate national trends in adherence to the omission of radiation therapy in patients with ductal carcinoma in situ (DCIS) and identify subgroup characteristics associated with compliance or non-compliance with these guidelines.
Methods: Using the National Cancer Data Base (NCDB), we identified female patients, aged≥70 years, who were diagnosed with DCIS, pathologically T0, clinicallynode negative, hormone receptor positive, and had undergone surgical treatmentbetween 2010 and 2021. Subgroup analysis evaluating Choosing Wisely compliance (CWC) inomission of radiation therapy (xRT) for patients 70 years or older was performed.
Results: Our analysis of the NCDB resulted in a total of 57,132 patients. We divided thepatients into Group 1- pre-CW (years 2010-2015) and Group 2- post-CW (years 2017-2021).Group 1 had a total of 24,658 patients and Group 2 had 32,474 patients. The average age was75 in both groups, and 84% and 81% of patients were White in Group 1 and 2, respectively. TheCharlson-Deyo Score (CDS) was similar between the two groups with a score of zero for 79% ofpatients in Group 1 and 76% in Group 2. Eighty-seven percent of patients in each group weretreated in metro areas. Fifty-one percent in both groups were classified as being treated in a community facility. In Group 1, 45% of patients had omission of radiation therapy compared to
50.3 % in Group 2. A multivariate logistic regression model analysis was performed to evaluate factors associated with a higher rate of CWC. Increased age (OR: 1.10, 95% CI 1.10-1.11, p<0.001), CDS of 3 (OR:1.31, 95% CI 1.11-1.53, p = 0.001), and treatment at an academic facility (OR:1.20, 95% CI 1.12-1.28, p <0.001), were associated with higher CWC. The papillary and cribriform histological subtypes were significantly more likely to be compliant, (OR:1.77, 95% CI 1.55-2.02, p <0.001) and (OR:1.40, 95% CI 1.19-1.64 p <0.001), respectively. The comedo subtype was 31% less likely to be compliant (OR:0.69, 95% CI 0.55-0.87, p = 0.002) compared to the DCIS non otherwise specified subtype. Race and an urban versus rural setting did not have a significant impact on compliance.
Conclusions: In this NCDB analysis of patients meeting Choosing Wisely (CW) eligibility criteria after 2017, 50.3% were compliant with the omission of radiation therapy. These findings highlight an opportunity to improve guideline implementation, particularly in non-academic settings. Ultimately, adherence to CW recommendations should be guided by shared decision-making between patients and physicians.Keywords: DCIS, Breast Cancer; Choosing Wisely; Elderly Females; Early-Stage Breast Cancer
Figure 1: Compliance Trend of omission of radiation across years 2010 – 2021
Presentation numberPS3-08-19
Evaluation of stable calcium isotope ratios in women undergoing neoadjuvant chemotherapy (NACT) with and without denosumab (GeparX)
Marion T. van Mackelenbergh, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
M. T. van Mackelenbergh1, T. Link2, C. Doering3, T. Karn4, S. Rachakonda5, M. Thill6, C. Denkert7, B. Heinrich8, V. Müller9, P. Krabisch10, F. Marmé11, C. Salat12, A. Noske13, V. Nekljudova5, B. Felder5, J. Blohmer14, S. Loibl15, A. Eisenhauer16; 1Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, GERMANY, 2Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, National Center for Tumor Diseases, Technische Universität Dresden, Dresden, GERMANY, 3Department of Obstetrics and Gynecology, Kreiskrankenhaus Torgau, Torgau, GERMANY, 4Department of Obstetrics and Gynecology, University of Frankfurt, UCT Frankfurt-Marburg, Frankfurt am Main, GERMANY, 5Medicine and Research Departement, GBG c/o GBG Forschungs GmbH, Neu-Isenburg, GERMANY, 6Department of Gynecology and Gynecological Oncology, Agaplesion Markus-Krankenhaus, Frankfurt am Main, GERMANY, 7Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), Marburg, GERMANY, 8Department of Internal Medicine, Hematology and Oncology, Haematologie-Onkologie im Zentrum MVZ GmbH, Augsburg, GERMANY, 9Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, GERMANY, 10Breast center, Klinikum Chemnitz, Chemnitz, GERMANY, 11University Medical Center Mannheim, University of Heidelberg, Mannheim, GERMANY, 12Internal medicine, haematology and internal oncology Department, Zentrum für Hämatologie und Onkologie MVZ, Munich, GERMANY, 13Pathology Medica, Zurich, Switzerland, Institute of Pathology, School of Medicine and Health, TUM, Munich, GERMANY, 14Department of Gynecology with Breast Center, Charité – Universitätsmedizin Berlin, Berlin, GERMANY, 15Medicine and Research Departement, GBG c/o GBG Forschungs GmbH, Neu Isenburg, Goethe University Frankfurt, Frankfurt am Main, GERMANY, 16GEOMAR, Helmholtz-Centre for Ocean Research Kiel, Kiel, GERMANY.
Background: Blood and bone Ca isotope compositions are in isotopic equilibrium. During equilibrium between bone material absorption and resorption the blood Ca isotope composition remains stable. However, when bone resorption is persistently greater than bone absorption, as seen in osteoporosis, the blood Ca isotope ratio decreases. This indicates a net shift of calcium from the bone into the blood. The calcium isotope marker (CIM) is a novel biomarker that sensitively detects changes in bone calcium balance in humans. For the first time, this study aims to evaluate the effects of neoadjuvant chemotherapy (NACT), with and without denosumab (Dmab), on bone calcium balance in postmenopausal women with early breast cancer enrolled in the GeparX trial (NCT02682693). Methods: This analysis was conducted on a predefined cohort of the patients (pts) enrolled in the GeparX trial (Blohmer, JAMA Oncol. 2022), specifically including postmenopausal pts with hormone receptor-positive (HR+), HER2-negative (HER2-) breast cancer who received NACT with weekly nab-paclitaxel followed by epirubicin/cyclophosphamide. Patients included in this cohort were randomized to receive Dmab or not in addition to NACT. Eligible pts had two serum samples available: one prior to treatment and one after completion of NACT. In total, 20 pts included in this study were treated in the NACT + Dmab arm and 29 received NACT alone. Paired serum samples (collected before therapy and before surgery) were analyzed for CIM ratios. Results: A total of 98 serum samples were tested for CIM serum levels and valid measurements were obtained for all. Pre-treatment mean CIM values were comparable in both cohorts (mean±SD -0.87±0.16 ‰ vs. -0.94±0.19 ‰; p=0.115; without vs. with Dmab, respectively), indicating a net loss of calcium; however, the difference was not statistically significant. In pts treated with NACT alone, a significant decrease in CIM-serum levels to -1.02±0.17 ‰ was detected signifying bone calcium loss (p<0.001) and osteoporotic situation. In contrast, pts treated in the Dmab arm had a significant increase in CIM-serum levels reaching a mean of -0.61±0.18 ‰ (p<0.001), indicating a net accumulation of calcium and new bone formation. No relationship between CIM levels and pathologic complete response was observed. Pre-treatment levels of CIM showed a mild positive correlation with age (slope = 10.9; R² = 0.10) and body mass index (BMI) (slope = 4.3; R² = 0.04). Post-treatment, the CIM levels showed a reverse trend with a mild negative correlation for age (slope = -5.4; R2 =0.06) and no correlation with BMI (slope = 0.11; R2 = 0). Within the post-treatment cohort, however, age and BMI showed contrasting weak correlations with CIM levels in relation to treatment with Dmab: there was a negative correlation with age for pts who did not receive Dmab (slope = -5.54; R² = 0.03), and a positive correlation with age for pts who did (slope = 13.03; R² = 0.12); there was no correlation with BMI for pts who did not receive Dmab (slope = -0.02; R² = 0), and a positive correlation with BMI for pts who did (slope =9.82; R² = 0.19). Hence, Dmab was associated with improved bone mineralization post-treatment, particularly in older pts and those with higher BMI. Conclusion: Analyses of CIM serum levels provide critical insights into the extent of bone mineral loss induced by NACT in postmenopausal pts with HR+/HER2− breast cancer. Concurrent administration of Dmab appears to mitigate this effect by promoting bone mineralization. CIM serum levels may serve as a valuable, easily measurable biomarker for monitoring bone mineral balance in patients undergoing chemotherapy for breast cancer, enabling early intervention in case of bone mass loss and preventing osteoporosis.
Presentation numberPS3-08-20
Sex-based differences in hormone receptor-positive breast cancer treatment and outcomes: A Veterans Health Administration 20-year cohort study
Ariana Naaseh, Washington University in St. Louis School of Medicine, St. Louis, MO
A. Naaseh1, J. B. Gruber2, R. L. Aft1, K. N. Maxwell3, M. W. Schoen4; 1Section of Surgical Oncology, Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 2Department of Medicine, Veterans Affairs St. Louis Health Care System, St. Louis, MO, 3Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine, Philadelphia, PA, 4Division of Hematology and Medical Oncology, Department of Internal Medicine,, Saint Louis University School of Medicine, St. Louis, MO.
Background: Estrogen receptor-positive (ER+) breast cancer is the most commonly diagnosed subtype in both male and female patients. However, due to the rarity of male breast cancer overall, there remains limited understanding of how this specific subtype is treated and how outcomes compare between sexes. This study aims to compare survival and contemporary treatment for ER+ male breast cancer (MBC) and female breast cancer (FBC) patients within the Veterans Health Administration (VHA). Methods: We conducted a retrospective cohort study of patients diagnosed with ER+ MBC and FBC between 2000 and 2020 using national data from the VHA Informatics and Computing Infrastructure database. Demographics, tumor characteristics, treatment (surgery, chemotherapy, radiation), and survival were compared between MBC and FBC patients. Stage 0 and IV patients were excluded from our analysis. Descriptive statistics, t-tests, and chi-square tests were used to compare the cohorts. Cox proportional hazards regression models were utilized to examine overall survival, controlling for age, race, body mass index, and grade. Results: Of the 6,336 total patients identified, 17.9% (n=1,134) were male. ER+ MBC patients were 66.0% White (n=754), 23.0% Black (n=257), and 11.0% Other/Unknown race (n=123). Most ER+ MBC patients were Non-Hispanic (n=1024, 90.0%). The average BMI of ER+ MBC patients was 30.6. ER+ MBC patients were significantly more likely to be diagnosed at an older age (69.0 vs. 57.7, p <0.001). Significantly more MBC patients were diagnosed with stage II (46.5% vs. 33.2%, p <0.001) and stage III disease (20.9% vs. 10.1%, p <0.001) than FBC. Compared to ER+ FBC patients, MBC patients had significantly higher rates of ductal histology (87.0% vs. 82.0%, p <0.001), grade 2 disease (39.0% vs 32.0%, p <0.001), and grade 3 disease (24% vs. 17.0%, p <0.001). MBC and FBC ER+ patients had similar rates of undergoing surgery (MBC: 94.0% vs. FBC: 96.0%) and chemotherapy (40.0% vs. 43.3%). Significantly more ER+ FBC patients underwent radiation therapy than MBC patients (53.0% vs. 23.0%, p <0.001). In a Cox proportional hazard model, stage I (HR 1.41, 95% CI: 1.06-1.86) and stage II (HR 2.06, 95% CI: 1.64-2.58) ER+ MBC patients had significantly higher risk-adjusted hazard of all-cause mortality when compared to FBC patients. Conclusions: Despite receiving similar surgical and chemotherapy treatments, ER+ MBC were more often diagnosed at later stages and had significantly worse risk-adjusted survival compared to ER+ FBC. These findings highlight the need for increased awareness, earlier detection, and tailored treatment strategies for ER+ MBC patients.
Presentation numberPS3-08-21
Real-world survival outcomes in patients with HR+/HER2− early breast cancer meeting Monarche and Natalee criteria: analysis from a Brazilian cohort
Natalia Cristina C. Nunes, Americas oncologia, Rio de Janeiro, Brazil
N. C. Nunes1, M. Monteiro2, G. Carvalho1, J. Pecoraro1, T. Ferreira1, D. Azevedo1, P. Santos1, R. Barroso-Sousa3; 1Instituto Americas, Americas oncologia, Rio de Janeiro, BRAZIL, 2Instituto Americas, Americas oncologia, São Paulo, BRAZIL, 3Instituto Americas, Americas oncologia, Brasilia, BRAZIL.
Background: While adjuvant CDK4/6 inhibitors improve outcomes in selected patients with high-risk HR+/HER2− early breast cancer (EBC), real-world evidence from Latin America remains limited. We aimed to evaluate clinical characteristics and survival outcomes of patients treated in a private oncology facility in Brazil, stratified by eligibility for MonarchE and NATALEE criteria. Methods: We analyzed HR+/HER2− stage I-III EBC patients treated from 2012 to 2024. Patients who received adjuvant abemaciclib were excluded from the analysis. Eligibility followed clinical-pathological criteria from MonarchE and NATALEE (excluding Ki-67). Kaplan-Meier and Cox models were used to estimate overall survival (OS) and disease-free survival (DFS) across groups: MonarchE and NATALEE-eligible, NATALEE-low-risk (eligible for NATALEE but not MonarchE), and non-eligible. Results: Among 2,609 patients, 18% met MonarchE criteria, 56% NATALEE criteria, and 44% were non-eligible. The NATALEE-low risk subgroup accounted for 38% of the cohort. Most patients had stage I or II disease (84%) and breast-conserving surgery (64%). 67% of the population were post-menopausal, with a mean age of 59 years old. 24% of patients are black or mixed race. Patients in the MonarchE- and NATALEE-eligible groups were more frequently premenopausal (42% and 39%, respectively, vs. 25% in the ineligible group), more likely to have received chemotherapy (89% and 77%, respectively, vs. 21%), and more often underwent ovarian function suppression (8.3% and 7.5% vs. 1.5%). After a median follow-up of 37 months, the multivariable-adjusted hazard ratios (HR) for recurrence were 2.31 (95%CI 1.67-3.18; p < 0.001) for MonarchE and 2.23 (1.49-3.34; p < 0.001) for NATALEE. At 36 months, DFS rates were 86.0% in MonarchE-eligible patients, 90.2% in NATALEE-eligible patients, 92.0% in NATALEE-low-risk patients, and 96.0% in non-eligible patients; at 60 months, the estimated DFS was 80.0%, 85.1%, 87.5%, and 93.0%, respectively. OS was significantly worse in MonarchE- and NATALEE-eligible patients compared to non-eligible patients, with an HR of 4.16 (95% CI, 2.54-6.82; p < 0.001). In the NATALEE-low-risk subgroup, OS was also inferior compared to the non-eligible group, with an HR of 2.36 (95% CI, 1.47-3.80; p < 0.001). At 36 months, OS rates were 89.0% (MonarchE-eligible), 93.6% (NATALEE-eligible), 95.0% (NATALEE low-risk-only), and 97.0% (non-eligible); 60-month estimates were 84.5%, 89.2%, 91.3%, and 95.4%, respectively. Independent predictors of poorer OS included ≥3 positive lymph nodes, T3/T4 tumors, use of chemotherapy, and absence of endocrine therapy. Conclusions: A substantial proportion of HR+/HER2− EBC patients treated in Brazilian private practice met eligibility for adjuvant CDK4/6 inhibitors, especially under the broader NATALEE criteria. These patients experienced significantly worse DFS and OS compared to non-eligible patients. These findings support expanding access to adjuvant CDK4/6 inhibitors and underscore the need to address treatment disparities in Latin America.
Presentation numberPS3-08-22
Uncovering mechanisms of ILC glucocorticoid receptor activity in cell cycle regulation and bone metastasis using in vivo models and human samples
Baylee A Porter-Hansen, UT Southwestern Medical Center, Dallas, TX
B. A. Porter-Hansen1, A. Briceno1, H. Borges2, A. Devilbiss1, K. Dean2, L. Bennett1, S. D. Conzen1; 1Internal Medicine, UT Southwestern Medical Center, Dallas, TX, 2Bioinformatics, UT Southwestern Medical Center, Dallas, TX.
San Antonio Breast Cancer Conference Abstract 2025Title: Uncovering mechanisms of ILC glucocorticoid receptor activity in cell cycle regulation and bone metastasis using in vivo models and human samples. Estrogen receptor (ER+) positive invasive lobular carcinoma comprises 15% of all breast cancer and is the second most common hormone-sensitive breast cancer (BC). Uniquely, ILC lacks e-cadherin expression resulting in relatively discohesive and multi-focal tumors within the breast, making it difficult to detect with conventional imaging. ILC is also highly metastatic to bone and compared to triple-negative breast cancer or luminal B ER+ BC, ILC has a low proliferative rate <15% and slower growing phenotype. Previously we have observed in mammary intraductal nipple injection (MIND) in vivo models that high GR (versus low GR) expression in ILC is associated with markedly reduced ILC cell proliferation without reducing metastatic colonization ability. Here we show the anti-tumor growth effect of GR+ on ILC cells in the primary engrafted mammary glands and the pro-metastatic effect. We observed decreased tumor cell proliferation and tumor burden 90 days post-mammary gland injection of GR+ expressing ILC cells compared to GR null or GR low cells in the MDA-IV-134, and BCK4 cell lines. Remarkably the slowed tumor growth had equivalent metastatic burden to GR-negative ILC with much larger primary tumors. This suggests that reduced proliferation in the mammary gland tumor does not affect metastatic efficiency in distant organs. We hypothesize that tumor intrinsic GR may increase bone metastatic colonization through activation of “bone mineralization gene expression signaling pathways” and “IL-6 signaling pathways” associated with GR activation in parallel in vitro experiments. Furthermore, ongoing studies including advanced light sheet microscopy and fluorescent-activated cell sorting allow us to quantify ILC cells in the bone as early as 90 days post MG nipple injection. In addition, we are performing bulk RNA sequencing from MG tumors to identify specific in vivo GR+ activated gene expression pathways that promote GR+ ILC bone metastasis. Future studies will also include GR modulation to uncover GR-regulated genes that favor more efficient ILC bone metastases.
Presentation numberPS3-08-23
Endocrine resistance: prevalence and outcomes in hispanic women with early breast cancer
Areli Velazquez Martinez, Instituto Nacional de Cancerologia, Mexico City, Mexico
A. V. Martinez1, P. A. Cabrera-Galeana1, A. Mohar-Betancourt2, J. E. Hernandez-Hernandez3, C. A. Gonzalez-Reyes1, A. Maliachi-Diaz1, A. Garcilazo-Reyes1, E. Bargallo-Rocha1, C. H. Arce-Salinas1, D. F. Flores-Diaz1; 1Breast Cancer Unit, Instituto Nacional de Cancerologia, Mexico City, MEXICO, 2Unidad Biomedica de Inv en Cáncer, Instituto Nacional de Cancerologia, Mexico City, MEXICO, 3Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, MEXICO.
Background: Hormone receptor (HR)-positive early breast cancer (EBC) accounts for 60-70% of cases(1). However, EBC experience probability of recurrence rate of 3%, which reflects endocrine resistance (ER). The impact of ER on clinical outcomes has not been thoroughly examined in EBC, especially in the hispanic population, despite their increased vulnerability and disproportionate burden of disease.Methods: Retrospective analysis of 1,551 ER(+)-EBC patients at from 2007 to 2017. Data on clinico-pathologic and treatment variables were collected from electronic medical records. Recurrence cases classified: primary endocrine resistance (1ER) within 24 months,secondary endocrine resistance (2ER) between 24-60 m, and endocrine-sensitive recurrence (3ESR) after 60 m of relapse. Descriptive statistics defined the population’s general characteristics. Differences were analyzed with Chi-square for categorical variables and t-tests for continuous variables. The Kaplan-Meier method and log-rank test were applied for survival analysis, while Cox regression for multivariate analysis. A p-value < 0.05 indicated statistical significance.Results Patients had a median age of 52.2 years (IQR 45.0-62.0). 574 (37%) were premenopausal,and 343 (22%) had a high nodal burden (N2/N3). In a median follow-up of 9.1 years (IQR5.9-12.0), 392 patients (25%) experienced recurrence, with an annual rate of 3-5% during the first five years. Among them, 128 patients (33%) had 1ER, 182 (46%) had 2ER, and 82(21%) had 3ESR. Table 1 displays population characteristics at recurrence. Clinical stage III, Grade 3, and H-Score expression of estrogen and progesterone receptors varied significantly among the subgroups. Oncotype was conducted in 130 patients, with median scores of 29 for 1ER, 24 for 2ER, 19 for 3ESR, and 14 for the non-recurrent group(p<0.001). At 10 years, overall survival (OS) rates for 1ER, 2ER, and 3ESR were 13%, 32%, and 79%, respectively (p<0.0001). Even in cases of metastatic visceral disease, OS rates show significant differences: 37, 80, and 173 months in the 1ER, 2ER, and 3ESR subgroups respectively (p<0.0001). Multivariate analysis for OS show the risk of death for women with 1ER HR 12.7; 95% CI (8.12-20.00) and compared to secondary resistance HR 4.4; 95% CI (2.81-6.8), as well as in those patients with visceral metastatic disease HR 1.7 95% CI (1.33-2.2) p<0.001. Conclusions: Our study found higher relapse rates and endocrine resistance, potentially due to delays in diagnosis and clinical risk. Women with 1ER had a higher mortality risk. This analysis highlights the need to improve early diagnosis, equal access to adjuvant CDK4/6 therapies, and enhance surveillance strategies in high clinical risk EBC.
| <24 months (n=128) | 24-72 months (n=182) | >72 months (n=82) | p-value | |||||||||||||||||||||
| ER H-Score | 185.00 [87.50, 240.00] | 180.00 [120.00, 210.00] | 200.00 [160.00, 247.50] | 0.041 | ||||||||||||||||||||
| PR H-Score | 80.00 [10.00, 150.00] | 100.00 [40.00, 160.00] | 120.00 [45.00, 200.00] | 0.011 | ||||||||||||||||||||
| Hormonal status | 0.068 | |||||||||||||||||||||||
| Pre-menopause | 67 (52.3) | 90 (49.5) | 30 (36.6) | |||||||||||||||||||||
| Post-menopause | 61 (47.7) | 92 (50.5) | 52 (63.4) | |||||||||||||||||||||
| Nodal status (clinical) | 0.001 | |||||||||||||||||||||||
| N0 | 18 (14.1) | 42 (23.1) | 20 (24.2) | |||||||||||||||||||||
| N1 | 37 (28.9) | 77 (42.3) | 40 (48.8) | |||||||||||||||||||||
| N2 | 55 (43.0) | 48 (26.4) | 17 (20.7) | |||||||||||||||||||||
| N3 | 18 (14.1) | 15 (8.2) | 5 (6.1) | |||||||||||||||||||||
| Tumor size (clinical) | <0.001 | |||||||||||||||||||||||
| T1 | 8 (6.2) | 27 (14.8) | 18 (22.0) | |||||||||||||||||||||
| T2 | 48 (37.5) | 74 (40.7) | 36 (43.9) | |||||||||||||||||||||
| T3 | 24 (18.8) | 47 (25.8) | 16 (19.5) | |||||||||||||||||||||
| T4 | 48 (37.5) | 34 (18.7) | 12 (14.6) | |||||||||||||||||||||
| Tumor grading (IDC)(n=370) | 0.490 | |||||||||||||||||||||||
| G1 | 25 (19.8) | 39 (23.5) | 24 (30.8) | |||||||||||||||||||||
| G2 | 45 (35.7) | 54 (32.5) | 25 (32.1) | |||||||||||||||||||||
| G3 | 56 (44.4) | 73 (44.0) | 29 (37.2) | |||||||||||||||||||||
| Pathological response (n=248) | 0.927 | |||||||||||||||||||||||
| Incomplete | 91 (91.9) | 99 (90.8) | 36 (90.0) | |||||||||||||||||||||
| Complete | 8 (8.1) | 10 (9.2) | 4 (10.0) |
Presentation numberPS3-08-24
Association of Oncotype DX Recurrence Score with Germline Mutations in Cancer Susceptibility Genes Including BRCA1/2 in HR+/HER2− Early Breast Cancer
Vasileios Venizelos, Metropolitan Hospital, Piraeus, Greece
V. Venizelos1, K. Papazisis2, C. Markopoulos3, G. Xepapadakis4, R. Iosifidou5, G. Kapetsis6, S. Giannoulakis6, N. Tsoulos6, A. Meintani6, D. Bouzarelou6, G. Tsaousis6, D. Grosomanidis7, N. Bredakis8, F. Zagouri9, C. Christodoulou10, K. Anastasakou11, M. Paraskeva12, D. Mavroudis13, N. Michalopoulos14, D. Tryfonopoulos15, S. Papadopoulos16, A. Adamidis17, D. Dimas18, E. Angelidou19, E. Papadopoulou6, G. Nasioulas6; 1Breast Clinic, Metropolitan Hospital, Piraeus, GREECE, 23rd Dpt of Oncology, Interbalcan European Medical Centre, Thessaloniki, GREECE, 3Medical School, National and Kapodistrian University of Athens, Athens, GREECE, 42nd Breast Unit, IASO, General Maternity and Gynecology Clinic, Athens, GREECE, 5Breast Surgical Oncology Clinic, Theageneio Anticancer Hospital, Thessaloniki, GREECE, 6Medical Dpt, Genekor Medical S.A., Gerakas, GREECE, 74th Breast Clinic, Mitera Hospital, Athens, GREECE, 81st Breast Unit, IASO, General Maternity and Gynecology Clinic, Athens, GREECE, 9Department of Clinical Therapeutics,, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, GREECE, 102nd Oncology Department, Metropolitan Hospital, Piraeus, GREECE, 11Breast Clinic, Metropolitan General Hospital, Athens, GREECE, 12Oncology Dpt, General Hospital of Rhodes, Rhodes, GREECE, 13Department of Medical Oncology, University General Hospital of Heraklion, Crete, GREECE, 14Breast unit, 1st Propaedeutic Dept. of Surgery, “Hippocratio” General Hospital, Medical School, Athens, GREECE, 152nd Department of Medical Oncology, Agios Savvas Cancer Hospital, Athens, GREECE, 16Breast Clinic, Thessaloniki Hospital, Thessaloniki, GREECE, 17Oncology Dpt, Interbalcan European Medical Centre, Thessaloniki, GREECE, 18Breast Clinic, Athens Medical Center, Athens, GREECE, 19Breast Clinic Rhodes, Euromedica General Clinic Rhodes, Rhodes, GREECE.
Background: In early-stage estrogen receptor-positive (ER+), HER2-negative (HER2−) breast cancer, the Oncotype DX® Recurrence Score (RS) is a clinically validated 21-gene assay that informs prognosis and predicts the benefit of adding chemotherapy to endocrine therapy. While germline pathogenic variants (PVs) in cancer susceptibility genes may influence tumor biology, treatment response and surgical management, their relationship with RS remains insufficiently explored and has been primarily limited to BRCA1/2 genes. This study investigates the association between Oncotype DX RS and the presence of germline PVs across a broader spectrum of breast cancer predisposition genes. Methods: We retrospectively analyzed data from a Greek cohort of early-stage ER+, HER2- breast cancer pts who underwent both germline testing using a 52-gene panel and Oncotype DX testing between 2015 and 2025. Pts were stratified by Oncotype DX Recurrence Score (RS) into low (RS 0-10), intermediate (RS 11-25), and high-risk (RS >25) genomic groups. Germline testing results were categorized into three groups: (a) pathogenic/likely pathogenic (P/LP) variants identified, (b) variants of uncertain significance (VUS), and (c) no variants detected. The P/LP group was further subdivided based on the presence of variants in (a) BRCA1/2, (b) other high-risk genes, (c) moderate-risk genes, and (d) incidental findings in additional genes. Results: A total of 1,465 pts were analyzed. The distribution of Oncotype DX RS across the various germline testing groups is detailed in the table below.
| Group | N | Median RS | RS 0-10 N(%) | RS 11-25 N(%) | RS >25 N(%) | ||||||
| Overall cohort | 1465 | 16 | 261(18%) | 959(65%) | 245(17%) | ||||||
| P/LP variants (all) | 239 | 18 | 29(12%) | 156(65%) | 54(23%) | ||||||
| – BRCA1/2 | 42 | 24.5 | 1(2%) | 21(50%) | 20(48%) | ||||||
| – Other high-risk genes (CDH1, PALB2, PTEN) | 20 | 23.5 | 2(10%) | 9(45%) | 9(45%) | ||||||
| – Moderate-risk genes (ATM, MRE11A, NBN, RAD50, RAD51C, BRIP1, BARD1, CHEK2) | 111 | 16 | 16(14%) | 78(70%) | 17(16%) | ||||||
| – Other genes (incidental findings) | 66 | 17 | 16(15%) | 48(73%) | 8(12%) | ||||||
| VUS identified | 711 | 16 | 128(18%) | 470(66%) | 113(16%) | ||||||
| No P/LP or VUS detected | 515 | 15 | 104(20%) | 333(65%) | 78(15%) |
Conclusion: The distribution of RS varied notably across genetic variant groups. Pts with BRCA1/2 and other high-risk gene variants exhibited statistically significant higher median RS values (p25 (p<0.0001) compared to all other subgroups (moderate risk variants, incidental findings, VUS and cases without detected variants). These patterns suggest that pts with germline mutations in high-risk susceptibility genes are more likely to have an unfavorable prognosis for distant recurrence and a greater likelihood of receiving chemotherapy, as indicated by their elevated RS values
Presentation numberPS3-08-25
Integrated Prognostic and Chemotherapy Sensitivity Assessment Using Multiple Gene Assays in ER-positive, HER2-negative Early Breast Cancer
Maiko Nishida, Kyoto Second Red Cross Hospital, Kyoto, Japan
M. Nishida1, R. Tsunashima2, N. Hirotani3, S. Kitano3, C. Matsui3, S. Matsumoto3, A. Watanabe4, C. Kato3, M. Morita3, K. Sakaguchi3, Y. Naoi3; 1Breast Surgery, Kyoto Second Red Cross Hospital, Kyoto, JAPAN, 2Breast Surgery, Independent Researcher, Osaka, JAPAN, 3Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, JAPAN, 4Breast Surgery, Fukuchiyama City Hospital, Fukuchiyama-shi, JAPAN.
Background: To assess recurrence risk in ER-positive, HER2-negative early breast cancer, multigene assays (MGAs) are useful in addition to clinicopathological factors. In Japan, Curebest™ 95GC was developed in 2011 and introduced clinically in 2013. Oncotype DX® was added to national health insurance in 2023. While adjuvant chemotherapy is often considered for patients classified as high-risk by any MGA, treatment decisions can be complex. Furthermore, breast cancers identified as high-risk by MGAs often exhibit higher pathological complete response (pCR) rates, suggesting increased sensitivity to chemotherapy. This study aimed to evaluate MGAs from two perspectives—prognostic prediction and NAC sensitivity—to inform individualized treatment strategies. Materials and Methods:We analyzed microarray gene expression data from public databases to identify patients with ER-positive, HER2-negative early breast cancer. In this study, Curebest™ 95GC, developed in Japan, was used post-analysis for research purposes only. Based on microarray expression data, we calculated PAM50, 21GC (Oncotype DX® surrogate analyses), EndoPredict (EP), and either 95GC or 155GC. In Study 1(n=555), we assessed the prognostic value of each MGA and evaluated recurrence risk based on the number of high-risk classifications. In study2(n=550), we evaluated the predictive value of each MGA for pCR rate and analyzed the relationship between NAC sensitivity and the combined assessment of the number of high-risk classifications treated with NAC. Results: In Study 1, all four MGAs (PAM50, 21GC, EP, and 95GC) significantly stratified DRFS between high- and low-risk groups (p =8.93e-08, 2.31e-08, 3.64e-07, and 9.25e-10), with comparable prognostic performance. Patients were grouped by the number of high-risk classifications. A 5-year DRFS of 97% was observed in patients with 0 high-risk classifications, compared to 66% in those with 4. Patients were further stratified into two groups: those with 0 or 1 high-risk classification and those with ≥2. The 0-1 group showed a 5-year DRFS of 96.7%, while the ≥2 group had a significantly worse prognosis at 79.0% (p=2.04e-11). The combined assessment provided better prognostic accuracy than any individual MGA alone. Notably, patients with only one high-risk result showed excellent outcomes, suggesting endocrine therapy alone may suffice. Conversely, those deemed low-risk by all MGAs may not require adjuvant chemotherapy.In Study 2, high-risk patients had significantly higher pCR rates across all MGAs. 155GC showed the strongest stratification (p=1.62e-08). Patients classified as high-risk by all four MGAs had significantly higher pCR rates than those with 0-3 high-risk classifications (p=9.37e-11). 95GC provides CEL files containing expression data for all 20,000 genes. This allows concurrent calculation of multiple MGAs, facilitating evaluation and reducing cost. Use of CEL files supports integrated MGA analysis, and may support precise personalized treatment. Conclusion: Combining four MGAs improves prediction over single MGAs. This integrated genomic approach offers a promising strategy to optimize individualized treatment in ER-positive, HER2-negative early breast cancer.
Presentation numberPS3-08-26
Comparison of Breast Cancer Index Scores from Core-Needle Biopsies versus Surgical Excisions in Early-Stage Breast Cancer
Nimmi S. Kapoor, UCLA, Los Angeles, CA
N. S. Kapoor1, N. K. Siuliukina2, Y. Zhang2, A. K. Anderson2, B. O’Neal3, J. Roberts4, K. Church-Nguyen5, K. Treuner3, A. Bardia6; 1Surgery, UCLA, Los Angeles, CA, 2R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 3R&D, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 4Diagnostics, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 5Dx Oncology, Biotheranostics, Inc. A Hologic Company, San Diego, CA, 6Department of Medicine, UCLA, Los Angeles, CA.
Background: Breast cancers are typically diagnosed by core-needle biopsy (CNB) and managed with surgical excision. While CNB collects a small tissue sample for analysis, surgical excision involves removal of a larger amount of tissue, potentially affecting molecular profiling results due to differences in sample size and tissue processing. Breast Cancer Index (BCI) is a validated gene expression assay that provides the risk of overall (0-10 year) and late (5-10 year) distant recurrence and predicts the likelihood of benefit from extended endocrine therapy in hormone receptor positive (HR+) early-stage breast cancer. BCI validation in clinical studies has been performed largely from surgical excisions. This study compares BCI scores and its components, HOXB13/IL17BR ratio (H/I) and Molecular Grade Index (MGI), between CNBs versus surgical excisions submitted for routine BCI testing to evaluate the reliability of BCI results using CNB samples. Methods: Tissue samples were identified using the BCI clinical database, which contains anonymized gene expression data from commercial BCI cases from multiple centers in the U.S. and corresponding clinicopathologic variables. BCI, H/I, and MGI scores were retrieved from the database. The dataset was balanced for nodal status, tumor size, and tumor grade using stratified random sampling, yielding a cohort of CNB cases with a 1:5 matched excision group. Pairwise comparisons by sample type were conducted using BCI scores and delta CT values, which represent normalized gene expression levels adjusted for RNA input and RT-PCR efficiency, analyzed via the Wilcoxon signed-rank test. Results: Of 1,554 patients with HR+ early-stage breast cancer with BCI scores included, 262 (17%) were from CNB and 1292 (83%) samples were from surgical excisions. All patients had HR+ tumors, 92% were post-menopausal and average age was 68 years. Most tumors were T1 (n=1236, 80%) and Grade 2 (n=834, 54%). There were no significant differences in clinicopathologic variables between groups. Among all samples, 63% were classified as (H/I)-Low and 37% as (H/I)-High; 47% were BCI low risk and 53% high risk. In N0 patients (n=1,152), there were no significant differences between CNB and excision groups for BCI, H/I, or MGI scores. Expression of the two H/I genes HOXB13 and IL17BR was also not significantly different between both groups. Individual comparisons for the 5 MGI genes showed no significant differences, with the exception of RRM2 (p=0.0091). Similarly, in N1 patients (n=402) no statistical significance was observed in BCI, H/I and MGI score between the two groups. The comparison of H/I and MGI individual genes also showed no statistical significance between CNB and excision. Conclusion: From this large dataset, BCI, H/I and MGI scores, as well as individual gene expression levels, are comparable between CNB and surgical excision specimens in both N0 and N1 early-stage breast cancer patients, supporting the use of CNB tissue for genomic classification with BCI.
Presentation numberPS3-08-27
Repower: a real-world noninterventional study of outcomes and experiences in patients with hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2−) early breast cancer (EBC) treated with an adjuvant cyclin-dependent kinase 4 and 6 inhibitor (CDK4/6i) plus endocrine therapy (ET)
Maryam Lustberg, Yale University School of Medicin, New Haven, CT
M. Lustberg1, S. Wang2, A. Casas3, Y. Zhu4, N. Derakshan5, G. Mason6, M. Campone7, M. De Laurentiis8, M. Fernandez-Abad9, J. Sirieix10, M. Akdere11, F. Ye12, P. Dominguez Castro13, A. Ring14; 1Department of Medicine, Yale University School of Medicin, New Haven, CT, 2Medical Oncology, Cancer Center,Sun Yat-sen University, Guangzhou, CHINA, 3Medical Oncology, University Hospital Virgen del Rocio, Sevilla, SPAIN, 4Patient Advocate, Patient Advocate, Shanghai, CHINA, 5Head of the GRiT Centre (Growth and Resilience in Trauma), University of Reading, Reading, UNITED KINGDOM, 6Executive Director, Inflammatory Breast Cancer Research Foundation, West Lafayette, VA, 7Medicine, Institut de Cancérologie de l’Ouest, St. Herblain, FRANCE, 8Breast and Thoracic Oncology, National Cancer Institute “Fondazione Pascale”, Napoli, ITALY, 9Breast and Gynecological Tumors, Ramón y Cajal Hospital, Madrid, SPAIN, 10Global Medical Affairs, Novartis Pharma S.A.S, Rueil-Malmaison, FRANCE, 11Global Medical Affairs, Novartis Pharma AG, Basel, SWITZERLAND, 12RWE Solid Tumor, Novartis Pharmaceuticals Corporation, East Hanover,, NJ, 13RWE and Innovative Evidence, Novartis Ireland Limited, Dublin, IRELAND, 14Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Surrey, UNITED KINGDOM.
Background: Ribociclib plus a nonsteroidal aromatase inhibitor demonstrated a significant invasive disease-free survival (iDFS) benefit in a broad population of patients with HR+/HER2− EBC, including those with node-negative disease in the phase 3 NATALEE trial. Abemaciclib plus ET also showed a significant iDFS benefit in patients with high-risk node-positive HR+/HER2− EBC in the phase 3 monarchE trial. However, evidence gaps persist related to real-world effectiveness, patient experiences, and safety with CDK4/6is in EBC. REPOWER, a retrospective clinical charts review and prospective patient-reported outcome (PRO) assessment, aims to characterize patients, clinical benefit, treatment experiences, and safety associated with adjuvant CDK4/6i treatment in the real-world EBC setting. Study design: REPOWER is a multicenter, international (13 countries in North American, Europe, and Asia), noninterventional, hybrid study assessing effectiveness; adherence, treatment satisfaction, and quality of life through PROs; and safety in patients with HR+/HER2− EBC treated with a CDK4/6i + ET. Data will be collected from patients aged ≥18 years with HR+/HER2− stage II or III EBC who have initiated adjuvant treatment with a CDK4/6i, in accordance with its approved local label, within 14 days prior to enrollment. Patients who have had a local or distant breast cancer recurrence before the CDK4/6i initiation date, or who enrolled in clinical trials ≤12 months prior to the initiation date, will be excluded. This study has a hybrid design, including a retrospective analysis of patient data from electronic health records and prospective data from PRO questionnaires and interviews. Two groups will be included, patients treated with ribociclib + ET as the primary cohort and patients treated with abemaciclib + ET as an exploratory cohort. The index date (time [T] 0) will be defined as the initiation of adjuvant ribociclib/abemaciclib. Patients will be followed up from T0 until death, lost to follow-up, end of the study period, or enrollment in a clinical trial, whichever occurs first. The expected enrollment period will be between 12-18 months, with planned patient follow-up for ≥36 months and a maximum follow-up of 54 months. This trial aims to enroll approximately 2650 patients in the ribociclib + ET cohort and 250 patients in the abemaciclib + ET cohort. The primary and secondary objectives will assess patients in the ribociclib + ET cohort. Primary outcomes include iDFS and distant disease-free survival (defined using Standardized Definitions for Efficacy End Points criteria). Select secondary objectives include baseline characteristics, PROs (adherence, health-related quality of life, treatment satisfaction, work productivity, patient self-reported treatment experiences), safety, and treatment patterns in the adjuvant and metastatic settings if distant recurrences occur. Subgroup analyses by nodal status, menopausal status, stage, and chemotherapy use will be performed. The exploratory objectives will assess patients in the abemaciclib + ET cohort. Objectives include baseline characteristics, PROs, and adverse events.
Presentation numberPS3-08-28
Real-world evidence on the use of a 21-gene recurrence score assay in patients with HR+/HER2− early breast cancer in japan: a nationwide claims database analysis
Masamitsu Hihara, Exact Sciences K.K., Tokyo, Japan
M. Hihara1, H. Bando2, A. Shimomura3, N. Sugiyama1, N. Yoshii1, R. Kawai4, A. Shintani4, S. Saji5; 1Medical Affairs Japan, Exact Sciences K.K., Tokyo, JAPAN, 2Department of Breast and Endocrine Surgery, Institute of Medicine, University of Tsukuba, Ibaraki, JAPAN, 3Department of Breast and Medical Oncology, National Center for Global Health and Medicine, Tokyo, JAPAN, 4Department of Medical Statistics, Osaka Metropolitan University Graduate School of Medicine, Osaka, JAPAN, 5Department of Medical Oncology, Fukushima Medical University, Fukushima, JAPAN.
Background: The Oncotype DX® test, 21-gene recurrence score assay, is a commercially available genomic assay that calculates a Recurrence Score® (RS) result ranging from 0 to 100 based on the expression of 21-genes. The RS result provides prognostic information independent of clinicopathological factors and predicts the benefit of chemotherapy (CT) to guide adjuvant treatment decisions in patients (pts) with hormone receptor-positive (HR+), HER2-negative (HER2-) early breast cancer (BC). In Japan, the Oncotype DX test has been reimbursed under public health insurance since September 1, 2023, and is expected to help optimize CT decision-making. However, real-world evidence (RWE) on its clinical use remains limited. This study presents the first large-scale, real-world analysis in Japan using a nationwide claims database to describe clinical and demographic characteristics, as well as treatment patterns associated with the use of Oncotype DX testing. Methods: We conducted a retrospective cohort study of pts with BC who underwent Oncotype DX testing between the start of reimbursement (September 2023) and December 2024 using a nationwide claims database provided by Medical Data Vision Co., Ltd. (Tokyo, Japan), which includes data from approximately 230 acute care hospitals. All Oncotype DX-tested pts were included in the analysis of clinical and demographic characteristics. For treatment pattern analysis, the cohort was limited to those with at least 6 months of follow-up after testing. CT and endocrine therapy (ET) were classified as either neoadjuvant or adjuvant. Results: A total of 3,837 pts with BC who underwent ODX testing following reimbursement were identified. The median age was 57.0 [IQR: 49.0-67.0], including 17 male pts (0.4%), and the median BMI was 22.6 [IQR: 20.3-25.7].The majority of pts had T1 (55.1%) or T2 (40.4%) tumors and were node-negative(N0: 87.4%). Distant metastasis was rare (M1: 0.1%). Whereas 49.9%, 38.7% and 7.0% of pts had Stage IA, Stage IIA or Stage IIB disease, Stage III or IV disease was rare (<1%).Most pts (87.8%) were treated at designated cancer care hospitals. Over half received care at large hospitals with ≥500 beds (53.3%), and 45.8% were treated at mid-sized hospitals with 200-499 beds. Among pts with at least 6 months of follow-up after Oncotype DX testing, 23.2% received adjuvant CT. The most common regimen was ACT (anthracycline, cyclophosphamide, and taxane) (48.4%) followed by TC (taxane, cyclophosphamide) (43.4%). ET was administered to 9.1% of pts as neoadjuvant treatment and to 95.1% as adjuvant treatment. Among pts who received adjuvant ET, aromatase inhibitors were most frequently used (58.0%), followed by selective estrogen receptor modulators (SERMs) alone (21.8%), and a combination of luteinizing hormone-releasing hormone agonists and SERMs (15.9%). Additionally, 36.9% pts received radiation therapy. Conclusion: This study presents the first nationwide RWE on the clinical use of the Oncotype DX test in Japan. The findings indicate that the large majority of pts with BC who undergo Oncotype DX testing receive care at cancer-specialized or large medical centers. Cancer staging and treatment patterns suggest that the Oncotype DX test is being appropriately applied in pts with HR+/HER2- early BC, consistent with current guidelines for personalized treatment.
Presentation numberPS3-08-29
Retrospective study evaluating eligibility, treatment patterns, and clinicopathologic factors associated with adjuvant abemaciclib use in patients with high-risk estrogen receptor-positive (ER+) HER2-negative (HER2-) early breast cancer (EBC)
Chiara Corti, Dana-Farber Cancer Institute, Boston, MA
C. Corti1, N. Zhou2, A. R. Martin1, C. Stever1, T. Parker1, C. Snow1, G. Curigliano3, T. A. King4, E. A. Mittendorf4, N. U. Lin1, N. Tayob2, S. M. Tolaney1; 1Breast Oncology Division, Dana-Farber Cancer Institute, Boston, MA, 2Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, 3New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, ITALY, 4Department of Surgery, Brigham and Women’s Hospital, Boston, MA.
Background: 2 years of adjuvant (adj) abemaciclib (abema) and endocrine therapy (ET) is standard for high-risk ER+HER2- EBC. Although eligibility rates are known, real-world use remains unclear. We examined the gap between eligible and treated patients (pts) and their clinicopathologic differences. Methods: Pts with ER+HER2- EBC eligible for adj abema per monarchE cohort 1 criteria (≥4 positive nodes, or 1-3 nodes plus G3 or tumor ≥5 cm), who had surgery between the date of FDA approval (10/12/2021) and the data lock (04/15/2024), were retrospectively identified from a single institution database. Pts were classified as EAT (eligible and treated) if an abema start date was recorded, or ENT (eligible, not treated) if no abema initiation was documented. Clinicopathologic features were compared using chi-square or Wilcoxon tests (p<.05 significant). Results: 129 pts were identified; 78 (60.5%) received adj abema, 51 (39.5%) did not. Selected clinicopathologic characteristics are shown in the Table. No differences in race and ethnicity were found. ENT pts were older (median 65.5 vs 50.8 years; p<.001), more often pre-operatively node-negative (cN0, 78.4% vs 51.3%, p=.004) and had lower nodal burden postoperatively (pN1, 78.4% vs 44.9%, p<.001). Lobular histology was more frequent in EAT (26.9% vs 11.8%; p=.040). Clinical stage, pT category, tumor size ≥5 cm, multifocality, and ER status were similar. ET regimens varied (p<.001), with more EAT pts receiving AI+OFS (38.5% vs 11.8%). We explored the reasons why ENT pts did not receive abema. Among 51 ENT pts, 16 (31.4%) were not treated per provider clinical judgment. Of these, 12 (75.0%) had surgery before Ki67-based FDA indication language was removed, with factors including Ki67 unavailable (n=2), N1mi+Ki67 unavailable (n=5), low Oncotype+Ki67 unavailable (n=3), advanced age+low Oncotype+Ki67 unavailable (n=1), surgical ER-low+Ki67 unavailable (n=1). Four (25%) had surgery after the FDA removed Ki67-based indication language (surgical ER-low n=1, N1mi n=2, N1mi+age+comorbidities n=1). 13 (25.5%) pts were offered but declined abema, 4 (7.8%) were lost to follow-up, 3 (5.9%) started ribociclib. Other reasons: comorbidity+age (n=4, 7.8%), ≥3 comorbidities (n=1, 2%), severe comorbidity (n=2, 3.9% [Crohn’s disease and psychiatric disorder]), poor ET tolerance (n=2, 3.9%), ongoing olaparib (n=1, 2%), metastatic disease at post-surgical staging (n=1, 2%), pt hesitation despite abema prescription (n=2, 3.9%). The reason was unknown in 2 (3.9%) cases. Conclusions: In this cohort, a gap between EAT and ENT pts was seen. Older age and lower nodal burden were associated with not receiving adj abema. Pt choice and comorbidities also contributed to non-initiation.
| Characteristic |
Eligible, not treated (n = 51) |
Eligible, treated (n = 78) |
Total (n = 129) |
P value | |||
| Age, median (range) | 65.5 (30.3 – 82.7) | 50.8 (31.6 – 77.2) | 58.2 (30.3 – 82.7) | < 0.001 | |||
| gBRCA1-2 mutation present, n. (%) | 4 (7.8) | 4 (5.1) | 8 (6.2) | 0.712 | |||
| Clinical N category, n. (%) | 0.004 | ||||||
| cN0 | 40 (78.4) | 40 (51.3) | 80 (62.0) | ||||
| cN1 | 11 (21.6) | 36 (46.2) | 47 (36.4) | ||||
| Histology, n. (%) | 0.040 | ||||||
| Invasive ductal | 40 (78.4) | 41 (52.6) | 81 (62.8) | ||||
| Invasive lobular | 6 (11.8) | 21 (26.9) | 27 (20.9) | ||||
| Mixed | 4 (7.8) | 10 (12.8) | 14 (10.9) | ||||
| ER >= 10% | 51 (100.0) | 78 (100.0) | 129 (100.0) | ||||
| Upfront Surgery | 48 (94.1) | 78 (100.0) | 126 (97.7) | 0.053 | |||
| Pathological T category, n. (%) | 0.129 | ||||||
| pT1-2 | 37 (72.5) | 46 (59.0) | 83 (64.3) | ||||
| pT3-4 | 13 (25.5) | 31 (39.7) | 44 (34.1) | ||||
| Pathological N category, n. (%) | < 0.001 | ||||||
| pN1 | 40 (78.4) | 35 (44.9) | 75 (58.1) | ||||
| pN2 | 9 (17.6) | 30 (38.5) | 39 (30.2) | ||||
| pN3 | 2 (3.9) | 13 (16.7) | 15 (11.6) | ||||
| Adjuvant systemic treatment, n. (%) | 0.002 | ||||||
| Chemotherapy | 4 (7.8) | 0 (0.0) | 4 (3.1) | ||||
| ET | 22 (43.1) | 31 (39.7) | 53 (41.1) | ||||
| Both | 22 (43.1) | 47 (60.3) | 69 (53.5) | ||||
| Adjuvant olaparib, n. (%) | 1 (2.0) | 3 (3.8) | 4 (3.1) | 0.657 | |||
| Adjuvant ribociclib, n. (%) | 3 (4.3) | 0 (0.0) | 3 (2.3) | 0.120 |
Presentation numberPS3-08-30
Evaluation of Late Recurrence Risk in Premenopausal N0-N1 Breast Cancer Using the CTS5 Score
Ayumi Saito, National Cancer Center Hospital Japan, Tokyo, Japan
A. Saito1, T. Shimoi1, T. Murata2, M. Onshi1, M. Yoshida3, A. Ogawa2, A. Nakashoji2, H. Maeda2, C. Watase2, S. Takayama2, K. Yonemori1; 1Department of Medical Oncology, National Cancer Center Hospital Japan, Tokyo, JAPAN, 2Department of Breast Surgery, National Cancer Center Hospital Japan, Tokyo, JAPAN, 3Department of Diagnostic Pathology, National Cancer Center Hospital Japan, Tokyo, JAPAN.
Background: Estrogen receptor (ER)-positive, HER2-negative breast cancer has a prolonged risk of recurrence extending up to 20 years after surgery. Extended endocrine therapy can reduce late recurrence; however, optimal selection and treatment duration remain unclear. The Clinical Treatment Score post-5 years (CTS5) is a simple tool using clinicopathological factors to estimate late recurrence risk, and this score stratifies late recurrence risk into low (3.86) groups. Although validated primarily in postmenopausal patients, CTS5’s utility in premenopausal, N0-N1 patients has not been established. This study retrospectively assessed late recurrence risk by CTS5 and evaluated the impact of extended endocrine therapy in this lower-risk population.Methods: We analyzed ER-positive, HER2-negative, non-metastatic breast cancer patients with pathological N0-1 disease treated with curative surgery between 2003-2017, excluding those with early recurrence or insufficient follow-up. CTS5 scores were calculated and categorized into risk groups, and the duration of endocrine therapy was classified as standard (7 years) or extended (≥7 years). Distant recurrence-free survival was assessed using Kaplan-Meier and Cox regression analyses to identify prognostic factors.Results: A total of 879 patients were included in this study. Median age was 45 years; 75.9% had node-negative disease and 58.5% had pathologic stage I. Adjuvant chemotherapy was administered in 30.3%, and ovarian function suppression in 27.3%. SERM was used in 76.7% of patients. The mean CTS5 score was 2.73, with 70.3% low-risk, 22.4% intermediate, and 7.1% high risk. Extended endocrine therapy ≥7 years was given to 37.7% of patients.Over a median follow‐up of 8.9 years, 31 distant recurrences occurred. Eight-year distant recurrence-free survival was 99.1% (95% CI 98.2-99.9) in the low-risk group, 95.5% (92.3-98.9) in the intermediate, and 93.7% (87.1-100.0) in the high CTS5 groups (log-rank p<0.001). In multivariable analysis adjusting for age, adjuvant chemotherapy, ovarian suppression, and extended endocrine therapy, intermediate (HR 4.73, 95% CI 2.02—11.05, p<0.001) and high CTS5 groups (HR 5.44, 95% CI 1.29-22.92, p=0.021) were significantly associated with increased risk of distant recurrence. Extended endocrine therapy ≥7 years was associated with a trend toward reduced recurrence risk (HR 0.14, 95% CI 0.02-1.09, p=0.06).Conclusion: In this premenopausal N0-N1 cohort, CTS5 effectively stratified late recurrence risk despite the generally lower-risk population. While patients with intermediate or high CTS5 scores had significantly higher recurrence and are likely to benefit from extended endocrine therapy, patients in the low-risk group may avoid prolonged treatment, potentially sparing adverse effects and preserving quality of life.
$$MISSING OR BAD TABLE SPECIFICATION {ED6A501E-AA7F-4699-9BAB-DB67D3758F4B}$$
| HR | Lower CI | Upper CI | p-value | |
| Age | 0.95 | 0.89 | 1.01 | 0.13 |
| CTS5 groups Intermediate | 4.73 | 2.02 | 11.05 | <0.001 |
| CTS5 groups High | 5.44 | 1.29 | 22.92 | 0.021 |
| LHRH agonist use | 0.98 | 0.41 | 2.31 | 0.96 |
| Adjuvant chemotherapy | 0.60 | 0.22 | 1.60 | 0.31 |
| Extended endocrine therapy >=7 years | 0.14 | 0.02 | 1.09 | 0.061 |
Presentation numberPS3-09-01
Use of the OncotypeDX Recurrence Score® (RS) as a continuous prognostic biomarker in hormone receptor (HR) positive HER2neu negative early breast cancer with intermediate risk of recurrence may asses risk of recurrence or death more appropriately
Hans-Christian Kolberg, Marienhospital Bottrop, Bottrop, Germany
H. Kolberg1, S. Hildebrandt1, L. Akpolat-Basci1, A. Farag1, A. Maguz1, M. Stephanou1, C. Kolberg-Liedtke2; 1Dept OB/GYN, Marienhospital Bottrop, Bottrop, GERMANY, 2Dept of applied Health Sciences, Hochschule für Gesundheit, Bochum, GERMANY.
Background: The OncotypeDX Recurrence Score® (RS) is a validated multigene assay yielding prognostic and predictive information in patients with hormone receptor (HR) positive HER2neu negative early breast cancer and is part of guideline recommendations in national and international guidelines. The clinical utility of the RS has been prospectively investigated using risk levels of low risk (0-10), intermediate risk (11-25) and high risk (26-100). In patients older than 50 years the risk groups low and intermediate are indicating a low risk of recurrence (justifying omission of adjuvant chemotherapy), whereas an RS of 26-100 indicates high risk. In patients age 50 and younger the granularity is higher with risk levels of low (0-15), low to medium (16-20), intermediate (21-25) and high risk (26-100). However, a continuous interpretation of the score would be helpful in risk assessment and counseling given the fact that recurrence risk is a biological continuum. Here we are presenting an analysis of RS values as a continuous parameter from a real-world population. Methods: Patients included in this analysis had HR positive HER2neu negative early breast cancer with 0-3 involved lymph nodes assessed as intermediate risk by institutional guidelines including Ki67 and a complete follow-up. An RS was performed in all tumors with material from the core cut biopsy. Patients with a RS of 26 and higher received neoadjuvant chemotherapy. Disease free survival (DFS) events were defined as local or distant recurrence or death from any cause. Overall survival (OS) events were defined as death from any cause. Associations of Recurrence Score® and DFS or OS events were calculated by Cox Regression. Results: 79 female patients were included in this analysis. Median follow-up was 6.9 (0.9-11.0) years. Patient characteristics were as follows: median tumor size 1.4 (0.3-4.5) cm, median age at diagnosis 60.7 (34.7-82.5) years, median Ki67 20 (2-35) %, median RS 16 (0-46). Sixty-two patients (78.5%) were postmenopausal. 2 (2.5%), 74 (93.7%) and 3 (3.8%) patients had grade 1, 2 and 3 disease, respectively. 79 (100%) patients had ER positive, 73 (92.4%) patients had PR positive tumors. 20 patients (25.3%) had node positive (1-3 positive lymph nodes) disease. 9 (11.4%) patients received anthracycline- and taxane-containing neoadjuvant chemotherapy. During follow-up 5 (6.3%) patients experienced a locoregional recurrence and 8 (10.1%) patients a distant recurrence. 5 (6.3%) patients died, in 4 cases (5.1%) from breast cancer. The risk for a DFS event increased by 8.1% by every one-unit (integer) in RS (p=0.022). For OS events, every one-unit increase in RS increased the risk of death by 9.3% (p=0.027). Conclusion: In our population of patients with intermediate risk by traditional risk parameters including Ki67 survival was excellent with 93.7% of patients alive after a median follow-up of 6.9 years. However, within this selected risk group the continuous assessment of the RS performed using material from core cut biopsies indicated an increased risk for a DFS or OS event by every one-unit increase in RS. Our analysis is hypothesis-generating and justifies an approach in a multicenter registry population with greater statistical power allowing multivariate analysis and the assessment of confounding parameters. If our results can be reproduced in a large-scale analysis they could help calculating the risk of an individual patient with more granularity than by use of risk groups alone.
Presentation numberPS3-09-02
Retrospective observational study of premenopausal ER+/HER2- patients in Greece with axillary tumor burden but low Oncotype DX® Recurrence Score who did not receive chemotherapy
Lazaros Papadopoulos, IASO, General Maternity and Gynecology Clinic, Athens, Greece
L. Papadopoulos1, C. Markopoulos2, K. Papazisis3, F. Zagouri4, S. Triantafyllidou5, A. Tsiftsoglou6, A. Zavos7, A. Koumarianou8, M. Kontos9, A. Fokianou1, P. Ntasiou1, P. Kalogerakos1, S. D. Eleftheriadis1, F. Fostira1, C. Iosifidou1, C. Stefanou1, D. Maniatis1, S. Filippidou1, R. Alevizou1, A. Papadopoulou10, G. Kapetsis11, S. Giannoulakis11, D. Grigoriadis11, G. Xepapadakis1; 12nd Breast Unit, IASO, General Maternity and Gynecology Clinic, Athens, GREECE, 2Medical School, National and Kapodistrian University of Athens, Athens, GREECE, 33rd Dpt of Oncology, Interbalcan European Medical Centre, Thessaloniki, GREECE, 4Department of Clinical Therapeutics,, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, GREECE, 5Breast Clinic, Genesis Hospital, Thessaloniki, GREECE, 6Department of Surgery, St. Luke’s Hospital, Thessaloniki, GREECE, 7Breast Unit, University of Thessaly, Larissa, GREECE, 84th Department of Internal Medicine, Attikon University Hospital, Athens, GREECE, 91st Department of Surgery, National and Kapodistrian University of Athens, Athens, GREECE, 10Hematology Department, General University Hospital of Heraklion, Heraklion, GREECE, 11Medical Dpt, Genekor Medical S.A., Gerakas, GREECE.
Background The Oncotype DX® assay is a validated genomic test that aids in assessing the risk of distant recurrence and in predicting the potential benefit of adjuvant chemotherapy in patients with early-stage HR+, HER2- breast cancer. In premenopausal women with 1-3 positive lymph nodes (N1) and a Recurrence Score (RS) of 0-25, the RxPONDER trial demonstrated a 2.4% improvement in distant recurrence-free interval (DRFI) and a 4.9% improvement in invasive disease-free survival (IDFS) in pts receiving chemo-endocrine therapy compared to endocrine therapy alone. Pts who received only endocrine therapy had a DRFI of 93,9% and an IDFS of 89%. The aim of this observational study is to establish a registry and present data on DRFI and IDFS in premenopausal women with node-positive breast cancer and a low RS who, either by choice or due to comorbidities, did not receive chemotherapy and were treated with endocrine therapy alone (with/without ovarian function suppression – OFS).Methods We conducted a retrospective multicenter analysis across 9 institutions in Greece, focusing on pre- & peri-menopausal women diagnosed with ER+/HER2-, node-positive (pN1 or pN1mic) early breast cancer, who underwent Oncotype DX® testing between 01/2012-01/2025, had a low RS (0-25), and did not receive chemotherapy. Three additional pts with N0(i+) disease were not included in the cohort analysis. Results A total of 74 pts were included in the analysis. The median age of diagnosis was 47 (range 26-54) and the median RS was 12 (range 0-22). Cohort clinical data are summarized below.
| Category | Subcategory | Τotal cohort, n (%) | RxPONDER Premenopausal cohort, % | ||||
| T Stage | T1 | 60 (81.1%) | 56.3% | ||||
| T2 | 13 (17.5%) | 37.3% | |||||
| T3 | 1 (1.4%) | 6.4% | |||||
| N Stage | N1mic | 33 (44.6%) | Not reported in final analysis | ||||
| N1 (1 positive lymph node) | 34 (45.9%) | 65.3% | |||||
| N1 (2 positive lymph nodes) | 5 (6.8%) | 25.7% | |||||
| N1 (3 positive lymph nodes) | 2 (2.7%) | 9% | |||||
| Grade (G) | 1 | 17 (23.0%) | 21.7% | ||||
| 2 | 50 (67.6%) | 67.6% | |||||
| 3 | 7 (9.4%) | 9.6% | |||||
| Endocrine therapy* | + OFS | 39 (52.7%) | 19% (endocrine arm) | ||||
| – OFS | 35 (47.3%) | 81% (endocrine arm) | |||||
| Age | <50 | 53 (71.6%) | 69.3% | ||||
| ≥50 | 21 (28.4%) | 30.7% | |||||
| Radiotherapy (RT) | Yes | 59 (79.7%) | Not reported | ||||
| No | 15 (20.3%) | Not reported |
*1 patient did not receive endocrine therapy. With a median follow-up of 4.8 years (range: 1-150 months), one patient developed a distant recurrence in the lung (T1, N1mic, G3, no OFS, RS 17), while another was diagnosed with contralateral breast cancer (T1, N1, G2, with OFS, RS 15). The estimated 5-year DRFI and IDFS were 98.6% (95% CI, 92.5%-99.9%) and 97.3% (95% CI, 90.7%-99.5%), respectively. Among the three N0(i+)pts, no recurrences were observed during a mean follow-up of 3.4 years. Conclusion While the RxPONDER trial demonstrated a modest benefit from the addition of chemotherapy in premenopausal node positive pts independently of the RS, these real-world data suggest that selected pts may achieve favorable outcomes with endocrine therapy alone. A substantial proportion of pts received OFS (52.7%), with nearly half having N1mic disease (44.6%) and most presenting with T1 tumors (81.1%), suggesting a biologically distinct subgroup with potentially different treatment needs. This highlights the potential of individualized treatment planning using Oncotype DX in pre-/peri-menopausal pts with limited axillary tumor burden. Further studies with larger patient cohorts and longer follow-up are warranted to better define outcomes and optimize treatment strategies in this subgroup.
Presentation numberPS3-09-03
The Societal Cost Impact of Oncotype-DX<sup>®</sup> Testing in an Irish Healthcare Setting
Iseult M Browne, St Vincent’s University Hospital, Dublin, Ireland
I. M. Browne1, R. A. McLaughlin2, C. S. Weadick3, S. O’Sullivan4, D. K. Hadi5, S. J. Millen6, M. J. Higgins1, J. P. Crown1, C. M. Quinn7, R. S. Prichard8, D. P. McCartan8, H. K. Carroll1, K. E. Ronan1, A. D. Hill9, R. M. Connolly3, S. A. Noonan3, D. O’Mahony10, C. O’Hanlon-Brown5, B. T. Hennessy2, C. M. Kelly4, S. O’Reilly3, P. G. Morris2, J. M. Walshe1; 1Medical Oncology, St Vincent’s University Hospital, Dublin, IRELAND, 2Medical Oncology, Beaumont Hospital, Dublin, IRELAND, 3Medical Oncology, Cork University Hospital/University College Cork Cancer Centre, Cork, IRELAND, 4Medical Oncology, Mater Misericordiae University Hospital, Dublin, IRELAND, 5Medical Oncology, St James’s Hospital, Dublin, IRELAND, 6Exact Sciences, Exact Sciences UK Ltd, London, UNITED KINGDOM, 7Pathology, St Vincent’s University Hospital, Dublin, IRELAND, 8Surgery, St Vincent’s University Hospital, Dublin, IRELAND, 9Surgery, Royal College of Surgeons, Dublin, IRELAND, 10Medical Oncology, Bon Secours Hospital, Cork, IRELAND.
Title: The Societal Cost Impact of Oncotype-DX® Testing in an Irish Healthcare Setting Background The use of genomic testing in patients with node-negative (N0) and 1-3 lymph nodes positive (N1) early-stage breast cancer has had a significant impact on the delivery of chemotherapy in this setting. Our previous studies have identified a reduction in the use of chemotherapy of over 50% in N0 and N1 patients. This has substantial direct economic benefits for the third-party payer, and for the patient in the avoidance of chemotherapy. An area which is often overlooked is the considerable societal costs that are associated with chemotherapy, and there is a paucity of studies that have included an evaluation of these costs for early-stage breast cancer. These costs include lost productivity due to work absence, caregiver productivity losses, and out-of-pocket expenses. The objective of this study was to examine the societal implications of Oncotype DX® testing in N1 patients at a national level in 5 Irish cancer centres over an 11-year period. Methods A retrospective observational study was undertaken including patients with early-stage ER-positive breast cancer and 1-3 positive lymph nodes who underwent Oncotype DX® testing between March 2011 to October 2022 across five of Ireland’s cancer centres. A survey of Irish Breast Oncologists provided the assumption that all patients in our study with N1 disease who underwent Oncotype DX® testing would be recommended adjuvant chemotherapy without testing. Data was collected via electronic records and clinical chart review. The estimated societal costs of chemotherapy were based on UK cost estimates from a report published by the University of East Anglia which used an incidence-based-cost-of-illness model to estimate annual societal costs associated with chemotherapy for early-stage breast cancer. Sterling estimates were converted to euro costs based on the exchange rate of 1 GBP = 1.17025 EUR. Information regarding costing was provided by the National Healthcare Pricing Regulatory Authority and the economic analysis was adjusted for changing costs over the study time-period. Results We identified 828 patients for inclusion in this analysis of societal cost savings associated with the use of Oncotype DX® testing. The mean age was 58 years (range 22-81). 171 patients (21%) were <50 years at diagnosis, and 657 (79%) were ≥50 years. With the use of Oncotype DX® testing 58% of patients avoided chemotherapy. As previously published, this resulted in savings of over €6 million in treatment costs, leading to net savings of over €3.3 million euro when the assay cost was deducted. For this societal cost impact analysis, we estimated potential societal costs per patient with early-stage breast cancer undergoing chemotherapy to be €43,628. This figure includes €38,450 and €4,008 due to lost productivity from long-term and short-term work absence respectively, and €1,170 from lost productivity for caregivers. Per patient out-of-pocket expenses totalled €1,287. The total estimated impact of Oncotype DX® testing among the study cohort on societal costs of chemotherapy treatment was €22,053,500. Conclusion Societal costs are often overlooked as they are not as easily calculated as direct costs, however, it is evident that chemotherapy use carries considerable indirect costs for society. These wider costs alongside a comprehensive consideration of benefits and harms should be incorporated when deciding on a patient’s treatment paradigm. The use of Oncotype DX® testing in N1 patients has led to an estimated societal cost saving of over 22 million euro in across 5 of Ireland’s oncology centres for the period of our study.
Presentation numberPS3-09-04
The impact of axillary lymph node dissection on the indication of adjuvant abemaciclib in high-risk hormone receptor-positive breast cancer: Real-world analysis from a multicenter cohort in Brazil
Renata C Bonadio, Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
L. Holland1, L. Testa1, C. Cavalcanti1, J. F. Bessa1, K. Cayres1, P. H. Amor Divino1, R. Naves1, J. Bines2, R. C. Bonadio1; 1Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 2Oncology, Instituto D’Or de Pesquisa e Ensino, N/A, BRAZIL.
Background: Adjuvant CDK4/6 inhibitor abemaciclib combined with endocrine therapy has demonstrated significant improvement in invasive disease-free survival among patients with high-risk, hormone receptor-positive, HER2-negative (HR+HER2-) early-stage breast cancer. Lymph node involvement is a key determinant of recurrence risk and plays a critical role in therapeutic decision-making. However, the optimal surgical management of axillary lymph nodes remains unclear when the extent of nodal involvement may influence adjuvant therapy decisions. This study aimed to investigate the proportion of real-world cases in which axillary dissection was essential for determining eligibility for adjuvant abemaciclib. Methods: We conducted a multicenter retrospective study of patients diagnosed with early-stage breast cancer between 2018 and 2025 within a national Brazilian cancer network. We evaluated the proportion of patients eligible for adjuvant abemaciclib based on phase 3 monarchE trial criteria. Indications were stratified into three sequentially applied groups (patients meeting a prior criterion were not reassigned to subsequent ones):1.Positive axillary lymph node(s) nodes and either grade 3 disease or tumor size ≥5 cm;2.Positive axillary lymph node(s) and Ki-67 ≥20%;3.Four or more positive axillary lymph nodes.Patients meeting only criterion 3 were considered reliant on axillary dissection for abemaciclib indication. Results: Among 656 patients with HR+/HER2- breast cancer, 284 (43.3%) met monarchE criteria for adjuvant abemaciclib. Median age was 49 years (range 24-83), with 52.8% being premenopausal. Ductal and lobular histologies were present in 72.2% and 16.5%, respectively. Tumor grade was 7.0% grade 1, 53.2% grade 2, and 34.5% grade 3. Clinical T stages were: 22.2% T1, 37.7% T2, 23.6% T3, and 6.7% T4. Clinical N stage was N0 in 23.2%, N1 in 49.3%, and N2-3 in 15.5%. Most patients (80.6%) had a Ki67-index ≥20%. Neoadjuvant chemotherapy was used in 46.8% of cases, and 51.1% received adjuvant chemotherapy. Axillary surgery comprised sentinel lymph node biopsy (51.4%) and axillary dissection (44.0%), with data missing in 4.6%. The distribution of criteria leading to abemaciclib indication is shown in Table 1. Discussion: This real-world study demonstrates that only a small subset of patients eligible for adjuvant abemaciclib met the criteria based solely on having four or more positive axillary lymph nodes. These findings support the notion that axillary dissection should not be pursued solely to establish eligibility for adjuvant CDK4/6 inhibitor therapy.
| Adjuvant abemaciclib criteria | N | % | |||
| 1.Positive axillary lymph node(s) and either grade 3 disease or tumour size of 5 cm or larger | 151 | 53.2% | |||
| 2.Positive axillary lymph node(s) and Ki-67 of at least 20% | 117 | 41.2% | |||
| 3.Four or more positive axillary lymph nodes | 16 | 5.6% |
Presentation numberPS3-09-05
Real-world clinical characteristics and treatment patterns among UK and French patients with a recent diagnosis (2020 onwards) of early-stage, estrogen receptor-positive, human epidermal growth factor receptor 2-negative breast cancer (ER+/HER2-eBC)
Paul Cottu, Institut Curie AND Université Paris Cité, Paris, France
P. Cottu1, E. Sawyer2, J. Frenel3, J. R. Earla4, K. M. Hirshfield5, G. Gooud6, D. Ntais6, A. Jamotte7, C. Perkins8, A. Babonneau9, E. Kitetere10, M. Vallet11, M. Riaz12, J. Timbres13, L. Haroun14, L. Cirneanu15, R. Hermans15, V. Saglimbene16, K. Dushkin15, A. Ajmal15, S. Oikonomou17, A. Vladimirova17, K. Desai4, F. Bocquet18, P. S. Hall11; 1Medical Oncology, Institut Curie AND Université Paris Cité, Paris, FRANCE, 2Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Kings College London, Guys Cancer Centre, London, UNITED KINGDOM, 3Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-herblain, FRANCE, 4V&I Outcomes Research Oncology, Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, 5Oncology Clinical Development, Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, 6HTA & Outcomes Research, MSD UK, London, UNITED KINGDOM, 7HEDS Oncology, MSD Innovation & Development GmbH, Zürich, SWITZERLAND, 8Medical Oncology, MSD France, Puteaux, FRANCE, 9HEOR, MSD France, Puteaux, FRANCE, 10Oncology Medical Affairs, MSD UK, London, UNITED KINGDOM, 11Genetics and Cancer, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UNITED KINGDOM, 12Data Department, Leeds Teaching Hospitals NHS Trust, Leeds, UNITED KINGDOM, 13Breast Cancer Genetics, King’s College London, London, UNITED KINGDOM, 14Data Department, Institut Curie, Paris, FRANCE, 15Real World Solutions, IQVIA, London, UNITED KINGDOM, 16Real World Solutions, IQVIA, Milan, ITALY, 17Real World Solutions, IQVIA, Sofia, BULGARIA, 18Data Factory and Analytics Department, Institut de Cancérologie de l’Ouest, Nantes, FRANCE.
Background: ER+/HER2-eBC is heterogeneous, with a subset of patients at high risk of recurrence and poorer outcomes. The treatment landscape for the high-risk group has shifted recently, with the addition of targeted therapies to clinical guidelines. This study aimed to describe the clinical characteristics and treatment patterns for recently diagnosed high-risk patients. Methods: This was an observational, retrospective cohort study using electronic health records from five oncology centers across France and the UK. The study included patients with recently diagnosed (2020-2024), high-risk (Grade III and T1c-T2/N1-N2 or T3-T4/N0-N2), ER+/HER2- eBC, with a minimum follow up of 12 months. Initial data on baseline characteristics and real-world treatment patterns (surgery, chemotherapy [CT], radiotherapy [RT] and other treatments) from first diagnosis to the time of appearance of metastasis are presented. Results: The study included 421 patients (68% between 35 and 65 years old); 239 (57%) from France and 182 (43%) from the UK. Of the 267 patients with known status, 164 (61%) were post-menopausal women. Patients were diagnosed with T1c-T2 and N1-N2 (309, 73%) or T3-T4 and N0-N2 (112, 27%) BC (including American Joint Committee on Cancer, version 7 stages IB, II, IIIA, IIIB). At baseline, 357 (85%) were ER-positive (≥10% expression). The majority were lymph node positive (369, 88%): 307 (73%) had 1-2 positive lymph nodes and 62 (15%) had ≥3. BRCA testing at diagnosis was performed in 104 (25%) patients with 17 (16%) positive. As expected, prognostic genomic testing was rarely conducted in this population. Of 277 patients with ECOG available at the start of treatment, 223 (81%) had a score of 0. Almost all patients (413, 98%) underwent eBC surgery and, among these, lumpectomies alone (191, 45%) and mastectomies alone (155, 37%) were the most common surgeries. Of the 421 patients included, many received adjuvant CT (204, 48%) and RT (303, 72%) post-surgery. Neoadjuvant CT was received by 178 patients (42%) and RT by 5 patients (1%). Based on initial findings, adjuvant use of abemaciclib increased from 3/66 patients (5%) diagnosed in 2020 to 51/108 patients (47%) in 2023 (Table). Conclusions: These results showcase recent real world clinical practice in France and the UK for patients with high-risk ER+/HER2- eBC. Most patients received surgery with (neo)adjuvant CT and other treatments; specifically, there was a substantial increase in the use of adjuvant abemaciclib during the study period. Detailed insights on specific treatment agents, regimen sequences received, and breakdowns by country will be provided in the symposium publication.
Presentation numberPS3-09-06
Comparative Predictive Utility of Oncotype DX and MammaPrint Using a Clinicopathologic Model in Matched HR+/HER2− Early Breast Cancer Cohorts: The BRAIN Study
Joo Heung Kim, Yonsei University College of Medicine, Yongin, Korea, Republic of
J. Kim1, S. Lee2, S. Lee3, I. Lee3, J. Ahn4, S. Park4, S. Gwon5, N. Son5; 1Surgery, Yonsei University College of Medicine, Yongin, KOREA, REPUBLIC OF, 2Catholic Kwandong University College of Medicine, catho, Incheon, KOREA, REPUBLIC OF, 3surgery, Catholic Kwandong University College of Medicine, Incheon, KOREA, REPUBLIC OF, 4Surgery, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 5Statistics, Keimyung University, Daegu, KOREA, REPUBLIC OF.
Oncotype DX (ODX) and MammaPrint (MMP) are two widely adopted multigene assays used to guide adjuvant chemotherapy decisions in patients with hormone receptor-positive (HR+), HER2-negative early breast cancer. Despite their similar clinical roles, ODX and MMP often produce discordant risk classifications. While head-to-head comparison in the same patient would be ideal, it is rarely feasible in practice due to cost, clinical utility, and reimbursement constraints. Therefore, we designed a pilot study to indirectly compare the predictive performance of ODX and MMP by applying a clinicopathologic model—the Tennessee nomogram—to matched cohorts of patients who received either test. A total of 2,542 patients who underwent ODX (n = 2,011) or MMP (n = 531) testing were retrospectively selected. Patients were matched 1:1 based on age and nodal status to control for key prognostic variables. Risk distribution was compared across both cohorts, followed by application of the Tennessee model to assess its predictive concordance with each assay’s reported risk classification. Model performance was evaluated using AUC, specificity, positive predictive value (PPV), and negative predictive value (NPV). The proportion of patients classified as high-risk differed between the two cohorts (ODX: 12.22% (8.21 – 17.28), MMP: 33.94% (27.72-40.59). The Tennessee model showed an AUC of 85.57 (77.49-93.65) for predicting high-risk ODX results and 70.70 (63.32-78.08) for MMP. Specificity and NPV were 90.21%, 94.59% in the ODX cohort, compared to 89.04% and 73.45% in the MMP cohort. Our findings indicate that the Tennessee model aligns more closely with ODX, suggesting that each assay may capture distinct biological dimensions of tumor behavior. This study highlights the importance of understanding the limitations of multigene tests when used interchangeably and supports the development of cost-effective, data-driven prediction tools that integrate clinicopathologic variables. These pilot results lay the groundwork for future AI-based models capable of refining treatment stratification in early HR+/HER2− breast cancer.
Presentation numberPS3-09-07
Adherence to adjuvant endocrine treatment in women 36-56yrs with a hormone receptor-positive breast cancer: Patient compliance and impact on prognosis
Maxime Van Houdt, University Hospital Leuven, Leuven, Belgium
M. Van Houdt1, L. De Nys1, A. Laenen2, S. Han1, I. Huys3, H. Wildiers4, F. Derouane4, G. Floris5, M. Keupers5, C. Van Ongeval5, Y. Van Herck4, A. Smeets6, I. Nevelsteen6, C. Weltens7, H. Janssen7, J. Verhoeven7, A. Baten7, V. Celis5, R. Prevos5, C. Remmerie8, H. De Boodt5, A. Coessens5, P. Neven1; 1Department of Gynaecological Oncology, University Hospital Leuven, Leuven, BELGIUM, 2Leuven Biostatistics and Statistical Bioinformatics Centre, KU Leuven, Leuven, BELGIUM, 3Department of Clinical Pharmacology and Pharmacotherapy, KU Leuven, Leuven, BELGIUM, 4Department of General Oncology, University Hospital Leuven, Leuven, BELGIUM, 5Department of Imaging & Pathology, University Hospital Leuven, Leuven, BELGIUM, 6Department of Surgical Oncology, University Hospital Leuven, Leuven, BELGIUM, 7Department of Radiotherapy, University Hospital Leuven, Leuven, BELGIUM, 8Department of Oncology, University Hospital Leuven, Leuven, BELGIUM.
BackgroundAdjuvant endocrine therapy (AET) is essential in treating early hormone receptor-positive (HR+) breast cancer, significantly reducing recurrence and mortality. However, side effects often lead to poor adherence, with discontinuation rates up to 50% within five years. This study assessed adherence rate (AR) and the impact of AET compliance on prognosis in women aged 36-56 treated at UZ Leuven. Methods This retrospective cross-sectional study analyzed AET adherence in all high-risk early HR+ breast cancer patients diagnosed at UZ Leuven between 01/01/2008 and 31/12/2018 and who received chemotherapy. Data were obtained from clinical records and pharmacy dispensing information from public and hospital pharmacies, provided by Belgian health insurance. AR was measured by medication possession ratio (MPR), defined as the number of pills dispensed per year divided by 365.25 days. Non-adherence and poor adherence were defined as MPR <80% and <95%. Covariate analyses evaluated clinical factors potentially influencing adherence, including age (<45 vs ≥45 years), tumor grade (1-3), pathological nodal status (pN0 vs pN1), and BMI (30). Logistic regression with generalized estimating equations estimated non-adherence over time. The relationship between adherence and overall survival (OS) and recurrence-free survival (RFS) was assessed using Cox proportional hazards models, linking yearly AR to survival outcomes starting after each treatment year.Results Data from 649 patients (mean age 48) were included (Table 1). Over five years, an absolute decrease of 12% in mean AR was observed. The proportion of patients maintaining an AR > 95% dropped from 90% (628) in year 1 to 44% (589) in year 5. Non- adherence (AR 30) had lower adherence compared to those with BMI 25-30. Poor adherence (<95%) and non-adherence (<80%) were not significantly predictive of RFS and OS in this cohort.Conclusion Our study showed a gradual decline in adherence over five years, consistent with global data showing 40-60% of patients patients experience poor long-term adherence. In Belgium’s universal healthcare system, where cost barriers are low, factors like side effects, treatment fatigue, and psychosocial issues likely drive non-adherence. While no significant link to survival was found, adherence remains clinically important, as breast cancer recurrence risk persists beyond five years. Longer follow-up is needed to fully assess the impact. Table 1: characteristics of the study population collected from the medical records of UZ Leuven.
Presentation numberPS3-09-09
Validation and Enhancement of the Tennessee Nomogram Incorporating Ki-67 for Predicting Oncotype DX Risk in Hormone Receptor-Positive, HER2-Negative Breast Cancer in a Korean Cohort
Suk Jun Lee, Catholic Kwandong University College of Medicine, Seo-gu, Incheon, Korea, Republic of
S. Lee1, H. Lee2, S. Lee1, J. Ahn3, J. Kim4, I. Lee1, S. Park5, N. Son6; 1Department of Surgery, Catholic Kwandong University College of Medicine, Seo-gu, Incheon, KOREA, REPUBLIC OF, 2Department of Statistics, Keimyung University,, Dalseo-gu, Daegu, KOREA, REPUBLIC OF, 3Department of Surgery, Yonsei University College of Medicine, Seodaemun-gu, Seoul, KOREA, REPUBLIC OF, 4Department of Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, Gyeonggi-do, KOREA, REPUBLIC OF, 5Department of Surgery, Yonsei University College of Medicine, Seoul, Seodaemun-gu, Seoul, KOREA, REPUBLIC OF, 6Department of Statistics, Keimyung University, Dalseo-gu, Daegu, KOREA, REPUBLIC OF.
Validation and Enhancement of the Tennessee Nomogram Incorporating Ki-67 for Predicting Oncotype DX Risk in Hormone Receptor-Positive, HER2-Negative Breast Cancer in a Korean CohortPurpose: Oncotype DX (ODX) is widely used for predicting recurrence risk and guiding adjuvant therapy in hormone receptor-positive (HR+), HER2-negative breast cancer. However, access limitations have prompted the development of alternative tools, such as the Tennessee nomogram. Previous studies in a Korean population reported moderate accuracy but highlighted discrepancies, particularly among tumors with aggressive features. This study aimed to validate the predictive accuracy of the Tennessee nomogram in a Korean cohort and evaluate whether integrating Ki-67 could enhance its performance.Methods: We retrospectively analyzed data from 1,324 patients with invasive ductal or lobular carcinoma, HR-positive, HER2-negative, and node-negative breast cancer who underwent ODX testing. Based on an analysis of discordant cases from the original Tennessee model, we developed a new nomogram incorporating Ki-67 as an additional variable. We then compared the performance of this revised nomogram with the original Tennessee model using sensitivity, specificity, accuracy, PPV, NPV, and area under the curve (AUC). Additional analyses were performed by histologic grade.Results: The original Tennessee nomogram demonstrated an accuracy of 85.8%, with sensitivity of 12.8%, specificity of 99.1%, and an AUC of 0.784. Discordant predictions were often associated with aggressive tumor features such as high histologic grade, PR negativity, and elevated Ki-67, especially among false negatives whose ODX scores clustered between 25 and 30. Incorporating Ki-67 into the nomogram improved sensitivity to 29.2% and slightly increased overall accuracy to 86.7%, although specificity declined modestly to 97.3%. The number of false negatives decreased from 197 to 160, indicating better identification of high-risk patients. However, false positives rose from 11 to 34, reflecting a trade-off between higher sensitivity and somewhat reduced specificity. Despite this, the revised model achieved a higher AUC of 0.812, suggesting improved overall discrimination and better clinical utility when ODX testing is not readily available.In subgroup analyses by histologic grade, the revised model showed notable gains. For grade 2 tumors, sensitivity increased from 4.2% to 15.3% while maintaining high specificity (>99%), leading to higher overall accuracy and an improved AUC from 0.675 to 0.739. In grade 3 tumors, sensitivity nearly doubled from 35.9% to 68.8%; however, this was accompanied by a significant drop in specificity (87.5% to 48.2%) and only a modest improvement in AUC.Conclusions: The Tennessee nomogram remains a useful tool when ODX testing is unavailable, but its sensitivity is limited, particularly in tumors with aggressive features. Incorporating Ki-67 improves predictive performance overall and offers benefits in grade 2 tumors, where sensitivity improves without substantial loss of specificity. These results underscore the clinical value of Ki-67 as a biomarker in intermediate-grade breast cancer and support its inclusion in future predictive nomograms.
Figure 1. AUC curve with Tennessee amd Ki67 added model
Figure 2. Comparison of Tennessee and Ki-67-Model by Histologic Grade![[{037a8f2d-45b6-44fc-9e35-1176f1968aa4}saf805.html_g�0003.gif]]({037a8f2d-45b6-44fc-9e35-1176f1968aa4}saf805.html_g3.gif)
Presentation numberPS3-09-10
Higher Breast Tumor Grades Could More Likely Benefit from Extended Adjuvant Endocrine Therapy
Yetunde Ogunsesan, Medical College of Wisconsin, Milwaukee, WI
Y. Ogunsesan1, M. Lin2, R. Dutta3, R. Brazauskas3, A. Szabo3, L. N. Chaudhary1, Y. C. Cheng1, S. Kamaraju1; 1Division of Hematology/Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 2Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 3Department of Biostatistics, Medical College of Wisconsin, Milwaukee, WI.
Higher Breast Tumor Grades Could More Likely Benefit from Extended Adjuvant Endocrine Therapy Background: The Breast Cancer Index (BCI) (Biotheranostics, Inc. A Hologic Company.) is a gene-expression based biomarker utilized in both node negative and node positive, early-stage HR-positive breast cancer patients who may benefit from extended adjuvant endocrine therapy (AET) based on the tests’ comprehensive predictive and prognostic evidence. However, the association between BCI and clinicopathological factors remains unclear. We aimed to identify associations between predictive BCI scores of HR-positive breast cancer patients with various clinicopathological factors. Methods: We conducted a single center retrospective study using the BCI to evaluate differences in clinicopathologic factors among HR-positive breast cancer patients. Variables of interest included age, tumor grade, race, node status, number of nodes involved, HER-2 status, and ER/PR staining intensity. BCI scores to predict recurrence were classified as “Yes” (likely to benefit from extended AET) or “No.” Fisher’s exact test was used to compare categorical variables between patients with positive and negative BCI test result. The Wilcoxon rank sum test was applied to compare ordinal variables. A p-value of <0.05 was statistically significant. All statistical analyses were performed with R software. Results: 52 patients with HR-positive breast cancer had BCI testing from 2014-2025 at our institution. Most patients were >55 years old, identified as White, both ER and PR positive (compared with ER or PR positive only), and HER2 negative, and had moderately high to high intensity ER/PR staining. Most patients were node negative (71%). 25.5% of patients had grade 3 tumor. Of this study cohort, 25 (48%) had a predictive BCI score. Patients with higher tumor grades had more predictive BCI scores (p=0.0293). Age, intensity of ER/PR staining, and number of nodes involved had no significant effect on the BCI score (Table 1,1Wilcoxon Rank Sum test, 2Fisher’s Exact test). Conclusion: Higher tumor grades were more likely to have a predictive BCI score compared with lower tumor grades, indicating higher recurrence risk and/or more benefit from extended AET. Nodal status was not a statistically significant factor. Larger, multicenter studies with varied patient populations are needed to further validate these findings. Table 1; Clinicopathologic Features by BCI Result BCI Result Predictive
| Variable | Total
N=52(%)
|
No
N=27(%)
|
Yes
N=25(%)
|
p-value |
| BCI Score | 5.8 (1.1-19.9) | 3.9 (1.1-14) | 8.6 (1.4-19.9) | 0.03522 |
| Age Group | 0.0822 | |||
| <55 years | 17 (33.3%) | 12 (46.2%) | 5 (20%) | |
| >55 years | 34 (66.7%) | 14 (53.8%) | 20 (80%) | |
| Race | 12 | |||
| White | 51 (98.1%) | 26 (96.3%) | 25 (100%) | |
| Black | 1 (1.9%) | 1 (3.7%) | 0 (0%) | |
| ER/PR status | 0.1672 | |||
| ER only | 10 (19.2%) | 3 (11.1%) | 7 (28%) | |
| PR only | 0 (0%) | 0 (0%) | 0 (0%) | |
| both ER and PR | 42 (80.8%) | 24 (88.9%) | 18 (72%) | |
| HER2 status | 0.3412 | |||
| HER2 -ve | 48 (92.3%) | 26 (96.3%) | 22 (88%) | |
| HER2 +ve | 4 (7.7%) | 1 (3.7%) | 3 (12%) | |
| Node status | 0.2272 | |||
| Node +ve | 15 (28.8%) | 10 (37%) | 5 (20%) | |
| Node _ve | 37 (71.2%) | 17 (63%) | 20 (80%) | |
| No of nodes | ||||
| <3 | 14 (100%) | 9 (100%) | 5 (100%) | |
| >3 | 0 (0%) | 0 (0%) | 0 (0%) | |
| Tumor grade | 0.02931 | |||
| 1 | 15 (29.4%) | 9 (33.3%) | 6 (25%) | |
| 2 | 23 (45.1%) | 16 (59.3%) | 7 (29.2%) | |
| 3 | 13 (25.5%) | 2 (7.4%) | 11 (45.8%) |
Presentation numberPS3-09-11
Acceptability of adjuvant chemotherapy in women aged 70 and older after surgery for ER-positive HER2-negative breast cancer selected with a high genomic grade index: results from the ASTER 70s/Unicancer trial
Etienne Brain, Institut Curie, Saint-Cloud, France
E. Brain1, J. Henriques2, F. Rollot3, O. Mir4, E. Bourbouloux5, O. Rigal6, J. M. Ferrero7, S. Kirscher8, D. Allouache9, V. d’Hondt10, A. M. Savoye11, X. Durando12, F. P. Duhoux13, L. Venat Bouvet14, E. Blot15, J. L. Canon16, H. Bonnefoi17, T. Roque18, J. Lemonnier18, A. Latouche19, M. Lacroix-Triki20, D. Vernerey2; 1Medical Oncology, IHU des Cancers des Femmes (ANR-23-IAHU-0006), Institut Curie, Saint-Cloud, FRANCE, 2Biostatistics, Besançon University Hospital, Besançon cedex, FRANCE, 3Geriatric oncology, IHU des Cancers des Femmes (ANR-23-IAHU-0006), Institut Curie, Paris, FRANCE, 4Medical Oncology, Institut Gustave Roussy, Villejuif, FRANCE, 5Medical Oncology, Institut de Cancérologie de l’Ouest/René Gauducheau, Saint-Herblain, FRANCE, 6Medical Oncology, Centre Henri Becquerel, Rouen, FRANCE, 7Medical Oncology, Centre Antoine Lacassagne, Nice, FRANCE, 8Medical Oncology, Institut Sainte Catherine, Avignon, FRANCE, 9Medical Oncology, Centre François Baclesse, Caen, FRANCE, 10Medical Oncology, Institut du Cancer de Montpellier, Montpellier, FRANCE, 11Medical Oncology, Institut Jean Godinot, Reims, FRANCE, 12Medical Oncology, Centre Jean Perrin, Clermont-Ferrand, FRANCE, 13Medical Oncology, Cliniques Universitaires Saint-Luc, Brussels, BELGIUM, 14Medical Oncology, CHU Dupuytren, Limoges, FRANCE, 15Medical Oncology, ELSAN, Hôpital Privé Océane/Centre Saint-Yves, Vannes, FRANCE, 16Medical Oncology, Grand Hôpital de Charleroi, Saint-Cloud, BELGIUM, 17Medical Oncology, Institut Bergonié, Bordeaux cedex, FRANCE, 18R&D, Unicancer, Le Kremlin-Bicêtre, FRANCE, 19U1331 Computational Oncology, Institut Curie, Saint-Cloud, FRANCE, 20Pathology, Gustave Roussy, Villejuif, FRANCE.
Background: The first results of ASTER 70s were presented at the 2022 ASCO annual meeting and have been accepted for publication in the Lancet. After a median follow-up of 8 years, women aged 70 years and older who underwent surgery for luminal breast cancer did not derive any significant benefit in overall survival from adjuvant chemotherapy added to hormonotherapy when selected based on a high-risk genomic grade index (GGI) tumour. However, the acceptability of chemotherapy may vary with age. Methods: We report here the acceptability of chemotherapy in women randomized to receive chemotherapy (4 cycles of anthracycline- or taxane-based chemotherapy administered every 3 weeks) in addition to hormonotherapy, as assessed using a self-administered questionnaire designed based on a previous experience in a similar setting (GERICO 06 trial). After the last cycle of chemotherapy, patients were asked ‘to what extent the treatment was acceptable given the absolute additional benefit of 7.5% in terms of cure sought with chemotherapy”, as stated in the patient information sheet and informed consent form, and the main reason if they found the treatment unacceptable. They were also asked whether they would recommend such treatment to relatives or close friends facing a similar situation. The questionnaires were then administered then yearly for 4 years. Results: Of the 1,969 women screened for tumour GGI between April 2012 and May 2016, 58 (2.9%) withdrew their consent before GGI results and randomisation, while 1,089 (55%) with high-risk GGI tumours were randomized between chemotherapy and no chemotherapy. Of the 541 (49.7%) assigned to chemotherapy, 111 (20.5%) did not receive it for the following reasons: 23 (20.7%) refused chemotherapy, 22 (19.8%) withdrew consent, 8 (7.2%) for medical reasons, and 58 (52.3%) for unknown reasons. Of the 430 patients (79.5%) who started chemotherapy [44 (10.2%) stopped before the fourth cycle], 299 (69.5%) completed the first acceptability questionnaire at the end of chemotherapy, with 266 (95.3%) and 13 (4.7%) rating the chemotherapy experience acceptable and unacceptable, respectively, an assessment that was strongly associated with the occurrence of a serious adverse event during chemotherapy (15% and 46%, respectively, p<0.05). In addition, 242 (94.2%) said they would recommend the treatment to relatives if they were in the same situation. These rates remained stable over time until the fourth year of follow-up. Patient and tumour characteristics, quality of life scores at baseline, overall survival and invasive disease-free survival did not differ between patients who completed the acceptability questionnaire and those who did not. Factors reducing the acceptability of chemotherapy over time (from acceptable à not acceptable versus stable acceptable since the first post-chemotherapy assessment) were advanced age, a high Lee score (4-year mortality estimation) and low IADL. At baseline, low role functioning, low cognitive or social functioning, pain, dyspnoea, and loss of appetite had some impact on the acceptability of chemotherapy. Conclusions: It is difficult to assess the acceptability of a treatment, which depends largely on its safety and, consequently, its intensity in relation to increasing frailty with age. The acceptability of chemotherapy appears also to be influenced by factors such as age and quality of life, which must be taken into account when developing hypotheses in clinical trials, especially in elderly subjects.
Presentation numberPS3-09-12
Thrombophilia Screening Prior to Tamoxifen Therapy in Early-Stage Hormone Receptor-Positive Breast Cancer: A Three-Year Retrospective Study
Stefania Gkoura, Metropolitan Hospital, Neo Faliro, Athens, Greece
S. Gkoura1, I. Binas1, E. Aravantinou – Fatorou1, T. Bilidas1, I. Evmorfiadis2, C. Poziopoulos2, E. Klouva1, K. Koutsoukos1, G. Oikonomopoulos1, G. Karkaletsos1, E. Vasili1, G. Tsakalos1, A. Vassias1, G. Klouvas1, M. Stathoulopoulou3, V. Venizelos3, S. Epameinondas1, C. Christodoulou1, E. Galani1; 12nd Oncology Department, Metropolitan Hospital, Neo Faliro, Athens, GREECE, 22nd Hematology Department, Metropolitan Hospital, Neo Faliro, Athens, GREECE, 3Breast Cancer Unit, Metropolitan Hospital, Neo Faliro, Athens, GREECE.
Background: Tamoxifen remains a cornerstone in the treatment of early-stage hormone receptor-positive breast cancer and is also used as prophylaxis in high-risk women. However, its use has been associated with an increased risk of venous thromboembolism (2-8%). In Greece, the prevalence of thrombophilic factors (inherited or acquired) is approximately 15%, and these factors may contribute to thrombosis when additional risk factors are present. Methods: We conducted a three-year retrospective analysis, from April 2022 until March 2025, of medical records from breast cancer patients in our department who were evaluated by a hematologist and underwent thrombophilia screening before tamoxifen initiation. Results: Among fifty-nine patients, eighteen (31%) were found to have clinically significant inherited thrombophilic factors, while another twenty (34%) had high-risk acquired thrombophilic conditions. Twelve patients (20%) had no detectable thrombophilic factors, and nine (15%) had non-clinically significant mutations that did not require therapeutic intervention. Following hematological assessment, treatment plans were modified in thirty-eight out of fifty-nine patients (64%). Specifically, four patients received tamoxifen with the addition of aspirin, twenty-five were treated with tamoxifen combined with prophylactic oral anticoagulation (apixaban or rivaroxaban), and in nine cases, an aromatase inhibitor was prescribed instead of tamoxifen due to high thrombotic risk, in combination with antiplatelet or anticoagulant therapy. Conclusions: Pre-treatment hematological evaluation and thrombophilia screening are considered of significant clinical value in patients considered for tamoxifen therapy. Based on their individualized thrombotic risk profile, a substantial proportion of patients may benefit from antithrombotic prophylaxis or a change in hormonal therapy. We intend to further evaluate these findings with a prospective cohort of these patients.
Presentation numberPS3-09-13
Adjuvant Abemaciclib in Male Breast Cancer: A UK Multicentre Case Series from the CONCISE Study
Olubukola Ayodele, University of Leicester, Leicester, United Kingdom
A. Ghose1, H. Abdallah2, Y. Owoseni3, A. Maniam4, B. Baraka5, S. Horne6, A. Nazir6, L. Mooney7, F. Nazeer8, N. Atsumi4, L. McAvan9, M. Abraham10, D. Morgan11, G. Langford11, E. Bean12, E. Morris13, R. Muhammad14, E. Daniels15, R. Pravinkumar16, S. Mohammed17, S. Raza18, C. Blair19, R. Khan20, M. Tsalic6, R. Kussaibati6, R. Douglas7, K. Panagiotis21, S. Waters11, J. Smith12, E. Papadimitraki13, C. Michie16, C. Wilson20, A. Konstantis14, O. Ayodele22; 1Barts Cancer Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UNITED KINGDOM, 2Cambridge Clinical Research Centre, Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UNITED KINGDOM, 3Oncology Service, University Hospitals of Leicester NHS Trust, UK, Leicester, UNITED KINGDOM, 4Cancer Centre, Portsmouth Hospitals University NHS Trust, Portsmouth, UNITED KINGDOM, 5Oncology Department, Nottingham University Hospitals NHS Trust, Nottingham, UNITED KINGDOM, 6Oncology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UNITED KINGDOM, 7Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast, UNITED KINGDOM, 8Cancer Services, Surrey and Sussex Healthcare NHS Trust, Redhill, UNITED KINGDOM, 9Cancer Services, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UNITED KINGDOM, 10Dorset Cancer Centre, University Hospitals Dorset NHS Foundation Trust, Poole, UNITED KINGDOM, 11Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UNITED KINGDOM, 12Northern Centre for Cancer Care, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UNITED KINGDOM, 13University College Hospital Macmillan Cancer Centre, University College London Hospitals NHS Foundation Trust, London, UNITED KINGDOM, 14University College London Hospital Cancer Collaborative, Princess Alexandra Hospital NHS Trust, Harlow, UNITED KINGDOM, 15Dyson Cancer Centre, Royal United Hospitals Bath NHS Foundation Trust, Bath, UNITED KINGDOM, 16Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, UNITED KINGDOM, 17Cancer Services, Great Western Hospitals NHS Foundation Trust, Swindon, UNITED KINGDOM, 18Queen’s Centre for Oncology and Haematology, Hull University Teaching Hospitals NHS Trust, Cottingham, UNITED KINGDOM, 19Bristol Haematology and Oncology Centre, University Hospitals of Bristol and Weston NHS Trust, Bristol, UNITED KINGDOM, 20The Christie, The Christie NHS Foundation Trust, Manchester, UNITED KINGDOM, 21Royal Surrey Cancer Centre, Royal Surrey NHS Foundation Trust, Guildford, UNITED KINGDOM, 22Leicester Cancer Research Centre, University of Leicester, Leicester, UNITED KINGDOM.
Background Male breast cancer (MBC) is rare, comprising approximately 1% of all breast cancer diagnoses. Due to its low incidence, treatment strategies are largely extrapolated from studies in female patients, and prospective data in men are limited. The monarchE trial, which established the benefit of Adjuvant Abemaciclib (AA) in high-risk hormone receptor-positive (HR+), HER2-negative early breast cancer (EBC), did not provide sex-specific outcomes. To our knowledge, this is the first report of real-world data on the use of AA in male breast cancer patients. Methods This case series is a subset of the CONCISE study, a retrospective, multicentre UK audit evaluating AA use across 21 centres between July 2022 and April 2025. Male patients with HR+/HER2- EBC who received AA were included. Data were collected using the Ledidi platform and analysed descriptively using Jamovi. Variables assessed included disease characteristics, systemic therapies received, abemaciclib duration, dose modifications, and adverse events (AEs). Results Of 1,026 patients in the CONCISE dataset, 13 (1.3%) were male. Median age at diagnosis was 72 years (range 28-81), and most patients (69%) were White. Staging at diagnosis included: Stage IB (n=6), IIA (n=1), IIB (n=2), IIIA (n=2), and IIIB (n=1). All tumours were ER-positive, HER2-negative (IHC 0-2+). Tumour grade distribution: G1 (n=1), G2 (n=6), and G3 (n=5).Neoadjuvant chemotherapy was administered in 2 patients; neoadjuvant endocrine therapy (tamoxifen) in 1. Breast-conserving surgery was performed in 1 case; the remainder underwent mastectomy. Adjuvant chemotherapy was delivered in 8/13 patients (62%), with 75% completing ≥6 cycles. Adjuvant ET included tamoxifen (n=8), aromatase inhibitors with ovarian function suppression (OFS) (n=5).Median AA duration was 12.3 months (range 1.9-24). At the time of analysis, 4 patients (31%) remained on therapy. Among those who discontinued, 5 (56%) stopped prematurely due to toxicity, 1 due to disease progression, and 3 completed the full 24-month course. At least one dose reduction occurred in 6 patients (46%), with a second reduction in 1.AEs included diarrhoea in 7 patients (54%, ≥G2 in 1), anaemia in 4 (31%, ≥G2 in 2), neutropenia in 6 (46%, ≥G2 in 5; including G3 in 3), thrombocytopenia in 3 (23%, ≥G2 in 2), and transaminitis in 2 (15%, G1 only). No treatment-related deaths were reported. Conclusion This is the first UK real-world cohort reporting on AA use in male breast cancer, an underrepresented population. Compared to the monarchE AA+ET cohort, male patients were older (median age 72 vs. 51 years), had slightly shorter treatment duration (12.3 vs. 14 months), and similar rates of dose reduction (46% vs. 41.2%). However, discontinuation due to toxicity was higher in this cohort (56% vs. 16.6%), highlighting the need of larger, prospective cohorts for tailored toxicity monitoring and management strategies in men receiving AA. While these findings should be generalised with caution due to limited sample size, treatment overall appears manageable and associated with toxicity profiles seen in other populations.
Presentation numberPS3-09-14
Caught in the Middle: Discrepancies Between Immunohistochemistry and Oncotype DX in Hormone Receptor Assessment Leading to Triple Negative Reclassification—Tumour Heterogeneity or Just Low ER Expression?
Vasileios Angelis, Barts Health NHS Trust, London, United Kingdom
V. Angelis1, A. Grobbelaar1, A. Zamani1, P. Hall1, M. Phillips1, P. Schmid1, K. Hawkesford1, M. Ullah1, L. Begolli1, L. Johnson1, S. Ledwidge1, M. Thorat2, F. Irakleidis3, K. Sidlauskas4, D. Ryan4, M. Warren4; 1Breast Unit, Medical Oncology, Barts Health NHS Trust, London, UNITED KINGDOM, 2Breast Unit, Medical Oncology, Homerton Healthcare NHS Trust, London, UNITED KINGDOM, 3Breast Unit, Euromedical General Hospital, Rhodes, Greece, Rhodes, UNITED KINGDOM, 4Department of Cellular Pathology, Barts Health NHS Trust, London, UNITED KINGDOM.
BackgroundDiscrepancies in the assessment of ER, PR, and HER2 status by immunohistochemistry (IHC) and Oncotype DX (ODX) can substantially impact treatment decisions. Variations in assay results that alter tumour molecular subtype may lead to changes in recommended adjuvant therapy. We present a series of such cases and explore whether these findings reflect tumour heterogeneity or misclassification of ER expression. We show how reclassification influenced systemic treatment choices and eligibility for genetic testing, underscoring the practical implications of such discrepancies for the personalisation of breast cancer care. MethodsRetrospective analysis was conducted of 682 patients who underwent ODX testing between 2020-2025 at Barts Health NHS Trusts hospitals. Data extracted from the ODX reports included the recurrence score and the qualitative and quantitative assessments of mRNA levels of ESR1, PGR, and ERBB2 receptor genes. These results were compared to ER, PR, and HER2 receptor protein status determined by standard IHC staining and scoring on the diagnostic biopsy specimens1. The standard ER IHC scoring categorises ER status as ER negative (score 0,2) ; ER low positive (score 3,4) and ER positive (score 5-8)1. All ER biopsy slides were reviewed blind by a breast pathologist (MW). Receptor IHC was repeated on the resection specimen tumour block used for ODX testing and the results compared to the ER IHC score on the diagnostic biopsy. Results were compared with 5 control cases in which the ODX as well as ER IHC on biopsy was positive. ResultsFrom a total of 682 ODX tests performed, 12 patients (1.7%; 12/682) demonstrated discordance, with a negative ESR1 gene expression score on ODX and ER IHC positive score (5-8). Upon repeat histological review, all 12 cases (100%) were confirmed to be either ER low-positive (75%; 9/12) or ER negative (25%; 3/12) on ER IHC on the resected tissue. On review of the ER IHC biopsy slides, in 7/12 cases (58.3%) the tumour displayed heterogeneous ER IHC staining on biopsy, with predominantly ER negative tumour being present on the resection block. 3/12 biopsies (25%) originally reported as ER positive on the basis of highest-staining hotspots were reclassified as overall ER low-positive on review. 2/3 (66.7%) of these cases also exhibited heterogeneity of ER IHC staining. 1/12 cases had multifocal breast cancer with one ER positive (score 8) and one ER low positive tumour (score 3) with the Oncotype test performed on the ER low positive tumour. 1/12 (8.3%) of cases was an external referral case and could not be reviewed. All 5 control cases with ODX/biopsy IHC positive were also positive by ER IHC on the resection block. All discordant tumours were grade 3 invasive ductal carcinomas presenting in women aged 30-77 years. 3 patients (25%) were BRCA1 mutation carriers. Regarding adjuvant chemotherapy, 4 patients (33.3%) were treated with ER+ regimens, while 7 patients (58.3%) received triple-negative regimens. ConclusionsDiscordance between IHC and ODX ER status is rare (1.7%). Most discrepancies were attributable to intratumoural heterogeneity of ER protein expression in biopsies with predominantly ER negative sub-clones in resection samples. Repeat IHC confirmed these resected cases as ER low-positive or negative, in concordance with ODX, confirming a higher-risk subgroup. Our findings as well as data from several breast receptor RT-PCR studies 2,3,4 also support the potential for subtype reclassification initially classified as ER low positive (score 3,4), with low or negative ODX ESR1 gene scores as similar in biological behaviour and chemotherapy response to ER negative tumours. This could help guide personalisation of adjuvant treatment strategies and prompt germline mutation testing.
Presentation numberPS3-09-15
Ribociclib drug-drug interaction and concomitant medication management in early and advanced breast cancer patients
Liz Santarsiero, Novartis Pharmaceutical Corporation, East Hanover, NJ
Y. Ji1, F. Huth2, L. Santarsiero1, J. P. Zarate1, H. Schiller2; 1Oncology, Novartis Pharmaceutical Corporation, East Hanover, NJ, 2Regulatory & Registration Readiness, Novartis Pharma AG, Basel, SWITZERLAND.
Background: Ribociclib is approved for the treatment of HR+/HER2- advanced or metastatic breast cancer (ABC) in combination with an aromatase inhibitor or fulvestrant, and more recently, for the adjuvant treatment of HR+/HER2- stage II and III early breast cancer (EBC) at high risk of recurrence, with the starting dose of 600 mg in ABC and 400mg in EBC. The DDI profile of ribociclib has been previously characterized 1,2. Ribociclib is a strong to moderate cytochrome P450 (CYP) CYP3A4 inhibitor at 600 mg and 400 mg, respectively, and thus caution is recommended for concomitant use with sensitive CYP3A4 substrates with a narrow therapeutic index. Ribociclib is also a CYP3A4 substrate, and concomitant use with strong inhibitors or inducers of CYP3A4 should be avoided. Methods: The DDI profile of ribociclib and physiology-based pharmacokinetic modeling (PBPK) modeling were utilized to assess the DDI between ribociclib and the commonly used concomitant medications that are statins, antidepressants/antipsychotics, and corticosteroids. Results: There is no overall DDI risk for the class of statins with ribociclib. The effect of ribociclib on rosuvastatin, pravastatin, pitavastatin, and fluvastatin is considered minor with no clinical relevance, since these statins are not sensitive CYP3A4 substrates. Only the statins with substantial metabolism by CYP3A4, such as simvastatin, lovastatin and atorvastatin, may have increased exposure upon co-medication with ribociclib 400 or 600 mg. No clinically relevant DDI is expected for commonly used antidepressants/antipsychotics, such as citalopram, escitalopram, fluoxetine, mirtazapine, sertraline, trazodone, venlafaxine, aripiprazole, olanzapine and cariprazine, when co-administered with ribociclib 400 mg or 600 mg. Quetiapine is a CYP3A4 substrate and its exposure may increase approximately 2-3-fold when co-administered with ribociclib 400 or 600 mg. Systemically administered corticosteroids which induce CYP3A4, such as dexamethasone, may reduce ribociclib exposure if they are used chronically. The extent of the decrease of ribociclib exposure depends on the dexamethasone dose and doses of 5 mg or lower are not expected to have clinically relevant DDI. Dexamethasone is also a CYP3A4 substrate, and its exposure may be increased (<2-fold) when co-administered with 400 or 600 mg ribociclib. DDI with ribociclib is not expected for corticosteroids that are administered non-systemically (e.g. topical, inhaled, eye drop, or local injection). Conclusion: No clinically relevant DDI with ribocicilib is anticipated for commonly used statins or antidepressants/antipsychotics, except simvastatin, lovastatin, atorvastatin and quetiapine, which are substantially metabolized by CYP3A4. Systemic corticosteroids that are CYP3A inducers and/or substrates such as dexamethasone should be used for a short duration and monitored for their adverse effects when co-administrated with ribociclib. References: 1. Untch M. et al, Ribociclib in Early Breast Cancer: Drug-Drug Interaction and Organ Impairment Assessments, European Society for Medical Oncology (ESMO) Breast Cancer Conference, Munich, Germany, May 14-17, 2025 2. Samant T. et al, Ribociclib Drug-Drug Interactions – Clinical Evaluations and PBPK Modeling Guiding Current Drug Labeling, Clin Pharmacol Ther. 2020, 108 (3): 575-585.
Presentation numberPS3-09-16
Prognostic impact of human epidermal growth factor receptor 2 (HER2) expression level in hormone receptor-positive/HER2-negative, non-metastatic premenopausal breast cancer patients: A retrospective cohort study
Takeshi Murata, National Cancer Center Hospital, Tokyo, Japan
T. Murata1, T. Shimoi2, A. Saito2, M. Onishi2, M. Yoshida3, N. Ogi1, M. Chen1, A. Ogawa1, A. Nakashoji1, H. Maeda1, C. Watase1, K. Sudo2, K. Yonemori2, S. Takayama1; 1Breast Surgery, National Cancer Center Hospital, Tokyo, JAPAN, 2Medical Oncology, National Cancer Center Hospital, Tokyo, JAPAN, 3Diagnostic Pathology, National Cancer Center Hospital, Tokyo, JAPAN.
Aims: With the advent of new antibody-drug conjugates, human epidermal growth factor receptor 2 (HER2) low expression is a new therapeutic category for breast cancer. However, the prognostic differences between categories corresponding to 1+ on immunohistochemistry (IHC) and 2+ on IHC with no amplification by ISH have not been sufficiently investigated. This study aimed to evaluate the prognostic impact of different HER2 expression levels in patients with hormone receptor-positive/HER2-negative, premenopausal breast cancer. Methods: Using data from our institutional database from 2008 to 2017, we enrolled hormone receptor-positive/HER2-negative, non-metastatic premenopausal breast cancer patients with pathological N0-1 disease treated with curative surgery. Hormone receptor-positive was defined as either estrogen receptor (ER)- or progesterone receptor (PR)-positive. ER and PR were considered positive if the IHC staining was positive in more than 1% of tumor cells. A HER2-negative result corresponded to a score of 0, 1+ on IHC, or 2+ on IHC with no amplification by ISH. We divided enrolled patients into three groups according to their HER2 expression levels: patients with HER2-zero (Group 1), HER2-1+ (Group 2), and HER2-2+ with no amplification by ISH (Group 3). Distant relapse-free survival (DRFS) among the 3 groups were estimated using the Kaplan-Meier method, and survival estimates were compared using the log-rank test. Cox proportional-hazards model with hazard ratio (HR) and 95% confidence interval (CI) was used to evaluate the independent prognostic effects of each variable on DRFS. Results: The median age was 45 years. Among 1011 patients enrolled, 269 patients (26.6%) had pN1 disease, 318 (31.5%) had pT2 disease, 193 (19.1%) had histological grade 3 disease. Group 1, 2, and 3 had 415 (41.1%), 465 (46.0%), and 129 (12.8%) patients, respectively. The median follow-up time after surgery was 8.2 years. During the follow-up period, 23 patients died from breast cancer, 7 patients died from causes other than breast cancer, and 58 patients had distant metastases. There were no differences in adjuvant treatment among the 3 groups, with mastectomy, chemotherapy, endocrine therapy (ET), and radiotherapy given to approximately 40%, 30%, 90%, and 60%, respectively. The duration of ET more than 5 years was given to approximately 50%. Patients in Group 3 had significantly worse DRFS than patients with Group 1 and 2 (8y-DRFS, 89.8% vs. 95.6%, P=0.0042, 89.8% vs. 94.9%, P=0.0048, respectively).Multivariable analysis revealed that the significant risk factors associated with worse DRFS were pT2 disease (HR 2.67; 95% CI 1.46-4.88; P=0.001), pN1 disease (HR 3.48; 95% CI 1.76-6.86; P<0.001), and HER2-2+ with no amplification by ISH (HR 3.26; 95% CI 1.61-6.61; P=0.001). HER2-1+ was not associated with worse DRFS (HR 1.18; 95% CI 0.64-2.16; P=0.587). The duration of ET more than 5 years was significantly associated with favorable DRFS (HR 0.11; 95% CI 0.06-0.23; P<0.001). Conclusions: The results from this single institutional database study suggested that HER2-2+ with no amplification by ISH may be one of the poor prognostic factors in hormone receptor-positive/HER2-negative, non-metastatic premenopausal breast cancer patients with pathological N0- 1 disease. Patients with HER2-2+ with no amplification by ISH may require a different treatment strategy than patients with HER2-zero and HER2-1+. Further evaluation in a larger cohort is needed.
Presentation numberPS3-09-17
De-escalation of systemic therapy for breast cancer in the Indian Setting: A Proteomics and artificial intelligence based Approach to Risk Stratification beyond Ki67, Tumour Grade, and NPI for HR+/HER2-Negative Breast Cancer.
Tanay N Shah, HCG AASTHA cancer hospital, Ahmedabad, India
T. N. Shah1, V. Devanhalli1, M. Shah2, M. Bakre3; 1Surgical Oncology, HCG AASTHA cancer hospital, Ahmedabad, INDIA, 2Medical Oncology, HCG AASTHA cancer hospital, Ahmedabad, INDIA, 3R&D, Oncostem Diagnostics, Ahmedabad, INDIA.
Objective Determining the need for adjuvant chemotherapy in early-stage hormone receptor-positive (HR+), HER2-negative breast cancer remains a clinical challenge, particularly in stage I/II disease. This study evaluates the prognostic performance of the CanAssist Breast (CAB) test a proteomics-based risk stratification tool against conventional markers such as tumour grade, Ki67 index, and Nottingham Prognostic Index (NPI), with a focus on validation in the Indian population. Notably, the CAB test is cost-effective, priced below USD 1000.Materials and MethodsA retrospective observational analysis was conducted on over 160 patients diagnosed with stage I/II HR+/HER2-negative invasive breast carcinoma, who underwent the CAB test at HCG Cancer Centre, Ahmedabad up to June 2025. Patients were classified into low- or high-risk groups using a CAB score cut-off of 15.5. Risk categorization was compared with histopathological parameters: tumour grade (1, 2, 3), NPI (low, intermediate, high), and Ki67 index (low, moderate, high). Associations between CAB scores and traditional prognostic markers were assessed using Chi-square statistical analysis. Ongoing follow-up is being carried out to update patient outcomes and recurrence data.Results Over 60% of patients were categorized as low-risk based on the CAB score. A statistically significant association was observed between CAB scores and tumour grade: most grade 3 tumours exhibited high CAB scores, whereas grade 2 tumours predominantly showed low CAB scores. Similarly, a significant correlation was found between NPI and CAB scores, with most intermediate NPI cases falling into the low-risk CAB group. Interestingly, a substantial proportion (>50%) of patients with moderate or high Ki67 indices also exhibited low CAB scores. Additionally, a subset of node-positive (N1) patients fell into the CAB low-risk category. Importantly, none of the low-risk patients who were spared chemotherapy had reported recurrence at the time of the last follow-up. Follow up ananlysis is ongoing currently. Final results will be further substantiated upon completion of follow-up data analysis.Conclusion The CanAssist Breast test effectively refines risk stratification in early-stage HR+/HER2-negative breast cancer, especially in patients with ambiguous traditional markers (e.g., grade 2, intermediate NPI, moderate Ki67). By reducing the reliance on adjuvant chemotherapy in low-risk groups, the CAB test helps avoid unnecessary treatment-related toxicities. These findings support the utility of CAB as a cost-efficient, clinically meaningful decision-making tool in the Indian oncological landscape.
Presentation numberPS3-09-18
Early AB-ITALY: Evaluating Real-World Use of Adjuvant Abemaciclib and Drug Interaction Risk in HR+/HER2- early Breast Cancer
Andrea Botticelli, Sapienza University of Rome; AOU Policlinico Umberto 1, Rome, Italy
S. Scagnoli1, M. Verrico1, S. Pisegna2, R. Caputo3, F. Pantano4, P. Falbo5, G. D’Auria6, A. Orlandi7, D. Alesini8, I. Portarena9, P. Vici10, M. Fabbri11, L. Rossi12, T. Di Palma13, G. Gentile14, M. Bonomo1, L. Sisca4, C. Bonadonna1, L. Strigari15, G. Ricciardi16, M. Pizzoli1, E. Giordani17, A. Irelli18, M. Sanò19, M. Palleschi20, R. Preissner21, A. Daneri22, O. Garrone23, G. Bianchini1, C. Martinelli3, C. Criscitiello24, M. Lambertini22, A. Fabi25, M. De Laurentiis3, L. Del Mastro22, G. Curigliano26, P. Marchetti27, A. Botticelli1; 1Department of Radiological, Oncological and Pathological Sciences; UOC Oncologia, Sapienza University of Rome; AOU Policlinico Umberto 1, Rome, ITALY, 2Department of Experimental Medicine; UOC Oncologia, Sapienza University of Rome; AOU Sant’Andrea, Rome, ITALY, 3Department of Breast and Thoracic Oncology, Istituto Nazionale Tumori-IRCCS-” Fondazione G.Pascale”, Naples, ITALY, 4Department of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, ITALY, 5Department of Oncology, Azienda Ospedaliera San Giovanni Addolorata, Rome, ITALY, 6Medical Oncology, Department of Medical Oncology, Azienda Ospedaliera Sandro Pertini Roma, Rome, ITALY, 7Department of Oncology, Comprehensive Cancer Center, Fondazione Policlinico Uni versitario Agostino Gemelli IRCCS, Rome, ITALY, 8Medical Oncology Department, Ospedale Santo Spirito, Rome, ITALY, 9Oncology Unit, Policlinico Tor Vergata, Rome, ITALY, 10Phase IV Clinical Studies Unit, IRCCS Istiuto Nazionale Tumori Regina Elena (IRE), Rome, ITALY, 11Department of Oncology, belcolle hospital ASL Viterbo, Viterbo, ITALY, 12UOS Oncologia Formia, UOC Oncologia Territoriale; Università Sapienza di Roma Polo Pontino, Formia, ITALY, 13Oncology Unit, Ospedale Santa Maria Goretti – ASL Latina, Latina, ITALY, 14Academic Trials Promoting Team, Université libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B), Institut Jules Bordet, Bruxelles, BELGIUM, 15Department of Medical Physics, University of Bologna- Alma Mater Studiorum, Bologna, ITALY, 16Department of Onco-hematology, Papardo Hospital, Messina, ITALY, 17Oncology Unit, Ospedale Santa Maria Goretti- ASL Latina, Latina, ITALY, 18Medical Oncology Unit, “Giuseppe Mazzini” Hospital AUSL 04 Teramo, Teramo, ITALY, 19Department of Medical Oncology, Humanitas Istituto Clinico Catanese, Catania, ITALY, 20Medical Oncology, Breast & GYN Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, ITALY, 21Institute of Physiology and Science-IT, Charité Universitaetsmedizin Berlin, Berlin, GERMANY, 22Department of Internal Medicine and Medical Specialties (DIMI); Department of Medical Oncology, University of Genova; Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 23Medical Oncology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, ITALY, 24Department of Oncology and Hematology-Oncology, European Institute of Oncology, IRCCS; University of Milan, Milan, ITALY, 25Precision Medicine in Senology, Scientific Directorate-Department of Woman and Child Health,, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, ITALY, 26Division of New Drugs and Early Drug Development for Innovative Therapies; Department of Oncology, European Institute of Oncology, IRCCS; University of Milan, Milan, ITALY, 27Oncology Unit, Istituto Dermopatico dell’Immacolata IRCCS, Rome, ITALY.
Introduction Abemaciclib is a selective CDK4/6 inhibitor approved for adjuvant treatment in patients with high-risk hormone receptor-positive, HER2-negative (HR+/HER2-) early breast cancer (eBC). The MONARCH-E trial demonstrated significant increase in invasive disease-free survival (iDFS) when combined abemaciclib with endocrine therapy. Nonetheless, real-world data regarding safety, treatment adherence, and drug-drug interactions (DDIs), particularly among elderly and comorbid populations often excluded from clinical trials, remain limited. Evaluating these real-world factors is critical to optimizing treatment and patient outcomes in clinical practice. Methods This retrospective study analyzed consecutive patients with HR+/HER2- eBC treated at 21 Italian oncology centers who received adjuvant abemaciclib combined with an aromatase inhibitor (AI). Comprehensive clinical data, medication usage, and adverse events (AEs) were collected. Drug-PIN®, a platform integrating clinical (age, sex, race, smoking and alcohol habits), laboratory (renal and hepatic function), and pharmacological parameters (chronic concomitant treatments), was utilized to assess DDIs, providing a numeric interaction score (Drug-PIN score) and qualitative risk tiers (green: none; yellow/dark yellow: intermediate; red: high risk). Statistical analyses, including univariate and multivariate approaches, identified predictors of AEs. Results We enrolled 714 patients with a median age of 56 years (range 30-86); 44% were premenopausal, and 51% had at least one comorbidity, while 13% had multiple comorbidities. Notably, 60 patients (8.4%) were aged ≥75 years. Genetic testing performed in 350 patients identified 29 mutated. Disease characteristics included node-positive (c/pN2) status in 389 (54%), carcinoma of no special type in 73%, lobular carcinoma in 14%, and high-grade tumors (G3) in 57%. Chemotherapy was omitted in 10% of cases. Oncotype DX testing was conducted in 42 patients (6%), with a mean recurrence score (RS) of 26 (range 8-50); 20 had an RS ≤25. With a median follow-up of 13.2 months (range 1-33), 85% of patients experienced any-grade AEs, primarily diarrhea (76%), asthenia (41%), and neutropenia (27%). Grade ≥3 AEs occurred in 30%, with severe diarrhea reported in 11%, mostly during the first two cycles. Dose reductions were required in 47% and discontinuation due to toxicity occurred in 9.5%. Patients aged ≥75 showed comparable diarrhea rates (p=0.20) but a trend toward increased dose reductions (p=0.09). Concomitant medications were reported in 50% of patients, and 8% had polypharmacy. The median Drug-PIN interaction score was 12.0 (range 3-100), with at least one DDI identified in 17.3% of the patients, with 10.5% categorized in the intermediate-high or high-risk tiers. Notably, high-risk DDIs occurred more frequently among patients who discontinued treatment due to toxicity (25% vs 10%, p=0.04). Conclusions Adjuvant abemaciclib is safe and manageable in real-world settings, even among elderly and comorbid patients, showing slightly higher but comparable AE rates and dose adjustments relative to clinical trials. Overall, abemaciclib treatment presents a low incidence of drug-drug interactions. However, the significant association observed between DDIs and dose reductions emphasizes the necessity for further prospective investigations to better define these interactions and inform clinical practice.
Presentation numberPS3-09-19
Correlation Between Breast Cancer Index (BCI) and RSClin Late in Assessing 5-10 Year Recurrence Risk in Early-Stage Hormone Receptor-Positive Breast Cancer
Karishma Vijay Rupani, Icahn School of Medicine at Mount Sinai, New York, NY
K. Rupani1, S. Wang2, M. Diniz2, P. Klein1; 1Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 2Division of Biostatistics, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY.
Background: Hormone receptor-positive (HR+), HER2-negative early-stage breast cancer constitutes approximately 70-80% of all breast cancers. However, despite favorable short-term outcomes with adjuvant endocrine therapy, a subset of patients remain at risk for late recurrence beyond 5 years, necessitating personalized decisions about extending endocrine therapy. Genomic assays have emerged as valuable tools for refining risk estimates and guiding clinical decision-making in this setting. Two such tools—Breast Cancer Index (BCI) and RSClin Late—are often used to assess the likelihood of distant recurrence from years 5 to 10, avoiding unnecessary endocrine therapy in patients with low risk and extending it in those with high risk. BCI combines two independent biomarkers, HOXB13:IL17BR (H:I) and the 5-gene molecular grade index (MGI), that assess estrogen-mediated signaling and tumor grade, respectively, to provide both prognostic and predictive information for late recurrence and extended endocrine therapy benefit. RSClin Late integrates the Oncotype DX 21-gene recurrence score with clinical and pathologic features (age at surgery, tumor size, tumor grade) to generate individualized 5- to 10-year risk estimates. Despite overlapping indications, the degree of concordance between these two tools remains unclear. Methods: We conducted a retrospective analysis of all patients within our practice with HR+, HER2-negative early-stage breast cancer without lymph node involvement who underwent BCI testing between January 1, 2024 to June 6, 2025. A total of 27 eligible patients were identified. The BCI Prognostic Results (risk of recurrence with 5 total years of adjuvant endocrine therapy) and RSClin Late scores were extracted from the electronic medical record. The Spearman rank correlation coefficient was calculated with a 95% confidence interval (CI) to assess the monotonic relationship between the two scoring systems. This nonparametric method was selected to account for potential outliers, non-linear relationships, and skewed distributions. Results: The Spearman correlation coefficient (ρ) between the BCI Prognostic Result and RSClin Late was 0.413 (95% CI 0.039 to 0.685), with a p-value of 0.032, indicating a moderate, statistically significant positive correlation between the two measures. These findings suggest that while the tests may grossly align in terms of risk stratification, clinically meaningful discordance may occur in individual cases. Discussion: While our study is limited by a small sample size and larger analyses are needed to better characterize the degree and clinical impact of discordance between these tools, the moderate but statistically significant correlation observed highlights that BCI and RSClin Late, although both designed to predict late recurrence, are complementary but not interchangeable. BCI is a purely genomic assay focused on tumor biology, while RSClin Late integrates genomic (Oncotype DX) and traditional clinical and pathologic data, leading to possible variation in risk assignment. Both assays have been independently validated to inform decisions regarding extended endocrine therapy, but their moderate correlation raises questions about how to interpret conflicting results. Conclusion: In this real-world cohort of patients with early-stage HR+ N0 breast cancer, BCI and RSClin Late scores demonstrated a moderate but statistically significant correlation. This supports a degree of concordance but also underscores that the tests may not be interchangeable. Incorporating both genomic and clinical-pathologic perspectives into shared decision-making discussions may help personalize extended endocrine therapy decisions, avoid unnecessary toxicity, and improve long-term outcomes.
Presentation numberPS3-09-20
Not all pN1 ER-pos HER2-neg breast cancers are equal: why careful patient selection is key for adjuvant ribociclib
Babette Salaets, University Hospitals Leuven, Leuven, Belgium
B. Salaets1, A. Deblander1, L. De Sutter2, K. Van Baelen1, M. Van Houdt1, N. Willers1, T. Baert1, A. Baten3, F. Derouane4, G. Floris5, S. Han1, H. Janssen3, I. Nevelsteen2, C. Remmerie6, A. Smeets2, J. Verhoeven3, Y. Van Herck4, C. Van Ongeval5, C. Weltens3, H. Wildiers4, P. Neven1; 1Department of Gynaecological Oncology, University Hospitals Leuven, Leuven, BELGIUM, 2Department of Surgical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 3Department of Radiotherapy, University Hospitals Leuven, Leuven, BELGIUM, 4Department of General Medical Oncology, University Hospitals Leuven, Leuven, BELGIUM, 5Department of Imaging and Pathology, University Hospitals Leuven, Leuven, BELGIUM, 6Department of Oncology, University Hospitals Leuven, Leuven, BELGIUM.
Introduction:For patients with high-risk ER-pos, HER2-neg breast cancer, adding abemaciclib (2 years) or ribociclib (3 years) to standard endocrine therapy improves invasive disease-free survival (IDFS). We hypothesize that not all patients who meet the eligibility criteria of the NATALEE trial will derive a clinically meaningful benefit from 3 years of adjuvant ribociclib. Previously, we demonstrated that patients with grade 3 pT2N0 tumors have a high risk of recurrence and might benefit from this additional treatment. Contrarily, patients with a pT0-2N1 tumor with more favorable tumor biology (grade 1-2), might have a low risk of recurrence irrespective of adjuvant treatment with a CDK4/6 inhibitor. The RxPONDER trial showed that 2497 patients with pN1, genomically low-risk tumors achieved a 94% 5-year distant disease-free survival (DDFS) with endocrine therapy alone. Here, we present real-world outcomes for patients with grade 1-2, pT0-2N1, ER-pos, HER2-neg tumors.Methods: We retrospectively reviewed the UZ Leuven-database of patients with early-stage ER-pos, HER2-neg breast cancer who underwent surgery between 2000 and 2023. All patients included met the NATALEE trial eligibility criteria for adjuvant ribociclib following local therapy. Eligibility (Cohort 1) was defined as stage III disease (AJCC 8th edition) or stage II including either pN1 or pT2-3N0 tumors with grade 3, or grade 2 tumors with Ki-67 >20% and/or a MammaPrint high-risk profile. We assessed both IDFS and DDFS in this NATALEE-eligible population and compared (Cohort 1) to a predefined lower-risk subgroup (Cohort 2), characterized as patients with stage II, pT0-2N1, grade 1 or 2 tumors (excluding pT1N1mi). Kaplan-Meier curves were used for the estimation of the DDFS and IDFS for both cohorts. Analyses have been performed using SPSS software. The 4-year outcome was chosen in accordance with the ESMO 2024 updated analysis of NATALEE with a 44.2 median follow-up.Results: We identified 3980 patients eligible for adjuvant ribociclib according to NATALEE-criteria (Cohort 1); 66,8% were stage II and 1357 of them met the lower-risk subgroup criteria (Cohort 2). Median age at diagnosis was 58 years for both cohorts and median follow-up time was 17,6 years for Cohort 1 and 18,8 years for Cohort 2. Analysis showed a 4-year IDFS for Cohort 1 of 87,6% as compared to 93,5% for patients for Cohort 2. DDFS was 86.4 % and 94.5% respectively. At 10 years of follow-up the absolute difference in DDFS increased to 9,9% (70,1% vs. 80,0%).Discussion: We found that lower-risk, NATALEE eligible patients, already have an encouraging 4-year IDFS and DDFS. These data emphasize the need for careful patient selection, to derive a clinically meaningful benefit, balancing efficacy against potential toxicity.
| Cohort1 | Cohort2 | |
| All NATALEE eligible (n= 3980) | Stage II N+ grade 1-2 (n= 1357) | |
| MEDIANAGEINYEARS | 58 | 58 |
| ANATOMICALSTAGE – no. (%) | ||
| Stage II | 2658 (66,8%) | 1357 (100%) |
| Stage III | 1322 (33,2%) | 0 (0%) |
| TUMORSIZE – NO. (%) | ||
| T1 | 732 (18,4%) | 497 (36,6%) |
| T2 | 2385 (59,9%) | 860 (63,3%) |
| T3 | 592 (14,9%) | 0 (0%) |
| T4 | 264 (6,6%) | 0 (0%) |
| T0 or Tx | 7 (0,2%) | 0 (0%) |
| NODALSTATUS – NO. (%) | ||
| N0 | 819 (20,6%) | 0 (0%) |
| N1 | 2327 (58,5%) | 1357 (100%) |
| N2-3 | 831 (20,9%) | 0 (0%) |
| NX | 3 (0,1 %) | 0 (0%) |
| HISTOLOGICGRADE – NO. (%) | ||
| 1 | 337 (8,5%) | 261 (19,2%) |
| 2 | 2084 (69,9%) | 1096 (80,8%) |
| 3 | 1543 (38,8%) | 0 (0%) |
| UNKNOWN | 16 (0,4%) | 0 (0%) |
| CHEMOTHERAPY – NO. (%) | ||
| NEO-ADJUVANT | 402 (10,1%) | 37 (2,7%) |
| ADJUVANT | 1957 (49,1%) | 493 (36,3%) |
Presentation numberPS3-09-21
Evaluation of Sensitivity to Endocrine Therapy (SET) as a Prognostic Biomarker in the Latin American Breast Cancer Cohort Study
Marcela Mazo Canola, UT Health San Antonio, san antonio, TX
M. Mazo Canola1, A. Llera2, K. Zhang3, K. M. Tran4, M. Jeon5, D. Alves da Quinta2, E. Abdelhay6, N. Artagaveytia7, A. Daneri-Navarro8, B. Müller9, O. Podhajcer2, J. Retamales10, W. Symmans11, J. Gomez12; 1Medicine/ Division of Hematology and Oncology, UT Health San Antonio, san antonio, TX, 2Laboratorio de Terapia Molecular y Celular, Laboratorio de Terapia Molecular y Celular, Fundación Instituto Leloir-CONICET, Buenos Aires, ARGENTINA, 3Center for Epigenetics & Disease Prevention, Texas A&M University, Health Science Center, Houston, TX, 4MD Anderson Cancer Center, University of Texas, Houston, TX, 5Center for Epigenetics & Disease Prevention, Health Science Center, Texas A&M University, Houston, TX, 6Instituto Nacional de Câncer, Instituto Nacional de Câncer, Rio de Janeiro, Rio de Janeiro, BRAZIL, 7Oncology, Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, URUGUAY, 8Oncologia, Universidad de Guadalajara, Guadalajara, MEXICO, 9Oncology, Instituto Nacional del Cáncer, Santiago de Chile, Santiago, CHILE, 10Oncologia, Grupo Oncológico Cooperativo Chileno de Investigación, Santiago, CHILE, 11Department of Anatomical Pathology., UT MD Anderson Cancer Center, Houstin, TX, 12Center for Genomic and Precision Medicine, Health Science Center Texas A&M Univeristy, College Station, TX.
Sponsored by: Breast Cancer Research Foundation: BCRF-19-187 Background:The SET2,3 index, which combines the SETER/PR index—a measure of transcriptional activity related to estrogen and progesterone receptor (ER, PR) —with the baseline prognostic index (BPI), has been validated as prognostic and predict endocrine treatment sensitivity in hormone receptor-positive (HR+) breast cancers from U.S. and European patients. However, its prognostic utility remains to be determined in Latin American (LA) breast cancer patients, who possess a unique and diverse genetic admixture.Methods:Clinical and pathological retrospective data were collected from a multi-country Latin American cohort of breast cancer patients. Tumor subtypes were previously characterized using PAM50 classification, Ki67%, and immunohistochemical analysis for ER, PR, and Human Epidermal Growth Factor Receptor 2 (HER2) status. The SET2,3 index was measured using the QuantiGene Plex Assay on archival tumor tissue samples from the HR+/HER2 Negative (-) cancers, blinded to clinical outcomes. Disease-free survival (DFS) was compared across patient subgroups using the log-rank test, and hazard ratios (HRs) were estimated through Cox proportional hazards models.Results: A total of 292 patients were included, with 38 disease-free survival (DFS) events observed over a median follow-up of 7.15 years. In a multivariable Cox model, use of adjuvant endocrine therapy (Yes vs. No; HR 0.27; p <0.001) and SET2,3 index (Low vs High; HR 3.45; p <0.001) were independently prognostic for longer DFS. Among patients who underwent primary surgery, SET2,3 index was significantly associated with DFS (Low vs. High; HR 3.20; 95% CI: 1.48-6.91; p =0.002). SET2,3 index also predicted outcomes for those receiving adjuvant chemotherapy (Low vs. High; HR 3.66; 95% CI: 1.25-10.78; p =0.01) or endocrine therapy alone (Low vs. High; HR 3.13; 95% CI: 1.21-8.10; p = 0.01). When stratified by PAM50 subtype, SET2,3 was significantly prognostic in non-Luminal A tumors (n = 9084; Low vs. High; HR 5.19; 95% CI: 1.20-22.51; p =0.01) but not in Luminal A tumors (n =162;0 p =0.3). Similarly, SET2,3 was prognostic in patients with low Ki67 index (<20%) (n=97 Low vs High; HR 4.79; 95% CI: 1.20-19.17; p =0.01), but not in those with a high Ki67 index (n=155 p=0.06). The SETER/PR index of endocrine transcriptional activity (continuous variable) was also prognostic in 198 patients who received adjuvant endocrine therapy (HR 0.31 per unit, 95%CI 0.17-0.59, p <0.001). It was not significantly prognostic in the subset of 46 patients who did not receive endocrine therapy (HR 0.59, 95%CI 0.26-1.38, p =0.23). Conclusions:Our preliminary results confirm the SET2,3 as a strong predictor of disease-free survival in breast cancer of Latin American patients, with high SET2,3 index indicating longer DFS. SETER/PR index of endocrine transcriptional activity was strongly prognostic for patients who received adjuvant endocrine therapy. Some limitations: Small sample size in some subsets, retrospective nature of the analysis and differences in use of endocrine therapy between countries.These findings highlight the importance of including diverse patients in clinical trials to better characterize population-specific factors influencing breast cancer outcomes. .
Presentation numberPS3-09-22
Er expression and age as key determinants of pathologic complete response (pcr) following neoadjuvant chemotherapy in patients with hormone receptor-positive, her2-negative early breast cancer. real world data
Faris Tamimi, King Hussein Cancer Center, Amman, Jordan
M. Horani, F. Tamimi, T. Al-Batsh, O. El Khatib, B. Sharaf, Y. Al-Masri, A. Ghanem, Q. Jawarneh, M. AL-yag’oub, A. Jaffal, R. Khader, S. Abdel-Razeq, A. Alanani, M. El-Atrash, M. Abunasser, H. Abdel-Razeq; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: Neoadjuvant chemotherapy remains a cornerstone in the treatment of patients with early-stage breast cancer and improves survival outcomes. Achievement of pathological complete response (pCR) have always been considered as a surrogate marker for better survival in patients with breast cancer and has been used as a primary endpoint in multiple clinical trials. However, patients with hormone receptor-positive (HR+) and human epidermal growth factor receptor 2 (HER2)-negative disease typically achieve low rates of pCR. Better understanding of the potential clinical and pathological variables affecting pCR in such population is important. Methods: This was a retrospective cohort analysis we conducted using electronic clinical and pathological data from patients diagnosed with early-stage breast cancer at one institution. All eligible patients included in the analysis had early HR+/HER2-negative disease: stage I, II, or III as per the American Joint Committee on Cancer (AJCC) 8th edition. Estrogen receptor (ER) expression by immunohistochemistry was stratified as: High (ER > 50%), intermediate (11-50%), low (1-10%) and negative (0%). Multivariable logistic regression was performed to identify independent predictors of pCR. We used multivariable logistic regression analysis to identify independent clinical and/or pathological predictors for pCR. Results: Out of 2724 patients included in the analysis; 1039 patients were treated with neoadjuvant chemotherapy. Median age was 50 (22-83) years. Most (n= 864, 83.1%) patients had invasive ductal carcinoma (IDC), while (n= 92, 8.8%) patients had invasive lobular carcinoma (ILC). Majority (n=870, 83.7%) patients had high ER-expression; 44 patients (4.2%) intermediate ER-expression, 59 patients (5.6%) low ER-expression, while 63 patients (6%) had ER-negative (but PR-positive) disease. 758 (72.9%) had clinical node-positive disease. The overall pCR rate in this population was 10.9% (n=114). Younger age was significantly associated with higher pCR rates: 15.0% in patients <45 years, compared to 8.7% in those aged 45-64 (adjusted OR: 0.52; 95% CI: 0.33-0.81; p=0.004), and 4.9% in patients ≥65 years. Histological subtype was not a significant predictor of pCR. Patients with ILC had a lower pCR rate (5.4%) compared to those with IDC (10.5%), but this difference was not statistically significant (adjusted OR: 0.90; 95% CI: 0.30-2.19; p=0.838). ER expression was a strong independent predictor of pCR. Compared to tumors with high ER expression (pCR rate: 6.4%), significantly higher pCR rates were observed in tumors with low ER expression (42.4%; OR: 10.35; p<0.001) and ER-negative/PR-positive tumors (39.7%; OR: 8.96; p<0.001). Tumors with intermediate ER expression also showed an increased pCR rate (18.2%; OR: 2.40; p=0.042). Clinical nodal status was not independently associated with pCR. Additionally, patients with HER2-zero tumors had higher pCR rates compared to those with HER2-low disease (IHC 1+ or 2+ with negative FISH), although this difference did not reach statistical significance. Conclusion: Our analysis showed consistent findings with previously published data worldwide informing a low rate of pCR in this population. This confirms that HR+ breast cancer carries a significant resistance to chemotherapy and validates ER-expression as an important biomarker for responsiveness to chemotherapy. Younger age and lower ER-expression are strong independent predictors of response. Such findings could influence tailored treatment approaches and the use of personalized therapeutics, as well as exploring different neoadjuvant strategies using targeted agents among others.
Presentation numberPS3-09-23
Neoadjuvant Aromatase Inhibitor Therapy Decreases Radiographic Tumor Size: Initial Results from the NAOMI Trial
Lillian Lawrence, Geisel School of Medicine at Dartmouth, Hanover, NH
L. Lawrence1, R. diFlorio-Alexander2, J. Marotti3, E. Demidenko4, M. Chamberlin5; 1School of Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, 2Department of Radiology, Dartmouth Hitchcock Medical Center, Lebanon, NH, 3Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, NH, 4Biostatistics, Geisel School of Medicine at Dartmouth, Hanover, NH, 5Department of Medical Oncology, Dartmouth Hitchcock Medical Center, Lebanon, NH.
Background: Nearly 70% of breast cancers are ER positive, and anti-estrogen therapy, such as tamoxifen and aromatase inhibitors (AI), are a mainstay of treatment. Neoadjuvant AI therapy is effective at reducing tumor burden, and may be used to improve surgical outcomes. Non-invasive assessment of response may be helpful in treatment planning, and radiographic treatment response has not been evaluated. Additionally, questions remain as to the social effects of neoadjuvant therapy. Preliminary data from this trial presented at Northern New England Clinical Oncology Society in 2024 showed an 85% adherence to aromatase inhibitor therapy at 1 year in patients who were treated in the neoadjuvant setting, with ongoing data collection, compared to 62% in the adjuvant setting, according to one study. Objective: Determine the ability of tumor imaging to detect the effect of neoadjuvant aromatase inhibitor (AI) therapy on tumor size. Methods: 52 post-menopausal women with Stage I-III ER+, HER2- breast cancer were treated with an aromatase inhibitor for 1-24 weeks (mean 6.7 weeks) prior to surgery and had pre-neoadjuvant imaging available for analysis. 12 patients did not have an imaging exam after neoadjuvant endocrine therapy and prior to surgery, or it was not possible to assess tumor size on pre-operative imaging, resulting in 40 patients for final analysis. Tumor measurements prior to neoadjuvant endocrine therapy were compared to tumor measurements after neo-endocrine therapy at the time of surgery using mammography or ultrasound to determine response to treatment. Post neo-endocrine treatment measurements were compared with pathologic tumor size. Results: Neoadjuvant aromatase inhibitor therapy decreased tumor size evaluated by imaging (mean pre-treatment size 19.4mm, mean post-treatment 14.8mm, p<.001), with size reduction in 68% of patients (AUC=.68). The larger the initial tumor size, the greater the reduction in tumor size (p=0.0017): on average, an additional 10mm increase in initial tumor size results in a 2mm decrease in post-treatment tumor size. The correlation coefficient between pathologic tumor size and post neoadjuvant AI therapy tumor size determined by specimen radiographs or localization imaging (with mammography or ultrasound) was 0.69. Discussion: AI therapy in the neoadjuvant setting could improve overall response to therapy and may improve long-term adherence. Response to therapy can be evaluated with mammography and ultrasound imaging obtained at the time of needle localization or specimen radiograph. Further work is needed to evaluate long-term outcomes with this treatment, and correlation with histology is ongoing. Due to the discordant results in lobular carcinomas, separate analysis of a large subset of lobular carcinomas is planned.
Presentation numberPS3-09-24
Impact of Competing Mortality on Survival Outcomes in Patients Aged ≥75 With HR+/HER2- Early Breast Cancer
Faris Tamimi, King Hussein Cancer Center, Amman, Jordan
F. Tamimi, M. Horani, T. Al-Batsh, O. El Khatib, B. Sharaf, Y. Al-Masri, A. Ghanem, Q. Jawarneh, M. AL-yag’oub, A. Jaffal, R. Khader, S. Abdel-Razeq, A. Alanani, M. El-Atrash, M. Abunasser, H. Abdel-Razeq; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: In older adults with early-stage breast cancer, clinical decisions must weigh the benefits of systemic therapy against age-related comorbidities and competing causes of death. We assessed age-stratified treatment patterns and outcomes in a Jordanian cohort, where national life expectancy approximates 75 years. Methods: We retrospectively analyzed 716 women aged ≥60 years with stage I-III hormone receptor-positive, HER2-negative early breast cancer treated at King Hussein Cancer Center (2015-2021). Patients were grouped as follows: 328 (45.8%) aged 60-65 years, 296 (41.3%) aged 66-74 years, and 92 (12.8%) aged ≥75 years. We compared tumor features, systemic therapy use, and survival. Kaplan-Meier methods estimated overall survival (OS) and breast cancer-specific survival (BCSS). Causes of death were classified as breast cancer-related or non-cancer-related. Results: Tumor characteristics were comparable across age groups. Most patients (91.9%) had ER-high tumors, and grade 2 histology predominated (62.6%). Clinical T and N stages, including cT2 tumors (52.5%) and cN+ nodal status (39.1%), were similarly distributed. Use of chemotherapy decreased significantly with age: 70.7% in those aged 60-65, 50.0% in 66-74, and 10.9% in ≥75 (P 97%), with aromatase inhibitors used in 79.4% and tamoxifen in 17.9%. Among patients receiving endocrine therapy, 51.8% completed or exceeded 5 years of treatment, while 14.4% relapsed before 5 years. The median follow-up was 66.7 months. Five-year BCSS remained high across age groups, at 96.2%, 94.2%, and 91.8% for patients aged 60-65, 66-74, and ≥75 years, respectively, with no significant difference (P = .40). However, 5-year OS declined significantly with age: 91.1%, 88.8%, and 78.0%, respectively (P = .0012). Breast cancer-related mortality was relatively stable across age groups, occurring in 7.6%, 9.1%, and 9.8% of patients aged 60-65, 66-74, and ≥75 years, respectively. while non-cancer-related mortality significantly increased: 6.1%, 10.1%, and 18.5% (P = .005), contributing substantially to the decline in OS among the oldest patients. There was a statistically significant difference in the distribution of causes of death across age groups (P = .005). Non-breast cancer-related mortality increased notably with age, occurring in 6.1%, 10.1%, and 18.5% of patients aged 60-65, 66-74, and ≥75 years, respectively, accounting for 67 deaths (9.4%) overall. In comparison, breast cancer-related deaths were relatively stable across age groups, occurring in 7.6%, 9.1%, and 9.8% of patients in the same age categories, totaling 61 deaths (8.5%) in the cohort. Conclusions: In our cohort, the decline in overall survival among patients aged ≥75 years was primarily driven by non-cancer-related mortality, rather than deaths from breast cancer. Despite this, breast cancer-specific survival remained excellent in this age group, even with markedly reduced use of chemotherapy. These findings support age-tailored treatment strategies that allow for safe de-escalation of systemic therapy while preserving oncologic outcomes, with careful consideration of national life expectancy in treatment planning.
Presentation numberPS3-09-25
Factors Associated With Real-World Use of Ovarian Function Suppression Versus Tamoxifen Alone in Premenopausal Women With Early-Stage HR+/HER2- Breast Cancer
Poornima Saha, Endeavor Health, Evanston, IL
P. Saha1, C. Sanchez2, O. Israel3, L. Eldridge4, K. Kuchta5, K. A. Yao5; 1Oncology, Endeavor Health, Evanston, IL, 2Medicine, Endeavor Health, Evanston, IL, 3Medicine, University of Michigan, Ann Arbor, MI, 4Research Institute, Endeavor Health, Evanston, IL, 5Surgery, Endeavor Health, Evanston, IL.
BackgroundApproximately 16% of new breast cancer diagnoses occur in premenopausal women. Young women with estrogen receptor-positive (ER+) human epidermal growth factor receptor 2-negative (HER2–) breast cancer may be considered for adjuvant endocrine therapy (ET) with tamoxifen (Tam) alone or with ovarian function suppression (OFS) plus Tam or aromatase inhibitor (AI). Optimal adjuvant therapy for this population is of significant interest because young age at diagnosis is associated with a worse prognosis. The SOFT and TEXT trials have demonstrated a benefit of OFS compared to Tam alone in premenopausal women with high risk. Use of OFS, however, may be associated with increased toxicity and may be under-utilized in clinical practice. This study evaluated the real-world use of OFS in premenopausal women aged <50 in a large multidisciplinary community breast oncology center. The goal of this study was to investigate factors associated with the use of Tam alone, and to evaluate treatment discontinuation and recurrence rates in women who received Tam alone versus OFS. MethodsWe analyzed the medical records of premenopausal women (defined as age < 50) with Stage 1-3 HR+/HER2– breast cancer from January 2012 to December 2023. We collected demographic, clinicopathologic characteristics, and treatment information. Patients with non-invasive breast cancer, those who did not begin ET, and those who received an aromatase inhibitor (AI) alone were excluded. ResultsA total of 874 women were diagnosed with early-stage breast cancer at age < 50 at our institution between 2012 and 2023. Of these, 723 met eligibility criteria. Median age was 44. The majority were Caucasian (77%), followed by Asian Pacific Islander (10%), Hispanic (5%), Black (5%), and Unknown (3%). 396 (54.8%) underwent mastectomy. 468 (64.7%) received radiation. A genomic assay was used in 49.0% of women. Chemotherapy was administered to 302 (41.8%) women. Adjuvant ET was administered in all women, with 185 patients treated with OFS and 538 treated with Tam alone. When a GnRH agonist was used for OFS, goserelin was used in the majority. Estradiol monitoring on OFS was inconsistently performed but increased in more recent years. A bilateral salpingo-oophorectomy (BSO) was completed in 88 women during their treatment course. Those treated with OFS were more likely to have tumors >3.0 cm, lymph node-positive disease, higher grade tumors, and to have received chemotherapy. A multivariable logistic regression analysis revealed that age at diagnosis and grade were factors significantly associated with the choice of Tam alone versus OFS for adjuvant therapy. ET was discontinued early due to toxicity in 14.5% and 20 patients discontinued ET due to recurrence (16 in the Tam alone group and 4 in the OFS group). At a median follow-up of 66 months, a total of 64 breast cancer recurrences had occurred, 13 in the OFS group and 51 in the Tam alone arm. Recurrence was not statistically different between the OFS and Tam groups when controlling for age, BMI, grade, and stage (HR 0.12 [95% CI: 0.60–2.09], p=0.7237). ConclusionData from the SOFT and TEXT trials support consideration of maximal hormone suppression with OFS in premenopausal women. Our study highlights the likely underutilization of OFS, even in this high-risk population in the community setting, likely due to increased concern for toxicity. However, these concerns are not supported by the increased rate of early discontinuation in the OFS group. Although not statistically significant, a higher number of recurrences were noted in the tamoxifen alone group. Additional research is warranted to optimize the selection of patients who might benefit most from maximal hormonal blockade with OFS compared to Tam alone.
Presentation numberPS3-09-26
Real-world clinical characteristics, treatment patterns and outcomes among UK and French patients with early-stage, estrogen receptor-positive, human epidermal growth factor receptor 2-negative (ER+/HER2-) breast cancer treated with neoadjuvant chemotherapy
Paul Cottu, Institut Curie AND Université Paris Cité, Paris, France
P. S. Hall1, E. Sawyer2, J. Frenel3, K. Desai4, K. M. Hirshfield5, G. Gooud6, D. Ntais6, A. Jamotte7, C. Perkins8, A. Babonneau9, E. Kitetere10, M. Vallet1, M. Riaz11, J. Timbres12, L. Haroun13, L. Cirneanu14, R. Hermans14, V. Saglimbene15, K. Dushkin14, A. Ajmal14, S. Oikonomou16, A. Vladimirova16, J. Earla4, F. Bocquet17, P. Cottu18; 1Genetics and Cancer, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UNITED KINGDOM, 2Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Kings College London, Guys Cancer Centre, London, UNITED KINGDOM, 3Medical Oncology, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 4V&I Outcomes Research Oncology, Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, 5Oncology Clinical Development, Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, 6HTA & Outcomes Research, MSD UK, London, UNITED KINGDOM, 7HEDS Oncology, MSD Innovation & Development GmbH, Zurich, SWITZERLAND, 8Medical Oncology, MSD France, Puteaux, UNITED KINGDOM, 9HEOR, MSD France, Puteaux, FRANCE, 10Oncology Medical Affairs, MSD UK, London, UNITED KINGDOM, 11Data Department, Leeds Teaching Hospitals NHS Trust, Leeds, UNITED KINGDOM, 12Breast Cancer Genetics, King’s College London, London, UNITED KINGDOM, 13Data Department, Institut Curie, Paris, FRANCE, 14Real World Solutions, IQVIA, London, UNITED KINGDOM, 15Real World Solutions, IQVIA, Milan, ITALY, 16Real World Solutions, IQVIA, Sofia, BULGARIA, 17Data Factory and Analytics Department, Institut de Cancérologie de l’Ouest, Saint-Herblain, FRANCE, 18Medical Oncology, Institut Curie AND Université Paris Cité, Paris, FRANCE.
Background: ER+/HER2-breast cancer (BC) is heterogeneous, with a subset of patients at high risk of recurrence and poorer outcomes. The treatment landscape for the high-risk group is evolving, with growing emphasis on survival benefits from adjuvant and neoadjuvant treatments. This study aimed to describe the real-world characteristics, treatment patterns, and outcomes for high-risk patients who received neoadjuvant chemotherapy (NACT). Methods: This was an observational, retrospective cohort study using electronic health records from five oncology centers across France and the UK. The study included patients with high-risk (grade III and clinical stage T1c-T2/N1-N2 or T3-T4/N0-N2), early-stage, ER+/HER2-BC initiating NACT (index date) between Jan 2013 and Dec 2020 (Jan 2014-Dec 2020 at one site). Patients participating in clinical trials were included. Demographics, prescribed treatments, and real-world pathological complete response (rwpCR) were detailed using descriptive statistics. Results: The study included 281 patients, 167 French (59.4%) and 114 British (40.6%). Patients were diagnosed with T1c-T2 and N1-N2 (157, 55.9%) or T3-T4 and N0-N2 (124, 44.1%) BC (including American Joint Committee on Cancer, version 7 stages II, IIA and IIB). At diagnosis, 235 patients (83.7%) were ER-positive (≥10% expression) and 84 (49.7%) of 169 patients with known status were post-menopausal women. Only 58 patients (20.6%) were tested for BRCA, with 11 (19.0%) positive. PD-L1 and prognostic genomic tests were rarely conducted. Of 229 patients with ECOG score at index, 207 (90.4%) had a 0 score. Prescribed agents are presented in the Table: 263 patients (93.6%) received NACT alone (i.e., without radiotherapy or other treatments). The most common (271 patients, 96.4%) NACT agents were anthracycline (A) with cyclophosphamide (C), mostly combined with a taxane (T; 257 patients, 91.5%). The most common reasons for NACT initiation (available for 92 patients) were clinical node involvement (46, 50.0%), large unifocal primary tumor (30, 32.6%), or multifocal disease (15, 16.3%). The median (Q1, Q3) duration of NACT was 4.4 (3.5, 4.8) months. At the time of surgery, rwpCR (ypT0/Tis ypN0) was achieved by 29 patients (10.3%). Overall, 221 patients (78.7%) were alive at the time of data provision (median follow up: 68.8 months). Conclusions: These initial results provide insights into the real-world characteristics and prescribed treatments of patients with stage II- III ER+/HER2- early BC initiating NACT. Treatment was primarily based on A, C and T. The recorded low rwpCR confirms the high unmet need for newer therapies in this setting. Further analyses will focus on treatment sequencing and real-world effectiveness.
| Treatment | Category/class |
Number (N=281) |
% |
| Neoadjuvant regimens | NACT only | 263 | 93.6 |
| NACT + RT | 6 | 2.1 | |
| NACT + Other | 12 | 4.3 | |
| Neoadjuvant agents received | AC + T | 154 | 54.8 |
| AC + T + Othera | 103 | 36.7 | |
| AC | 11 | 3.9 | |
| AC + Non-T Othera | 3 | 1.1 | |
| Other combinationsa | 10 | 3.6 | |
|
A, anthracyclines; C, cyclophosphamide; OFS, ovarian function suppression; NACT, neoadjuvant chemotherapy; RT, radiotherapy; T, taxanes. aTwo patients received OFS. |
Presentation numberPS3-09-28
Clinical investigations into the metabolic pathways of early stage hormone receptor positive/HER2 negative breast cancer.
Coral Omene, Rutgers Cancer Institute, New Brunswick, NJ
C. Omene1, S. Kumar2, M. George1, L. Potdevin2, J. Smith2, A. Shah3, K. Matsuda4, D. Toppmeyer1, M. Gupta5, K. Olszewski5, E. White6, J. Rabinowitz5; 1Division of Medical Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 2Division of Surgical Oncology, Rutgers Cancer Institute, New Brunswick, NJ, 3Division of Endocrinology, Rutgers RWJ Medical School, New Brunswick, NJ, 4Division of Pathology, Rutgers RWJ Medical School, New Brunswick, NJ, 5Ludwig Princeton Branch, Princeton University, Princeton, NJ, 6Molecular Biology and Biochemistry, Rutgers Cancer Institute, New Brunswick, NJ.
Background: One of the recognized hallmarks of cancer cells is deregulated cellular metabolism, characterized by enhanced metabolic autonomy compared with non-transformed cells. Tumor cells display an overall increase in glucose metabolism, enhanced aerobic glycolysis and decreased oxidative phosphorylation, accompanied by a requirement for a high rate of protein, nucleotide, and fatty acid synthesis to provide the raw materials for cell division. Approximately 70-80% of all breast cancers (BC) are hormone receptor positive (HR+), and most HR+ breast cancers are HER2-negative (Her2-). The metabolic dependencies of HR+/Her2- BC and in particular, high-risk HR+ BC, are poorly understood. In depth analyses of in vivo metabolic processes by HR+ BC using carbon-13 labeled glucose (13C-glucose) infusions have never been done in humans. 13C-glucose is a stable isotope tracer that has been widely used in vitro, in vivo, and in patients in a variety of disease settings to study glucose, amino acid, and lipid metabolism. We hypothesize that HR+ breast cancers in vivo have altered use of metabolic pathways that can be determined by in vivo 13C isotope glucose labelling. The primary objective is to describe and discover new insights into the glucose, TCA cycle, amino acid, and lipid metabolic dependencies of HR+/Her2- BC, via liquid chromatography-mass spectrometry (LC-MS) and matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry analysis of in vivo U-13C-glucose-labeled biopsy of tumor and benign adjacent tissue. Methods: This is an ongoing pilot study that will enroll 16 patients with early stage I, II or III HR+/Her2- breast cancer who do not require neoadjuvant therapy and have planned curative primary resection. They will receive intraoperative 13C-glucose intravenous infusions followed by core biopsies of tumor and normal breast tissue and blood will be collected at time of resection. Tumor samples will be analyzed for metabolite labeling by LC-MS and spatial distribution of metabolite levels and labeling patterns by MALDI-MS. Genomic and proteomic analyses will be performed and related to observed metabolic fluxes. Demographic and clinicopathological information will be collected. ClinicalTrials.gov ID NCT05736367. Results: Preliminary results from LC-MS of tumors from the first four patients show avid glucose metabolism in HR+/Her2- BC. These tumors locally produce lactate from glucose, with higher 13C-lactate labeling observed in the tumor than in circulation. The contribution of glucose-derived lactate to the TCA cycle is approximately 30%, which is similar to what has previously been observed in hormone receptor-negative (HR-) BC. Interestingly, there was little to no labeling of the amino acid serine, demonstrating minimal activity of the de novo serine synthesis pathway in these tumors. This contrasts with previous results demonstrating extensive serine synthesis in HR-/Her2- BC and a common genomic amplification of the rate-limiting enzyme PHGDH in HR-/Her2- BC. This trial is ongoing, and further results will be presented at a future date. Conclusion: This study has the potential to discover new insights into the glucose, TCA cycle, amino acid, and lipid metabolic dependencies of HR+/Her2- breast cancer and will allow these critical pathways to be resolved spatially within the tumor, allowing analysis such as the effects of distance from blood vessels and of specific cell subpopulations. Grant Support: Ludwig Institute for Cancer Research, Ludwig Princeton Branch.
Presentation numberPS3-09-29
Comparing Functional Fibroblasts in Tumor vs. Adjacent Normal Tissue in Young Hormone Receptor-Positive Breast Cancer
Jiayi Tan, SUNY Upstate Medical University, Syracuse, NY
J. Tan1, H. Hackbart2, Y. Li3, N. Bercovici2, X. Cui4, Y. Yuan2, J. Bitar2; 1Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, 2Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 3Spatial Molecular Profiling Core, Cedars-Sinai Medical Center, Los Angeles, CA, 4Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA.
Background: Breast cancer incidence among young women has risen markedly over the past decade, particularly hormone receptor-positive (HR+) breast cancer (BC), yet the tumor microenvironment (TME) in this young population remains poorly characterized. Cancer-associated fibroblasts (CAFs) are abundant stromal cells that contribute to extracellular matrix remodeling, cytokine signaling, and immune regulation; their persistent activation and phenotype shift support tumor growth and treatment resistance. To delineate fibroblast heterogeneity and functional alterations in young HR+BC, we conducted spatially resolved single-cell analyses on matched tumor and adjacent normal tissue using image mass cytometry (IMC). Methods: We collected formalin-fixed, paraffin-embedded (FFPE) specimens from ten treatment-naïve, stage I-II HR+ BC patients diagnosed at age ≤40. Patients with germline pathogenic mutations such as BRCA1/2 and PALB2 were excluded. From each case, two regions of interest (ROIs)—one within the invasive tumor and one from adjacent normal tissue—were selected and imaged at 1 μm resolution using a 40-marker IMC panel targeting hormone receptors, stromal components, adhesion/migration, and immune populations. Key markers were used to identify different phenotypes of fibroblasts. Image preprocessing and cell segmentation were performed with imctools and DeepCell; single-cell feature extraction, clustering, and differential expression analyses employed Scanpy, while spatial neighborhood and interaction analyses utilized Squidpy. Results: Across 20 ROIs, we identified 109,097 individual cells, including 71,406 from tumor ROIs and 37,691 from normal ROIs. Cluster annotation was based on differential marker expression to identify cancer, stromal, and immune populations, and 5 distinct fibroblast subsets were identified. When comparing fibroblast phenotypes between tumor and normal ROIs, Ki67⁺PD-L1⁺ and MHCII⁺ fibroblasts in tumor tissue exhibited more active functional states, as indicated by higher expression of FAP, αSMA, and increased collagen secretion. Additionally, hormone receptors such as ERα and ERβ, were significantly upregulated in tumor-associated fibroblasts compared to those in normal tissue. MHCII⁺ fibroblasts in tumors also displayed a more pro-tumorigenic phenotype, with elevated expression of MMP9 and CXCL12. Spatial analysis revealed that both Ki67⁺PD-L1⁺ and MHCII⁺ fibroblasts were actively engaged in cell-cell interactions with T cells and epithelial/cancer cells. Specifically, Ki67⁺PD-L1⁺ fibroblasts showed increased interactions with CD8⁺ T cells in tumor ROIs, while MHCII⁺ fibroblasts were more involved with epithelial cells in normal tissue. Conclusions: Our study uncovers functionally activated fibroblast subsets in young HR+ BC compared to adjacent normal tissues. The distinct fibroblast phenotypes and their spatial interactions with immune and epithelial cells highlight stromal PD-L1 and MHCII pathways as potential therapeutic targets. Future studies with larger sample size, including older HR+ cases, are required to establish the age-associated specificity of these stromal alterations.
Presentation numberPS3-09-30
Real-World Outcomes of Neoadjuvant Chemotherapy vs Endocrine Therapy in ER+/HER2− and HER2-Low Early Breast Cancer: A Single-Institution Retrospective Study
Abdullah A Almazyad, King Fahad Medical City, Riyadh, Saudi Arabia
A. A. Almazyad, M. Rudainee, M. H. Alharbi, A. A. Alwohaibi, N. Algazlan, A. R. altamimi, F. H. Faqihi, A. K. Altwairgi; Medical Oncology, King Fahad Medical City, Riyadh, SAUDI ARABIA.
Real-World Outcomes of Neoadjuvant Chemotherapy vs Endocrine Therapy in ER+/HER2− and HER2-Low Early Breast Cancer: A Single-Institution Retrospective Study Abdullah A Almazyad, Abdullah A Alwohaibi, Mojahed Rudainee, Marwa M Alharbi, Abdulaziz R Altamimi, Najd Algazlan, Fatima T Faqihi, Abdullah K Altwairgi. BackgroundNeoadjuvant chemotherapy (NAC) and endocrine therapy (NET) are both used in ER+/HER2− or HER2-low breast cancer, yet comparative real-world data remain limited. This study aimed to evaluate treatment patterns, pathological response, and clinical outcomes between NAC and NET in this setting. MethodsWe retrospectively analyzed 211 ER+/HER2− or HER2-low early breast cancer patients who received neoadjuvant systemic therapy at King Fahad Medical City. Patients were grouped by treatment type: NAC (n=169) and NET (n=42). Luminal subtypes were defined by histologic grade and Ki-67 index. Key outcomes included pathologic complete response (pCR; defined as ypT0/is and ypN0), nodal and tumor downstaging, breast-conserving surgery (BCS), and recurrence-free survival (RFS). ResultsThe NAC and NET groups represented distinct clinical populations. Patients receiving NET were older (median 66.5 vs 47.0 years), more often postmenopausal (90.5% vs 50.9%), and had a higher burden of comorbidities and ECOG 2+ status. NAC was more commonly used in patients with Grade 3 tumors, high Ki-67, and Luminal B subtype. pCR was observed in 9.5% of NAC and 4.8% of NET patients. Nodal downstaging occurred in 16.6% of NAC patients and 7.1% of NET patients. Tumor downstaging was observed in 68.3% of NAC patients and 74.2% of NET patients who underwent surgery. BCS was achieved in 17.8% of NAC patients and 9.5% of NET patients. Distant recurrence was reported in 6.5% of NAC and 2.4% of NET patients. Among those who recurred, the median RFS was 20.6 months for NAC and 13.3 months for NET. ConclusionsIn this real-world study, NAC was favored in younger, fitter patients with aggressive tumor features, while NET was reserved for older patients or those with more favorable disease biology. pCR and nodal response rates were higher in the NAC group, but treatment selection reflected inherent baseline differences. Tumor downstaging and low recurrence rates were observed with both strategies.
Presentation numberPS3-10-01
Impact of ER Reduction Rate on Survival Outcomes in ER-Positive/HER2-Negative Breast Cancer Post-Neoadjuvant Chemotherapy
Shunyi Liu, Fujian Medical University Union Hospital, Fuzhou, China
S. Liu1, Y. Qiu1, Y. Wang1, W. Cai1, H. Peng1, Q. Cai1, L. Wu1, J. Li2, L. Yu1, M. Chen1, L. Chen1, C. Wang1, F. Fu1; 1Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA, 2Department of Breast Surgery, Zhongshan Hospital Xiamen University, Xiamen, CHINA.
Background: The conversion of estrogen receptor (ER)-positive to ER-negative status after neoadjuvant chemotherapy (NAC) is often considered a poor prognostic indicator in ER-positive/HER2-negative breast cancer (BC). However, the prognostic impact of the extent of ER reduction remains unclear.Methods: This study included ER-positive/HER2-negative BC patients without pathological complete response after NAC. Cox regression assessed the relationship between ER reduction rate (ERr) and invasive disease-free survival (iDFS), distant disease-free survival (DDFS), and overall survival (OS). Time-dependent receiver operating characteristic curves (ROC) and concordance index compared model discrimination.Results: A total of 519 patients were followed up for a median of 81 months. Participants were categorized into two groups based on the restricted cubic spline analysis, using a cut-off value of 10%. A total of 415 patients (79.96%) had ERr < 10%, while 104 patients (20.04%) had ERr ≥ 10%. In the multivariate Cox regression model, participants with ERr ≥ 10% had a significantly higher risk of iDFS (hazard ratio [HR], 2.405; 95% confidence interval [CI], 1.703–3.395), DDFS (HR, 2.278; 95% CI, 1.575–3.294), and OS (HR, 2.224; 95% CI, 1.450–3.411) compared to those with ERr < 10%. Multiple sensitivity analyses yielded consistent results. Based on time-dependent ROC, adding the ERr to the prognostic model significantly improved predictions of iDFS (p = 0.025) and DDFS (p = 0.044).Conclusions: ERr ≥ 10% is associated with poorer outcomes in ER-positive/HER2-negative BC patients treated with NAC, highlighting the need for novel therapeutic strategies for this population.
Presentation numberPS3-10-02
Factors influencing the number of adjuvant TC – docetaxel and cyclophosphamide – cycles in hormone receptor-positive early-stage breast cancer
Wesley Antônio Lopes de Lima, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil, São Paulo, Brazil
W. A. Lima1, R. Naves2, R. C. Bonadio2, B. Pego1, L. Amorim1, G. J. Filheiro1, C. Cavalcanti2, L. Holland2, J. Bessa2, P. H. Divino2, M. Nishimuni2, L. G. Torres2, J. Bines2, L. Testa2; 1Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil, São Paulo, BRAZIL, 2Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL.
Introduction Docetaxel and cyclophosphamide (TC) is a widely used anthracycline-free regimen for hormone receptor-positive (HR+) early-stage breast cancer. While typically administered over four to six cycles, the optimal number of cycles for various risk profiles remains a topic of clinical debate. This study aimed to identify clinical and pathological factors associated with the number of adjuvant TC cycles prescribed in a real-world setting. Methods We conducted a retrospective analysis of patients with HR+ early-stage breast cancer treated between 2018 and 2025 across a multicenter oncology network in Brazil. Eligible patients had undergone surgery and received adjuvant TC chemotherapy. Variables such as age, menopausal status, tumor size and nodal status, histologic grade, and Ki-67 index were examined in relation to the number of TC cycles administered (six versus four). For patients who received four cycles, we recorded whether this was based on the original treatment plan, early discontinuation due to adverse events, or other reasons. Logistic regression was used to evaluate associations between baseline characteristics and the likelihood of receiving fewer versus more TC cycles. Results A total of 372 patients who received adjuvant TC were evaluated; 206 (55.4%) received four cycles and 166 (44.6%) received six cycles. Among those receiving four cycles, the majority (87.9%) had this number planned at the outset. Only four patients (1.9%) had the number of cycles reduced due to treatment-related toxicities. The reason for choosing four cycles was not documented in the remaining 10.2%. Patients who received six cycles were slightly younger (median age 50 years [range 27-81]) than those receiving four cycles (median age 52 years [range 29-83]; P=0.025). The six-cycle group had a higher proportion of patients with positive lymph nodes (42.2% pN1 and 3.6% pN2) compared to the four-cycle group (19.9% pN1 and 0.9% pN2; P<0.001), and a greater proportion of grade 3 tumors (34.3% vs. 28.6%; P=0.048). No significant differences were observed in primary tumor size, menopausal status, or Ki-67 index. In multivariable logistic regression, lymph node status, tumor grade, and age were significantly associated with receiving six TC cycles (Table). Increasing age was associated with a lower likelihood of receiving six cycles, while pN1, pN2, and grade 3 tumors were associated with higher odds. Conclusion These findings suggest that treatment duration with TC is influenced not only by formal staging criteria but also by individualized clinical judgment reflecting perceived recurrence risk and patient tolerance. Further studies are needed to determine whether six versus four cycles of TC differ in efficacy when compared in cohorts balanced for these baseline factors.
| Variable | OR | 95% CI | P-value | ||||
| Age (continuous) | 0.96 | 0.94 – 0.98 | 0.001 | ||||
| pN0 (reference) | – | – | – | ||||
| pN1 | 3.35 | 2.01 – 5.58 | < 0.001 | ||||
| pN2 | 9.06 | 1.54 – 53.27 | 0.015 | ||||
| Grade 1-2 (reference) | – | – | – | ||||
| Grade 3 | 1.76 | 1.07 – 2.89 | 0.024 |
Presentation numberPS3-10-03
Genomic Platform (GP) Testing and Adjuvant Chemotherapy (CT) Use in Early-Stage hormone receptor-positive (HR+) /HER2 negative (HER2-) Breast Cancer (BC): Real-World Insights and Prognostic Factors
Rocio Urbano, Hospital Universitario de Jaén, Jaen, Spain
R. Urbano, R. García, A. Beltran, M. Martin-Salvago, A. Cano, A. Jaén, P. Sánchez-Rovira; Medical Oncology, Hospital Universitario de Jaén, Jaen, SPAIN.
BACKGROUND Genomics platform (GP) has transformed the management of patients with early stage, HR+, HER2 negative (HER2-) BC by guiding personalised treatment decisions regarding adjuvant CT. However, real world patterns of CT used base of recurrence score (RS) remain a topic interest. Using our hospital-based database we evaluated testing patterns, factors associated with GP testing and examined predictor of CT use among patients by RS. METHODS This retrospective observational study included patients ≥ 18 years with early-stage, HR+/HER2- BC diagnosed from 2021-2024 who underwent surgery with pT1-T3, pN0-N1 staging and meet clinical guideline criteria for requesting a genomic study were included. Descriptive analyses were used, including demographic and clinical characteristics of patients. Multivariable logistic regression was used to examine factors associated with receipt of GP testing and predictors of CT use, stratified by Low risk, intermediate risk and high risk RESULTS A total of 196 patients were included, with a median age of 60 years. The majority had stage IA (54%), grade 2 (79%) tumors, and infiltrating ductal histology (84.7%), with 36% presenting Ki67 ≥20%. Twenty-six percent were premenopausal, and 27% had axillary involvement. Oncotype DX was the most commonly used genomic platform (81%) followed by MammaPrint (17.1%) and Prosigna (1.7%). Risk stratification showed 72% low risk, 12% intermediate risk, and 15% high risk. All high-risk patients, except one, received adjuvant chemotherapy, while no intermediate-risk patients received chemotherapy. With a median follow-up of 34 months, only two local recurrences were observed, both in low-risk patients. Low progesterone levels, Ki67 >20%, and premenopausal status were significantly associated with high risk (p20%, and premenopausal status with high risk underscores their role as prognostic factors, consistent with prior literature. The trend toward significance for grade 3 tumors (p=0.057) and the higher proportion of high-risk patients among HER2 1+/2+ cases (20% vs. 4% in HER2 0, p=0.1) warrant further investigation, as these may indicate subgroups with distinct biological behavior. Limitations include the single-center design and relatively short follow-up, which may limit generalizability and detection of late recurrences. Larger, multicenter studies with longer follow-up are needed to validate these findings and refine the role of HER2 status in risk stratification.
Presentation numberPS3-10-04
Characteristics of patients with HR+/HER2− early breast cancer eligible for adjuvant treatment with iCDK4/6: real-life evidence in a Colombian cancer center.
Alfredo Sebastian Golemba, Centro Cancerologico del Caribe LTDA, Barranquilla, Colombia
A. S. Golemba1, S. P. Rodriguez Torres1, R. F. Vargas Moranth1, P. Mandó2, A. A. Aguirre Santamaria1; 1Atlantico, Centro Cancerologico del Caribe LTDA, Barranquilla, COLOMBIA, 2Unidad de Tumores Mamarios y Ginecologicos, CEMIC, CABA, ARGENTINA.
Background: The introduction of CDK4/6 inhibitors (iCDK4/6) in the adjuvant setting for hormone receptor-positive (HR+)/HER2-negative (HER2-) early breast cancer has expanded treatment options for high-risk patients. However, real-world data on eligibility for these therapies in Latin American populations remain limited. Methods: This retrospective study analyzed 334 patients with HR+/HER2- early breast cancer (stages I-III) treated at a Colombian cancer center (2022-2024). Eligibility criteria for adjuvant abemaciclib (monarch-E) and ribociclib (NATALEE) were applied. Clinicopathological characteristics and treatment patterns were assessed. Results: Among abemaciclib-eligible patients (36.2%, n=121), 54.5% had ≥4 positive nodes and 45.5% met alternative high-risk criteria (1-3 nodes plus tumor size ≥5 cm or grade 3 or Ki67 ≥20%). Ribociclib-eligible patients (59%, n=197) included 53.8% with stage III disease and 12.7% with node-negative, grade 3 tumors. Compared to pivotal trials, our cohort had older median age (61 vs 51-58 years) and lower prevalence of grade 3 tumors (17.1% vs 30-40%). Conclusion: A substantial proportion of Colombian patients with early HR+/HER2- breast cancer qualify for adjuvant CDK4/6i, particularly those with nodal involvement. These findings highlight both the clinical opportunity and economic challenges of implementing these therapies in resource-limited settings, while underscoring the need for improved early detection to reduce advanced presentations.
Presentation numberPS3-10-05
Indications and Outcomes of Anthracycline-Based Regimens for Early-Stage Grade 3 Hormone Receptor-Positive Breast Cancer in a Real-World Scenario
Luiz Gustavo Torres, Instituto D’Or de Pesquisa e Ensino, São Paulo, Brazil
L. G. Torres1, J. Bines1, L. Testa1, D. Negrini1, V. Petry1, A. C. Baptista1, C. R. Victor1, W. A. Lima2, R. Naves1, C. A. Cavalcanti1, L. Holland1, P. H. Divino1, M. Nogueira1, J. d. Bessa1, R. Colombo Bonadio1; 1Oncology, Instituto D’Or de Pesquisa e Ensino, São Paulo, BRAZIL, 2Oncology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil, São Paulo, BRAZIL.
Background: In hormone receptor-positive (HR+) breast cancer, anthracycline-free chemotherapy regimens—most commonly the TC regimen (docetaxel and cyclophosphamide)—have been widely adopted, particularly for early-stage, node-negative disease. However, certain HR+ patients, such as those with higher genomic risk, may still benefit from anthracycline-based chemotherapy. This study aimed to evaluate real-world treatment practices in an intermediate- to high-risk population, specifically patients with grade 3 tumors. Methods: We retrospectively reviewed records of patients with early-stage HR+ grade 3 breast cancer treated between 2018 and 2025 across a Brazilian cancer network. Eligible patients had received systemic therapy (neoadjuvant or adjuvant) for early-stage disease. Patients were categorized by chemotherapy regimen into anthracycline-based and anthracycline-free groups. We used logistic regression to assess the odds of receiving an anthracycline-based regimen based on clinical and demographic features. Event-free survival (EFS) was estimated using the Kaplan-Meier method, and factors associated with EFS were evaluated using Cox regression. Results: A total of 215 patients were included; 92 (42.8%) received anthracycline-based and 123 (57.2%) received anthracycline-free chemotherapy. Patients were more likely to receive anthracycline-based regimens if they were younger, premenopausal, had higher T or N stage, or had a Ki-67 index ≥ 20%. The distribution of these characteristics and their associated odds ratios (ORs) are shown in the Table (reference categories: age ≥ 50 years, postmenopausal, T1, N0, Ki-67 < 20%). Nearly half of the anthracycline group (46.7%) received the regimen as neoadjuvant therapy, whereas almost all patients in the anthracycline-free group (97.6%) were treated in the adjuvant setting. With a median follow-up of 23 months, 15 patients experienced disease recurrence or death. The 2-year EFS was 97.1% in the anthracycline group and 97.4% in the anthracycline-free group. In multivariable Cox regression adjusting for age and stage, there was no significant difference in EFS between the groups (HR 1.51, 95% CI 0.44-5.16; P = 0.508). Discussion: In this real-world cohort, anthracycline-based chemotherapy was more frequently used in patients with higher-risk features, including younger age and more advanced stage. No difference in short-term EFS was observed between patients who received anthracycline and those who did not, although the analysis is limited by the small sample size, low event rate, and relatively short follow-up. We plan to expand the cohort and extend follow-up to further investigate this question.
| Anthracyclin-based CT (%) | Anthracyclin-free CT (%) | OR | 95% CI | P-value | |||||||||||||||||||
| Age < 50 years | 55.4% | 38.2% | 1.98 | 1.14-3.43 | 0.014 | ||||||||||||||||||
| Premenopause | 55.4% | 43.9% | 1.83 | 1.03-3.23 | 0.037 | ||||||||||||||||||
| T2 | 48.9% | 45.5% | 2.94 | 1.47-5.89 | 0.002 | ||||||||||||||||||
| T3-4 | 25% | 2.4% | 28.11 | 7.42-106.47 | < 0.001 | ||||||||||||||||||
| N1 | 39.1% | 13.8% | 6.42 | 3.17-12.99 | < 0.001 | ||||||||||||||||||
| N2-3 | 13.0% | 1.6% | 18.19 | 3.85-85.80 | < 0.001 | ||||||||||||||||||
| Ki67 ≥ 20% | 85.9% | 78.0% | 4.32 | 1.42-13.10 | 0.010 |
Presentation numberPS3-10-06
Comparison between the LRP model and Adjuvant! Online in predicting 9-year distant disease-free survival
Ling Xu, Peking University First Hospital, Beijing, China
M. Ding1, H. Zhang2, Q. Wu3, Y. Cheng1, L. Xu1; 1Thyroid and Breast Surgery, Peking University First Hospital, Beijing, CHINA, 2Department of Pathology, 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 Interventional Medicine, China-Japan Friendship Hospital, Beijing, CHINA.
Background: MammaTyper® is a RT-qPCR kit that quantitatively detects the mRNA level of the four genes ESR1, PGR, ERBB2, and MKI67 in breast cancer FFPE tissue. The MammaTyper® Low Risk Prediction (LRP) model has demonstrated high predictive value for identifying Oncotype DX Recurrence Score (ODX-RS) <=25 in patients with T1-2, N0-1, ER+/HER2− breast cancer. However, the prognostic value of the LRP model remains unclear. This study aims to compare the prognostic performance of the LRP model with Adjuvant! Online in predicting distant disease-free survival (DDFS). Methods: This retrospective study included women diagnosed with T1-2, N0-1, ER+/HER2- BC who received ET±CT between 2014 and 2018 at Peking University First Hospital. After laboratory testing with MammaTyper®, the LRP model was applied in this cohort (Available online at: https://mtrg-ruo.com/LRP). A modified version of Adjuvant! Online was used to determine clinical risk groups. DDFS was compared using log-rank tests and Cox proportional hazards models. Results: A total of 352 samples were successfully classified into clinical high-risk (C-high) and low-risk (C-low) groups by the modified version of Adjuvant! Online, and into LRP high-risk (LRP-high) and low-risk (LRP-low) groups using the MammaTyper® LRP model. The median follow-up time was 85 months. The LRP model showed greater statistical significance in differentiating low-risk from high-risk groups in terms of 9-year DDFS compared to Adjuvant! Online (log-rank p = 0.0015 for the LRP model vs. log-rank p = 0.016 for Adjuvant! Online). Patients were categorized into four combined risk groups: C-low/LRP-low (33.5%, 118/352), C-low/LRP-high (15.9%, 56/352), C-high/LRP-low (23.3%, 82/352), and C-high/LRP-high (27.3%, 96/352). Patients in the C-low/LRP-low group had the highest 9-year DDFS rate of 97.3% (95% CI: 94.3-100.0%), whereas the lowest rate of 81.3% (95% CI: 70.6-93.6%) was observed in the C-high/LRP-high group. Patients in the C-high/LRP-low group did not show a significant benefit from adjuvant chemotherapy in terms of DDFS (HR = 0.33, 95% CI: 0.03-3.65).Conclusions: The MammaTyper® LRP model is comparable to Adjuvant! Online in predicting 9-year DDFS in patients with T1-2, N0-1, ER+/HER2- breast cancer treated with ET ± CT. No significant DDFS benefits was observed from adjuvant chemotherapy in patients classified as C-high/LRP-low. Our results suggest that MammaTyper® may be useful as a locally available kit for identifying ER+/HER2- BC patients with low risk of recurrence.
Presentation numberPS3-10-07
Addressing unmet clinical need in HR-positive/HER2-negative early breast cancer: baseline profile of Italian patients included in a compassionate use program ribociclib plus NSAI
Armando Lunello, Novartis Farma Italy, Milan, Italy
I. Viganò1, A. Lunello1, D. Valsecchi1, N. Viapiana1, A. Martinez-Fernandez2; 1Medical Department, Novartis Farma Italy, Milan, ITALY, 2Global Medical Affairs, Novartis Farmaceutica SA, Barcelona, SPAIN.
Background: Hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative early breast cancer (EBC) patients at high risk of recurrence have limited treatment options. The phase 3 NATALEE trial1 showed that adjuvant ribociclib plus non-steroidal aromatase inhibitors significantly improved invasive disease-free survival in stage II-III HR-positive/HER2-negative EBC, supporting ribociclib for a broader population. On these data, the corresponding ribociclib label extension was approved by the Food and Drug Administration and the European Medicines Agency in September and November 2024, respectively. A Managed Access Program (MAP) was opened to allow access to ribociclib for eligible patients with HR-positive/HER2-negative EBC (stages II and III, based on NATALEE inclusion/exclusion criteria and no alternative authorized option). We assessed the baseline profile of patients included in the MAP of ribociclib in Italy. Methods: This retrospective analysis uses real-world data from Italian patients with HR-positive HER2-negative EBC stages II and III, approved for ribociclib access. The requesting physician submitted an unsolicited request, which was evaluated against eligibility criteria. The descriptive analysis reports their baseline profiles -demographics, clinical characteristics, and (neo-)adjuvant treatment patterns. Results: From December 2024 to April 2025, 634 requests for ribociclib were approved, and 87 patients started ribociclib adjuvant therapy. Local ethical committee approval was pending for 253 requests. The mean age of patients requiring treatment access was 54.6 years (SD 10.9; range 28-87). Most were female (629; 99.2%), and 162 (25.6%) had at least one comorbidity. Concomitant medications were reported in 308 (48.6%) patients. According to the MAP inclusion criteria, which allows only patients without an alternative treatment available, at treatment access most patients had stage II disease (602; 95%), stage IIA 366 (57.7%) and IIB 234 (36.9%). Stage III disease was reported in 31 patients (4.9%), with 1.9% each in stages IIIA and IIIC, and 1.1% in stage IIIB. The most common tumor classifications were T0-1N1 (34.9%) and T2N1 (33.3%). Among patients in stage T2N0 (22.9%), 91 (62.7%) were G3. 72.5% of patients had nodal involvement, mainly N1 (68.6%), and 27% had N0 disease. 296 (46.7%) patients received previous chemotherapy, 227 (76.7%) in the adjuvant setting and 69 (23.3%) in the neoadjuvant setting. Prior chemotherapy was administered to 41.5% of patients with lymph node involvement, compared to 61.4% of those without nodal metastases. Adjuvant radiotherapy was received by 312 (49.2%) patients, while an adjuvant CDK4/6 inhibitor was previously received by 15 (2.4%). Four patients originally eligible for abemaciclib were included in the MAP due to clinical conditions (50%) or because the treatment window had elapsed (50%). Conclusions: These real-world data underscore the need for additional adjuvant treatment options in patients with HR-positive/HER2-negative EBC in Italy. The population treated within the compassionate use program included a broad spectrum of disease stages, such as patients with stage II disease and those without nodal involvement—subgroups with limited current therapeutic alternatives.1Hortobagyi GN, et al. Ann Oncol. 20252 Fashing PA, et al. Ann Oncol 2024
Presentation numberPS3-10-08
Patterns of OncotypeDX use in early HR+/HER2- Breast Cancer: clinical-pathological comparison of tested vs. untested patients followed at the Breast Unit of Parma
Chiara Tommasi, University Hospital of Parma, Parma, Italy
C. Tommasi1, M. Corianò1, O. Serra2, I. Leccardi2, A. Loschiavo3, D. Zanoni1, D. Boggiani1, F. Pratticò4, M. Baronchelli4, F. Sica4, A. Lazzarin4, G. Di Maria1, G. Caruso4, S. Rossi3, C. Mancini5, F. Gussago6, M. F. Arcuri6, E. M. Martella5, A. Musolino7, B. Pellegrino1; 1Medical Oncology & Breast Unit, University Hospital of Parma, Parma, ITALY, 2Department of Medicine and Surgery, University of Parma, Parma, ITALY, 3Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, ITALY, 4Medical Oncology, University Hospital of Parma, Parma, ITALY, 5Anatomy and Pathological Histology Unit, Department of Oncohematology, University Hospital of Parma, Parma, ITALY, 6Breast Surgery and Thoracic-Vascular Department, University Hospital of Parma, Parma, ITALY, 7Medical Oncology, Breast & GYN Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola FC, ITALY.
Backgrounds: OncotypeDX (ODX) test in early-stage hormone receptor-positive (HR+)/ Epidermal growth factor receptor2 negative (HER2-) breast cancer (BC) helps clinicians in evaluating patients’ prognosis and defining adjuvant treatment. In Emilia-Romagna (Italy), the indication for conducting ODX is determined by the multidisciplinary breast unit team. However, the criteria for test reimbursement remain broad. Methods: We performed a comparative analysis of the clinical and pathological characteristics of patients treated at the Parma Breast Unit from July 2020 to July 2024, comparing those who underwent ODX testing with those who, despite being potentially eligible, were not offered the test. Results: ODX test was proposed to 139 patients (pts), with a median age at diagnosis of 56 years (range 36-74). Among these, 60% (84 pts) presented with T1 (75 years (including 171 aged ≥80 years). Among the remaining 654 patients, 64% (536 pts) had T1 tumors, 78% (512 pts) were N0, 62% (405 pts) had G2 tumors, and 57% (375 pts) exhibited a low Ki-67 (<15%). 30% (41 pts) of the patients who underwent ODX testing received adjuvant chemotherapy due to a high recurrence score (RS).Among patients who were not tested with ODX, only 7% (45 pts) received adjuvant chemotherapy; of these, 62% (28 pts) were premenopausal and 64% (29 pts) had nodal involvement (N1).Patients in the ODX-tested group typically presented with multiple intermediate-risk features, whereas those not tested generally had fewer risk factors. Conclusions: Our real-world analysis reveals a substantial discrepancy between potential eligibility for ODX and its actual use in early-stage HR+/HER2- BC patients. Patients who underwent ODX testing more frequently exhibited multiple risk factors, which justified the genomic assessment and allowed for a more personalized decision-making process regarding adjuvant chemotherapy.Considering the high cost of the assay and the limited availability of healthcare resources, a more precise selection of patients is essential to ensure cost-effective and clinically appropriate use of genomic testing, avoiding unnecessary expenses and improving treatment personalization.
Presentation numberPS3-10-09
Improving Timeliness of Adjuvant Therapy Through Reduction of Oncotype DX Turnaround Time: A Multi-Site Quality Improvement Initiative at a Rural-Academic Cancer Network in West Virginia
Michelle Hartzell, West Virginia University Cancer Institute, Morgantown, WV
M. Hartzell1, S. Nethagani1, N. Holley2, O. Chintuya3, S. Wen4, K. Barnett1, D. Safi1, S. Kurian1, M. Hafez5, B. Jiang1; 1Hematology and Medical Oncology, West Virginia University Cancer Institute, Morgantown, WV, 2Internal Medicine, West Virginia University Department of Medicine, Morgantown, WV, 3School of Osteopathic Medicine, West Virginia School of Osteopathic Medicine, Lewisburg, WV, 4Department of Epidemiology and Biostatistics, West Virginia University School of Public Health, Morgantown, WV, 5Hematology and Medical Oncology, Sidney Kimmel Medical College, Philadelphia, PA.
Introduction: Timely initiation of adjuvant therapy is critical for patients with early-stage, HR-positive, HER2-negative breast cancer and is optimally achieved within 4 to 8 weeks post-surgery. At the West Virginia University Cancer Institute (WVUCI), a single health system serving both rural and urban patients at 16 cancer centers, delays in Oncotype DX Recurrence Score testing and prolonged time intervals between testing results and adjuvant therapy initiation can lead to suboptimal care delivery. This quality improvement (QI) initiative targeted Oncotype DX turnaround time (TAT) as a modifiable factor to facilitate timely, risk-adapted adjuvant therapy, and increase the rate of treatment initiation within guideline-recommended timeframes. Methods: Using a Plan–Do–Study–Act (PDSA) model, a multi-phase QI project was implemented at 15 WVUCI sites. Interventions included: (1) EMR integration of Oncotype DX ordering and reporting via Epic Aura (August 2025) and (2) insurance pre-authorization bypass for specimen processing and release of results (January 2025). An IRB-approved protocol was used to collect patient demographics and treatment history for all patients that underwent Oncotype DX testing between January 2024 and May 2025. Patients were divided into three groups by intervention phase: (1) pre-intervention, (2) post-EMR integration, and (3) post-full interventions. Primary outcomes included Oncotype TAT, time from surgery to adjuvant chemotherapy, and time from surgery to first adjuvant therapy with chemotherapy or radiation. Statistical analyses included the Kruskal-Wallis test due to non-normal data distribution, the post-hoc Dunn’s test, and Chi square test. Results: A total of 339 Oncotype DX orders were analyzed across the system during the QI initiative. The patient population was 97% female and 57% were aged ≥65 years. There were 89% of patients with stage I disease and 11% with stage II. A total of 75% of patients received adjuvant chemotherapy and/or radiation, with 15% treated with chemotherapy and 69% treated with radiation. Following implementation of the interventions, median TAT decreased from a median of 12 days (IQR 8-20) at baseline to 9 (IQR 8-13) and 8 days in groups 2 and 3, respectively. A significant difference in TAT between groups was observed (p<0.001, Kruskal-Wallis test) with significant differences in all pairwise comparisons in a post-hoc analysis (p<0.001, Dunn’s test). Time from surgery to initiation of first adjuvant treatment decreased from a baseline median of 63 days (IQR 46-77) to 62 (IQR 48-86) and 54 days (IQR 41-75) in groups 2 and 3, respectively. There were significant differences in time from surgery to initiation of adjuvant treatment between groups (p<0.05, Kruskal-Wallis test). The proportion of patients who initiated adjuvant treatment within 8 weeks of surgery increased from 40.0% at baseline to 42.4% and 55.0% in groups 2 and 3, respectively (p=0.12, Chi-square test). This increase reflected an approximately 38% relative improvement from baseline. Conclusions: This QI initiative led to significant and sustained reductions in Oncotype DX turnaround time and improved timeliness of adjuvant therapy initiation. Although the increased proportion of patients who initiated adjuvant treatment within 8 weeks of surgery did not reach statistical significance, the trend suggests improved timeliness of care delivery. System-level process redesign with Oncotype DX EMR integration and insurance streamlining proved feasible and impactful across a rural-academic network. Ongoing efforts will focus on reducing the interval between result availability and therapy initiation, with a goal to increase the rate of adjuvant therapy initiation within 8 weeks from 55% to 80%.
Presentation numberPS3-10-10
Decision-making on adjuvant chemotherapy in early hr+ breast cancer: a prospective evaluation of oncotype dx<sup>®</sup> utility among swiss breast cancer experts
Marcus Vetter, Cantonal Hospital Baselland, Liestal, Switzerland
M. Vetter1, S. Ebner2, E. Kralidis3, C. Kurzeder4, A. Oseledchyk5, E. Chiru1, J. Landin6, K. Schmutz-Kober1; 1Cancer Center Baselland, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 2Department of Gynecology and Obstetrics, Cantonal Hospital Baselland, Liestal, SWITZERLAND, 3Breast Center Seefeld ZH, Breast Center Seefeld ZH, Zürich, SWITZERLAND, 4Breast Center, University Hospital Basel, Basel, SWITZERLAND, 5Medical Oncology, University Hospital Basel, Basel, SWITZERLAND, 6Cancer Center Baselland, Breast Center, Cantonal Hospital Baselland, Liestal, SWITZERLAND.
Background: The Oncotype DX® test (21-gene Recurrence Score (RS) test) is widely used to guide adjuvant chemotherapy (CT) decisions in hormone receptor-positive (HR+), HER2-negative early breast cancer. However, variability in clinical interpretation persists in real-world settings. This study prospectively assessed how the Oncotype DX test influences CT decision-making among Swiss breast cancer experts. Methods: From a cohort of 325 patients with HR+/HER2− early breast cancer and available Recurrence Score® results, 50 anonymized randomly selected cases were chosen. Ten board-certified Swiss medical oncologists, all specialized in breast cancer, independently evaluated each case and made CT recommendations before and after disclosure of the Recurrence Score results. Changes in treatment decisions were assessed using McNemar’s test and ANOVA. Results: The Oncotype DX test led to changes in treatment recommendations in 19.8% of cases. CT was recommended in 35.4% of pre-testing decisions, compared to 21% post-testing, representing a 14.4% net reduction. Changes in CDK4/6 inhibitor and osteoprotection recommendations were minimal (0.4% and 0.2%, respectively). The most pronounced impact was observed in cases with intermediate clinical risk, particularly tumors 2-4.5 cm and N0 status. Notably, 23 (46%) patients had tumors with node-positive disease. One-way ANOVA showed no significant difference in mean treatment recommendations before and after Oncotype DX testing (F(1,58)=0.29, p=0.59). However, McNemar’s test revealed a highly significant directional shift (p<0.0001), primarily reflecting de-escalation of CT. Only one oncologist escalated treatment in a single case. Inter-observer variability decreased following Recurrence Score result disclosure. Conclusions: The Oncotype DX test significantly influenced adjuvant treatment decisions, primarily leading to chemotherapy de-escalation. It also increased decision consistency among Swiss breast cancer specialists, underscoring its value not only in guiding individual care but also in harmonizing expert judgment in routine practice.
Presentation numberPS3-10-11
Feasibility of Telemonitoring of Personalized Circadian Rhythms in Patients with Early-Stage HR-Positive Breast Cancer Receiving Endocrine Therapy
Sarah Poland, The University of Chicago Medicine, Chicago, IL
S. Poland1, N. Chen1, X. Li2, B. Tucker3, F. Lévi2, E. Tasali3, R. Nanda1; 1Hematology/Oncology, The University of Chicago Medicine, Chicago, IL, 2UPR Chronotherapie, Cancers et Transplantation, Université Paris Saclay, Villejuif, FRANCE, 3Sleep Medicine, The University of Chicago Medicine, Chicago, IL.
Background: Endocrine therapy (ET) is an integral treatment for early-stage hormone receptor-positive (HR+) breast cancer, but significant side effect burden can lead to decreased adherence and increased risk of recurrence. Chronotherapy, or aligning medication timing with circadian rhythms, has shown promise in reducing toxicity and improving efficacy of medications in other malignancies, but remains underexplored in breast cancer and ET. We conducted a pilot study to evaluate the feasibility of monitoring circadian rhythms in patients receiving ET using wearable digital health sensors and validated patient-reported outcomes (PROs). Methods: Patients with early-stage HR+ breast cancer already receiving or planning to initiate standard ET were eligible to enroll in a 14-day pilot study. Primary endpoint was to establish the feasibility of novel telemonitoring techniques to identify personalized circadian rhythms. Patients wore digital health devices including a wristwatch and a chest sensor to capture continuous physiologic data, including rest-activity, sleep-wake cycles, skin temperature, and heart rate continuously for 24 hours a day. Baseline symptom surveys were administered using the Pittsburgh Sleep Quality Index (PSQI) and Munich Chronotype Questionnaire (MCTQ). Patients completed daily PROs using the Karolinska Sleepiness Scale (KSS) and Functional Assessment of Cancer Therapy-Endocrine Subscale (FACT-ES) to capture ET-related symptoms and sleep-wake patterns. Results: As of 7/9/25, 11 of 20 patients (pts) have completed the study. Average pt age is 59.2 years (range: 32-86 years). 2/11 of pts self-identified as Black and 9/11 as White; 2 were premenopausal. All pts were on endocrine monotherapy: 5 anastrozole, 2 tamoxifen, 2 letrozole, and 2 exemestane. On average, pts had taken their ET for 9.5 months (range 0 – 43) prior to study entry. Five patients took their ET “early” (between 05:00 – 13:00) and 6 took their ET “late” (between 20:00 – 22:00). Pts completed greater than 96% of all surveys. The average FACT-ES score was 70.9 in the “early” group versus 63.1 in the “late” group (p<.05). The average KSS was 2.7 in the “early” group versus 5.6 in the “late” group (p<.05). At the end of the study, 10 of 11 pts (91%) reported it was “very easy” or “easy” to participate in the study while continuing daily life. Thus far, descriptive data of rest-activity, sleep-wake, and temperature rhythms highlight the individual variability in circadian rhythms between patients. Analyses to derive the circadian metrics are ongoing. Conclusions: Telemonitoring of circadian rhythms using wearable sensors is feasible and well-tolerated in patients receiving ET for HR+ breast cancer. Preliminary data suggest excellent patient adherence and feasibility of such telemonitoring approach, as well as increased ET-related symptoms and sleepiness in the “late” as compared to “early” ET groups. Further analysis will explore circadian rhythm metrics and associations with ET-related symptom burden. These findings support the feasibility of designing future research aimed at personalizing ET timing based on individual circadian profiles to improve tolerability and adherence.
Presentation numberPS3-10-13
Treatment outcomes of patients with early-stage breast cancer eligible, but not treated, with adjuvant ribociclib. a real-world cohort study
Hikmat Abdel-Razeq, King Hussein Cancer Center, Amman, Jordan
H. Abdel-Razeq, M. Horani, F. Tamimi, T. Al-Batsh, O. El Khatib, Y. Al-Masri, A. Ghanem, Q. Jawarneh, M. AL-yag’oub, A. Jaffal, R. Khader, S. Abdel-Razeq, A. Alanani, B. Sharaf, M. El-Atrash, M. Abunasser; Internal Medicine, King Hussein Cancer Center, Amman, JORDAN.
Background: Adjuvant CDK4/6 inhibitors, such as ribociclib and abemaciclib, have demonstrated significant disease-free survival (DFS) benefits in early-stage hormone receptor-positive (HR+)/HER2-negative breast cancer within randomized trials. However, real-world implementation remains limited, and outcomes for eligible-but-untreated populations are poorly characterized. This study evaluates treatment outcomes in patients retrospectively classified as eligible for adjuvant CDK4/6 inhibition under NATALEE criteria, all of whom were managed without such agents in routine practice. Methods: A retrospective cohort analysis was conducted using clinical data from patients with early-stage HR+/HER2-negative breast cancer. Eligibility for adjuvant ribociclib was assigned using trial-based criteria: stage IIA (high risk), IIB, or III disease as per the American Joint Committee on Cancer (AJCC) 8th edition. Kaplan-Meier methods and Cox proportional hazards models were employed to evaluate DFS and overall survival (OS), stratified by eligibility status. Results: Of the 2,730 patients included, 1,955 (71.6%) met the NATALEE eligibility criteria. Median (range) age was 50 (22-91) years and majority (n= 2,213, 81.1%) had invasive ductal carcinoma (IDC). A significant proportion of patients had high-risk disease with 1410 (51.6%) patients had pathologic node-positive disease, 602 (22.1%) had clinical T3 or T4, and 741 (27.1%) had grade 3 disease. Almost all patients were treated with adjuvant endocrine therapy with or without ovarian function suppression (OFS) with over half (n=1422, 52.1%) received an aromatase inhibitor (AI). However, none of the patients received adjuvant CDK4/6 inhibitors, allowing for real-world benchmarking of outcomes in a CDK4/6i-untreated setting. Across the cohort, patients eligible for CDK4/6 inhibitors had significantly worse 3-year DFS (85.2% [95% CI, 83.6%-86.8%]) compared to ineligible patients (95.1% [95% CI, 93.5%-96.6%]; HR = 2.75 [95% CI, 2.06-3.68]; p < 0.001). Stratified multivariable analysis revealed the following independent predictors of worse outcomes: Hazard Ratio (HR) for ER-negative vs. high-expression = 1.62 (95% CI: 1.10-2.39; p=0.015), high tumor grade (Grade 3): HR = 1.64 (95% CI: 1.20-2.25; p=0.002), clinical nodal involvement (cN+): HR= 1.27 (95% CI: 0.99-1.64; p=0.064), tumor size ≥cT3: HR = 2.23 (95% CI: 1.60-3.12; p<0.001). Although median OS was not reached in either group, separation of survival curves became apparent by 36 months and continued to widen thereafter, consistent with early relapse clustering in the higher-risk, untreated cohort. At 5 years, OS was 87.9% (95% CI, 86.4%-89.5%) in eligible patients compared with 95.9% (95% CI, 94.4%-97.4%) in the ineligible group. Together, these findings reveal a consistent and clinically meaningful survival gap between eligible-but-untreated patients and their lower-risk counterparts, highlighting the real-world impact of not receiving adjuvant CDK4/6-targeted therapy. Conclusions: A significant proportion of patients in this real-world cohort met criteria for adjuvant CDK4/6 inhibition but were not treated with these agents. These untreated, yet eligible individuals, experienced markedly worse survival outcomes compared to their ineligible counterparts. The data underscore a critical gap between evidence-based guidelines and clinical adoption. Closing this implementation gap could meaningfully impact recurrence rates and long-term survival in early-stage HR+ breast cancer.
Presentation numberPS3-10-14
Survival Outcome in Premenopausal ER+/PR+ HER2− Breast Cancer Stratified by Nodal Status: a National Cancer Database (NCDB) Experience
Lan Lei, Winship Cancer Institute of Emory University, Atlanta, GA
L. Lei1, A. Marra1, M. Canning1, J. Switchenko2, S. Gandhi1; 1Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, 2Department of Biostatistics and Bioinformatics at Rollins School of Public Health, Emory University, Atlanta, GA.
Background: The comparative benefit of chemotherapy (CT) versus endocrine therapy (ET) in premenopausal women with stage I-III estrogen receptor-positive (ER+), progesterone receptor-positive (PR+), and HER2-negative (HER2-) breast cancer (BC) remains unclear. This study examined differences in survival outcomes among patients with node-negative (N0) and node-positive (N1) disease stratified by treatment modality.Methods: We analyzed data from NCDB for women <50 years with ER+ and/or PR+, HER2−, stage I-III BC from 2005 to 2018. Collected variables included tumor size, grade, nodal status, lymphovascular invasion (LVI), Charlson-Deyo comorbidity score, and treatment history (ET, CT, radiation therapy [RT]). Overall survival (OS) was estimated using Kaplan-Meier analysis and compared between CT and ET groups using the log-rank test. Multivariable Cox proportional hazards models were used to identify independent predictors of OS. Statistical significance was assessed using a two-sided α level of 0.05.Results: Among 376,303 eligible patients, 168,741 (44.8%) had N0 and 83,428 (22.2%) had N1 disease. Median age was 45 years (range: 18-50). In the N0 group, 71,733 (42.5%) received ET alone and 97,008 (57.5%) received CT. Compared to ET-only recipients, CT-treated patients were younger and more likely to have grade 3/4 tumors, larger tumor size, LVI, PR-negative disease, and no RT (P < 0.01). In the N1 group, 5,658 (6.8%) received only ET and 77,770 (93.2%) received CT. Similar trends were observed, although RT was more commonly given in CT recipients (P < 0.01).OS was superior in patients treated with ET alone versus CT across both nodal cohorts. Among N0 patients, 4-year OS was 99.2% with ET vs. 96.4% with CT, and 6-year OS was 97.3% vs. 92.7% (P < 0.0001 for both). In the N1 group, ET was associated with 4-year OS of 96.9% vs. 93.5% for CT, and 6-year OS of 91.0% vs. 87.0% (P < 0.0001 for both). On multivariable analysis (Table 1), larger tumor size, high grade malignancy, PR-negative status, heavy comorbidity burden, and absence of RT were associated with worse OS in all patients (P < 0.01). Interestingly, CT was independently associated with worse OS in N0 cohort but improved OS in N1 population.Conclusions: In premenopausal women with early-stage ER+/PR+, HER2- BC, ET alone was associated with superior OS in N0 disease. In contrast, CT improved OS in patients with N1 disease. These findings support the consideration of ET monotherapy as a potential alternative to CT in carefully selected N0 premenopausal patients. This work was limited by the inability to evaluate the role of ovarian function suppression. Future studies should explore the addition of ovarian suppression to ET as a strategy for safely omitting CT in N0 population.
| N0 | N1 | |||
| Covariates | Univariate | Multivariate | Univariate | Multivariate |
| Age | 0.98 (0.98-0.98) | 1.00 (1.00-1.00) | 0.98 (0.98-0.98) | 0.99 (0.98-0.99) |
| Grade (4 vs.1) | 8.29 (6.86-9.90) | 1.86 (1.38-2.51) | 3.00 (2.22-3.98) | 4.66 (4.18-5.20) |
|
Tumor size (T4 vs. T0-1) |
3.93 (2.90-5.19) | 5.31 (4.34-6.49) | 4.96 (4.47-5.50) | 1.89 (1.40-2.54) |
| PR-negative status | 2.38 (2.27-2.44) | 1.54 (1.45-1.61) | 2.08 (2.04-2.17) | 1.78 (1.69-1.85) |
|
Comorbidities (score 2 vs. 0) |
2.96 (2.53-3.43) | 2.84 (2.43-3.31) | 2.02 (1.70-2.37) | 2.07 (1.74-2.46) |
| CT vs. ET | 2.61 (2.48-2.75) | 1.43 (1.34-1.52) | 1.47 (1.34-1.62) | 0.91 (0.82-1.00) |
| RT | 0.67 (0.63-0.72) | 0.70 (0.65-0.75) | 0.71 (0.67-0.75) | 0.73 (0.68-0.77) |
Presentation numberPS3-10-15
Smarcd3 co-suppresses erα signaling pathway mediates tumor progression and its role in anti-estrogen resistance in breast cancer
Haoran Dong, First Hospital of China Medical University, Shen Yang, China
H. Dong, Y. Xu; Breast surgery, First Hospital of China Medical University, Shen Yang, CHINA.
Breast cancer is one of the most prevalent malignant tumors globally and a leading cause of cancer-related mortality among women. Estrogen receptor (ER) positive breast cancer accounts for approximately 70% of all cases. Endocrine therapy targeting estrogen receptors is a cornerstone of clinical treatment for ER-positive breast cancer; however, the emergence of resistance during therapy significantly undermines its therapeutic efficacy ERα is a key factor in the initiation and progression of ER-positive breast cancer and serves as the primary target for endocrine therapies. In this study, we identified a novel ERα co-regulator, SMARCD3, a member of the SMARCD family within the SWI/SNF chromatin remodeling complex. SMARCD3 acts as a tumor suppressor in ER-positive breast cancer, inhibiting tumor progression and enhancing sensitivity to endocrine therapy. Notably, SMARCD3 expression is significantly downregulated in ER-positive breast cancer, and its reduced expression correlates with poor prognosis. Overexpression of SMARCD3 inhibits the proliferation of ER-positive breast cancer cells. Mechanistically, SMARCD3 recruit histone deacetylases HDAC1 and HDAC2, as well as the E3 ubiquitin ligase TRIM21, to form a complex that downregulates ERα-mediated transcriptional activity and reduces ERα protein stability, thus inhibiting ERα function through both transcriptional and post-translational mechanisms. Additionally, we observed that the SWIB domain of SMARCD3 participates in the ubiquitin-mediated degradation of ERα. In summary, our findings identify SMARCD3 as a novel co-suppressor of ERα that inhibits the progression of ER-positive breast cancer and enhances sensitivity to endocrine therapy, providing new insights for therapeutic strategies in ER-positive breast cancer.
Presentation numberPS3-10-16
Clinical and Histopathological Prognostic Factors and Long-Term Outcomes in Early Stage Luminal Breast Cancer: A Real-World Cohort from Brazil
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.
The rising incidence of early-stage breast cancer has prompted critical discussion regarding the true benefit of adjuvant chemotherapy (ACT) in luminal tumors, a subtype characterized by favorable prognosis and high sensitivity to endocrine therapy (ET). While multigene assays have become increasingly central to therapeutic decision-making, their limited availability in developing countries—including within private healthcare systems—represents a significant barrier. In many cases, patients must cover the high cost of testing themselves. This highlights the pressing need to identify reliable and accessible clinicopathological markers to guide ACT decisions in resource-constrained settings. We conducted a retrospective observational study using electronic medical records from a referral cancer center in Brazil. Eligible patients were women with ER-positive, HER2-negative, clinical stage I (T1N0M0) breast cancer treated between 2016 and 2024. Patients received either ET alone or in combination with ACT. Event-free survival (EFS)—defined as time from diagnosis to recurrence or death—was evaluated using Kaplan-Meier analysis and compared using the log-rank test. Variables with p-values <0.20 were included in the multivariate analysis using the Cox proportional hazards model, with significance set at p <0.05. SPSS version 23 was used for all analyses. In this cohort (n=134), the mean age was 59.3 years, and 69.2% of patients were postmenopausal. Most tumors were invasive ductal carcinoma (78.4%); 20.3% were histological grade 3, and 47.7% had Ki67 ≥14%. ET alone was administered to 81.3%, while 18.7% received ACT before ET, mostly with docetaxel and cyclophosphamide regime (60.0%). After a median follow-up of 47.3 months, no classical clinicopathological variable, such as body weight and histological grade, was independently associated with EFS. Mean EFS did not significantly differ between the ET-only group (132.5 ± 10.4 months) and the ACT group (114.4 ± 8.9 months; p = 0.705). In this real-world cohort of stage I luminal breast cancer, endocrine therapy alone provided excellent long-term outcomes. The addition of adjuvant chemotherapy conferred no significant benefit, and conventional clinicopathological features lacked predictive power. These findings reinforce TNM as the most important prognostic factor among luminal breast cancer, support a de-escalation approach in selected low-risk patients, and emphasize the importance of treatment personalization, particularly in settings where molecular testing is unavailable.
Presentation numberPS3-10-17
Residual Cancer Burden in ER+/HER2− Breast Cancer: Predictive Role of Hormone Receptor Expression and Clinical Stage
Gemma Viñas, Catalan Institute of Oncology Girona, Girona, Spain
A. Brotons1, H. Pla1, R. Fort1, A. Roqué-Lloveras1, S. del Barco1, C. Montañés-Ferrer1, J. Paz1, A. Ribera1, Í. López-Rovira2, E. Polonio-Alcalá2, F. León3, I. Oliveras3, I. Ramos4, J. Ribas5, E. Vila6, J. Ferrer7, M. Negre7, J. Martínez-Sancho8, G. Viñas1; 1Medical Oncology, Catalan Institute of Oncology Girona, Girona, SPAIN, 2Precision Oncology Group (OncoGIR-Pro), Institut d’Investigació Biomèdica Dr. Josep Trueta de Girona (IDIBGI), Salt, SPAIN, 3Radiotherapy Department, Catalan Institute of Oncology Girona, Girona, SPAIN, 4Pathological Anatomy Department, Dr. Josep Trueta University Hospital, Girona, SPAIN, 5Plastic Surgery Department, Dr. Josep Trueta University Hospital, Girona, SPAIN, 6Gynecology and Obstetrics Department, Dr. Josep Trueta University Hospital, Girona, SPAIN, 7Diagnostic Imaging Institute, Dr. Josep Trueta University Hospital, Girona, SPAIN, 8Statistics Unit, Institut d’Investigació Biomèdica Dr. Josep Trueta de Girona (IDIBGI), Salt, SPAIN.
Neoadjuvant chemotherapy (NACT) plays a critical role in managing locally advanced breast cancer by reducing tumor burden before surgery and allowing for early initiation of systemic treatment. In estrogen receptor-positive/HER2-negative (ER+/HER2−) tumors, response to NACT is limited and unpredictable. Identifying predictors of response is essential to optimize treatment and minimize toxicity. The main aim of this study is to investigate clinicopathological factors associated with residual cancer burden (RCB). Secondary objectives included evaluating body mass index (BMI) and surgical outcomes. We conducted a retrospective single-center study at Girona’s Catalan Institute of Oncology, including ER+/HER2− breast cancer patients (stage I-III) treated with anthracycline- and/or taxane-based NACT between 2010 and 2023. Clinical and pathological data – age, BMI, estrogen and progesterone receptor (ER, PR) status, nodal stage, surgical procedures- were analyzed. Associations with RCB were analyzed using chi-square tests and linear regression analysis. A total of 111 patients with stage II-III breast cancer were included (mean age 47.6 years; BMI 25.8 kg/m²). Surgery involved mastectomy in 52.3% and axillary dissection in 74.8% of cases, with 17.5% converted to mastectomy after NACT. Only 13 (11.7%) achieved a favorable pathological response (RCB 0-1). Tumors with low ER expression (1-10%) were significantly more likely to respond to NACT (p=0.034). In contrast, BMI and menopausal status were not significantly associated with RCB. Advanced clinical nodal status (cN3) was strongly associated with chemoresistant tumors (p=0.030), while older age (p=0.058) and larger tumor size (cT4) (p=0.089) showed a non-significant trend toward worse outcomes. PR and HER2 expression and Ki67 index did not show significant associations with RCB in this cohort. Our findings highlight lower ER expression and limited nodal burden as predictive biomarkers of improved response to NACT in ER+/HER2− breast cancer. Considering the low response rate, these results support a more tailored therapeutic approach and underscore the need for predictive tools in optimizing patient selection within this biologically heterogeneous subgroup.
Presentation numberPS3-10-18
Early Discontinuation of Adjuvant Endocrine Therapy in Patients with Triple Positive Early Stage Breast Cancer after Pathologic Complete Response (pCR) to Neoadjuvant Therapy: A Retrospective Review
Agreen Hadadi, Emory Winship Cancer Institute, Atlanta, GA
A. Hadadi1, W. Yang2, X. Yi2, E. A. Sakach1, J. Meisel1; 1Medical Oncology, Emory Winship Cancer Institute, Atlanta, GA, 2Pathology, Emory Winship Cancer Institute, Atlanta, GA.
Title:Early discontinuation of adjuvant endocrine therapy in patients with triple positive early stage breast cancer after pathologic complete response (pCR) to neoadjuvant therapy: A retrospective reviewAgreen Hadadi,MD, Wei Yang MD, Xiaoxian Li, MD, PhD, Elizabeth Sakach, MD, Jane Meisel, MD, FASCO Purpose:The current standard of care for patients (pts) with stage 2 or 3 hormone receptor positive (HR+)/ HER2 + breast cancer is neoadjuvant chemotherapy plus HER2-directed therapy (NACT-HP) followed by surgery. For pts who achieve a pCR, anti-HER2 therapy is continued to complete 1 year and pts are recommended to take adjuvant endocrine therapy (ET) for a minimum of 5 years. Adherence to ET can be challenging due to multiple adverse effects that can impair quality of life (QOL). Here we evaluate whether early discontinuation of ET impacts clinical outcomes in a diverse real-world population who achieve pCR following NACT-HP. Methods:A retrospective chart review was conducted for pts with stage 2/3 HR+/HER2+ breast cancer who were treated between 2009 -2020 at the Winship Cancer Institute of Emory University. HR+ was defined as estrogen and progesterone receptor (ER/PR) >1% and HER2 + as IHC 3+ or IHC 2+/FISH positive. Eligible patients received NACT-HP and achieved a pCR. Endpoints were adherence to endocrine therapy (ET) and disease-free survival (DFS), local or distant recurrence, and death. Descriptive statistics were used to summarize demographics and clinical characteristics. Results: Of 40 pts who achieved pCR, 10 (25%) were Black, 28 (70%) were White, 2 (5.0 %) were identified as other. The median follow-up time was 86 months (range: 9-184 months). Of the 40 pts, 24 were age >50 years, and 16 were 10%. 13 (32.5%) pts underwent lumpectomy, 16 (40%) underwent bilateral mastectomy, and 11 (27.5%) unilateral mastectomy. Regarding adjuvant ET, 22 (55.0%) pts received an aromatase inhibitor (AI), 9 (22.5%) received tamoxifen, 2 (5.0%) received AI plus ovarian suppression, and 6 (15.0%) received both AI and tamoxifen at various time points. One pt declined ET.Three (7.5%) individuals self-discontinued ET prior to completing 5 years of treatment due to side effects. At the time of follow-up, 36 (80%) patients were disease-free, 3 (7.5%) had recurrence with survival, and 1 (2.5%) had recurrence which led to death. All 3 patients who discontinued ET before 5 years remained disease-free, while the one patient who declined ET died from distant recurrence. Conclusions:In this small, real-world cohort, early discontinuation of adjuvant ET was not associated with disease recurrence. This observation is hypothesis generating and raises the question of whether the recommended 5-year duration of adjuvant ET is necessary in pts who achieve pCR with NACT-HP. With the advent of trials like the LoTamTrial exploring de-escalation of adjuvant ET, it is prudent to highlight real-world evidence of distant recurrence following early ET discontinuation. While our findings are limited by a small cohort size and retrospective nature, a larger cohort will be updated prior to the meeting, also including patients treated between 2020 to present.
Presentation numberPS3-10-19
Prevalence of HER2-low status in Ecuadorian women with Hormone Receptor Positive Breast Cancer
Lorena Estrada, Sociedad de Lucha Contra el Cancer – SOLCA Guayaquil, Ecuador, Guayaquil, Ecuador
L. Estrada, W. Morquecho, V. Bastidas, R. Escala; Education & Research, Sociedad de Lucha Contra el Cancer – SOLCA Guayaquil, Ecuador, Guayaquil, ECUADOR.
Prevalence of HER2-low status in Ecuadorian women with Hormone Receptor Positive BreastCancerBACKGROUNDBreast cancer (BC) is the most frequent neoplasia in women worldwide. According to GLOBOCAN(2022), 9.6% of new cases globally occur in the Caribbean and Latin America. The prevalence ofHER2-low status in South America and particularly in Ecuador is limited. Although many HER2-low tumors coexpress hormone receptors (HR), their exact global prevalence remainsundetermined. Therefore, this study aims to determine the prevalence of HER2-low status inEcuadorian women with HR-positive BC.METHODSA retrospective, cross-sectional study of BC patients’ medical records (2011-2021) fromEcuador’s SOLCA oncological hospital. Simple random sampling included 508 patients (p). HR-positive patients express estrogen receptors; HR-negative do not. HER2-low status is defined asHER2 1+ or HER2 2+/SISH- (silver in situ hybridization). Univariate analyses measuredfrequencies and percentages.RESULTSAmong the 723p evaluated, 215 were omitted from the analysis owing to missing data on HER2status. Of 508p with evaluable HER2 expression, 144 (28.3%) were HR-negative and 364 (71.7%)HR-positive. Of the 364p HR-positive, 135 (26.6%) presented with HER2 0+, 66 (13.0%) with HER21+, 64 (12.6%) with HER2 2+ (42 SISH-, 11 SISH+ and 11 SISH not available), and 99 (19.5%) withHER2 3+. Therefore, 108p (21.3%) had tumors classified as HER2-low and HR-positive. Clinicalstage II was the most prevalent among HER2-low HR-positive patients.CONCLUSIONSThe prevalence of HER2-low status in Ecuadorian women with HR-positive BC was 21.3%. AmongHR-positive patients, 108 of 364 (29.7%) exhibited HER2-low status, which is comparable to the34% reported in a Thai cohort of HR-positive cases. Conversely, among HER2-low patients, 108of 132 (81.8%) were HR-positive, exceeding the 71.1% reported in a ten-country study spanningEurope, Asia, and North America. Table 1. Baseline characteristics of patients n=508; results in frequency and (%) Age years, mean (s.d.)54.4 (12.7) Initial Clinical Stage I59 (11.6) II241 (47.4) III157 (30.9) IV22 (4.3) Progesterone receptors Positive345 (67.9) Negative163 (32.1) Estrogen receptors (ER) Positive364 (71.7) Negative144 (28.3) Ki-67 < 14100 (19.7) ≥ 14403 (79.3) HER2 0+172 (33.9) 1+84 (16.5) 2+ SISH-48 (9.4) 2+ SISH+13 (2.6) 2+ SISH (NA)13 (2.6) 3+178 (35.0) HER2-low y ER+108 (21.3)s.d.: standard deviation, SISH: silver in situ hybridization, ER: estrogen receptor, NA: not available
Presentation numberPS3-10-20
Immunological aspects that determine the risk of breast cancer recurrence in patients with early luminal subtype
Aleksandr Sultanbaev, Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Bashkortostan, Ufa, Russian Federation
A. Sultanbaev1, I. Tuzankina2, I. Kolyadina3, N. Sultanbaeva1, D. Kudlay4, S. Musin1, K. Menshikov1, M. Sultanbaev1; 1Department of Chemotherapy, Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Bashkortostan, Ufa, RUSSIAN FEDERATION, 2Immunology, Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, RUSSIAN FEDERATION, 3Department of oncology, Russian Medical Academy of Continuous Professional Education, Moscow, RUSSIAN FEDERATION, 4Department of Pharmacognosy and Industrial Pharmacy, Lomonosov Moscow State University, Moscow, RUSSIAN FEDERATION.
Breast cancer is the most common disease among malignant neoplasms. Some patients develop relapses and metastases even after radical treatment. There are still no accurate methods for determining the risk of such an outcome; recommendations for tactics in this case are based on statistical data on survival in different observation groups. We assumed that the risks of breast cancer progression are closely related to insufficient functioning of the immune system. One of the promising areas in the study of immunodeficiency states is the analysis of TREC and KREC (sections of T- and B-lymphocyte DNA that remain after the maturation of these cells). Objective. The aim of the study was to determine the level of TREC in the blood of patients with and without relapse in the luminal subtype of breast cancer. Materials and methods. The study included 196 patients with luminal Her2-negative subtype with stages I-III. Venous blood of patients was used to assess the level of TREC and KREC. Quantitative analysis of TREC and KREC was performed using the IMMUNO-BIT reagent kit. The analyzer used was: Real-time CFX96 amplifier, Bio-Rad Laboratories, USA. Detecting amplifier DT-96, DNA-Technology, Russia. Statistical analysis was performed using the StatTech v. 4.6.1 program (developer – StatTech LLC, Russia). Results. The median TREC level in the group without relapse was 22.43, in the relapse group – 3.0 copies per 105 cells p < 0.001. The results of the study showed a low level of TREC in the blood of patients with disease progression within 1 year, which was regarded as a deficiency of the T-cell link, which plays a major role in controlling tumor growth. Conclusions. The obtained data confirm the opinion that breast cancer is an immune-mediated oncological disease, in which there is a decrease in the activity of the body’s immune system. Thus, the assessment of the TREC level against the background of adjuvant therapy may become a new risk factor for relapse in patients with breast cancer.
Presentation numberPS3-10-21
Should genomic risk guide the use of adjuvant CDK4-6i in N0-N1 HR+ and HER2− early breast cancer?
Marcos Iglesias Campos, Hospital Universitario Virgen de la Victoria de Malaga, Malaga, Spain
M. Iglesias Campos1, J. Pascual López1, A. Sánchez-Muñoz1, N. Ribelles Entrena1, M. J. Bermejo Pérez1, A. Márquez Aragonés1, B. Pajares Hachero1, M. E. Domínguez Recio1, A. Godoy Ortiz1, B. Jiménez Rodríguez2, F. Carabantes Ocón2, T. Díaz Redondo2, E. Villar Chamorro2, P. Cantizani Maíllo2, E. Alba Conejo1; 1Medical Oncology, Hospital Universitario Virgen de la Victoria de Malaga, Malaga, SPAIN, 2Medical Oncology, Hospital Regional Universitario de Málaga, Malaga, SPAIN.
Background: Systemic adjuvant treatment for hormone receptor-positive, HER2-negative (HR+,HER2−) breast cancer has been undergoing a major paradigm shift in recent years. On the one hand, multigene prognostic genomic assays (MPGA) have enabled improved selection of patients who are likely to benefit from chemotherapy (CT). On the other hand, the addition of CDK4-6 inhibitors (CDK4-6i) to endocrine therapy (ET) has demonstrated a significant improvement in invasive disease-free survival (iDFS) in high-risk patients, as shown in the MONARCH-E and NATALEE trial (median follow-up of 33.3 months in the last analysis). However, there is currently no standardized definition of high clinical risk (for instance, NATALEE trial included both node negative and positive patients), and correlation between clinicopathological features and genomic risk classification is not consistently observed. This lack of concordance introduces uncertainty regarding the clinical benefit of adding CDK4-6i to ET in this setting. Objectives: The aim of this study is to describe a population of patients with early-stage (N0-N1) HR+,HER2- breast cancer who underwent MPGA (either the Prosigna® or Oncotype DX®) and met the inclusion criteria of the NATALEE trial, with the aim of determining whether the addition of a CDK4-6i to ET provides a clinical benefit in this setting. Methods: This retrospective study included patients diagnosed with early-stage (N0-N1) HR+,HER2- breast cancer between 2014 and 2023 across two hospitals in Málaga, Spain (Hospital Virgen de la Victoria and Hospital Regional Universitario). Efficacy outcomes were iDFS, DDFS and OS. The statistical analysis was performed using the R software (R Core Team. (2019). R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. Version 3.6.1). Results: 1.508 patients had a genomic platform, of whom 367 met the Natalee inclusion criteria (254 and 113 in the low-intermediate and high risk subgroup, respectively). The most frequent subgroup in both the low-intermediate and high-risk groups according to Prosigna® was T2N0 with Ki67 ≥20% and grade 2 (n = 41 and n = 32, respectively). Regarding Oncotype DX®, the most common low-risk subgroup was T2N0 with Ki67 ≥20% and grade 2 (n = 22), while in the high-risk group, it was T2N1 (n = 13). Among the most relevant clinicopathological features, 14.2%, 53.1%, and 31.3% of patients presented with stage IB, IIA, and IIB disease, respectively; 44.6% had no nodal involvement, 46.3% received adjuvant chemotherapy, and 20% and 10.1% were classified as high risk according to the Prosigna® and Oncotype DX® assays, respectively. Of these high-risk patients, 85.6% and 97.3% received chemotherapy. With a median follow-up of 37.4 months, there were 30 iDFS events (8,2%). The 3-year iDFS rate was 94.9% (95% CI, 92% to 96.8%) in the overall population, 94.8% (95% CI, 91.1% to 97.1%) in the low/intermediate genomic risk subgroup, 95.2% (95% CI 88.8% to 98%) in the high genomic risk subgroup, 94.3% (95% CI 90.8% to 96.5%) in stage II disease, 94.1% in N0 (95% CI, 89% to 97%), and 95.6% (95% CI, 91.2% to 97.7%) in N1 disease. DDFS and OS events occurred in 11 (2.9%) and 12 patients (3.26%) respectively. 3-year OS rates were 98.7% (95% CI 96,7% to 99.5%) in the overall population, 98.2% (95% CI 95.3% to 99.3%) in the low/intermediate genomic risk subgroup and 98.6% (95% CI 90.4% to 99.8%) in the high genomic risk subgroup. Conclusions: Despite the size of our sample, these findings suggest that the use of adjuvant CDK4-6i in N0 or N1 HR+,HER2− breast cancer should be guided not only on clinicopathological features but also on genomic risk, due to the favourable outcomes in low-intermediate risk patients who received ET alone and high-risk patients treated with ET and CT. Therefore, prospective studies are needed to better validate this hypothesis.
Presentation numberPS3-10-22
An Online Tool to Assist Decisions on Chemotherapy for Men with Luminal Early Breast Cancer
Yuxuan Gao, First Affiliated Hospital of Sun Yat-sen Univsersity, Guangzhou, China
Y. Gao1, M. Zhang1, J. Nie2, G. Ye3, G. Sun4, L. Ma5, Z. Jiang6, Y. Lin1; 1Breast Disease Center, First Affiliated Hospital of Sun Yat-sen Univsersity, Guangzhou, CHINA, 2Breast Cancer Institute, Yunnan Cancer Hospital, Kunming, CHINA, 3Department of Breast Surgery, The First People’s Hospital of Foshan, Foshan, CHINA, 4Department of Breast and Thyroid Surgery, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, CHINA, 5Department of Breast Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, CHINA, 6Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, CHINA.
Background: Due to the rarity of male breast cancer (MBC), prognosis prediction and chemotherapy indication remain understudied. We aimed to develop a generalized model to predict outcomes and identify candidates for chemotherapy omission in luminal early-stage MBC.Methods: Luminal MBC patients from the Surveillance, Epidemiology, and End Results (SEER) program (2010-2018, N=2996) and a Chinese cohort (2000-2020, N=599) were included. SEER-derived patients (2010-2016) were split into training (N=1356) and test (N=905) sets in a 6:4 ratio; others formed the US validation set (2017-2018, N=735). Chinese patients constituted the CHN validation set. The primary outcome was overall survival (OS). Four machine learning models and a Cox-based nomogram were evaluated. Risk stratification (low-, intermediate-, and high-risk groups) was based on the tertiles of 5-year risk scores in the training set predicted by the optimal model. Survival analysis employed Kaplan-Meier, log-rank tests, and Cox regression.Results: The optimal model showed robust performance (C-indices: training set=0.715, test set=0.713, US validation set=0.742, CHN validation set=0.748). OS of three risk groups differed significantly across all datasets (all P<0.001). Chemotherapy conferred no OS benefit in low-risk patients [Univariate: training set, hazard ratio (HR)=1.87, 95% confidence interval (CI), 1.24 to 2.80, P=0.002; other sets, P=0.176, P=0.147, P=0.864, respectively. Multivariate: training set, HR=1.80, 95% CI, 1.04 to 3.10, P=0.035; test set, P=0.211; US validation set, HR=3.24, 95% CI, 0.98 to 10.73, P=0.054; CHN validation set, P=0.449]. This tool has been deployed online for free access.Conclusions: Our model accurately predicts 5-year OS rates of American and Chinese male patients with luminal early breast cancer. Its risk stratification may aid in identifying low-risk patients who may be safe to omit chemotherapy.
Presentation numberPS3-10-23
Retrospective study on breast cancer index testing in a community hospital and analyzing its impact on physician-decision making for extended endocrine therapy in early breast cancer
Mumta Essarani, Bayhealth Medical Center, Dover, DE
M. Essarani, S. Muhammad, R. Sawhney, S. McLellan; Internal Medicine, Bayhealth Medical Center, Dover, DE.
Background:The Breast Cancer Index (BCI) is a biomarker assay which provides objective data on benefit of extended endocrine therapy (EET) and risk of late recurrence in hormone receptor-positive early-stage breast cancer. Clinical trials have demonstrated the importance of its utilization in practice, however data on its influence on treatment decisions in a community hospital setting remains limited.Methods: We conducted a retrospective study from 2012 to 2020 involving 100 patients with early-stage breast cancer who underwent BCI testing at Bayhealth Medical Center. Data collected included basic demographic information, tumor pathology results, endocrine agent, BCI results, oncotype scores, physician recommendations for EET, recurrences, and survival outcomes. The primary objective was to assess the impact of BCI results on physician recommendations for EET. Physician decisions pre- and post-test were abstracted from clinic documentation and compared. Secondary outcomes included recurrence-free survival and treatment tolerability. Results: Analysis of the study included 100 patients (ER+/PR+) with a median age at diagnosis of 63 years and a median tumor size of 1.3 cm. Of the total cases, approximately 87% were HER2- and 29% were node positive. Endocrine therapy consisted of anastrozole (32%), letrozole (31%), tamoxifen (9%), and other agents. Approximately, 33% patients were noted to have high predictive benefit from EET and 65% were noted to have low benefit from EET. Before Breast Cancer Index (BCI) testing, 56% of patients had no recommendation regarding EET, 26% were advised to continue, and 18% were advised against it. Following BCI testing, 50% were advised to discontinue EET, 36% were advised to continue, and 14% did not follow the post-BCI recommendation due to patient preference. Overall, 75% of cases had a change in recommendation after BCI testing. Among those with no initial recommendation, 30% were later advised to discontinue EET and 20% to continue. Additionally, an initial recommendation to continue changed to discontinue in 8% of cases, while an initial recommendation to discontinue changed to continue in 3%. During a median follow-up of 7.9 years, there were seven recurrences and four deaths. Notably, no recurrences occurred among patients who adhered to more than five years of EET. Conclusions: In this retrospective analysis, BCI testing significantly influenced physician decision-making, leading to changes in EET recommendations in 75% of cases. These findings support the clinical utility of BCI in guiding personalized treatment strategies and minimizing unnecessary therapy in early breast cancer management. It also highlights the importance of its standardization in a community hospital to prevent over-treatment as well as encourage long-term adherence in patients with EET recommendations.
Presentation numberPS3-10-24
Biologic and Pathologic Correlations of the HER2DX Genomic Test in HER2+ Disease
Esther Sanfeliu, Hospital Clínic of Barcelona, Barcelona, Spain
E. Sanfeliu1, A. Martinez-Romero2, M. Marín-Aguilera3, S. Cobo2, B. González-Farré1, E. Hernandez4, P. Jares4, J. Puig-Butillé4, M. Muñoz5, R. Gómez-Bravo5, M. Tapia6, C. Tebar6, C. Saura7, S. Escrivà-de-Romaní7, J. Soberino8, J. Cortés9, S. Morales10, K. Amillano11, L. Paré3, P. Villagrasa3, W. Buckingham3, F. Pardo3, J. Parker12, F. Brasó-Maristany2, E. Ciruelos13, R. Sánchez-Bayona13, O. Martinez-Sáez5, J. Cejalvo6, A. Prat5; 1Pathology, Hospital Clínic of Barcelona, Barcelona, SPAIN, 2Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, SPAIN, 3Reveal Genomics, S.L., Reveal Genomics, S.L., Barcelona, SPAIN, 4Molecular Biology Core, Hospital Clínic of Barcelona, Barcelona, SPAIN, 5Medical Oncology, Hospital Clínic of Barcelona, Barcelona, SPAIN, 6Medical Oncology, Hospital Clínic of Valencia, Valencia, SPAIN, 7Medical Oncology, University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 8Medical Oncology, IOB-QuirónSalud, Barcelona, SPAIN, 9Medical Oncology, IBCC, Barcelona, SPAIN, 10Medical Oncology, Hospital Arnau de Vilanova, Lleida, SPAIN, 11Medical Oncology, Hospital Universitari Sant Joan de Reus, Tarragona, SPAIN, 12Medical Oncology, Reveal Genomics, S.L., Barcelona, SPAIN, 13Medical Oncology, Hospital Universitario 12 Octubre, Madrid, SPAIN.
HER2DX is a genomic test developed to guide treatment decisions in newly diagnosed HER2-positive (HER2+) breast cancer. It analyzes the expression of 27 genes grouped into 4 biological signatures: immune/immunoglobulin (IGG), luminal differentiation, proliferation, and HER2 amplicon expression. The test provides three distinct outputs: a prognostic relapse risk score, a pathologic complete response (pCR) likelihood score, and a quantitative ERBB2 mRNA score. This study investigates the relationship between HER2DX-derived outputs and standard histopathologic features in early-stage HER2+ breast cancer. The standardized HER2DX genomic test provided by Reveal Genomics was performed on paraffin tumor samples from newly diagnosed patients with early-stage HER2+ breast cancer, treated across 24 hospitals from January 2022 to June 2025. Histopathologic evaluation of hematoxylin-eosin-stained slides included tumor grade, stromal tumor-infiltrating lymphocytes (TILs) percentage, spatial TIL distribution, and presence of tertiary lymphoid structures (TLS). Hormone receptor (HR) status and Ki67 proliferative index were assessed locally by immunohistochemistry (IHC), while HER2 status was determined using IHC and in situ hybridization. Associations between HER2DX scores and pathologic features were assessed using appropriate statistical tests for continuous variables and categorical comparisons. HER2DX scores and their underlying biological signatures were analyzed in 410 HER2+ tumors. HR+ disease represented 75.7% of cases. The distribution of tumor grade, HER2 IHC status, HR status, Ki67 index, stromal TILs, TLS, and TIL spatial patterns is summarized in the Table. The HER2DX pCR score was significantly associated with all histopathologic features evaluated, the relapse-risk score five of these features (including Ki67 and TILs), and the HER2DX ERBB2 score with HER2 and HR status. Higher stromal TIL levels were moderately correlated with the IGG signature (r=0.59, p<0.001), and the inflamed TIL distribution was associated with increased IGG signature expression and higher HER2DX pCR scores (p<0.001). TLS presence also correlated with elevated IGG signature expression (p<0.001). HR+ tumors exhibited higher luminal signature expression (p<0.001), while HER2 IHC 3+ tumors had increased HER2 amplicon signature and HER2DX ERBB2 scores (p<0.001). Finally, higher Ki67 levels were moderately correlated with the proliferation signature (r=0.50, p<0.001). The HER2DX genomic test shows significant associations with multiple histopathologic features in early-stage HER2+ breast cancer, although no single feature can fully explain the outputs of the test. These findings support the notion that HER2DX integrates complex biological and histological dimensions, offering complementary information that may enhance personalized treatment decision-making.
Presentation numberPS3-10-26
Long-term outcomes of two her2 vaccines to prevent recurrence in patients with her-2 positive early stage breast cancer
Hyo Sook Han, Moffitt Cancer Center, Tampa, FL
H. S. Han1, A. Aldrich1, R. Weslowski2, C. Fisher3, S. Gandhi4, W. R. Gwin III5, M. Kowzun6, K. Gogineri7, H. Soliman1, R. Costa1, H. Liu1, J. Whiting1, M. Schell1, M. L. Disis5, B. Czerniecki1; 1Breast Oncology, Moffitt Cancer Center, Tampa, FL, 2Medical Oncology, Ohio State University Comprehensive Cancer Center, Columbus, OH, 3Surgery, Indiana University Shool of Medicine, Indianapolis, IN, 4Medicine, Rosewall Park Comprehensive Cancer Center, Buffalo, NY, 5Medical Oncology, University of Washington, Seattle, WA, 6Surgery, Rutgers Cancer Institute, Brunswick, NJ, 7Medical Oncology, Emory University School of Medicine, Atlanta, GA.
Background: Patients (pts) with residual invasive disease following neoadjuvant therapy for HER2-positive early stage breast cancer (EBC) have an increased risk of recurrences compared to pts with a complete pathologic response (pCR). It also has been observed that pts who do not achieve a pCR have low or absent anti-HER-2 CD4 Th1 responses. We hypothesize that boosting anti-HER-2 CD4 Th1 response using vaccines is safe, induces anti-tumor immunity in HER2-positive breast cancer and reduces the risk of recurrence. We conducted a multi-center, phase 2, randomized study to determine the safety, immunogenicity and recurrence free survival of two HER2 vaccines (multivalent anti-oncodriver DNA vaccine (WOKVAC) or HER-2-pulsed dendritic cell vaccine (DC1)). Methods: Pts with HER2-positive EBC were eligible if they had residual invasive disease at surgery after receiving neoadjuvant chemotherapy plus HER2-targeted therapy. Pts were randomly assigned in a 1:1 ratio, stratified by residual cancer burden (RCB) (1+2 vs 3) to receive adjuvant HER2 vaccination with either DC1 or WOKVAC. For the initial vaccination phase, DC1 was delivered via US guided inguinal lymph node administration, weekly x 6 weeks and WOKVAC was given intradermally on weeks 1, 4, and 7. Booster vaccines were given at months 6, 9 and 12. The primary end points were the safety and immunogenicity of each vaccine and the secondary end point was recurrence-free survival (RFS). Exploratory analyses include the assessment of prognostic and predictive biomarkers including circulating tumor cells, serum HER2 levels, and others immune correlative biomarkers. Here we report the immune response and updated long-term outcome data. RESULTS: 110 eligible pts (55 WOKVAC vs 55 DC1) were accrued from 2/2018 to 12/2022 from 7 academic institutions and received at least one dose of treatment. Most pts had clinical stage II/III 56/35 (83%) and 50% had node positive disease at diagnosis. 82% had hormone receptor positive disease. 37/38/8 patients had RCB 1/2/3. For patients with unknown RCB status, ypTNM was used to randomize (yp stage I/II/III=13/13/1 pts). 70.9% received adjuvant trastuzumab emtansine (T-DM1) following FDA approval in 2019. Most treatment related adverse events (TRAE) were grade 1 and 2 (previously reported). Eleven pts (6 in DC1 and 5 in WOKVAC) had recurrence with a median follow-up of 51.8 months. Of those 2/6 patients in the DC1 arm developed brain metastasis shortly after study enrollment during the induction phase. Of the remaining 9 pts with recurrence, 1 had a new primary cancer, 3 had local recurrence and 5 had distant recurrence. Three pts died from recurrent disease. RFS was 94.5% and 90.5% at 3 and 5 years respectively. Among pts who received adjuvant T-DM1, RFS was 95.9% at 3 and 5 years. HER2 immune response measured by ELISPOT (N=80) following vaccination demonstrated an average two-fold increase across both treatment groups at 12 months. CONCLUSION: Both DC1 and WOKVAC HER2 vaccines were well tolerated without significant toxicities. Both HER2 vaccines were immunogenic and associated with persistence of immunity at 12 months follow up. Our results (RFS) compare favorably with the KATHERINE trial suggesting potential additional benefit of adjuvant vaccines for these high-risk pts populations. Several studies are ongoing to evaluate the addition of neoadjuvant DC1 and WOKVAC to HER2 targeted therapies. (NCT 05325632/04329065) ClinicalTrials.gov Identifier: NCT03384914
Presentation numberPS3-10-28
Randomized phase II trial evaluating three anti-diarrheal prophylaxis strategies in patients (pts) with HER2+/HR+ early breast cancer (EBC) treated with extended adjuvant neratinib (dianer geicam/2018-06)
Juan Miguel Gil-Gil, ICO L’Hospitalet; GEICAM Spanish Breast Cancer Group, Barcelona, Spain
J. Gil-Gil1, M. Ruíz-Borrego2, E. Carrasco3, Y. Izarzugaza4, C. Martínez-Vila5, E. Galve6, B. López-Barajas7, E. Adrover8, T. Silovski9, M. Valero-Arbizu10, Á. Guerrero-Zotano11, S. González-Santiago12, M. Echarri13, A. Cano-Jiménez14, A. Vethencourt-Casado1, M. De Laurentis15, C. Bueno-Muiño16, B. Adamo17, R. Andrés18, C. Villanueva19, F. Rojo20, M. Casas21, P. Wilson22, J. Suissa23, V. Adams24, M. Martin25; 1Medical Oncology, ICO L’Hospitalet; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 2Medical Oncology, Hospital Universitario Virgen del Rocío; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 3Scientific department, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 4Medical Oncology, Hospital Universitario Fundación Jiménez Díaz; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN, 5Medical Oncology, ALTHAIA Xarxa Asistencial de Manresa; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 6Medical Oncology, Hospital Universitario de Basurto; GEICAM Spanish Breast Cancer Group, Bilbao, SPAIN, 7Medical Oncology, Hospital Universitario Clinico San Cecilio; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 8Medical Oncology, Complejo Hospitalario Universitario de Albacete; GEICAM Spanish Breast Cancer Group, Albacete, SPAIN, 9Medical Oncology, Department of Oncology, University Hospital Center Zagreb; School of Medicine, University of Zagreb, Zagreb, CROATIA, 10Medical Oncology, Hospital Quirónsalud Sagrado Corazón; GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 11Medical Oncology, Instituto Valenciano de Oncologia (IVO); GEICAM Spanish Breast Cancer Group, Valencia, SPAIN, 12Medical Oncology, Hospital Universitario San Pedro de Alcántara; GEICAM Spanish Breast Cancer Group, Cáceres, SPAIN, 13Medical Oncology, Hospital Universitario Severo Ochoa; GEICAM Spanish Breast Cancer Group, Leganés, SPAIN, 14Medical Oncology, Hospital Universitario de Jaen; GEICAM Spanish Breast Cancer Group, Jaén, SPAIN, 15Medical Oncology, Chair, Dept. Breast and Thoracic Oncology, Istituto Nazionale Tumori IRCCS “Fondazione Pascale”, Napoli, ITALY, 16Medical Oncology, Hospital Universitario Infanta Cristina; GEICAM Spanish Breast Cancer Group, Parla, SPAIN, 17Medical Oncology, Hospital Clinic de Barcelona; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 18Medical Oncology, Hospital Clinico Universitario Lozano Blesa; GEICAM Spanish Breast Cancer Group, Zaragoza, SPAIN, 19Medical Oncology, Clinique Clementville Montpellier, Montpellier, FRANCE, 20Medical Oncology, Hospital Universitario Fundación Jiménez Díaz; GEICAM Spanish Breast Cancer Group, Barcelona, SPAIN, 21Statics department, GEICAM Spanish Breast Cancer Group, San Sebastián de los Reyes, SPAIN, 22Medical Oncology, Puma Biotechnology Inc, San Francisco, CA, 23Medical Oncology, Pierre Fabre Medicament, Boulogne-Billancourt, FRANCE, 24Medical Oncology, Breast International Group, Woluwe-Saint-Lambert, BELGIUM, 25Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón, Medicine Department, Universidad Complutense; GEICAM Spanish Breast Cancer Group, Madrid, SPAIN.
Background: In the ExteNET clinical trial, extended adjuvant treatment with neratinib (NER), reduced theabsolute risk of recurrence or death by 5.1% at 5 years versus (vs.) placebo in HR+/HER2+ EBC pts whoinitiated treatment ≤1 year post trastuzumab (T) (Chan 2021). Diarrhea was the main reason for NERdiscontinuation in the first 3 cycles (c) (1 c=28 days [d]). DIANER further studied 3 previously reportedstrategies for managing diarrhea (CONTROL: Chan 2023) in pts receiving NER.Methods: DIANER is a European, controlled, randomized, phase II study in pts with HER2+/HR+ EBC (stageIB-IIIC) who completed prior neo/adjuvant T-based therapy within 1 year. Pts were randomized 1:1:1 toArm A: NER (240 mg/d x 1 year) + loperamide (LP) (12 mg/d x 14 d → 8 mg/d till end of c 2 → as needed[PRN]), Arm B: NER dose escalation (120 mg/d x 7 d → 160 mg/d till d 14 → 240 mg/d x 13 c) + LP PRN,or Arm C: NER (like Arm A) + LP (12 mg/d x 14 d → 8 mg/d x 14 d → PRN) + colesevelam (3,750 mg/d x 28d). Pts were stratified by menopausal status and prior anti-HER2 therapy (T vs. T + pertuzumab). Theprimary endpoint was the rate of early (in the first 3 c) NER discontinuations due to diarrhea. Simon’soptimal two-stage design was used with a target of 5% (H1) vs. 13% (H0). In the first stage, up to 36 ptsper arm were to be enrolled. Arms with ≥ 4 early discontinuations due to diarrhea were planned to beclosed, otherwise recruitment would continue up to 105 pts per arm. Secondary endpoints were the rateof NER discontinuations (any reason); AEs and hospitalizations; duration, severity, and treatments fordiarrhea; NER exposure.Results: From Sept 2022 to Oct 2024, 177 pts were randomized (41 arm A, 107 arm B, 29 arm C) at 47sites. Median age was 51 years (range: 30-80), 50.3% of pts were postmenopausal, and 39.0%, 28.8% and15.3% had stage IIA, IIB and IIIA disease, respectively. Prior pertuzumab and T-DM1 were received by76.3% and 43.5% of pts, respectively. Arms A and C were closed due to 5 and 4 pts with earlydiscontinuation due to diarrhea, respectively. Early discontinuations due to any reason/diarrhea were25.6%/17.9% in arm A, 13.3%/8.6% in arm B and 27.6%/13.8% in arm C. For the first 3 c, mean (standarddeviation)/median (range) duration of NER exposure was 67 d (32)/84 d (4-101) in arm A, 78 d (19)/84 d(12-105) in arm B and 65 d (34)/84 d (1-91) in arm C. Median (range) relative NER dose intensity was 98%(14-107) in arm A, 99% (28-109) in arm B and 95% (4-105) in Arm C. Most common TEAEs within the first3 c are shown in Table 1.Conclusions: Although none of the 3 strategies for reducing the impact of NER-associated diarrheaassessed in DIANER reduced early treatment discontinuation due to diarrhea to ≤5%, the incidence ofearly NER discontinuation observed in the study suggests that NER dose escalation is the best strategy formanaging diarrhea and other AEs.
Presentation numberPS3-10-29
Ner-tree study interim analysis of safety and patient-reported outcomes in patients with human epidermal growth factor receptor 2 positive (HER2+) early breast cancer (eBC) treated with neratinib in China
Xiaojia Wang, Zhejiang Cancer Hospital-Cancer Research Institute, Hangzhou, China
X. Wang1, X. Liu2, H. Liu3, S. Ma4, Y. Zhang5, J. Gao6, J. Ye7, J. Ma8, A. Zhang9, Y. Wang10, X. Wu11, J. Wu12, Y. Chen13, S. Zhang14, Q. Ouyang15, G. Jiang16, J. Liu17, F. Tian18, S. Yang19, J. Zhang20; 1Department of Medical Oncology (Breast), Zhejiang Cancer Hospital-Cancer Research Institute, Hangzhou, CHINA, 2Department of Medical Oncology, General Hospital of Ningxia Medical University, Yinchuan, CHINA, 3Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, CHINA, 4Breast Center, Zhongshan City People’s Hospital, Zhongshan, CHINA, 5Department of Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, CHINA, 6Department of Breast Surgical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, CHINA, 7Department of Breast Oncology, Zhaoqing First People’s Hospital, Zhaoqing, CHINA, 8Department of Breast Surgery, Tangshan People’s Hospital, Tangshan, CHINA, 9Breast Disease Control Center, Maternal and Child Health Care Hospital of Guangdong Province, Guangzhou, CHINA, 10Breast Cancer Center, Shandong Cancer Hospital, Shandong University, Jinan, CHINA, 11Department of Breast Cancer, Hubei Cancer Hospital, Wuhan, China, Wuhan, CHINA, 12Department of Breast, Cancer Hospital of Shantou University Medical College, Shantou, CHINA, 13Department of Breast Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, CHINA, 14Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, CHINA, 15Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, CHINA, 16Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, CHINA, 17Department of Breast Surgery, The First Affiliated Hospital of Soochow University, Suzhou, CHINA, 18Breast Diagnosis and Treatment Center, Shanxi Provincial Cancer Hospital, Taiyuan, CHINA, 19Department of Breast Surgery, Gansu Cancer Hospital, Lanzhou, CHINA, 20Third Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, Tianjin, CHINA.
Background Neratinib, an oral irreversible pan-HER tyrosine kinase inhibitor, was approved in 2020 in China for the extended adjuvant treatment of HER2+ early breast cancer (EBC) adult patients. Diarrhea is the most common treatment-related adverse event associated with neratinib and an important factor impacting patients’ quality of life during therapy. Evaluating the effectiveness of anti-diarrheal prophylaxis measures and patient-reported outcomes in real-world settings is essential. Method NER-Tree is an ongoing, prospective, multi-center, non-interventional study (NCT05491057) that aims to enroll 500 patients with HER2+ early breast cancer (EBC) starting 1-year extended adjuvant neratinib. Diarrheal prophylaxis for neratinib was not required. Prespecified interim analyses are planned after the recruitment of 250 and 500 patients. Primary and secondary objectives are to characterize real-world treatment patterns and assess neratinib’s safety profile including diarrhea, respectively. The exploratory objectives include patient-reported quality of life (QoL), and pattern of breast cancer recurrences. Here we report safety data in the second interim analysis, focusing on diarrhea management and patient-reported outcomes. Results As of 28 October 2024, 500 patients were included in the analysis. Diarrhea (86.4%), nausea (18.2%), fatigue (15.6%), and decreased appetite (14.4%) were the most common AEs of any grade. Serious adverse events (SAEs) were reported in 3.4% of the patients. Diarrhea was predominantly reported as Grade 1 or 2 (69.6%), while Grade 3 diarrhea was reported in 16.8% of the patients. No grade 4 diarrhea was observed. The median time from the initiation of neratinib treatment to the onset of diarrhea was 3.0 days (interquartile range [IQR] 2.0,6.0). Diarrhea was the most common adverse event leading to dose reduction (14.4% of cases vs. 18.2% for all adverse events combined), dose interruption (18.0% vs. 33.6%), and permanent discontinuation (4.2% vs. 6.8%). Patients who initiated neratinib at <240 mg experienced a lower incidence of Grade 3 diarrhea compared to those who started at 240 mg (12.2% vs. 26.1%). Anti-diarrheal strategies were adopted at least once in 297 patients (59.4%), including drug prophylaxis (loperamide or other agents; 18.0%), dose escalation (28.0%), or a combination of both strategies (13.4%). Compared to no prophylaxis (28.6%, 32/112), all strategies reduced the incidence of Grade ≥3 diarrhea: most substantially with the combination approach (9.0%, 6/67), followed by dose escalation (17.9%, 25/140) and drug prophylaxis (20.8%, 11/53).Neratinib initiation induced concomitant transient deteriorations in patient-reported outcomes, peaking at Day 30 across all instruments. The EQ-5D-5L showed maximal declines in Health Status (-4.5) and Utility Index (-0.0492), while FACIT instruments revealed greater disease-specific impacts: FACIT-F Trial Outcome Index (fatigue/function) decreased by 10.31 points, FACIT-G Total (general QoL) by 4.85, and FACIT-D Total (breast cancer-specific) by 11.58. All metrics demonstrated progressive recovery through Day 60 toward near-baseline levels by Day 180. Conclusion This study provides further insights on the pattern of diarrhea associated with neratinib and preventive anti-diarrheal measures. Dose escalation and drug prophylaxis combined strategy was the most effective approach in reducing the incidence of Grade 3 diarrhea associated with neratinib. Patient-reported outcomes showed an initial deterioration until Day 30 during neratinib treatment initiation, possibly related to the occurrence of treatment-emergent adverse events, followed by a functional/QoL recovery afterward.
Presentation numberPS3-10-30
A clinical model to predict pathologic complete response using early peripheral absolute lymphocyte dynamics in HER2+ breast cancer
Colin Bergstrom, Stanford, Stanford, CA
C. Bergstrom1, I. Luo2, E. Kotler1, M. Satoyoshi3, S. Philip1, C. Curtis1, A. Kurian1, S. Han3, J. Caswell-Jin1; 1Department of Medicine, Stanford, Stanford, CA, 2Quantitative Sciences Unit, Stanford, Stanford, CA, 3Department of Epidemiology and Population Health, Stanford, Stanford, CA.
Introduction: Immune activation and lymphocyte infiltration into the tumor microenvironment are known to predict treatment response in HER2-positive (HER2+) breast cancer. Our work has shown that dynamic changes in peripheral absolute lymphocyte count (ALC) following initial neoadjuvant therapy (NAT) are also associated with a higher likelihood of achieving pathologic complete response (pCR). In this study, we aimed to develop and validate a predictive model for pCR based on clinically obtained pre-treatment ALC and ALC following a single dose of NAT, with the goal of creating a tool for early response prediction in HER2+ breast cancer. Methods: We assembled a retrospective discovery cohort of patients with HER2+ breast cancer treated with trastuzumab-based NAT from the Oncoshare database, which integrates electronic medical records and California Cancer Registry data from patients diagnosed between 2000 and 2020 at Stanford University. Our non-overlapping validation cohort consisted of patients with HER2+ breast cancer who underwent NAT on clinical trials of HER2-targeted therapy and/or who were enrolled in prospective institutional tissue collection studies. In the discovery cohort, we constructed a multivariable logistic regression model to identify predictors of pCR, including baseline ALC, early ALC change, days between baseline and on-treatment ALC draws, age at diagnosis, estrogen receptor (ER) status, stage, grade, and self-reported race/ethnicity. Early ALC change was defined as 1 minus the ratio of cycle 1 ALC to baseline ALC. To develop an ALC-based risk grouping strategy, we used logistic regression to create a composite risk score from baseline ALC and early ALC change, followed by receiver operating characteristic curve analysis. We then applied grid search optimization to test combinations of cutoff points across the 10th to 90th percentiles of the risk score distribution and identified the pair of cutoffs that maximized area under the curve (AUC) when patients were stratified into three risk categories. Model performance was evaluated using AUC analysis in both the discovery and validation cohorts. Results: In the discovery cohort (n = 154), higher baseline ALC and greater early ALC decline were independently associated with increased odds of pCR. The composite ALC risk score, using baseline ALC and early ALC change, stratified patients into three groups: low (25% of patients, n = 39), intermediate (45%, n = 69), and high (30%, n = 46) probability of pCR. Observed pCR rates in these groups were 15%, 51%, and 76%, respectively. When this ALC-based risk grouping was integrated with the other covariates (including time interval between ALC draws, age, ER status, clinical stage, tumor grade, and race/ethnicity), the full multivariable model demonstrated strong predictive performance with an area under the curve (AUC) of 0.81 (95% CI 0.74-0.88). In the separate validation cohort (n = 83), the composite ALC risk score stratified patients into low (30% of patients, n=25), intermediate (48% of patients, n=40), and high (22% of patients, n=18) probability of pCR groups, with observed pCR rates of 12%, 63%, and 78%, respectively. The model showed similar discriminative ability in the validation cohort, with an AUC of 0.81 (0.71-0.90). Conclusion: Early ALC dynamics, with no other covariates, effectively stratify patients with HER2+ breast cancer based on their likelihood to achieve pCR after NAT. We developed and validated a clincal model using routinely collected laboratory values, which achieved strong predictive performance in both discovery and validation cohorts. This ALC-based tool offers a cost-effective and scalable alternative for early response prediction, with potential to inform treatment adaptation strategies in real time.
Presentation numberPS3-11-01
Steroidogenic acute regulatory protein related lipid transfer domain containing-3 n-terminal like(STARD3NL) expression in clinical breast cancer and its connection with metastatic lymph node 64 (MLN64) in breast cancer
Hong Yu Xiang, Cardiff University, CARDIFF, United Kingdom
H. Y. Xiang1, H. Y. Xiang2, Y. R. Chen1, C. Wang1, X. Y. Li1, F. Askr1, T. A. Martin1, K. Mokbel1, W. G. Jiang1; 1School of Medicine, Cardiff University, CARDIFF, UNITED KINGDOM, 2Department of Thyroid and Breast Surgery, Peking University First Hospital, Beijing, CHINA.
Introduction StarD3NL (StAR Related Lipid Transfer Domain Containing-3 N-Terminal Like), also known as MENTHO, is a tethering protein. It allows the endoplasmic reticulum (ER) and late endosomes to make contact and to intracellularly transport cholesterol from ER to other endosomes and cytoplasmic membrane where cholesterol known to play a role in organising membrane integral proteins for their function. This is a role similarly held by MLN64 (Metastatic Lymph Node Gene 64 Protein) (also known as StarD3, StAR Related Lipid Transfer Domain Containing 3). We have previously established that MLN64 has an important role in the cell adhesion of cancer cells and a clinical role in clinical breast cancer. There is little knowledge on the expression of StarD3NL in breast cancer. Methods Quantitative PCR (qPCR) was used to assess transcript levels in a Cardiff cohort of breast cancer and normal tissues (151 samples) with a 10 year follow up. Clinical and pathological outcomes were compared and results analysed using Mann Whitney U test for comparisons, logistic regression and Kaplan-Meier’s methods for survival analysesResults StarD3NL levels were seen to be lower in patients with moderate and poor prognosis compared to those with good prognosis and lower in node positive tumours (p=0.046). When assessed against clinical outcome, it was found that patients who developed breast cancer related incidence had significantly lower levels of StarD3NL when compared with those who remained disease free (p=0.0086). This was reflected in the survival analysis in which patients with lower levels of StarD3NL had a significantly shorter disease free survival (DFS) (p=0.025, Hazard Ratio (HR)=0.329 (95% confidence internal 0.116-0.933)). A similar trend was seen with overall survival (OS) although this did not reach statistical significance. The link between StarD3NL and DFS was more prominent in HER2 positive and HER3 negative tumours, and in non-TNBC breast cancers. StarD3NL expression appears to be significantly correlated with MLN64 in both breast cancer (r=0.410, p<0.001) and in normal mammary tissues (r=0.679, p<0.001). This was observed in that the subgroup with low levels of MLN64 also demonstrated that StarD3NL had a more striking connection with DFS (p=0.009) and indeed overall survival (p=0.043). Conclusion StarD3NL appears to be significantly related to breast cancer patient disease free survival, which was pronounced in HER2 positive and HER3 negative tumour types; moreover, this change was linked to MLN64 expression. These data suggest that these two proteins could be an interesting target for future prognostic development.
Presentation numberPS3-11-02
Exploring heterogeneity of HER2 immunohistochemistry staining in patients undergoing neoadjuvant breast cancer treatment in two cancer centers of South America.
Federico Waisberg, Instituto Alexander Fleming, Buenos Aires, Argentina
M. Ancao1, M. Costanzo2, M. Gonzalez Donna3, M. Pombo4, M. Nally4, M. Amat4, A. Ostinelli2, S. Rivero2, L. Lapuchesky2, A. Nervo2, J. Nadal2, C. Loza1, F. Colo1, V. Fabiano1, A. Perazzollo1, L. Sabatini1, T. Vargas2, A. Pavetti3, M. Cabañas Leon5, F. Waisberg2; 1Breast Surgery, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 2Medical Oncology, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 3Medical Oncology, INCAN, Asuncion, PARAGUAY, 4Pathology, Instituto Alexander Fleming, Buenos Aires, ARGENTINA, 5Pathology, INCAN, Asuncion, PARAGUAY.
Background Intra-tumoral heterogeneity (ITH) is a biological characteristic associated with various key tumor biomarkers in different cancer models. Heterogeneous expression of HER2 has been observed in around 10-40% of patients with early breast cancer (eBC), and its determination has been associated with a worse response to HER2-directed therapies. While ITH characterization has commonly been addressed using gene amplification tests, international guidelines recommend identifying it in immunohistochemistry (IHC) evaluations of HER2 staining Materials and Methods We included patients with HER2-3+ eBC diagnosis that underwent neoadjuvant treatment in two cancer centers: Instituto Alexander Fleming (Buenos Aires, Argentina) and Instituto Nacional del Cáncer (Asunción, Paraguay). ITH was retrospectively evaluated in the initial tumor biopsies by two different pathologists in each center, and defined as the presence of at least 10% of tumor cells without complete or intense membrane staining. Univariate and multivariate models were used to understand the role of ITH to predict pathological complete response (pCR). Results 116 patients were incorporated in our cohort (female 100%; median age 49[IQR 43-56.3]; clinically axillary involvement 51.7%). ITH was observed in 15 cases (12.9%), and associated with numerically higher median estrogen receptor (ER) expression (90 [IQR 0-98] vs. 5 [IQR 0-90]; p=0.07). The pCR in our sample was 68.9%, with rates of 26.7% and 68.3% in patients with and without ITH diagnosis, respectively (p=0.004). pCR remained negatively associated with ITH (OR 0.15; CI95% 0.03-0.54; p=0.006) and ER expression (OR 0.24; CI95% 0.09-0.6; p=0.003) in multivariate models after other key variables such as axillary involvement, age and tumor size were concomitantly analyzed. Conclusions ITH defined per IHC analysis was one of the main factors that explained residual disease in our cohort. Our results support the routinary determination of ITH for HER2-staining in initial biopsy results, and the evaluation of this biomarker in novel prognostic assays, as well as in modern clinical trials exploring the role of conjugated antibodies in the eBC setting.
Presentation numberPS3-11-03
Pathologic Complete Response and Surgical Outcomes, in a Brazilian Cohort.
Fabiana Makdissi, Accamargo Cancer Center, São Paulo, Brazil
M. Canal, R. Santos, S. Santos, M. Tavares, A. Bitencourt, M. Andrade, S. M. Sanches, F. Makdissi; Breast Cancer, Accamargo Cancer Center, São Paulo, BRAZIL.
Pathological Complete Response and Surgical Outcome, in a Brazilian Cohort.Introduction: Neoadjuvant chemotherapy (NACT) is a treatment strategy for locally advanced tumors and even for operable tumors in situations that can determine prognosis, modify adjuvant treatment and reduce surgical intensity.Method: This study evaluated Brazilian patients undergoing NACT from 2013-2023 in a Cancer Center in Brazil, through a retrospective cohort, analyzing the chemotherapy (CT) performed, pathological complete response rate (pCR) and surgical outcomes, the latter particularly in the HER2 positive (+) and Triple Negative (TN) population.Results: A total of 1,626 female patients were included, median age at diagnosis 46. The most frequent tumor subtype was luminal B (38.4%), followed by Her2 + (29.5%, including luminal) and TN (27.4%). Locally advanced tumors (T3-T4) accounted for 53.5% of cases and 67.2% were cN+ (cN1- 45.2%, cN2 -16.1% and cN3- 5.9%). Of the breast surgeries, 33.9% were conservative (BCS), 61.4% mastectomies, 4.7% nipple-sparing. Of the axillary surgeries, 38.0% were sentinel lymph node biopsies (SLNB), 67.1% lymphadenectomies (LA). The pCR occurred in 38.3% of cases, but was higher depending on the treatment modality, as shown in the table 1. The high-volume disease also influenced the pCR: N1 (16.4%) compared to N3 (1.6%, p<0.05). BCS were more frequent in the TN group than in the luminal group (11.7% vs. 0.4%; p<0.05). In HER2+ sample, BSC was significantly higher (23.6%) in the group that received double target therapy (DTT) (trastuzumab only, 12.2%; p<0.05) , as well more number of SLNB (27.6%/DTT group vs. 14.7%/trastuzumab group; p<0.05). Of the total patients, 80.1% were alive without disease, 20.6% showed progression and 11.4% died of BC.Discussion: Achieving a pCR is one of the objectives of NACT, especially for TN/HER2 + tumors with an impact on survival (Cotazar P et al, 2014.) and on the definition of adjuvancy. In addition, another objective of NACT is to promote BCS, as seen in our series, which was also higher in the TN group than in the Luminal group. Mattar et al. evaluated patients treated with NACT in Brazil, and showed an overall pCR rate of 22.7%, lower than in this study (38.3%), probably due to the disparity in access between public and private treatments in the country. The pCR rate in this study was also higher than in other studies published in Latin America, such as the study by Acevedo F et al. and VillaReal Garza C et al., ranging from 24.1% to 28.1%. Anti-HER2 DTT started (2015), we observed in our series an absolute increase in pCR of 11%, and after the brazilian approval of pembrolizumab, an absolute increase of 8% in pCR of TN, a fact that contributes to the higher number of BCS.Conclusion: The reproduction of NACT regimens carried out in pivotal studies in the general population achieves comparable results in terms of pCR and contributes to promoting more BCS, especially in the TN/HER2 + group.
| Treament / subtypes | n | pCR | RCB=1 | RCB=2 | RCB=3 |
| cT / pertuzumab + trastuzumab | 236 | 63.6% | 13.1% | 22.9% | 8.9% |
| cT / trastuzumab | 175 | 53.1% | 11.4% | 22.9% | 12.6% |
| NACT alone (to TN) | 322 | 50.6% | 8.7% | 26.7% | 14.0% |
| NACT / pembrolizumab (to TN) | 86 | 58.1% | 15.1% | 22.1% | 4.7% |
| Luminal A | 48 | 4.2% | 6.3% | 47.9% | 41.7% |
| Luminal B | 584 | 16.6% | 9.2% | 45.2% | 28.9% |
Presentation numberPS3-11-04
Adjuvant Ado-Trastuzumab Emtansine (T-DM1) for Older Patients with HER2+ Breast Cancer – Results from the ATOP Study
Rachel A. Freedman, Dana-Farber Cancer Institute, Boston, MA
R. A. Freedman1, H. Heiling2, K. J. Ruddy3, L. Rogak4, M. Sedrak5, A. Dueck6, H. Liu7, A. Fraettarelli1, S. Atanasov1, M. DeMeo1, P. Lange1, A. Perilla Glen1, S. Sinclair8, D. Dillon9, J. Mortimer10, M. Fenton11, J. Walsh12, L. Spring13, H. Muss14, N. Sinclair15, E. Hamilton16, C. Dang17, S. Giordano18, M. Faggen19, S. Lo20, C. Kuzma21, S. Lemke22, P. Stella23, R. Mowat24, M. Wilkinson25, J. Rauch26, N. Tayob2, E. Winer27; 1Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Data Science, Dana-Farber Cancer Institute, Boston, MA, 3Medical Oncology, Mayo Clinic, Rochester, MN, 4Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, 5Medical Oncology, UCLA, Santa Monica, CA, 6Department of Quantitative Health Sciences, Mayo Clinic, Phoenix, AZ, 7Alliance Statistical and Data Management Center, Mayo Clinic, Rochester, MN, 8Medical Oncology, Northern Light Cancer Care, Brewer, ME, 9Pathology, Brigham and Women’s Hospital, Boston, MA, 10Medical Oncology, City of Hope, Duarte, CA, 11Medical Oncology, Warren Alpert Medical School of Brown University, Providence, RI, 12Medical Oncology, Dana-Farber New Hampshire Oncology, Londonderry, NH, 13Medical Oncology, Mass General Brigham Cancer Center, Boston, MA, 14Medicine, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, 15Medical Oncology, Dana-Farber Brigham Cancer Center – Foxborough, Foxborough, MA, 16Medical Oncology, Sarah Cannon Research Institute, Nashville, TN, 17Medical Oncology, Memorial Sloan Kettering Cancer Center, West Harrison, NY, 18Medical Oncology, Dana-Farber Cancer Institute at Boston Medical Center- Brighton, Brighton, MA, 19Medical Oncology, Dana-Farber Cancer Brigham Cancer Center at South Shore Health, Weymouth, MA, 20Medical Oncology, Stamford Health, Stamford, CT, 21Medical Oncology, First Health of the Carolinas Cancer Center, Pinehurst, NC, 22Medical Oncology, Upstate University Hospital, Syracuse, NY, 23Medical Oncology, Trinity Health, Ypsilanti, MI, 24Medical Oncology, Toledo Clinic Cancer Centers, Toledo, OH, 25Medical Oncology, Sibley Memorial Hospital, Johns Hopkins, Washington, DC, 26Medical Oncology, Rex Hematology Oncology Asssociates-Garner, University of North Carolina, Garner, NC, 27Medical Oncology, Smilow Cancer Hospital at Yale, New Haven, CT.
Purpose: There are limited prospective data on the efficacy, tolerability, and functional impact of treatments for older and frail adults with HER2+ early breast cancer. We report the first-ever, US-based adjuvant trial dedicated to this population. Methods: We conducted a single arm, phase II, multicenter, investigator-initiated study of adjuvant ado-trastuzumab emtansine (T-DM1) in patients aged 60 and older with stage I-III HER2+ breast cancer who either declined standard adjuvant therapy or were deemed by their physician not to be candidates for standard treatment for any reason. Protocol therapy included postoperative administration of T-DM1 (3.6 mg/kg) every 21 days for one year (17 cycles). The primary endpoint was 5-year invasive disease-free survival (iDFS) with a 2-sided 90% confidence interval. Secondary endpoints presented here are toxicity and 3-year iDFS, invasive breast cancer-free survival (iBCFS), and overall survival (OS). All survival analyses were evaluated using Kaplan Meier methods and began at the time of C1D1 of T-DM1. Results: ATOP enrolled 111 evaluable patients during 2015-2020; median follow-up is 4.9 years (IQR 4.2-5.5). The median age was 71 (range 60-88); 50.4% were aged >70 and 11.7% were aged >80. Overall, 69.4% had stage I, 26.1% had stage II, and 4.5% had stage III disease; 70.3% had hormone receptor-positive tumors. The 5-year iDFS = 84.2% (90% CI 77.1-92.0%). The 3-year iDFS = 91.2% (90% CI 85.9%, 96.9%) and iBCFS = 94.1% (90% CI 89.7-98.8%) (see Table for event details); 3-year OS = 96.1% (90% CI 92.4-99.9%). In total, there were 3 distant recurrences and 2 breast cancer-related deaths. Overall, 82% reported grade 2+ toxicity during treatment; the most common grade 2+ events were fatigue (30.6%) and peripheral sensory neuropathy (19.8%). The most frequent grade 3 events were liver function abnormality, anemia, decreased neutrophil count, and neuropathy (<5% for each); one patient (0.9%) had grade 4 thrombocytopenia. Sixty-five (58.6%) completed all 17 cycles of protocol therapy; 27 (24.3%) completed 10-16 cycles, and 19 (17.1%) completed 1-9 cycles. Reasons for early treatment discontinuation in the remaining 41.4% included toxicity (28.8%), patient withdrawal (6.3%), other (3.6%), disease progression (0.9%), death on study (0.9%, deemed unrelated to treatment), and other complicating disease (0.9%). Conclusions: The ATOP trial demonstrated a favorable 5-year iDFS and manageable toxicity among older adults with early stage HER2+ breast cancer. Although iDFS was numerically worse than historical controls (i.e., 5-year iDFS in the ‘ATEMPT’ trial by Tolaney et al. J Clin Oncol, 2024 was 97% [95% 95.2-98.7]), ATOP’s participants had higher-risk cancers, advanced age, and more competing non-breast cancer events. Future endpoints will include in-depth analyses of patient-reported outcomes, biomarkers of aging, and longitudinal functional status.
| iDFS events (all invasive recurrences, any new invasive cancer, any-cause deaths) | n (%) |
| Ipsilateral invasive breast cancer | 1 (0.9) |
| Contralateral invasive breast cancer | 3 (2.7) |
| Distant recurrence | 3 (2.7) |
| New primary non-breast cancer | 7 (6.3) |
| Death due to breast cancer | 2 (1.8) |
| Death due to other reason (pulmonary infection, deemed not related to treatment) | 1 (0.9) |
| Alive at last follow-up | 94 (84.7) |
| iBCFS events (invasive breast cancer-related events or any-cause deaths) | n (%) |
| Ipsilateral invasive breast cancer | 1 (0.9) |
| Contralateral invasive breast cancer | 3 (2.7) |
| Distant recurrence | 3 (2.7) |
| Death due to breast cancer | 2 (1.8) |
| Death due to other reason | 1 (0.9) |
| Alive at last follow-up | 94 (84.7) |
| Censored at new primary non-breast invasive cancer | 7 (6.3) |
| Site of distant recurrence | n (%)
[n=3]
|
| Chest Wall | 2 (66.7) |
| Liver | 1 (33.3) |
| Site of new primary non-breast cancer | n (%)
[n=7]
|
| Skin | 3 (42.9) |
| Colon | 1 (14.3) |
| Malignant tumor of extrahepatic bile duct | 1 (14.3) |
| Pleura mesothelioma | 1 (14.3) |
| Rectal | 1 (14.3) |
Presentation numberPS3-11-05
Trastuzumab Combined with Pyrotinib and Capecitabine as Postoperative Adjuvant Therapy in Non-Pathological Complete Response HER2-Positive Early Breast Cancer Following Neoadjuvant Therapy: A Multicenter Phase II Study
Jie Zhang, Fujian Medical University Union Hospital, Fuzhou, China
C. Wang1, F. Fu1, J. Zhang1, S. Lin1, Z. Song2, Z. Ouyang3, X. Chen4, M. Lin5, G. Cui6, G. Han7, W. Guo1, X. Chen8, L. Jia9, C. Song8, S. Yang10, R. Luo11, Y. Yan12, Y. Zeng8, D. Chen13, J. Lin14, F. Yang15, R. Huang16, Y. Wang17, Z. Zeng18, Z. Zhang19; 1Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, CHINA, 2Department of Breast Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, CHINA, 3Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, CHINA, 4Department of Breast Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, CHINA, 5Department of Breast Surgery, Fujian Provincial Hospital, Fuzhou, CHINA, 6Department of Breast Surgery, Cangzhou Central Hospital, Cangzhou, CHINA, 7Department of Breast Surgery, Shanxi Cancer Hospital, Taiyuan, CHINA, 8Department of Breast Surgery, Fujian Cancer Hospital, Fuzhou, CHINA, 9Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, CHINA, 10Department of Breast Surgery, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, CHINA, 11Department of Breast Surgery, Cangzhou Hospital of Integrated Traditional Chinese and Western of Hebei Province, Cangzhou, CHINA, 12Department of Breast Surgery, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, CHINA, 13Department of Breast Surgery, Quanzhou First Hospital, Quanzhou, CHINA, 14Department of Breast Surgery, The Second Attached Hospital Of Fujian Medical University, Quanzhou, CHINA, 15Department of Breast Surgery, Fuzhou First General Hospital Affiliated with Fujian Medical University, Fuzhou, CHINA, 16Department of Breast Surgery, Maternity & Child Care Center of Qinhuangdao, Qinhuangdao, CHINA, 17Department of Breast Surgery, Nanchang People’s Hospital, Nanchang, CHINA, 18Department of Breast Surgery, Zhangzhou Municipal Hospital of Fujian Province, Zhangzhou, CHINA, 19Department of Breast Surgery, Mindong Hospital Affiliated to Fujian Medical University, Ningde, CHINA.
Background: HER2-positive early breast cancer patients achieving a pathological complete response (pCR) during neoadjuvant therapy demonstrate a more favorable prognosis. The KATHERINE study indicated that for HER2-positive early breast cancer patients who did not achieve pCR (the presence of residual tumor lesions) after neoadjuvant therapy, the administration of TDM-1 during postoperative adjuvant therapy significantly reduces the risk of disease recurrence compared to trastuzumab. This study aims to evaluate the efficacy and safety of trastuzumab in combination with pyrotinib and capecitabine in HER2-positive early breast cancer patients who did not achieve pCR following neoadjuvant therapy.Methods: This single-arm, open-label, multicenter study enrolled patients with HER2-positive early breast cancer who did not achieve pCR (defined as residual invasive tumor >1 cm or lymph node metastasis) following neoadjuvant therapy. Eligible patients received trastuzumab (initial loading dose of 8 mg/kg followed by 6 mg/kg every three weeks for one year, encompassing both neoadjuvant and adjuvant settings), pyrotinib (400 mg/day for one year post-surgery), and capecitabine (1000 mg/m² twice daily for six 3-week cycles, with continuation at the investigator’s discretion beyond six cycles). Primary prophylaxis with loperamide was administered to mitigate pyrotinib-induced diarrhea. The primary endpoint was 3-year invasive disease-free survival (iDFS), with secondary endpoints including distant disease-free survival (DDFS), overall survival, and safety. This study is registered with ClinicalTrials.gov (identifier NCT05292742).Results: From July 2021 to November 2023, a total of 102 patients were enrolled, all of whom did not achieve pCR following neoadjuvant therapy. As of June 30, 2025, with a median follow-up of 25 months, 2-year iDFS rate was 91.3% (95%CI, 83.3-95.6), and 2-year DDFS rate was 96.8% (95%CI, 90.5-99.0). Regarding safety, 22 patients (21.6%) experienced grade ≥3 adverse events, with the most common being diarrhea (13.7%), white blood cell count decreased (2.0%), and hand-foot syndrome (1.0%). Adverse events led to the treatment interruption of any agent in 66 patients (64.7%), discontinuation of any agent in 8 patients (7.8%), and dose reduction of any agent in 37 patients (36.3%).Conclusions: For patients with HER2-positive early breast cancer who did not achieve pCR (residual invasive tumor >1 cm or lymph node metastasis) following neoadjuvant therapy, the combination of pyrotinib, trastuzumab and capecitabine as postoperative adjuvant therapy appears to improve prognosis. The overall safety profile of the regimen was acceptable, with no new safety signals identified. Primary prophylaxis with loperamide effectively reduced the incidence of grade ≥3 diarrhea during adjuvant therapy with pyrotinib.
Presentation numberPS3-11-07
Real-world use of neratinib in her2-positive early breast cancer in korea: updated follow-up from a multicenter observational study
Seul-Gi Kim, CHA Bundang Medical Center, CHA University, Gyeonggi-do, Seongnam-si, Bundang-gu, Korea, Republic of
S. Kim1, S. Kim2, K. Shin3, J. Lee3, J. Kim4, Y. Moon1; 1Division of Hematology and Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Gyeonggi-do, Seongnam-si, Bundang-gu, KOREA, REPUBLIC OF, 2Division of Hematology and Oncology, Department of Internal Medicine, 2Division of Hematology and Oncology, Department of Internal Medicine, Samsung Changwon Hospital, Sungkyunkwan University, Gyeongsang-do, Changwon-si, Masanhoewon-gu, KOREA, REPUBLIC OF, 3Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul, KOREA, REPUBLIC OF, 4Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Gangnam-gu, Seoul, KOREA, REPUBLIC OF.
Background: Extended adjuvant neratinib improves invasive disease-free survival (iDFS) in patients with HER2-positive, hormone receptor-positive (HR+) early breast cancer (eBC) following adjuvant trastuzumab-based therapy, as shown in the ExteNET trial. However, real-world evidence in the post-pertuzumab/T-DM1 era is limited. We report updated outcomes from a Korean multicenter cohort. Methods: We retrospectively analyzed 48 HR+/HER2+ eBC patients who received neratinib between May 2022 and June 2025 at four academic hospitals in Korea. Patients had completed trastuzumab-based neoadjuvant or adjuvant therapy within 12 months prior to initiating neratinib. Data on prior regimens, neratinib administration, adverse events (AEs), and recurrence were collected. The primary endpoint was treatment pattern and tolerability; the secondary endpoint was iDFS. Results: The median age was 50 years (range, 37-66), and all patients were hormone receptor-positive. Most patients (75%) received the TCHP regimen. Post-neoadjuvant HER2-targeted therapies included: trastuzumab emtansine in 20 patients (41.7%), trastuzumab plus pertuzumab in 16 (33.3%), trastuzumab alone in 11 (22.9%), and trastuzumab deruxtecan in 1 patient (2.1%). Neratinib was initiated at 240 mg/day in 46 patients (95.8%), and at 120 mg/day with subsequent dose escalation in 2 patients. The median treatment duration was 11.2 months (range, 1.0-12.0). Diarrhea prophylaxis was administered in 85.4% of patients. The most common treatment-emergent adverse event (TEAE) was diarrhea (88.6%), followed by fatigue (29.5%) and skin reaction (20.5%), mostly grade 1 or 2. Grade ≥3 toxicities occurred in 12.5% of patients. Adverse events led to dose modifications in 9 patients (18.8%), including both dose interruptions and reductions. Permanent discontinuation due to AEs occurred in 4 patients (8.3%); AEs leading to dose modification included: diarrhea (n=6), nausea (n=2), oral mucositis (n=1), and duodenal ulcer (n=1). AEs leading to discontinuation included: vomiting (n=1), COVID-19 (n=1), diarrhea (n=1), nausea (n=1), fever (n=1), and duodenal ulcer (n=1). At a median follow-up of 11.8 months, no invasive disease recurrence was observed. Among the 48 patients, 31 completed the full 1-year course of neratinib, 7 remained on treatment, and 7 were lost to follow-up. Conclusion: In this real-world Korean cohort, neratinib was commonly used after pertuzumab or T-DM1. Treatment was feasible and generally well tolerated with appropriate supportive care. No recurrence was observed during early follow-up. To our knowledge, this represents the first Asian real-world data on neratinib in the extended adjuvant setting. Funding Source: This work was supported by BIXINK Therapeutics Co., Ltd.
Presentation numberPS3-11-08
Subcutaneous pertuzumab and trastuzumab fixed-dose combination versus dual intravenous pertuzumab and trastuzumab or subcutaneous trastuzumab plus intravenous pertuzumab in HER2-positive breast cancer adjuvant therapy: A time and motion study in a lean operating day care oncology unit in China
Amei Huang, Cancer Hospital of Shandong First Medical University, Jinan, China
Y. Meng1, A. Huang2; 1Nursing Management Department, Cancer Hospital of Shandong First Medical University, Jinan, CHINA, 2Breast surgery ward 2, Cancer Hospital of Shandong First Medical University, Jinan, CHINA.
Background: The fixed-dose combination of pertuzumab (P) and trastuzumab (H) for subcutaneous injection (PH FDC SC) significantly reduces drug preparation and administration time compared to intravenous (IV) formulations. However, real-world time efficiency data comparing PH FDC SC with dual IV P-H (P-H IV) and sequential SC H plus IV P (H-SC + P-IV) in HER2-positive breast cancer (HER2+ BC) are limited. Methods: This prospective observational study comprises two components: a cohort study and a patient (pt) survey. HER2+ BC pts receiving P plus H therapy and their healthcare professionals (HCPs) were recruited from a day-care unit at the Cancer Hospital of Shandong First Medical University. In the cohort study, administration records were divided into five groups by the route of administration and venous access (peripheral intravenous catheter [PIVC] or implantable venous access port [IVAP]). The groups are as follows: PH FDC SC, H-SC + P-PIVC, H-SC + P-IVAP,P-H PIVC and P-H IVAP. The survey encompassed three groups: PH FDC SC, H-SC + P-IV, and P-H IV. Time metrics were collected during cycles 7 through 18. Pt time included SC injection, IV infusion, and observation time. HCP time included drug preparation, SC injection, IV line setup/infusion, and observation time. Observation time was additional to total time. The total time differences and percentage time savings were analyzed. Pt preference questionnaires were collected. Results: The cohort study included 100 administration records from 14 pts receiving PH FDC SC, 100 records from 13 pts receiving H-SC + P-IV, and 100 records from 13 pts receiving P-H IV. H-SC + P-IV records were further categorized into H-SC + P-PIVC (n=29) and H-SC +P-IVAP (n=71), while the P-H IV records were divided into P-H PIVC (n=46) and P-H IVAP (n=54). The survey included 137 pts (PH FDC SC vs. H-SC +P-IV vs. P-H IV: 24 vs. 13 vs. 100). The PH FDC SC group demonstrated significant reductions in both patient and HCP total times compared to all IV-containing regimens (all p<0.001), with absolute time savings exceeding 3000 seconds and relative reductions of 78-89%. Observation time remained consistent across groups (Table 1). Questionnaire data revealed significantly higher preference for PH FDC SC in convenience-related domains including procedure time reduction, privacy protection, and venous access avoidance (all p<0.001). Conclusions: This real-world time-motion study demonstrates that PH FDC SC significantly reduces pt treatment burden and healthcare resource utilization in HER2+ BC adjuvant therapy. Pt preference data strongly favored PH FDC SC across convenience dimensions. These findings support PH FDC SC implementation as a favorable option in day-care settings.
| Variables | PH FDC SC (N=100) | H-SC + P-PIVC (N=29) | H-SC + P-IVAP (N=71) | P-H PIVC (N=46) | P-H IVAP (N=54) | P-value | |
| Patient time | Total time, s | 665.00 (656.75, 1080.00) | 3738.00 (3174.00, 4551.00) | 3831.00 (3590.00, 4505.50) | 6249.50 (5954.00, 6837.00) | 7721.50 (6312.25, 7939.00) | <0.001 |
| Mean difference, s, 95% CI | / | 3097.78 (2867.72, 3327.84) | 3157.79 (3032.70, 3282.89) | 5716.63 (5458.00, 5975.26) | 7151.68 (6412.44, 7890.92) | ||
| Time-savings, % | / | 78.51 | 78.83 | 87.08 | 89.4 | ||
| SC injection time, s | 665.00 (656.75, 1080.00) | 293.00 (279.00, 305.00) | 274.00 (249.50, 285.00) | / | / | <0.001 | |
| Peripheral intravenous catheter, s | / | 3459.00 (2869.00, 4258.00) | / | 6249.50 (5954.00, 6837.00) | / | <0.001 | |
| Infusion time, s | / | 3301.00 (2764.00, 4162.00) | / | 6103.00 (5828.00, 6635.25) | / | <0.001 | |
| Pre and duration of catheter management, s | / | 101.00 (97.00, 151.00) | / | 134.00 (126.00, 160.00) | / | 0.002 | |
| IVAP, s | / | / | 3512.00 (3352.50, 4232.50) | / | 7721.50 (6312.25, 7939.00) | <0.001 | |
| Infusion time, s | / | / | 3090.00 (2915.00, 3700.00) | / | 7305.50 (5901.00, 7490.00) | <0.001 | |
| Pre and duration of catheter management, s | / | / | 452.00 (432.50, 489.00) | / | 7305.50 (5901.00, 7490.00) | 0.022 | |
| Observation time, s | 900.00 (900.00, 900.00) |
900.00 (840.00, 900.00) (n=21) |
900.00 (600.00, 900.00) (n=63) |
720.00 (600.00, 900.00) | 600.00 (600.00, 900.00) | <0.001 | |
| HCP time | Total time, s | 693.50 (676.00, 1092.50) | 3870.00 (3297.00, 4665.00) | 3893.00 (3694.00, 4637.50) | 6371.50 (6069.00, 6953.25) | 7834.00 (6437.00, 8064.00) | <0.001 |
| Mean difference, s, 95% CI | / | 3181.43 (2953.41, 3409.45) | 3224.80 (3096.19, 3353.41) | 5798.27 (5539.31, 6057.23) | 7234.53 (6494.78, 7974.28) | ||
| Time-savings, % | / | 78.22 | 78.45 | 86.75 | 89.09 | ||
| Drug preparation, s | 50.00 (48.00, 53.25) | 113.00 (113.00, 122.00) | 113.00 (113.00, 122.00) | 120.00 (116.25, 124.00) | 124.00 (117.75, 125.00) | <0.001 | |
| SC injection time, s | 639.00 (621.00, 1041.25) | 298.00 (285.00, 315.00) | 283.00 (251.00, 290.00) | / | / | <0.001 | |
| Peripheral intravenous catheter, s | / | 3459.00 (2869.00, 4258.00) | / | 6249.50 (5954.00, 6837.00) | / | <0.001 | |
| Pre-administration, s | / | 3301.00 (2764.00, 4162.00) | / | 6103.00 (5828.00, 6635.25) | / | <0.001 | |
| Patient monitoring during administration, s | / | 101.00 (97.00, 151.00) | / | 134.00 (126.00, 160.00) | / | 0.002 | |
| IVAP, s | / | / | 3512.00 (3352.50, 4232.50) | / | 7721.50 (6312.25, 7939.00) | <0.001 | |
| Pre-administration, s | / | / | 3090.00 (2915.00, 3700.00) | / | 7305.50 (5901.00, 7490.00) | <0.001 | |
| Patient monitoring during administration, s | / | / | 452.00 (432.50, 489.00) | / | 449.00 (412.00, 458.00) | 0.022 | |
| Observation time, s | 900.00 (900.00, 900.00) |
900.00 (840.00, 900.00) (n=21) |
900.00 (600.00, 900.00) (n=63) |
720.00 (600.00, 900.00) | 600.00 (600.00, 900.00) | <0.001 | |
|
Note: PIVC, peripheral intravenous catheter; IVAP, implantable venous access port. Continuous variables were summarized as medians with quartiles (quartile 1 [Q1], quartile 3 [Q3]). |
Presentation numberPS3-11-09
Alignment between commonly measured and patient-elicited toxicities and impacts of human epidermal growth factor receptor 2-positive (HER2+) breast cancer therapies
Jonathon Gable, Jazz Pharmaceuticals, Philadelphia, PA
J. Gable1, M. Wraight1, K. Roat2, E. Kim3, R. Nair4, J. Buzaglo5; 1Patient-Centered Outcomes, Jazz Pharmaceuticals, Philadelphia, PA, 2Patient Experience Solutions, IQVIA, Durham, NC, 3Strategic Consulting, IQVIA, New York, NY, 4Patient Centered Solutions, IQVIA, Bengaluru, INDIA, 5Scientific Services, IQVIA, Wayne, PA.
Background. Successful development of oncology treatments relies on optimizing drug dosing by maximizing both efficacy and tolerability, thereby enhancing patients’ health-related quality of life (HRQoL). This analysis aimed to assess the extent of alignment between symptomatic toxicities and impacts measured in RCTs and those elicited as important by patients receiving neoadjuvant and adjuvant therapies for high- and low-risk stage II/III HER2+ breast cancer. Methods. A targeted literature search was conducted using the Ovid platform and the IQVIA Clinical Outcome Assessment Accelerator to identify publications reporting HRQoL data (randomized clinical trials [RCTs]) and patient-elicited experiences with breast cancer (qualitative studies). English-language publications (2013-August 2024) involving participants ≥18 years of age were included. Frequency (number of RCTs measuring the symptomatic toxicities and impacts [concepts]) was evaluated, and patient-reported prevalence values, where mentioned in at least one qualitative study, were extracted. Concepts were grouped post hoc by frequency and patient-reported prevalence. Results. Of 980 articles identified, 13 were eligible for inclusion (10 RCTs, 2 qualitative studies, and 1 systematic literature review). From these, 27 symptomatic toxicities and impacts (concepts) were extracted and grouped (Table). 5 of 13 concepts that had a prevalence of >25% in the qualitative literature were measured in ≥5 RCTs, whereas the remaining 8 were measured in <5 RCTs. Of these, 6 (distortion of taste, hot flashes, joint pain, memory loss, runny nose, and tingling sensation) were not measured by any patient-reported outcome (PRO) instrument. Inversely, 10 outcomes that were assessed in ≥5 RCTs were reported by 1 PRO instrument, such as the European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core Questionnaire 30 (EORTC QLQ-C30); of these concepts, 5 were identified as bothersome. Conclusions. Data regarding tolerability-related concept frequency and prevalence in patients with stage II/III HER2+ breast cancer were limited. Several of the patient-elicited concepts—particularly those with a prevalence of >25%—were not measured by any PRO instrument, whereas others were redundantly assessed by >1 PRO instrument in ≥6 RCTs; this redundancy may be due to concept inclusion in frequently used long-form legacy PRO instruments, like the EORTC QLQ-C30. Concept mapping against PRO item libraries, such as the Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE), may be warranted to ensure that the most relevant symptoms and impacts are assessed in RCTs. Further qualitative research is needed to optimize item selection.
Presentation numberPS3-11-11
A comparative study of trastuzumab and pertuzumab combination subcutaneous and intravenous infusion in patients with HER2-positive breast cancer: a retrospective and prospective combined study of time efficiency, cost-effectiveness, quality of life, and socioeconomic impact
Junichi Mase, Gifu Prefectural General Medical Center, Gifu City, Japan
J. Mase1, A. Ogiso1, H. Kinomura2, K. Ishihara3, H. Ohno4, S. Mano4, Y. Nagao1; 1Breast Surgery Department, Gifu Prefectural General Medical Center, Gifu City, JAPAN, 2Medical Affair Department, Gifu Prefectural General Medical Center, Gifu City, JAPAN, 3Nursing Department, Gifu Prefectural General Medical Center, Gifu City, JAPAN, 4Pharmacy Department, Gifu Prefectural General Medical Center, Gifu City, JAPAN.
Background: Intravenous infusion of trastuzumab plus pertuzumab takes 60-150 minutes per dose, which restricts both patients and chemotherapy room beds for long periods of time. A subcutaneous (SC) formulation was approved for insurance coverage in Japan in 2023, but real-world evidence on time efficiency and cost-effectiveness is lacking. Methods: This is a single-center retrospective and prospective combined study conducted at our institution from March 1, 2024, to June 30, 2025, targeting HER2-positive breast cancer patients who received trastuzumab plus pertuzumab IV or SC administration. A total of 302 IV administrations and 269 SC administrations were extracted from electronic health records, and chemotherapy room-stay times were compared by regimen. A prospective questionnaire was administered to 54 patients in the IV group and 41 patients in the SC group (including 21 patients with experience in both groups), along with 18 questions similar to those asked of healthcare providers. The primary outcome measures were chemotherapy room-stay time and total costs associated with chemotherapy. Secondary outcome measures included patient treatment satisfaction, psychological burden, and convenience, as well as healthcare providers’ perceptions of workload, efficiency, safety, and satisfaction. Results: In the case of trastuzumab plus pertuzumab monotherapy, the median chemotherapy room-stay time was 133.5 minutes (95% CI 112.4-167.3) in the IV group and 44.0 minutes (95% CI 31.4-59.3) in the SC group. When combined with docetaxel, the median was 213.0 minutes (95% CI 181.6-256.8) in the IV group, SC group was 167.5 minutes (95% CI 134.6-236.6). When combined with eribulin, the median was 198.0 minutes (95% CI 160.8-266.2) in the IV group and 107.0 minutes (95% CI 83.6-164.7) in the SC group. Regarding costs, when calculating the costs of medical supplies and labor expenses for the perioperative one-year administration of trastuzumab plus pertuzumab monotherapy, the IV group was 88,690 yen, and the SC group was 2,733 yen, with the IV group being higher. On the other hand, patient out-of-pocket expenses remained unchanged due to the high-cost medical care system. Furthermore, according to a questionnaire survey, both patients and healthcare professionals reported higher satisfaction with the SC group, and patients were more likely to choose the SC group than healthcare professionals. Discussion: In clinical practice, SC formulations significantly reduced the length of stay in the outpatient chemotherapy room compared to IV formulations and demonstrated high satisfaction among both patients and healthcare professionals. The reduction in staying time was consistent not only with trastuzumab plus pertuzumab monotherapy but also with chemotherapy combination regimens. Costs for medical supplies and labor were lower in the SC group, suggesting potential for cost savings. However, patient out-of-pocket expenses remained unchanged due to the high-cost medical care system. These results provide real-world evidence supporting the pharmacokinetic non-inferiority of the FeDeriCa trial and the high patient preference demonstrated in the PHranceSCa trial. However, limitations include the single-center, short-term observation and the fact that cost analysis was limited to cases without complications. A comprehensive evaluation through multi-center, long-term observations is necessary. Conclusion: The SC formulation of trastuzumab and pertuzumab may be a useful treatment option for HER2-positive breast cancer, improving time efficiency and patient satisfaction.
Presentation numberPS3-11-12
Efficacy and safety of disitamab vedotin combined with pyrotinib as neoadjuvant therapy for HER2-positive breast cancer: a phase II trial
Tianyu Zeng, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
T. Zeng, X. Sun, J. Wang, Y. Yin; Oncology Department, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, CHINA.
Background: Dual HER2 blockade plus chemotherapy achieves significant responses in HER2+ breast cancer but carries toxicities. Disitamab vedotin (RC48), a HER2-targeting ADC, and pyrotinib, a pan-HER TKI, show synergistic antitumor activity. This trial evaluates the first chemo-free neoadjuvant combination. Methods: This multicenter, single-arm phase II trial enrolled patients with operable or locally advanced HER2+ breast cancer (T2-4/N0-3; IHC 3+ or ISH+). Patients received RC48 at a dosage of 2.0 mg/kg intravenously every 2 weeks, combined with pyrotinib at a dosage of 400 mg orally once daily (cycles 1-4). Post-surgery, adjuvant therapy (epirubicin 90 mg/m² + cyclophosphamide 600 mg/m² Q3W, cycles 5-8) was administered, followed by maintenance therapy (cycles 9-22): trastuzumab + pertuzumab for pCR patients, T-DM1 or trastuzumab + pertuzumab for non-pCR patients. The primary endpoint was pathological complete response (pCR, defined as ypT0/Tis ypN0); the secondary endpoints included overall response (OR), assessed per RECIST 1.1 every two cycles, event-free survival (EFS), invasive disease-free survival (IDFS), and treatment-related adverse events (TRAEs). For binary endpoints, 95% confidence intervals (CIs) were calculated using the Clopper-Pearson exact method. Furthermore, a biomarker analysis was conducted to explore HER2 dynamics and alterations within the tumor microenvironment. Results: At data cutoff for this analysis on June 25, 2025, 17 patients were evaluable with a median follow-up of 5.2 months, ranging from 1.9 to 8.5 months. All patients were female, with a mean age of 51.9 years (range 29.0- 67.0), and a mean BMI of 21.8 kg/m² (range 17.5-30.5). All had HR-/HER2+ disease (17/17, 100%). Of the 15 patients who underwent surgery, the neoadjuvant regimen achieved a pCR in 53.3% (8/15), with a 95% CI of 26.6-78.7%. The ORR among the 17 evaluable patients assessed before surgery was 88.2% (15/17) with a 95% CI of 63.6-98.5%. No events related to EFS, including progressive disease per RECIST 1.1, failure to undergo surgery due to progression, toxicity, or death, or local or distant recurrence post-surgery, occurred. No death events occurred. As indicated in the table below, hepatic dysfunction was the most frequent TRAEs with a grade ≥ 3, and its median duration was 13 days. No treatment discontinuations occurred due to TRAEs.
| TRAEs | All Grade, n (%) | Grade ≥ 3, n (%) |
| Hepatic function abnormal | 16 (94.1) | 5 (29.4) |
| Hypokalemia | 9 (52.9) | 1 (5.9) |
| Hyperuricemia | 4 (23.5) | 0 (0.0) |
| Blood creatinine increased | 4 (23.5) | 0 (0.0) |
| Insomnia | 3 (17.6) | 0 (0.0) |
| Abdominal pain upper | 2 (11.8) | 0 (0.0) |
Conclusion: This first neoadjuvant study combining HER2-ADC and TKI demonstrated a chemo-free neoadjuvant regimen with high efficacy. The safety profile was manageable, with hepatic toxicity requiring longer follow-up. These results support randomized evaluation against standard chemotherapy-containing regimens. As the first disclosure of this regimen, the follow-up period remains limited for EFS/IDFS assessment, and exploratory biomarker data are pending. Another limitation was the single-arm design.
Presentation numberPS3-11-13
Fatty Acid Profile and Its Association with HER2-Positive Breast Cancer and Response to Neoadjuvant Chemotherapy
Benjamin Walbaum, pontificia universidad catolica de chile, santiago, Chile
F. Acevedo1, B. Walbaum1, K. Ramirez2, L. Medina1, F. Dominguez3, M. Camus3, C. Vargas3, M. Abud4, R. Valenzuela5, C. Farias6, c. G. sanchez1; 1Hematology-Oncology, pontificia universidad catolica de chile, santiago, CHILE, 2Oncology, Hospital Dr. Sotero del Rio, santiago, CHILE, 3Breast Surgery, pontificia universidad catolica de chile, santiago, CHILE, 4Pathology, pontificia universidad catolica de chile, santiago, CHILE, 5Nutrition, Universidad de Chile, santiago, CHILE, 6Hematology-Oncology, Universidad de Chile, santiago, CHILE.
Introduction Fatty acids (FAs), particularly omega-3 (n-3) polyunsaturated fatty acids, exhibit anti-inflammatory and antitumor properties. Preclinical models have demonstrated their ability to modulate HER2 expression and enhance the efficacy of targeted therapies. However, clinical evidence regarding the relationship between lipid profiles and the clinicopathological subtypes of breast cancer, as well as their utility as biomarkers of therapeutic response, remains limited. Methods Blood samples from 99 patients with stage I-III breast cancer treated with neoadjuvant chemotherapy (NAC) were analyzed. In a subset of 28 patients, paired plasma and erythrocyte samples were collected before and after treatment. Individual and grouped FAs (saturated FAs [SFAs], monounsaturated FAs [MUFAs], polyunsaturated FAs [PUFAs], n-3, and n-6) were quantified by gas chromatography. Associations between these profiles and HER2 status (immunohistochemistry and gene amplification) as well as pathological complete response (pCR) were evaluated. Statistical analyses included bivariate tests and linear and logistic regression models. Results Plasma MUFA levels (pMUFAs) were lower in HER2-positive patients (mean ± SD: 22.07 ± X vs. 25.00 ± Y; p = 0.0044), as were erythrocyte MUFA levels (eMUFAs; 19.95 ± X vs. 22.28 ± Y; p = 0.0107). In contrast, erythrocyte SFA levels (eSFAs) were higher in the HER2-positive group (54.89 ± X vs. 51.42 ± Y; p = 0.0069), as were plasma PUFA levels (pPUFAs; 41.10 ± X vs. 36.82 ± Y; p = 0.0166). A significant increase in plasma omega-6 fatty acids (linoleic acid + arachidonic acid) was also observed among HER2-positive patients (37.90 ± X vs. 34.21 ± Y; p = 0.0198). In the HER2 amplification analysis, higher plasma n-3 levels (ALA + DPA + EPA + DHA) were significantly associated with lower gene amplification (β = -0.73; 95% CI: -1.44 to -0.02; p = 0.045; R² = 0.13). Among the paired pre- and post-NAC samples, an increase in plasma n-3 fatty acids was correlated with higher rates of pCR, particularly in HER2-positive patients (p = 0.0321). Conclusions Blood FA profiles differ significantly according to HER2 status, especially regarding SFAs, MUFAs, PUFAs, and omega-6 fatty acids. Moreover, plasma omega-3 levels were inversely associated with HER2 amplification, and their increase during NAC may play a beneficial role in treatment response. These findings support the hypothesis that FAs, particularly omega-3s, could serve as complementary biomarkers for stratification and monitoring of patients with HER2-positive breast cancer.
Presentation numberPS3-11-14
Impact of HER2 amplification and Estrogen Receptor Expression on Pathologic Complete Response to Dual Blockade in HER2-Positive Breast Cancer
Benjamin V Walbaum, pontificia universidad catolica de chile, santiago, Chile
F. N. Acevedo1, B. V. Walbaum1, F. Gaete2, P. Peñaloza2, M. Olivares2, L. Medina1, M. Abud3, R. Gejman3, P. Zoroquiain1, F. Dominguez4, M. Camus5, C. Vargas4, M. Navarro6, C. Pinto6, M. Manzor6, c. G. sanchez1; 1Hematology-Oncology, pontificia universidad catolica de chile, santiago, CHILE, 2Pathology, Hospital Santiago Oriente Dr. Luis Tisné Brousse, santiago, CHILE, 3Pathology, pontificia universidad catolica de chile, santiago, CHILE, 4Breast surgery, pontificia universidad catolica de chile, santiago, CHILE, 5Breast Surgery, pontificia universidad catolica de chile, santiago, CHILE, 6Oncology, Hospital Dr. Sotero del Rio, santiago, CHILE.
Background: HER2-positive breast cancer (BC) accounts for 15-20% of cases and is defined by HER2 gene amplification and overexpression. Neoadjuvant chemotherapy (NAC) combined with HER2-targeted agents, particularly trastuzumab and pertuzumab, has improved outcomes. However, accessible biomarkers to guide treatment remain limited.Methods: A retrospective cohort of 446 stage I-III HER2-positive BC patients treated with NAC and trastuzumab ± pertuzumab from 2010 to 2023 was analyzed. The primary endpoint was pathological complete response (pCR; ypT0/isN0). HER2 was assessed via IHC (2+ vs. 3+), HER2/CEP17 ratio, and HER2 copy number, analyzed both continuously and by tertiles. Estrogen receptor (ER) was categorized as low (≤10%) or high (>10%). Other covariates included Ki-67, stage, BMI, and institution. Multivariable logistic regression evaluated pCR predictors. Analyses were stratified by pertuzumab use, with interaction terms tested. Propensity score modeling with inverse probability of treatment weighting (IPTW) adjusted for treatment selection bias. Internal consistency was assessed by bootstrap resampling (1,000 iterations).Results: The pCR rate was 51.8%. Independent pCR predictors included HER2 IHC 3+ (OR 4.54), HER2 amplification (medium: OR 3.11; high: OR 3.34), HER2/CEP17 ratio (mid: OR 2.57; high: OR 3.60), ER-low (OR 2.22), and higher Ki-67 (OR 1.011 per %). In pertuzumab-treated patients, HER2 amplification and HER2/CEP17 ratio were strongest (amplification high vs. low: OR 9.04; ratio high vs. low: OR 10.70), while HER2 IHC 3+ was more predictive without pertuzumab. ER-low tumors showed greatest benefit from dual blockade (OR 4.79). ER-pertuzumab interaction was significant (p<0.001). IPTW models confirmed ER and HER2 biomarkers as independent predictors. In ER ≤10% and medium/high HER2 amplification (32.6% of patients), pertuzumab significantly increased pCR (94.7% vs. 66.7%; p=0.013).Conclusions: Quantitative HER2 amplification and low ER expression are robust predictors of pCR in early HER2-positive BC. These biomarkers may help personalize neoadjuvant strategies and guide pertuzumab use.
Presentation numberPS3-11-15
Comprehensive Analysis and Prediction of HER2-targeted Therapy Insensitivity among HER2-Positive Breast Cancer Patients Undergoing Neoadjuvant treatment
Qingyao Shang, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
Q. Shang1, L. Zian2, L. Sheng3, W. Xin1; 1Breast Surgical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, CHINA, 2Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 3Department of Biostatistics & Bioinformatics, Duke University School of Medicine, Durham, NC.
Background: HER2-targeted therapy has been incorporated into the standard neoadjuvant treatment regimen for HER2-positive early-stage breast cancer, yet a subset of patients show limited pathological response. Early identification of these treatment-insensitive individuals may improve therapeutic decision-making. Methods: This retrospective study included 13,004 HER2-positive breast cancer patients from the National Cancer Database (2010-2022) who received neoadjuvant chemotherapy plus HER2-targeted therapy. Neoadjuvant treatment (NAT) insensitivity was defined as the absence of pathologic T-stage downstaging after treatment. Pathological complete response (pCR) was defined as no residual invasive carcinoma in the breast and axillary lymph nodes (ypT0/is, ypN0). Baseline characteristics and overall survival (OS) were compared between NAT-sensitive and NAT-insensitive groups. A multivariable logistic regression model was developed using significant clinicopathological variables. Categorical variables and interaction terms were also tested by the likelihood ratio test. Model performance was assessed using the area under the receiver operating characteristic curve (AUC), Hosmer-Lemeshow test, and calibration curves.Results: Among all patients, 22.5% (n = 2926) were classified as NAT-insensitive, 49.4% (n = 6418) were partial responders, and 28.1% (n = 3660) achieved pCR. NAT-insensitive patients were older and more likely to have HR-positive, low-grade, and non-ductal tumors. They experienced significantly worse OS compared to NAT-sensitive patients (log-rank p < 0.0001). Patients who achieved pCR were more likely to be younger, have poorly differentiated tumors, HR-negative status, and invasive ductal carcinoma, indicating a more responsive biological subtype. The predictive model showed good discrimination (AUC = 0.760 in training, 0.753 in validation) and calibration.Conclusion: Younger age, HR negativity, high tumor grade, and ductal histology were associated with higher pCR rates, whereas the opposite traits characterized treatment-insensitive patients. The predictive model developed in this study demonstrated good performance and may assist in early identification of individuals unlikely to benefit from neoadjuvant HER2-targeted therapy.
Presentation numberPS3-11-16
Choice in HER2-positive locally Advanced breast cancer of Neoadjuvant Taxane: a retrospective review of practice
Alexander Lozano, Gerald Bronfman, Montreal, QC, Canada
R. Barros1, B. Jiao2, A. Lozano2, A. Shah3, E. Bushatsky Andrade de Alencar1, M. Frija-Gruman4, J. Cools-Lartigue5, N. Gagnon-Choy6, I. Prakash7, K. Ma8; 1Oncology, Jewish General Hospital, Montreal, QC, CANADA, 2Oncology, Gerald Bronfman, Montreal, QC, CANADA, 3Oncology, University of British Columbia, Montreal, QC, CANADA, 4Internal Medicine, McGill University, Montreal, QC, CANADA, 5Thoracic Surgery, Mcgill University Health Center, Montreal, QC, CANADA, 6Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, CANADA, 7Surgery, Jewish General Hospital, Montreal, QC, CANADA, 8Medical Oncology, Jewish General Hospital, Montreal, QC, CANADA.
Background: The NeoSphere trial first established docetaxel-based dual HER2 blockade with pertuzumab and trastuzumab as an effective neoadjuvant regimen, yielding superior pathologic complete response (pCR) rates, though without conclusive long-term survival benefit. The subsequent PEONY trial confirmed the pCR advantage and demonstrated improved 5-year event-free survival with this regimen. Despite this, paclitaxel—administered weekly (P1) or every 3 weeks (P3)—is commonly used in clinical practice due to its more favorable tolerability profile. Whether the choice of taxane impacts efficacy or toxicity in the context of modern dual HER2-targeted therapy remains unclear, particularly in Canada, where pertuzumab reimbursement has been variable. Methods: We conducted a single-center retrospective cohort study at the Jewish General Hospital in Montreal, Quebec, Canada. Eligible patients were adults with HER2-positive stage II-III breast cancer who received at least one dose of pertuzumab and trastuzumab between December 2021 and December 2024, and underwent complete (R0) surgical resection. Patients were grouped by neoadjuvant taxane: docetaxel 75-100 mg/m² every 3 weeks (D3), paclitaxel 175 mg/m² every 3 weeks (P3), or paclitaxel 80 mg/m² weekly (P1). The primary endpoint was pCR; the secondary endpoint was the highest CTCAE v5.0 grade of adverse events. Fisher’s exact test was used to calculate odds ratios for pCR, and the Mann-Whitney U test compared toxicity grade distributions. Prism 10.5 was used in the analysis. Results: Cohort: We identified 87 patients: 60 received paclitaxel (45 weekly, 15 every 3 weeks) and 27 received docetaxel. The median age was 55 years (range 25-83) and median clinical stage was IIB (range IIA-IIIC); these were balanced between taxane cohorts (P = 0.37 and P = 0.59, respectively). Estrogen receptor status was similarly distributed (P = 0.48). Efficacy: Paclitaxel either every 3 or 1 week showed similar efficacy to Docetaxel every 3 weeks (P = 0.35), with a slight trend in our data towards better response with Paclitaxel. Safety: Paclitaxel either every 3 or 1 week and docetaxel every 3 weeks yielded safety profiles as measured by adverse events (CTCAE v5), P = 0.68 for paclitaxel every 3 weeks versus docetaxel, and P = 0.60 when comparing docetaxel against paclitaxel every 3 weeks and every 1 week. Conclusion: In routine clinical practice, paclitaxel—whether weekly or q3-weekly—achieves comparable pCR rates and acute toxicity to docetaxel when combined with pertuzumab and trastuzumab. Given paclitaxel’s logistical and tolerability advantages, these data support its use as an alternative taxane backbone in HER2-positive neoadjuvant therapy. Prospective randomized studies with survival endpoints are warranted. Adjustment for confounders will be addressed in future analyses.
Presentation numberPS3-11-18
Neoadjuvant Response in HER2+ Early Breast Cancer: A Comparative Study Between HER2-Enriched and Luminal HER2 Subtypes
Letícia Coelho de Mattos, AC Carmago Cancer Center, São Paulo, Brazil
L. Coelho de Mattos, B. Nelli Barbatto, L. Pivetta Genovez, L. Rodrigues Garcia, M. Assis da Escócia Fernandes, M. Celeste Tavares, S. Moraes Sanches, M. Goldner Cesca, F. Peterson Cavalher, L. de Moura Leite; Clinical Oncology, AC Carmago Cancer Center, São Paulo, BRAZIL.
Introduction: HER2-positive breast cancer represents approximately 15–20% of all breast tumors and is characterized by amplification of the ERBB2 gene, associated with aggressive biological behavior. However, this group includes biologically distinct subtypes based on hormone receptor (HR) status. Tumors lacking HR expression (HER2-enriched) typically show increased sensitivity to neoadjuvant therapy (NAT), while HR-positive tumors (Luminal HER2) often exhibit lower rates of pathological complete response (pCR), despite favorable long-term outcomes in some cases. We aimed to compare pathological complete response (pCR) and survival outcomes between Luminal HER2 (HER2+/HR+) and HER2-enriched (HER2+/HR−) breast cancer subtypes following neoadjuvant therapy (NAT) in the real world setting.Methods: All HER2-positive breast cancer treated with NAT and surgery from 2007 to 2018, who received at least 1 dose of neoadjuvant trastuzumab, in a single center were analyzed. Tumors were classified as Luminal HER2 or HER2-enriched based on immunohistochemistry. Endpoints included pCR, defined as absence of invasive disease in breast and axilla (ypT0/is ypN0), relapse-free survival (RFS) and overall survival (OS). Associations between categorical variables were assessed using chi-square and Fisher’s exact tests. Survival was estimated using Kaplan-Meier curves and factors associated with RFS were evaluated in a multivariate analysis. Bilateral p-values <0.05 were considered significant.Results: Among the 301 patients, 196 (65.1%) were HER2+/HR+ and 105 (34.9%) HER2+/HR-.The median age was 45 years vs. 52 years (p<.001), 68.2% vs. 39% were premenopausal (p<.001), 82.7% vs.94.3% had ductal histology (p=0.008), 40.8% vs. 52.5% had grade III tumors (p=0.09), 19.4% vs. 6.7% had HER2+2/FISH-amplified (p=0.003), 62.2% vs. 60% had cT3/4 (p=0.75), 73.5% vs.80% had cN+ (p=0.26) and 59.7% vs. 62.9% had clinical stage III (p=0.76), in the HER2+/HR+ vs. HER2+/HR-, respectively. Most patients in both groups were treated with anthracyclines plus taxanes (89.7%) and trastuzumab (62.5%). Thirty four percent of patients received trastuzumab plus pertuzumab, and only 9 patients didn’t receive anti-HER2 NAT, but all of those received adjuvant anti-HER2 blockade. One hundred fifty-eight patients (52.5%) achieved pCR, significantly higher in the HER2+/HR- group (65/105, 61.9%) compared to HER2+/HR+ (93/196, 47.4%) (p = 0.017). After a median follow up of 74 months, no significant difference in RFS was observed between subtypes (5-year RFS 79.2% vs. 76.3% in HER2+/HR+ vs. HER2+/HR-, p=0.47) or in OS (5-year OS 89.2% vs. 86.1% in in HER2+/HR+ vs. HER2+/HR-, p=0.65). On univariate analysis RFS was associated with higher cT (HR 1.33 95%CI 1.00 – 1.76, p=0.045), cN+ (HR 2.0 95%CI 1.03 – 3.92, p=0.041), pCR (HR 0.28 95%CI 0.16 – 0.48, p<.001), but not HER2+/HR- subtype (HR 1.19, 95%CI 0.73 – 1.94, p=0.48). On a multivariate analysis of these factors, only cN (HR 1.33, 95%CI 1.0 – 1.77, p=0.049) and pCR (HR 0.29, 95%CI 0.17 – 0.51, p=<.001) persisted as significant factors for RFS.Conclusion: HER2-enriched breast cancers showed a significantly higher pCR rate to neoadjuvant systemic therapy compared to Luminal HER2 tumors. However, this improved short-term response did not translate into superior recurrence-free survival. Clinical tumor burden remained the strongest prognostic factor. These findings highlight the importance of biological subtype in predicting treatment response and support its use in guiding neoadjuvant strategies.
Presentation numberPS3-11-19
Guiding the Axilla: Combining SUVmax and Monocytes to Predict Response to Neoadjuvant Therapy in HER2 positive Breast Cancer
Lucrezia Raimondi, Humanitas, Torino, Italy
L. Raimondi1, M. Cuzzocrea2, C. Oliva1, N. Peradze3, Y. Harder4, D. Schmauss4, R. Graffeo Galbiati5, L. Rossi6; 1Medical Oncology, Humanitas, Torino, ITALY, 2Division of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, SWITZERLAND, 3Division of Breast Surgery, European Institute of Oncology, Milano, ITALY, 4Department of Plastic, Reconstructive and Aesthetic Surgery, Ente ospedaliero cantonale, Lugano, SWITZERLAND, 5Institute of Oncology of Southern Switzerland (IOSI), Ente ospedaliero cantonale, Bellinzona, SWITZERLAND, 6Centro di Senologia della Svizzera Italiana, Ente ospedaliero cantonale, Lugano, SWITZERLAND.
Introduction: The prognostic significance of achieving axillary pathologic complete response (AxpCR) following neoadjuvant chemotherapy (NAT) in clinically node-positive (cN+) breast cancer (BC) is well established. However, consensus regarding the optimal strategy for axillary management—particularly in patients with HER2-positive BC—remains lacking. This study aims to evaluate the predictive value of the maximum standardized uptake value of axillary lymph nodes (N-SUVmax) obtained from baseline 18F-FDG PET/CT scans, in combination with the absolute monocyte count (AMC), for AxpCR following NAT in patients with cN+ HER2-positive BC. Methods: A retrospective analysis was conducted on HER2-positive cN+ BC patients treated with NAT. All patients received the same chemotherapy regimen, including dual HER2 blockade. ROC curves were used to determine cut-off values for N-SUVmax and AMC. Associations with AxpCR were analyzed using logistic regression models. Results: Among 285 patients diagnosed with HER2-positive BC, 215 presented with cN+ disease at baseline. Of these, 155 patients (72.1%) achieved AxpCR following NAT. All patients underwent axillary lymph node dissection (ALND) according to current guidelines, regardless of imaging or biomarker results. Notably, a large proportion showed no residual nodal disease at pathology, confirming AxpCR despite initial cN+ status.Patients with an N-SUVmax ≥ 3.5 had a 4.9-fold higher likelihood of achieving AxpCR (p = 0.039; odds ratio [OR] = 4.938 [95% CI: 1.081-1.904]). In contrast, patients with an AMC > 340/mm³ had a 90.4% lower probability of achieving AxpCR (p < 0.001; OR = 0.096 [95% CI: 0.028-0.324]). Notably, the interaction between N-SUVmax and AMC was significant: higher AMC levels diminished the predictive value of N-SUVmax for AxpCR (p = 0.002; OR = 0.999 [95% CI: 0.997-1.000]). Based on N-SUVmax and AMC values, patients were stratified into three groups. Group 1: High N-SUVmax – Low AMC; Group 2: High N-SUVmax – High AMC or Low N-SUVmax – Low AMC; Group 3: Low N-SUVmax – High AMC. Compared to Group 1, both Group 2 (p < 0.001; OR = 0.047 [95% CI: 0.009-0.237]) and Group 3 (p < 0.001; OR = 0.017 [95% CI: 0.002-0.137]) showed significantly reduced probabilities of achieving AxpCR. Conclusions: Assessing axillary response to NAT in HER2-positive BC provides a critical opportunity to tailor surgical management. The combination of baseline N-SUVmax and AMC improves prediction of AxpCR and may support more individualized axillary treatment strategies, ultimately contributing to better patient outcomes.
Presentation numberPS3-11-20
Tumor-intrinsic signatures associated with response to chemotherapy-free HER2/PD-L1 blockade in HER2-enriched early breast cancer
Mailin Li, Houston Methodist Research Institute, Houston, TX
M. Li1, J. Deng1, X. Hoi2, J. Zheng3, R. Hashmani1, W. Qian1, J. Zhou1, J. Guan4, K. Sun4, H. Mai4, T. Sheu5, S. Haley5, M. Schwartz5, S. Wong6, F. Nikolo2, K. Chan2, P. Niravath1, J. Chang1; 1Breast Oncology, Houston Methodist Research Institute, Houston, TX, 2Urology, Houston Methodist Research Institute, Houston, TX, 3Immunomonitoring Core, Houston Methodist Research Institute, Houston, TX, 4Hematology/Oncology, Houston Methodist Hospital, Houston, TX, 5Pathology, Houston Methodist Hospital, Houston, TX, 6Systems Medicine and Bioengineering, Houston Methodist Hospital, Houston, TX.
Background: HER2-positive breast cancers have demonstrated sensitivity to de-escalated regimens combining HER2-targeted therapy with immune checkpoint inhibition. In our recent Phase II trial evaluating a chemotherapy-free regimen of durvalumab, trastuzumab, and pertuzumab (DTP), we observed a 49% pathologic complete response (pCR) rate among patients with HER2-Enriched tumors (BluePrint ®). Understanding tumor-intrinsic and microenvironment factors driving response variability remains a critical goal. Methods: To investigate mechanisms of therapeutic response and resistance, we performed single nucleus RNA sequencing (snRNA-seq) on formalin-fixed, paraffin-embedded tumor specimens from all 37 evaluable DTP trial participants. Paired pre-treatment needle biopsies and post-treatment surgical resections were profiled to capture longitudinal transcription dynamics and changes in cellular composition. Patients were stratified by Residual Cancer Burden (RCB) following surgical pathology. While previous analyses focused on immune cell dynamics, this study presents preliminary findings from 201,788 total epithelial cells. Epithelial populations were initially classified as aneuploid (“tumor”) or diploid (“normal”) epithelial cells based on inferred copy number alterations, with tumor cell calling validated by concordance with RCB status. Results: Tumor cells from Responder patients (RCB0, n = 18), RCB1 patients (n = 7), and Non-Responder patients (RCB2/3, n = 6) exhibited Chromosome 17 amplification consistent with HER2 positive status. snRNA-seq of tumor samples collected at surgery revealed a stepwise increase in tumor content—absent in RCB0, low in RCB1, and high in RCB2/3—supporting the accuracy of epithelial cell classification. Notably, Non-Responders harbored multiple tumor subclones, whereas Responders retained a single dominant clone from pre- to post-DTP treatment. Gene Set Enrichment Analysis of pre-treatment tumor cells from Responders versus Non-Responders revealed enrichment of endoplasmic reticulum (ER) stress and unfolded protein response pathways, increased ER-Golgi trafficking, and altered translational programs in tumor cells in Responders, suggesting that DTP-sensitive tumors are undergoing apoptosis or immunogenic cell death. In contrast, tumor cells from Non-Responders were enriched for Wnt/β-catenin and PI3K/Akt/mTOR signaling, indicating activation of tumor-intrinsic resistance pathways. Conclusions: Single-cell profiling revealed epithelial gene expression programs with potential to predict response or resistance to dual HER2 blockade and immune checkpoint inhibition. Ongoing spatial transcriptomic studies aim to map immune-epithelial interactions and tissue architecture in HER2-enriched breast cancer. Clinical trial information: NCT03820141. Research Sponsor: Houston Methodist Hospital and Astrazeneca.
Presentation numberPS3-11-21
A real-world prospective observational multi-national study in adult patients with breast cancer treated with extended adjuvant neratinib: interim results from the NERLYFE study
Rupert Bartsch, Medical University of Vienna / Comprehensive Cancer Center, Vienna, Austria
D. Baerens1, A. Waqas2, G. Dagmar3, A. Jegannathen4, T. Sarkodie5, P. Seropian6, V. Bjelic-Radisic7, M. Elnaggar8, P. Staib9, B. Lex10, N. Harbeck11, C. Jackisch12, M. Schmidt13, J. Suissa14, A. Zkik15, O. Dialla16, R. Bartsch17; 1Gynecology/Obstetrics, Frauenarztpraxis, Ilsede, GERMANY, 2Oncology, United Lincolnshire Hospitals NHS Trust, Lincoln, UNITED KINGDOM, 3Gynecology/Obstetrics, Praxis Dr. Guth, Plauen, GERMANY, 4Cancer Centre, UHNM NHS Trust – Royal Stoke University Hospital, Stoke-on-Trent, UNITED KINGDOM, 5Oncology, The Mid and South Essex University Hospitals Group, Chelmsford, UNITED KINGDOM, 6Gynecology/Obstetrics, Freudenstadt Hospital, Freudenstadt, GERMANY, 7Senology, Helios University Hospital, Wuppertal, GERMANY, 8Hematology/Oncology, Royal Derby Hospital, Derby, UNITED KINGDOM, 9Oncology, St. Antonius Hospital, Eschweiler, GERMANY, 10Gynecology/Obstetrics, Klinikum Kulmbach, Kulmbach, GERMANY, 11Breast Cancer, Department of Obstetrics & Gynecology, LMU University Hospital, Munich, GERMANY, 12Obstetrics & Gynecology, Oncology Unit, Sana Klinikum Offenbach, Offenbach am Main, GERMANY, 13Obstetrics & Gynecology, University Medical Center of Johannes Gutenberg University, Mainz, GERMANY, 14Medical Affairs, Pierre Fabre Laboratories, Boulogne Billancourt, FRANCE, 15Real World Evidence, Pierre Fabre Laboratories, Boulogne Billancourt, FRANCE, 16R&D, Pierre Fabre Laboratories, Boulogne Billancourt, FRANCE, 17Division of Oncology, Department of Medicine I, Medical University of Vienna / Comprehensive Cancer Center, Vienna, AUSTRIA.
Background Neratinib, a pan-HER tyrosine kinase inhibitor, is approved in Europe as extended adjuvant therapy for HR+/HER2+ early breast cancer (EBC) in patients who have completed trastuzumab-based therapy ≤ 1 year ago. In this population, the ExteNET trial for patients with HR+/HER2+ EBC demonstrated a clinical benefit for neratinib vs. placebo, including significantly improved 5-year invasive disease-free survival (iDFS) (iDFS; Δ5.1%, HR 0.58, 95% CI 0.41-0.82). Neratinib-associated diarrhoea was the most common adverse event (39% Grade 3 without mandatory prophylaxis). Real-world data are essential for informing the management of diarrhoea and understanding patterns within its approved indications. Methods NERLYFE is a European, prospective, observational post-authorization safety study (PASS) conducted in routine clinical practice across Austria, Czech Republic, Germany, and the United Kingdom. Patients with HR+/HER2+ EBC were scheduled to receive neratinib as extended adjuvant therapy for up to 12 months in accordance with the EU SmPC. The primary objective is to describe the incidence of permanent treatment discontinuation due to diarrhoea during the first 3 months (mo) of neratinib treatment (core phase). Secondary objectives include describing the pattern of diarrhoea and maintenance of neratinib treatment. Results At data cut-off on 31 December 2024, 113 patients received at least one dose of neratinib and were observed for at least 3 months. Results of this pre-planned interim analysis are reported. Baseline characteristics were median age 55 years, 86% ECOG PS 0, 79% high risk of recurrence (AJCC stage > I or N+ or non-pCR after neoadjuvant treatment). Overall, 56%, 28% and 11% of patients received adjuvant/post-neoadjuvant trastuzumab monotherapy, trastuzumab-emtansine (T-DM1), and trastuzumab/pertuzumab, respectively, with 5% of patients receiving other anti-HER2 therapy. The initial dose of neratinib was 240 mg/day for 61% of patients and < 240 mg/day for 39%; 63% of patients received anti-diarrhoeal prophylaxis at least once. Fourteen percent of patients permanently discontinued neratinib due to diarrhoea within the first 3 mo of neratinib treatment, primarily within the first 2 mo of treatment. The diarrhoea-related neratinib discontinuation rate was lower in patients who received anti-diarrhoeal prophylaxis vs. those who did not (10% vs. 21%) and in those who started below vs. at 240 mg/day (12% vs. 15%). Any grade diarrhoea occurred in 88% of patients (20% Grade 3): 86% and 91% in the prophylaxis/non-prophylaxis groups respectively, and 82% vs. 91% in the below vs. at 240 mg/day group. No Grade 4 or 5 event was reported. Median time to first diarrhoea was within the first 7 days. Among patients receiving prophylaxis, the median cumulative duration of Grade ≥3 diarrhoea was 6.5 days versus 13 days without prophylaxis. Corrective treatment was used in 66% of patients. Treatment-emergent adverse events (TEAEs) other than diarrhoea were reported in 55% of patients, mostly Grade 1-2 gastrointestinal events. Three patients experienced serious TEAEs, including one diarrhoea event. Conclusion Anti-diarrhoeal prophylaxis was associated with a reduction in the incidence of diarrhoea occurrence (any grade), rate of neratinib discontinuation, and duration of Grade ≥3 diarrhoea. The overall safety profile of neratinib was consistent with the EU SmPC. These real-world findings support the benefit of anti-diarrhoeal prophylaxis and the use of neratinib in HR+/HER2+ early breast cancer patients in the current treatment landscape.
Presentation numberPS3-11-22
Efficacy and safety of 12-week adjuvant docetaxel plus trastuzumab in patients with node-negative HER2-positive breast cancer (tumors ≤1cm): results from the SOBER prospective single-arm trial
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.
Background There is no standard treatment regimen for HER2-positive node-negative patients with tumor smaller than 1 cm yet. Therefore, we designed a single-arm, open, prospective, phase II study to evaluate the clinical efficacy and toxicity of adjuvant 12-week trastuzumab combined with docetaxel in HER2-positive node-negative patients with tumor smaller than 1 cm.MethodsPatients having HER2-postive node-negative breast cancer with T≤1cm received docetaxel with concurrent trastuzumab for 12 weeks. The primary endpoint was 3-year disease-free survival (DFS). Th secondary endpoints included 3-year overall survival (OS) and breast cancer specific survival (BCSS).ResultsA total of 78 patients were enrolled in this trial. The percentage of T1mic [≤0.1 cm], T1a [>0.1 to ≤0.5 cm], and T1b [>0.5 to ≤1.0 cm]) tumors were 47.4%, 35.5% and 17.1%, respectively. After a median follow-up of 45.2 months, five DFS events were reported, with 96.9% of 3-year DFS rate and 89.8% of 5-year DFS rate. The 3-year and 5-year OS rate was 100.0% and 96.0%, respectively. No BCSS event was reported. Tumor size, molecular subtype or Ki-67 level was not associated with DFS (p>0.05) according to subgroup analyses.Fifteen (19.7%) patients had a drop in LVEF over 5% after treatment with no one having decreased LVEF over 10%. No grade 3 or 4 cardiac event was reported. The most frequently reported adverse events were neutropenia (23.7%).ConclusionsAmong HER2-postive node-negative breast cancer patients with T≤1cm, treatment with 12-week adjuvant docetaxel plus trastuzumab was associated with favorable prognosis and tolerable toxicity.
Presentation numberPS3-11-23
Cross-country Treatment Practices after pCR Following Neoadjuvant Trastuzumab (H) and Pertuzumab (P) in HER2+ Early Breast Cancer: Preliminary Results from the PEARL-HER2 Study
Soraia Lobo-Martins, Institut Jules Bordet, Brussels, Belgium
S. Lobo-Martins1, G. Gentile1, H. Gouveia2, N. Teixeira Tavares3, M. Gonzalez-Rodriguez4, E. Campôa5, B. Pereira6, T. Pina Cabral7, E. Agostinetto1, D. Cabuk8, D. Alpuim Costa9, F. P Duhoux10, P. Mandó11, T. Cunha Pereira12, P. Simões13, H. Wildiers14, L. Arecco1, R. Gerosa1, C. Dauccia1, S. Ramalho2, M. Carvalho Couto3, T. Pascual4, N. Cunha5, F. Sarmento6, A. Martins7, E. Kolemen8, M. Delgado da Silva15, C. van Marcke10, S. Rivero11, A. Garcia12, G. Nader-Marta16, M. Piccart1, P. Kristanto17, E. de Azambuja1; 1Academic Trials Promoting Team (ATPT), Institut Jules Bordet, Brussels, BELGIUM, 2Breast Unit, Champalimaud Foundation, Lisbon, PORTUGAL, 3Oncology Department, Unidade Local de Saúde de São João, EPE, Porto, PORTUGAL, 4Medical Oncology, Cancer Institute and Blood Diseases, Hospital Clinic de Barcelona, Barcelona, SPAIN, 5Medical Oncology, ULS Algarve, Faro, PORTUGAL, 6Medical Oncology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisbon, PORTUGAL, 7Oncology Department, Unidade Local de Saúde Lisboa Ocidental, Lisbon, PORTUGAL, 8Department of Medical Oncology, Kocaeli University, Kocaeli, TURKEY, 9Oncology Functional Unit, Hospital de Cascais, Lisbon, PORTUGAL, 10Medical Oncology, Cliniques universitaires Saint-Luc, Brussels, BELGIUM, 11(Grupo Cooperativa Argentino para el estudio y la investigación del cáncer de mama), SUMA, Ciudad de Buenos Aires, ARGENTINA, 12Medical Oncology Department, Portuguese Oncology Institute of Coimbra Francisco Gentil, Coimbra, PORTUGAL, 13Medical Oncology, Unidade Local de Saúde de Loures / Odivelas, Loures, PORTUGAL, 14Medical Oncology, UZ Leuven, Leuven, BELGIUM, 15Pharmaceutical Services, Hospital de Cascais, Lisbon, PORTUGAL, 16Breast Oncology Program, Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 17Clinical Biostatistics Unit (CBU), Institut Jules Bordet, Brussels, BELGIUM.
Background: Pts with HER2+ early BC who achieve pCR following neoadjuvant chemotherapy (NACT) with HP routinely continue dual anti-HER2 therapy, although the added benefit of continuing HP versus H alone in this setting remains uncertain, with added cost and toxicity. PEARL-HER2 aims to clarify this question by evaluating whether adjuvant HP provides additional clinical benefit compared to H alone. Here, we report a preliminary cross-country comparison of clinical practices. Method: PEARL-HER2 is an international, retrospective cohort study including pts with HER2+ early BC who achieved pCR (ypT0/isN0) after NACT and HP. Eligible pts started NACT between Jan-2014 and Dec-2023. This descriptive analysis, data cut-off of 20-Jun-2025, summarizes diagnostic, and treatment data stratified by country. Results: Of 1045 pts screened, 649 with pCR were eligible for this analysis. Country-level characteristics are shown in Table 1. Differences in baseline characteristics included a higher proportion of pre-/perimenopausal pts in Argentina (56%), higher ER positivity in Belgium (58%) and lower Ki-67 expression in Turkey (21%). Imaging practices also differed: breast MRI was routinely used in Spain, Portugal, and Belgium, but infrequent in Turkey (15%). Staging with FDG-PET was more commonly employed in Turkey (65%) and Belgium (44%), whereas CT and bone scan were predominant elsewhere. Anthracycline-free regimens were more frequent in Argentina (100%) and Belgium (34%), contrasting with near-universal anthracycline use in Portugal and Turkey. Adjuvant HP was continued in >90% of pts in Belgium vs. <9% in Iberian countries, highlighting disparities in access. Among ER+ patients, ET use was near-universal (97%), though OFS-based combinations were infrequent even in very young pts. With a median follow-up of 44 months (IQR 29-67), only 35 relapses (5.4%) were reported, 18 (51%) of which in the central nervous system (CNS). Conclusion: This preliminary analysis reveals marked international variability in post-pCR management of HER2+ early BC. These findings should be interpreted with consideration of the unequal distribution of patients across countries, with Portugal contributing over two-thirds of the cohort. Differences in imaging and tumor burden likely reflect national screening and staging practices, while variation in adjuvant P use likely reflects national funding policies, with routine access in Belgium but limited or no reimbursement in Iberian countries. Notably, over half of reported relapses occurred in the CNS. PEARL-HER2 continues to accrue and follow patients to clarify the role of adjuvant P in this setting.
| Characteristics |
Total
(n=649)
|
Argentina
(n=18)
|
Belgium
(n=86)
|
Portugal
(n=437)
|
Spain
(n=80)
|
Turkey
(n=28)
|
p-value |
| Age at diagnosis in years, median (IQR) | 51.9 (44.4-62.0) | 52.9 (40.9-63.2) | 52.7 (43.9-64.2) | 52.1 (45.0-61.4) | 53.8 (42.5-62.1) | 48.6 (45.2-57.9) | 0.729 |
| Pre-/peri-menopausal status, n (%) | 303 (46.7) | 10 (55.6) | 36 (41.9) | 208 (47.7) | 35 (43.8) | 14 (50.0) | 0.039 |
| NST subtype, n (%) | 595 (91.8) | 15 (88.2) | 81 (94.2) | 396 (90.6) | 75 (93.8) | 28 (100.0) | 0.788 |
| Lobular subtype, n (%) | 18 (2.8) | 1 (5.9) | 4 (4.7) | 11 (2.5) | 2 (2.5) | 0 (0.0) | – |
| Grade 3, n (%) | 322 (49.7) | 8 (47.1) | 54 (62.8) | 216 (49.4) | 31 (38.8) | 13 (46.4) | 0.318 |
| ER-negative, n (%) | 314 (48.4) | 14 (77.8) | 36 (41.9) | 207 (47.4) | 39 (48.8) | 18 (64.3) | <0.001 |
| Ki-67 <20%, n (%) | 58 (10.2) | 0 (0.0) | 7 (8.1) | 30 (8.3) | 15 (18.8) | 6 (21.4) | <0.001 |
| DCIS present, n (%) | 191 (29.5) | 0 (0.0) | 32 (37.2) | 113 (25.9) | 39 (48.8) | 7 (25.0) | <0.001 |
| Tumor size in mm, median (IQR) | 32.0 (23.0-50.0) | 52.0 (25.0-66.0) | 31.0 (22.0-47.0) | 32.0 (23.0-49.0) | 28.0 (21.0-52.5) | 30.0 (23.0-40.0) | 0.246 |
| Clinical N0, n (%) | 251 (38.8) | 2 (11.1) | 16 (18.6) | 185 (42.3) | 37 (46.3) | 11 (42.3) | <0.001 |
| Genetic testing performed, n (%) | 185 (28.5) | 5 (27.8) | 27 (31.4) | 122 (27.9) | 26 (32.5) | 5 (18.5) | 0.667 |
| Breast MRI performed, n (%) | 552 (85.2) | 13 (72.2) | 80 (93.0) | 375 (85.8) | 80 (100.0) | 4 (14.8) | <0.001 |
| FDG-PET used, n (%) | 144 (22.3) | 5 (27.8) | 38 (44.2) | 65 (14.9) | 19 (23.8) | 17 (65.4) | <0.001 |
| Anthracycline-based NACT, n (%) | 536 (82.6) | 0 (0.0) | 56 (65.1) | 396 (90.6) | 57 (71.3) | 27 (96.4) | <0.001 |
| Adjuvant pertuzumab, n (%) | 137 (21.1) | 14 (77.8) | 81 (94.2) | 37 (8.5) | 5 (6.3) | 0 (0.0) | <0.001 |
| ET use among ER+, n (%) | 336 (96.8) | 5 (83.3) | 45 (91.8) | 231 (97.9) | 44 (100.0) | 11 (91.7) | 0.028 |
| OFS use among pre/peri, n (%) | 74 (22.0) | 0 (0.0) | 10 (22.3) | 46 (19.9) | 16 (36.4) | 2 (18.2) | 0.042 |
Presentation numberPS3-11-24
The Combined Immune-Stromal Score (CISS): A Novel Prognostic Gene Signature in Early both ER-positive and HER2-positive Breast Cancer
qiao yang, Karolinska Institutet, Stockholm, Sweden
q. yang1, C. Rönnlund1, C. Schagerholm Stanev1, X. Chen1, T. Foukakis1, K. Wang1, I. Fredriksson2, R. Stephanie1, E. G. Sifakis1, J. Hartman1; 1Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SWEDEN, 2Breast Center, Theme Cancer, Karolinska University Hospital, Stockholm, SWEDEN.
Background: Despite therapeutic advances, patients with both estrogen receptor (ER)-positive and Human Epidermal growth factor Receptor 2 (HER2)-positive breast cancers have persistent clinical heterogeneity. We hypothesized that stromal-immune interactions within the tumor microenvironment (TME) drive differential treatment outcomes. Materials and methods: We analyzed 356 primary surgical HER2-positive tumors (treated with adjuvant trastuzumab) using gene expression panel profiling derived from formalin-fixed paraffin-embedded (FFPE) specimens. Unsupervised clustering of pathway enrichment levels was performed. The Combined Immune-Stromal Score (CISS) was developed, and we validated it for associations with recurrence-free survival (RFS) in both ER-positive and HER2-positive patients. Validation was performed using independent validation cohorts: PREDIX HER2 (neoadjuvant), SCAN-B, METABRIC, and TCGA. Results: Four biological subgroups were identified, characterized by differences in stromal markers (stromal), immune infiltration, cytokine and chemokine signaling, and antigen presentation (immune). Patients with Low_stromal and Low_immune tumors had significantly worse RFS compared to Low_stromal and High_immune (adjusted Hazard Ratio (HRadj) = 2.55, 95% confidence interval (CI) = 1.09-5.95, p = 0.03), and High_stromal and Low_immune (HRadj = 2.30, 95% CI = 1.18-4.48, p = 0.01). In both ER-positive and HER2-positive patients, each unit increase in CISS was associated with a 40% reduction in recurrence risk (HRadj = 0.60, 95% CI = 0.42-0.86, p = 0.00544), a finding consistently replicated across validation cohorts. Patients with low-CISS tumors (concurrent low stromal and low immune activity) demonstrated a worse prognosis (Log Rank p = 0.0069). Conclusions: The Combined Immune-Stromal Score (CISS) is a novel gene expression-based signature that stratifies recurrence risk in early both ER-positive and HER2-positive breast cancer. Tumors with low CISS, characterized by concurrently low stromal and immune pathway activity, were associated with significantly poorer outcomes. These findings suggest that a suppressed TME may contribute to recurrence risk and highlight the potential of CISS as a prognostic tool and a basis for future studies exploring therapeutic vulnerabilities.
Presentation numberPS3-11-25
Adjuvant ovarian function suppression (OFS) in HR+/HER2+ premenopausal breast cancer (BC) patients with pathologic complete response (pCR) or residual disease (RD) after neoadjuvant chemotherapy (NAC): a real-world study
Daniella Audi Blotta, Memorial Sloan Kettering Cancer Center, New York, NY
D. Audi Blotta1, N. Mai1, M. Bromberg2, Y. Chen2, F. Parvin-Nejad3, G. Plitas3, P. Razavi1, M. Robson1, S. Modi1; 1Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, 2Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, 3Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY.
Background: The HERA trial demonstrated survival benefits with the addition of OFS to adjuvant endocrine therapy (ET) in premenopausal patients (pts) with HR+/HER2+ early BC. However, the benefit of OFS over tamoxifen (TAM) alone remains unclear in the post-NAC setting, particularly among pts who achieve pCR or have RD. We present real-world survival outcomes comparing OFS-based ET vs. TAM alone in this specific population following NAC. Methods: We conducted a retrospective study of premenopausal pts ≤50 years with HR+/HER2+ BC who achieved pCR (ypTis/T0 ypN0) or had RD after NAC at Memorial Sloan Kettering (2008-2025). Pts were categorized by adjuvant ET: OFS+TAM or aromatase inhibitor (AI) vs. TAM alone. Pts receiving >1 ET type were classified according to the therapy received for >75% of their total ET duration. Primary endpoints were disease-free survival (DFS) and overall survival (OS), estimated using the Kaplan-Meier method and compared across ET groups using log-rank tests. Pt and treatment characteristics were compared between ET groups using chi-squared tests. Multivariable Cox proportional hazards models were used to evaluate associations between adjuvant ET and survival outcomes, adjusting for pathologic stage and adjuvant HER2 therapy. ET exposure was modeled as a time-dependent covariate to account for differences in exposure time among pts receiving >1 ET type. Results: A total of 309 pts were included: 157 with pCR and 152 with RD. Overall, 146 (47%) received OFS+TAM/AI and 163 (53%) received TAM alone. Median follow-up was 55.6 months (IQR 37-80). AC-THP was the primary NAC regimen (69%) in both ET groups. Compared to TAM alone, pts treated with OFS+TAM/AI were more likely to have clinical node-positive disease at baseline (63% vs. 51%, p=0.032) and to receive T-DM1 as adjuvant HER2-targeted therapy (36% vs. 15%, p<0.001). Five-year DFS was 89% with OFS+TAM/AI vs. 84% with TAM (p=0.12). No significant difference in OS was observed (p=0.20). In the full cohort, TAM was associated with inferior DFS compared to OFS+TAM/AI in multivariable analysis (HR 1.90; 95% CI, 0.91-3.98; p=0.088), with borderline statistical significance. Pathologic stage and adjuvant HER2-targeted therapy were significantly associated with DFS: stage III vs. pCR (HR 7.52; 95% CI, 2.83-20.0; p<0.001), and trastuzumab alone vs. T-DM1 (HR 3.60; 95% CI, 1.27-10.3; p=0.016). These results were consistent when analyzing the pCR and RD subgroups separately. In the pCR cohort, TAM showed a nonsignificant trend toward inferior DFS (HR 3.46; 95% CI, 0.72-16.6; p=0.12), while clinical stage III was associated with worse DFS (HR 4.67; 95% CI, 1.33-16.5; p=0.016). In the RD cohort, pathologic stage III (HR 4.09; 95% CI, 1.71-9.79; p=0.002) and trastuzumab monotherapy (HR 3.47; 95% CI, 1.16-10.4; p=0.026) were associated with worse DFS. TAM also showed a nonsignificant trend toward inferior DFS (HR 1.55; 95% CI, 0.65-3.67; p=0.30). Conclusions: In this real-world cohort of premenopausal pts with HR+/HER2+ BC who achieved pCR or had RD after NAC, OFS-based ET was more frequently prescribed to those with higher-risk features, such as clinical node-positive disease. Although this retrospective study was underpowered to detect definitive differences in DFS between ET strategies, a trend toward improved DFS was observed in pts treated with OFS+TAM/AI compared to TAM alone, independent of pathologic stage and adjuvant HER2-targeted therapy, with borderline statistical significance. These findings support the need for larger retrospective cohorts to better clarify the role of OFS for HR+/HER2+ BC in the post-NAC setting.
Presentation numberPS3-11-26
A real-world study of pyrotinib as neoadjuvant therapy for HER2-positive breast cancer
Hongmei Zheng, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty construction discipline, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast C, wuhan, China
H. Zheng1, Y. Tan2, Q. Zhang3, J. Li4, H. Shen5, G. Wang6, Y. Tu7, Y. Yang1, J. Liu1, J. Wang1, R. Tian1, X. Fu1, X. Wu*1; 1breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty construction discipline, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast C, wuhan, CHINA, 2Thyroid and Breast Surgery, The First College of Clinical Science of Three Gorges University, Yichang, CHINA, 3breast, Jingzhou Center hosipital, Jingzhou, CHINA, 4breast surgery, The Central Hospital of Enshi Tujia And Miao Autonomous Prefecture, Enshi, CHINA, 5Thyroid Gland Breast Surgery, Xiaogan Hospital,Wuhan University of Science and Technology, Xiaogan, CHINA, 6Breast and thyroid vascular surgery, Taihe Hospital, Affiliated Hospital of Hubei University of Medicine, wuhan, CHINA, 7Breast and thyroid surgery, Renmin Hospital of Wuhan University, wuhan, CHINA.
Background: A double-blind, randomized Phase 3 clinical study, PHEDRA, met its primary endpoint with a significantly higher tpCR rate than placebo in patients treated with pyrotinib, trastuzumab, and docetaxel in HER2-positive early or locally advanced breast cancer (41.0% vs. 22.0%, unilateral P< 0.0001). At present, trastuzumab combined with pyrotinib and docetaxel has become the standard neoadjuvant treatment strategy for HER2-positive early or locally breast cancer.We aim to evaluate the efficacy and safety of pyrotinib-containing regimen as neoadjuvant therapy for HER2-positive early or locally advanced breast cancer in a real-world setting.Methods: This is a prospective, real-world, one-arm study. Patients with untreated HER2-positive operable early or locally advanced breast cancer were enrolled. Eligible patients received pyrotinib-containing regimen. Surgery was performed after the completion of the last cycle of study treatment. The primary endpoint was tpCR (ypT0/Tis ypN0) and secondary endpoints included objective response rate (ORR), bpCR and safety. Results: Between Feb 13, 2022, and May 29, 2025, a total of 104 patients with a median age of 51 years (range:26-66) were enrolled. 45% patients received AC/EC-THPy, 38% patients received TCbHPy and 15% patients received THPy regimens. As of the data cutoff date on Jun12, 2025, 66 patients underwent surgery, tpCR rate was 58%. The tpCR rates of HR- and HR+ patients were 68% and 42% respectively .Out of the 84 patients with evaluable response, 2 achieved complete and 66 achieved partial response, resulting in an ORR of 81%. The grade 3 or 4 adverse events were 24.03%. The most common treatment-related adverse events (AEs) were diarrhea (50%), Vomiting (26.9%), and decreased appetite (11.5%). Most AEs were tolerable.Conclusions: In this real-world study, neoadjuvant pyrotinib- containing regimens showed promising efficacy and safety in patients with HER2-positive breast cancer, further verify its effectiveness and safety in the real world.
Presentation numberPS3-11-27
Twelve Weekly Neoadjuvant Paclitaxel and Carboplatin with Trastuzumab and Pertuzumab: A Novel De-escalation Strategy
Yasmin Leshem, Tel Aviv Medical Center, Tel Aviv, Israel
Y. Leshem1, I. Golomb2, Y. Bar1, S. Strulov Shachar1, A. Zubkov3, N. Khadmy1, A. Sonnenblick1; 1Oncology, Tel Aviv Medical Center, Tel Aviv, ISRAEL, 2Division of Oncology, Tel Aviv Medical Center, Tel Aviv, ISRAEL, 3Institute of Pathology, Tel Aviv Medical Center, Tel Aviv, ISRAEL.
Introduction: The standard treatment regimen for early HER2-positive disease consists of 18 weeks of carboplatin, docetaxel, trastuzumab, and pertuzumab. This regimen associated with significant toxicities. Therefore there is a pressing need for tailored de-escalation strategies that maintain efficacy while reducing treatment burden. Methods: We performed a retrospective analysis of a unique cohort of patients with stage II and III HER2 positive breast cancer prescribed a de-escalated neoadjuvant regimen of 12 weekly cycles of paclitaxel and carboplatin along with trastuzumab, and pertuzumab (12wTCHP). Results: Of the 43 patients who received 12wTCHP regimen, 31 (72%) had stage IIA disease, 27 (63%) were positive for estrogen receptor (ER), and 40 (93%) had invasive ductal carcinoma (IDC). The median age was 65 (IQR 51-75). The most prevalent grade 3-4 toxicities were neutropenia (17%) and diarrhea (17%). No treatment-related deaths occurred. The median relative dose intensity (RDI) of paclitaxel and carboplatin was 79%, with a higher RDI in the stage IIA stage compared to stage IIB and III (82% vs. 60%, p=0.003). The pathological complete response (pCR) rate was 63%. In the ER-positive and ER-negative subtypes, the pCR rate was 56% and 75% respectively. During a median follow-up of 20 months, invasive disease recurrence occurred in two patients (5%). Of these, none of the 29 patients with stage IIA IDC experienced a recurrence. Conclusions: A de-escalated neoadjuvant approach (12wTCHP) for early-stage HER2-positive breast cancer demonstrated a favorable safety profile, high pCR rate, and low recurrence rate.
Presentation numberPS3-11-28
EmpowHER-208: A phase 2 neoadjuvant study of zanidatamab in combination with chemotherapy in patients with stage II-III HER2-positive early breast cancer
Adrienne G. Waks, Dana-Farber Cancer Institute, Boston, MA
A. G. Waks1, S. Hurvitz2, E. Hamilton3, J. O’Shaughnessy4, V. Valero5, A. Combest6, B. Salim7, F. Herbst8, S. Faderl6, S. M. Tolaney1; 1Breast Oncology, Dana-Farber Cancer Institute, Boston, MA, 2Clinical Research Division, Fred Hutch Cancer Center, Seattle, WA, 3Breast Cancer Research Program, Sarah Cannon Research Institute, Nashville, TN, 4Breast Cancer Research, Baylor University Medical Center, Texas Oncology, US Oncology Network, Dallas, TX, 5Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 6Clinical Development, Jazz Pharmaceuticals, Palo Alto, CA, 7Biostatistics, Jazz Pharmaceuticals, Palo Alto, CA, 8Global Medical Affairs, Jazz Pharmaceuticals, Zug, SWITZERLAND.
Background: In human epidermal growth factor receptor 2-positive (HER2+) early breast cancer, pathologic complete response (pCR) is strongly associated with favorable long-term outcomes, with 93%-97.5% of patients who achieve pCR remaining event-free at 3-year follow-up. One standard-of-care (SOC) neoadjuvant regimen for patients with stage II-III HER2+ early breast cancer is docetaxel (T) and carboplatin (C) + dual HER2-directed therapy (trastuzumab [H] and pertuzumab [P]; collectively TCHP), with single-agent taxane (docetaxel or paclitaxel [T]) + HP regimens becoming increasingly common. Despite pCR rates >50% with these regimens, a substantial proportion of patients have residual disease at surgery, requiring toxic adjuvant therapies, and some patients will experience recurrence and death. TCHP is also associated with short- and long-term toxicities that substantially impact quality of life, hence the need for novel neoadjuvant regimens to increase pCR rate, improve long-term outcomes, and reduce toxicity. Zanidatamab is a bispecific HER2-directed antibody that binds 2 distinct sites on HER2 (juxtamembrane extracellular domain and dimerization domain), facilitating receptor clustering and exerting multiple antitumor mechanisms of action, including HER2 internalization and downregulation and immune-mediated effects (complement-dependent cytotoxicity and antibody-dependent cellular cytotoxicity and phagocytosis). Zanidatamab demonstrated promising antitumor activity with a manageable safety profile combined with chemotherapy in HER2+ metastatic breast cancer and as a neoadjuvant monotherapy in stage I HER2+ early breast cancer. The phase 2 EmpowHER-208 study is designed to evaluate neoadjuvant zanidatamab combined with chemotherapy and may inform further investigations in HER2+ early breast cancer. Methods: EmpowHER-208 is a phase 2, randomized, open-label study assessing the pCR rate of neoadjuvant zanidatamab + paclitaxel (ZT), zanidatamab + docetaxel + carboplatin (ZTC), or TCHP in patients with untreated, histologically confirmed stage II-III HER2+ early breast cancer (including inflammatory early breast cancer). Eligible patients must have a tumor ≥2 cm and no clinical or radiographic evidence of distant metastasis. Patients will be randomized (2:2:1) to receive six 3-week cycles of ZT, ZTC, or TCHP. Randomization will be stratified by local assessment of hormone receptor status (positive vs negative) and clinical stage (II-IIIA vs IIIB-IIIC). In the ZT and ZTC arms, patients will receive intravenous (IV) zanidatamab every 2 weeks during cycles 1 and 2, then every 3 weeks (Q3W) + a standard regimen of the assigned chemotherapy. Following neoadjuvant therapy, patients will proceed to surgery followed by response-tailored adjuvant therapy. Patients who achieve pCR will receive 12 additional 3-week cycles of zanidatamab IV Q3W (ZT and ZTC arms) or H ± P (TCHP arm). Patients who do not achieve pCR will receive fourteen 3-week cycles of SOC ado-trastuzumab emtansine (T-DM1). Adjuvant endocrine therapy, extended neratinib, radiotherapy, or additional chemotherapy (ZT arm only) may be administered based on investigator preference and institutional SOC. The primary endpoint is pCR rate by blinded local assessment. Key secondary endpoints include pathologic response by residual cancer burden score at time of surgery, breast-conserving surgery rate, safety and tolerability, event-free survival, and overall survival.
Presentation numberPS3-11-29
Impact of GLP-1 Receptor Agonist Use on Pathologic Complete Response in HER2-Positive Breast Cancer Treated with Neoadjuvant TCHP
Daniela Urueta Portillo, UT Health Mays Cancer Center, San Antonio, TX
D. Urueta Portillo1, M. Mazo Canola2; 1Hematology Oncology, UT Health Mays Cancer Center, San Antonio, TX, 2Hematology Oncology, UT Health Mays cancer center, San Antonio, TX.
Background: GLP-1 receptor agonists (GLP-1RAs) are commonly prescribed in the setting of obesity and diabetes. Weight gain is known to increase risk of breast cancer recurrence, and GLP-1RAs are being more commonly used to facilitate weight loss. GLP-1 is rapidly degraded by DPP4, leading to the development of DPP4 inhibitors. Mechanistically, GLP-1 receptor activation on tumor cells may activate several growth signaling pathways based on preclinical studies, and GLP-1RAs may have an inhibitory effect on inflammation. Both of these functions have the potential to alter rates of response to HER2 targeted neoadjuvant chemotherapy. The recent study by Santos et al. demonstrated lower rates of pCR among patients with triple-negative breast cancer that were on GLP-1RAs while on neoadjuvant chemotherapy regimens. Whether this holds true for patients with HER2-positive breast cancer is unknown. The purpose of this study is to determine the effect of GLP-1RA exposure on pCR rate in patients with HER2-positive breast cancer treated with neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP). This population is notable for a high proportion of Hispanic patients and an increased rate of obesity compared to national averages, which may impact treatment response.Methods: We retrospectively analyzed HER2-positive breast cancer patients treated at UT Health Mays Cancer Center (MCC) from January 2021 to June 2025 with standard of care neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP). Patients were stratified by exposure to GLP-1 receptor agonists during neoadjuvant therapy. The primary endpoint was pathologic complete response (ypT0/is, ypN0). Age, body mass index (BMI, calculated as weight in kilograms divided by height in meters squared), diabetes status, hormone receptor (HR) status, race, and clinical stage were collected as covariates. Fisher’s exact, chi-square, and descriptive statistics summarized group differences. Results: Among 42 patients, 6 (14%) used GLP-1 receptor agonists. The pCR rate was 33% (2/6) in GLP-1RA users compared to 58% (21/36) in non-users (p=0.21). Among patients on other DM2 medications (n=9), the pCR rate was 56%, while it was 55% among those on no DM2 medications (n=33). GLP-1RA users had similar mean age (54.2 vs 54.1 years, p=0.98) and a higher mean BMI (33.8 vs 29.6 kg/m², p=0.14) compared to non-users. GLP-1RA users were mostly HR-positive (83%), similar to non-users (71%). Pathologic complete response by HR status showed no significant difference: pCR was achieved in 69% (9/13) of HR-negative patients vs 48% (14/29) of HR-positive patients (p=0.32). The racial distribution was 40% Hispanic white (n=17), 36% non-Hispanic white (n=15), 14% non-Hispanic Black (n=6), 7% Asian (n=3), and 2% Hispanic Black (n=1), consistent with the diverse population served by our center. There was no significant association between race and pCR (p=0.44). Due to sample size, no multivariate analysis was performed. Conclusions: In this single-center cohort, GLP-1RA use during neoadjuvant TCHP was associated with a numerically lower pCR rate in HER2-positive breast cancer. Although not statistically significant, this trend supports the need for larger prospective studies to evaluate whether GLP-1RA exposure may reduce pCR rates.
Presentation numberPS3-11-30
Neoadjuvant pyrotinib, trastuzumab, dalpiciclib, and exemestane in triple-positive breast cancer: a multicenter phase II trial
Peifen Fu, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
P. Fu1, L. Chen1, M. Yao1, C. Du1, Y. Li2, J. Zhou3, S. Chen4, S. Cai4, Q. Feng5; 1Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, CHINA, 2Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, CHINA, 3Department of Breast Surgery, The Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, CHINA, 4Department of Breast Surgery, Lishui Central Hospital, Lishui, CHINA, 5Department of Breast Surgery, Yiwu Maternity and Children Hospital, Yiwu, CHINA.
Background: Triple-positive breast cancer (TPBC), defined by concurrent expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), accounts for 10-15% of breast cancers. Standard neoadjuvant regimens combining trastuzumab-based HER2-targeted therapy with chemotherapy show limited efficacy in this subtype. Resistance is attributed to HER receptor heterodimerization, HER2-ER signaling cross-talk, and activation of PI3K/AKT/mTOR pathways. Integrating HER2-targeted therapy with endocrine therapy and cyclin-dependent kinase 4/6 (CDK4/6) inhibitors may help overcome this resistance. The NA-PHER2 study demonstrated the potential benefit of a chemotherapy-free regimen, trastuzumab, pertuzumab, palbociclib, and fulvestrant, in TPBC. Pyrotinib, an irreversible pan-HER tyrosine kinase inhibitor, shows synergistic activity with CDK4/6 blockade in preclinical models. In the phase II MUKDEN 01 trial, pyrotinib combined with dalpiciclib and letrozole showed promise in TPBC, though efficacy remains suboptimal. Building on this rationale, we evaluated a novel, chemotherapy-free neoadjuvant regimen of pyrotinib, trastuzumab, dalpiciclib, and exemestane in patients with stage II-III TPBC.Methods: Eligible patients had previously untreated stage II-III TPBC, defined as ER >10%, PR >1%, and HER2 positivity (immunohistochemistry 3+ or in situ hybridization positive). Patients received pyrotinib (320 mg orally once daily, every 4 weeks, for 5 cycles), trastuzumab (intravenously: loading dose 8 mg/kg, then 6 mg/kg every 3 weeks for 6 cycles; or subcutaneously: 600 mg every 3 weeks for 6 cycles), dalpiciclib (125 mg orally once daily on days 1-21 of a 4-week cycle, for 5 cycles), and exemestane (25 mg orally once daily, every 4 weeks, for 5 cycles), followed by surgery. The primary endpoint was total pathological complete response (tpCR), defined as ypT0/is ypN0.Results: Between September 2022 and February 2025, 33 patients were enrolled. The median age was 48 years (range, 29-75). Disease stage at baseline included 17 patients (51.5%) with stage IIA, 11 (33.3%) with stage IIB, 4 (12.1%) with stage IIIA, and 1 (3.0%) with stage IIIC. Ki-67 expression at baseline was ≥30% in 25 patients (75.8%). As of June 10, 2025, 25 patients had undergone surgery with available pathological data. The tpCR rate was 24% (6 of 25; 95% confidence interval [CI], 10-46%). Residual cancer burden 0-1 was observed in 19 patients (76%; 95% CI, 54-90%), and 21 (84%; 95% CI, 63-95%) achieved a Miller-Payne grade 4-5 response. Among 31 patients with evaluable radiographic data, 12 achieved complete response and 19 had partial response, yielding an objective response rate of 100% (95% CI, 86-100%). Adverse events occurred most frequently during the first two treatment cycles. No grade ≥4 events were reported. The most common grade 3 adverse events were diarrhea (41.9%) and neutropenia (22.6%).Conclusions: This chemotherapy-free, quadruple-targeted neoadjuvant regimen demonstrated promising activity and manageable toxicity in early-stage TPBC. These data support further evaluation of this regimen as a potential alternative to standard chemotherapy-based neoadjuvant approaches in TPBC.
Presentation numberPS3-12-01
T-cell receptor (TCR) Repertoire Differences Between Stage III Inflammatory Breast Cancer (IBC) and Matched Non-IBC Controls
Ilana Schlam*, Dana Farber Cancer Institute, Boston, MA
I. Schlam*1, X. Cai*1, N. U. Lin1, C. Stever1, A. Giordano1, J. Rosenbluth2, C. C. Block1, C. Graser3, F. Nakhlis4, G. Kirkner1, A. Martin1, F. Michor3, S. T. Schumer1, E. Rotella1, M. Moore1, H. Heiling1, C. Snow1, A. Patel1, S. M. Tolaney1, K. Polyak*1, F. Lynce*1; 1Medical oncology, Dana Farber Cancer Institute, Boston, MA, 2Medical oncology, University of California San Francisco, San Francisco, CA, 3Human Cancer Genetics, Dana Farber Cancer Institute, Boston, MA, 4Surgery, Brigham and Women’s Hospital, Boston, MA.
Background: Inflammatory breast cancer (IBC) is a rare and aggressive subtype of breast cancer characterized by a distinct clinical presentation and poor prognosis. Despite multiple studies aimed at understanding the biological drivers of its aggressive behavior, the underlying mechanisms remain elusive. Emerging evidence suggests that the host immune system may play a role in the unique clinical course of IBC. Prior research has demonstrated that peripheral blood T-cell receptor (TCR) clonotype diversity declines with age, and it is associated with cancer risk and disease progression. However, the peripheral blood TCR clonotype repertoire has not been studied in IBC. Methods: We conducted a matched-cohort study using baseline peripheral blood samples from treatment-naïve patients. Stage III cases and controls were matched 1:1 by age such that the ages of each case-control pair were within 5 years of each other. The controls were identified from our institutional annotated datasets (COQD, EMBRACE). RNA was extracted from whole blood and subjected to TCR sequencing to characterize the clonotype repertoire. TCR diversity was quantified using established metrics, including Shannon entropy, Richness, and clonality indices. We compared the frequencies and uniqueness of CDR3 sequences between IBC and non-IBC patients. We also clustered all CDR3 (alpha or beta) sequences from patients using Geometric Isometry-based TCR AligNment Algorithm (GIANA) tool to evaluate CDR3 cluster differences between the two groups. Results: 41 patients with stage III IBC and age-matched 41 patients with stage III non-IBC were included (n = 82). The age ranged from 26 to 87 in the groups. A trend toward increased TCRα clonotype diversity was observed in non-IBC compared to IBC samples. This was evidenced by higher Shannon entropy and Richness scores in non-IBC samples, reflecting a more diverse and heterogeneous T-cell receptor repertoire. Conversely, IBC samples exhibited lower diversity, as indicated by higher Shannon and Simpson clonality values, consistent with a more oligoclonal T-cell population. The ratio of unique CDR3a sequences in individual patients was significantly higher in the non-IBC group (Mann-Whitney U test, p=0.036). Non-IBC samples also had more unique CDR3 clusters than IBC samples, indicating greater T-cell repertoire heterogeneity. TCR diversity was not significantly associated with overall survival in either group, however, the number of events was low. Conclusions: In this study, we evaluated TCR clonotype diversity in patients with stage III IBC and non-IBC. Our findings suggest that IBC is associated with a less diverse TCR repertoire, which may reflect a more restricted or less effective anti-tumor immune response. These results warrant further investigation to determine the clinical relevance and potential therapeutic implications of TCR characteristics in IBC.
Presentation numberPS3-12-02
Microbiota disruption with oral antibiotics has a detrimental effect on HER2+ breast cancer therapy efficacy
Romina E Araya, George Washington University, Washington, DC
R. E. Araya1, L. J. Parker1, P. Mitra1, N. Erlichman1, P. Chalasani2, R. Li1; 1Biochemistry and Molecular Medicine-SMHS, George Washington University, Washington, DC, 2Division of Hematology and Oncology, George Washington Cancer Center- George Washington University, Washington, DC.
Cancer patients are particularly susceptible to infections. Approximately 20-60% of patients may receive antibiotics at some point during their therapy, including perioperative care and periods of neutropenia. Retrospective studies suggest that antibiotics negatively impact outcomes in various cancers. A pilot study focused on breast cancer (BC) indicated that HER2⁺ BC patients experience greater reductions in survival following antibiotic exposure than HER2-negative cases. While this effect is thought to arise from disruptions in the host microbiota, the underlying mechanisms remain poorly defined. We previously demonstrated that microbiota signaling modulates innate immune cells in different tumor types, with consequences for the efficacy of chemotherapy and immunotherapy. However, HER2+ BC patients receive targeted therapies such as anti-HER2 antibodies with small-molecule tyrosine kinase inhibitors in case of therapy resistance. Therefore, the impact of microbiota specifically on HER2 therapies requires further study. To address this knowledge gap, we investigate how microbiota disruption, induced by antibiotics, affects responses to HER2-targeted therapies using mouse preclinical models. We utilized orthotopic HER2⁺ breast cancer mouse models, administered an oral broad-spectrum antibiotic cocktail (ABX) to deplete the microbiota, and employed HER2-targeted antibody (anti-HER2/neu) and small molecule (neratinib) therapies, along with high-throughput spectral flow cytometry, to assess treatment efficacy and the immune mechanisms of response. While antibiotic treatment alone did not alter tumor growth, it significantly impaired responses to HER2 therapies. Notably, the efficacy of anti-HER2 was partially diminished, while the response to neratinib was completely abrogated. To mimic clinical scenarios, we administered oral ciprofloxacin (cipro) or amoxicillin/clavulanic acid (amox/clav), examples of antibiotics commonly given to patients, and neratinib. Cipro mirrored the ABX effect, fully halting the neratinib response, whereas amox/clav induced a partial impairment. Given that cipro and amox/clav target fewer bacteria than the ABX cocktail, these findings suggest that both the abundance and composition of the microbiota play a role in the efficacy of HER2 therapy. Next, we examined whether disrupting microbiota with antibiotics influences the immune response linked to HER2+ BC treatments. We thoroughly profiled immune cell subsets in tumors and the circulation after HER2-targeted therapy. In control (H2O) mice, both HER2 therapies led to immune remodeling of the tumor microenvironment, characterized by a decrease in tumor-associated neutrophils and macrophages, along with increased infiltration of NK, CD8⁺, and CD4⁺ T cells. In contrast, ABX-treated mice failed to exhibit the same immune shifts, particularly in response to neratinib, with only marginal changes observed after treatment with anti-HER2/neu. Interestingly, a similar reduction in neutrophils post-therapy occurs in blood from H2O and ABX tumor-bearing mice. In contrast, NK cells are systemically reduced in untreated ABX animals compared to those in H2O-treated mice and do not recover after therapy, which could explain the lack of recruitment of these cells to the tumor bed in ABX mice and suggest a role of these cells in therapy efficacy. Overall, our results demonstrate that antibiotic-induced disruption of the gut microbiota impairs HER2⁺ breast cancer therapy and the immune response associated with treatment efficacy, both systemically and locally within the tumor bed. These findings underscore innate immune pathways and microbiota-based strategies as potential therapeutic targets to mitigate the detrimental effects of antibiotics in HER2+ BC patients.
Presentation numberPS3-12-03
Targeting ERβ signaling as a novel immunotherapeutic strategy in TNBC
Kristina Diana A Zambo, Houston Methodist, Houston, TX
K. A. Zambo, F. Nikolos, H. Nagandla, K. Cap, A. Phillips, W. Qian, J. Qian, K. Chan, J. Chang, C. Thomas; Cancer Center, Houston Methodist, Houston, TX.
Purpose: To assess the potential of an estrogen receptor beta (ERβ) agonist as an immunotherapy for triple negative breast cancer (TNBC). Background: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer characterized by the absence of ERα, PR, and HER2 expression with limited treatment options. Although immune checkpoint inhibitors (ICIs) have shown some benefit, response rates remain low in advanced-stage TNBC. This highlights a critical gap in our understanding of oncogenic mechanisms, especially those leading to immunosuppressive tumor microenvironment (TME), and underscores the urgent need for innovative therapeutic targets to improve patient outcomes. Estrogen receptor beta (ERβ), expressed in approximately 30% of TNBCs, is a subtype of estrogen receptor with known anti-tumor properties. While prior studies have established ERβ’s tumor-suppressive role, its effect on the TME remains poorly defined. Despite recently observed clinical associations of ERβ with improved prognosis and immune-related markers, the mechanisms by which ERβ impacts immune cell recruitment, phenotype, and response to immunotherapy have not been thoroughly studied. Here, we aim to define how ERβ regulates immune activity in TNBC and to evaluate its utility as an immunotherapeutic target and biomarker. Methods: To test whether tumor-specific ERβ activity contributes to an immunostimulatory tumor microenvironment, we generated unique syngeneic models and patient-derived xenografts (PDXs) of TNBC with altered ERβ expression and investigated differences in tumor growth and immunophenotype using flow cytometry and immunohistochemistry. Single-cell and spatial genomics were used to study how ERβ alters signaling in tumor cells, affecting effector cells to reduce immunosuppression. To determine whether activating ERβ with agonists further stimulates tumor immunity and inhibits tumor progression, we administered the ERβ agonist LY500307, which is clinically tested in other diseases. ERβ targets and downstream signaling were validated through transcriptional and functional studies in isolated immune cells and cocultures. Lastly, we correlated ERβ’s expression and function with immune signatures and clinical outcome by deconvoluting sequencing data from TCGA-profiled breast cancers and through spatial proteomics analysis of a tissue microarray with TNBC samples. Result: We observed that activated tumor-specific ERβ leads to slowed tumor growth and a less immunosuppressive TME, which is characterized by a significant reduction in tumor-associated M2-like macrophages and increased cytotoxic T cell activity. RNA-seq and cytokine profiling of LY500307-treated ERβ-expressing breast cancer cells identified key target cytokines as downstream mediators, particularly CXCL2, CCL5, CSF2, and IL4 that regulate macrophage function and antigen presentation during tumor adaptive immunity. Preliminary clinical analysis revealed an inverse correlation between ERβ expression and the frequency of myeloid cells that predicted a worse patient outcome, reinforcing the prognostic significance of the receptor. Conclusions: By integrating spatial and single-cell genomics, and immune-functional assays, this study uncovers important functional and mechanistic features of the immunomodulatory role of ERβ. Our findings aim to transform our understanding of ERβ as a tumor suppressor in the immunosuppressive TME of TNBC and could help identify predictive biomarkers and therapy targets to improve patient outcomes.
Presentation numberPS3-12-04
Integrated Tumor Microenvironment Profiling Reveals Distinct Immune, Stromal and MicroRNA (miRNA) Signatures Across Triple-Negative Breast Cancer (TNBC) Molecular Subtypes
Shivangi M Sengupta, Cleveland Clinic Lerner Research Institute, Cleveland, OH
S. M. Sengupta1, B. Maiti2; 1Clinical and Translational Oncology, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 2Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH.
Background: The breast tumor microenvironment is influenced by miRNAs, which can act as biomarkers for cancer subtyping, detection, and treatment. However, the relationship between microenvironment and miRNA in TNBC molecular subtypes is underexplored. We hypothesized that there are subtype-specific cell enrichment patterns, whose heterogeneity is reflected in immunomodulatory and stroma-modulatory miRNA expression diversity. Methods: Normalized gene expression data was analyzed for TCGA, METABRIC, and GSE31519 cohorts. Normalized MiRNA sequencing data (ppm) was obtained for TCGA. TNBCType was used to classify cases as basal-like 1 (BL1), basal-like 2 (BL2), luminal androgen receptor (LAR) and mesenchymal (M). XCell 2.0 was used to compute immune and stromal enrichment. For TCGA, normalized miRNA findings were integrated with XCell results through correlates. Krusal-Wallis tests, Bonferroni correction, and Dunn’s tests were performed in R. Results: Across cohorts, (n = 1038; n = 195 validation), TNBC subtypes showed heterogeneity in stromal (p < 0.000001), immune (p < 0.00001) and microenvironment scores (p = 2.59e-8). BL1 had enriched CD8+ T, NK and dendritic cells. BL2 exhibited elevated epithelial cells, neutrophils and M2-like macrophages (p < 0.01). M was marked by low T-cells, and high mesenchymal stromal cells. LAR had enriched B (p < 0.01) and plasma cells. The TCGA cohort (n = 175) showed differential composition in 13/15 tested cell types: adipocyte, endothelial, CD8+ T, CD4+ T, M1 and M2 macrophage, NK, B, dendritic, neutrophil, monocyte, mast (all p < 0.001), and fibroblast (p = 0.008). There was expression variation across subtypes for 12/17 profiled miRNAs, including miR-10b, miR-146a/b, miR-155, miR-181a-1/2, miR-222, miR-223 (all p < 0.001); and miR-27a (p = 0.003) (Table). MiR-155 and miR-146a/b correlated positively with immune scores, CD8+ T cells, and M1 macrophages. Elevated miR-146a was seen in BL1 compared to M, consistent with miR-146a status as an epithelial-to-mesenchymal transition (EMT) inhibitor. High miR-146a/b expression in BL2 reflected inflammatory modulation, and lower levels in M may allow for EMT. High miR-155 expression in BL1 aligned with the immune-enriched phenotype. EMT promoters, miR-181a-1 and miR-181a-2, were elevated in M. Elevated miR-222 and miR-223 in BL2 correlated with immune enrichment. MiR-200a/b levels did not significantly differ across subtypes, consistent with their shared epithelial features. Conclusion: Using the largest public miRNA-seq dataset, this study provides novel mechanistic insight into the TNBC microenvironment and reveals promising miRNA biomarkers: miR-146a for BL1, miR-146b and miR-222 for BL2, miR-3615 for BL 1/2, miR-10b for LAR, miR-181a/b for M, low miR-223 for M (Table), with implications for diagnostic and therapeutic use.
Presentation numberPS3-12-06
Immune microenvironment and survival differences among Hispanic and African American breast cancer cases by biopsy site
Robert Hsu, University of Southern California, Los Angeles, CA
R. Hsu1, S. Deshmukh2, N. Gyabaah-Kessie1, E. Rallos3, B. Al-Zubeidy1, E. Gonzalez1, A. G. Baugh1, S. Carrel4, M. Li5, D. Zavala1, A. Martynova1, P. Jayachandran1, D. Stewart1, D. Spicer1, S. Wu6, J. Xiu6, G. W. Sledge Jr.6, R. L. Mahtani7, A. C. Sandoval Leon7, S. Gandhi8, S. Chumsri9, M. Lustberg10, M. C. Stern11, E. T. Roussos Torres1; 1Medicine, University of Southern California, Los Angeles, CA, 2Clinical and Translational Research , Caris Life Sciences, Phoenix, AZ, 3Medicine, Eastern Virginia Medical School, Fairfax, VA, 4Medicine, University of South Carolina, Greenville, SC, 5Medicine, New York Medical College, Valhalla, NY, 6Clinical and Translational Research, Caris Life Sciences, Phoenix, AZ, 7Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 8Medicine, Emory University, Atlanta, GA, 9Medicine, Mayo Clinic, Jacksonville, FL, 10Medicine, Yale University, New Haven, CT, 11Population and Public Health Sciences, University of Southern California, Los Angeles, CA.
Background: Hispanic/Latinx (HL) and African American (AA) breast cancer (BC) patients (pts) have increased prevalence of high-risk features such as incidence of BC at age <50 and estrogen receptor negativity. We previously showed that AA and HL had a lower fraction of neutrophils and M2 macrophages (Mɸ) despite higher percentage of PD-L1 positive cases. This study aims to further characterize differences in the tumor immune microenvironment between racial and ethnic groups, with a focus on how these differences vary by site of biopsy.Methods: We analyzed 19,459 BC samples tested by NGS (592, NextSeq; WES, NovaSeq) and WTS (NovaSeq; Caris Life Sciences). Race/ethnicity data were self-reported by pts. Immune cell proportions were estimated using WTS deconvolution (Quantiseq). Gene expression was analyzed for T-cell inflammed score (TIS), MAPK Activation Score (MPAS) and interferon-gamma (IFNy) score. Real-world overall survival (OS) data were derived from insurance claims, calculated from tissue collection or treatment initiation to last contact, and analyzed using Kaplan-Meier. Statistical significance was assessed by chi-square and Mann-Whitney U test with p-values adjusted for multiple comparisons (q<0.05).Results: There were 9674 Non-Hispanic Whites (NHW) (primary BC, defined as biopsy (bx) from breast) pBC: n=3,537, 36.6%; (metastatic BC, defined as biopsy outside of the breast) mBC: n=6,137 63.4%, 1800 AA (pBC: n=750, 42%, mBC: n=1,050, 58%), and 1795 HL (pBC: n=842, 47%; mBC n=953, 53%) samples. There were 2364 TNBC (66% NHW, 19% NHB, 15% HL), 4318 HR+/HER2- (76% NHW, 11.6% NHB, 12.4% HL), and 893 HER2+ (68% NHW, 15% NHB, 17% HL). In pBC overall, AA had lower median cell fraction (%) of B cells (4.8% vs 5.3%), M2 Mɸ (3.4% vs 4.4%), but higher median cell fraction of dendritic cells (DC) (2.8% vs 2.6%) compared to NHW while HL had lower median cell fraction of M2 Mɸ (3.9% vs 4.4%) and higher median cell fraction of CD8+ T cell (0.4% vs 0.2%), all q<0.05. In pBC, HL had higher IFNy score (-0.3 vs -0.34), while AA had lower MPAS (-1.2 vs -0.96), all q<0.05. In lymph node bx, AA had lower median % of M2 Mɸ (3.4% vs 4.4%, q<0.05). No significant differences were seen in the liver bx by race but notably there were lower median cell fractions of CD4+, CD8+, and regulatory T cells (0.0 across NHW, AA, HL) across all race/ethnicities vs. other biopsy sites. By subtype, TNBC primary bx yielded much of the significant changes with AA having lower median cell fraction of M2 Mɸ (2.49% vs 3.04%, q= 0.01) compared to NHW. In primary bx, AA (35.6 m vs 48.5 m, HR 1.4, 95% CI 1.2-1.5, p<0.001) and HL (42.4 m vs 48.5 m, HR 1.2, 95% CI 1.1-1.4, p<0.001) had worse median OS versus NHW. In lymph node bx, AA also had worse OS (29.2 m vs 37.2 m, HR 1.2, 95% CI 1-1.5, p=0.03) compared to NHW. In liver bx, there was worse mOS overall but no difference by race (NHW 18.65m, AA 18.79m, HL 20.6m, p =0.73). In HR+/HER2- liver bx AA had worse OS (14.2 m vs 20.9 m, HR 1.3, 95% CI 1-1.7, p=0.03) versus NHW. In HER2+ primary bx, HL had worse OS (43.2m vs 66.9m, HR 1.7, 95% CI 1.1-2.6, p=0.009). There was a trend towards worse mOS in TNBC AA patients receiving pembrolizumab (18.42m vs HL 23.69m vs NHW 35.30m; p=0.16) and HER2+ AA patients receiving trastuzumab deruxtecan (AA: 16.06m vs HL: 26.81m vs NHW: not reached; p = 0.05). Conclusions: We found race and biopsy site specific differences in the breast cancer immune microenvironment. AA pts had lower M2 Mɸ in primary and lymph node bx while NHW pts showed higher CD8+ T cell in primary tumors. Liver biopsies exhibited low T cell infiltration across all groups. AA pts had worse mOS in primary and lymph node bx compared to NHW but not in liver bx of which we saw worse mOS in all races. These results highlight the need to consider both race and bx site in treatment decisions.
Presentation numberPS3-12-07
Tertiary Lymphoid Structures as Histopathologic Biomarkers in Invasive Breast Cancer: Subtype-Specific Patterns and Clinical Implications
Alyssa J. Cozzo, University of North Carolina at Chapel Hill, Chapel Hill, NC
A. J. Cozzo1, S. C. Van Alsten1, L. T. Olsson2, Q. Wang3, K. A. Hoadley1, M. A. Troester1; 1Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 2Department of Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, 3Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC.
Invasive breast cancer encompasses a range of tumor subtypes with variable immune landscapes that influence prognosis and treatment response. Tertiary lymphoid structures (TLS), organized immune aggregates resembling secondary lymphoid organs, may serve as histologic indicators of local antitumor immunity. We evaluated clinicopathologic and immune features associated with TLS presence and maturity using histology-based scoring and gene expression–based modeling. TLS and tumor-infiltrating lymphocytes (TILs) were scored on H&E-stained whole slide images (N = 842) from the Carolina Breast Cancer Study (CBCS). Associations with clinicopathologic features and progression-free survival (PFS) were assessed using multinomial logistic and Cox models. Immune expression profiles of TLS groups informed development of weighted centroid classifiers: a 10-gene TLS classifier based on Tfh genes enriched in TLS-positive tumors and B cell genes elevated in tumors with mature (mTLS) vs. only immature (iTLS), and an 18-gene TIL signature derived from remaining lymphocyte markers upregulated in TLS-positive cases. Classifiers were trained and tested on non-overlapping subsets of the CBCS cohort, then applied to the I-SPY2 neoadjuvant trial dataset to assess association with pathologic complete response (pCR). TLS were identified in 38% of tumors and varied in number and location. Subjects were assigned to one of three TLS groups: No TLS, iTLS Only, or mTLS Present (>93% of which also contained iTLS). mTLS Present tumors harbored approximately 2-fold the total TLS compared to iTLS Only and were over twice as likely to contain intratumoral TLS (54.2% vs. 26.3%). TLS presence and maturation were significantly associated with younger age (<50), Black race, and high grade (mTLS ORs 1.66-6.28) as well as ER-negativity and basal-like or ER−/HER2+ subtypes (mTLS ORs 6.70-17.00, all p < 0.001). TLS were also enriched in tumors with genomic instability (mutant-like vs. wild type-like p53, OR = 5.57; high vs. low homologous recombination deficiency scores, OR = 4.21) and high-risk ROR-PT scores (OR = 6.50; all p < 0.001). Stromal TIL scores were strongly correlated with TLS group and total TLS burden (mTLS OR = 21.06, p < 0.001; ρ = 0.54, p < 0.001, respectively). In unadjusted analyses of ER− tumors, both high TIL strength (HR = 0.52, 95% CI: 0.27–0.99, p = 0.047) and presence of mTLS (HR = 0.34, 95% CI: 0.15–0.79, p = 0.012) were significantly associated with improved PFS. However, in multivariable models adjusting for both histologic TLS group and TIL score, only mTLS remained independently associated with improved PFS (HR = 0.39, p = 0.036), while TIL score was not (HR = 0.67, p = 0.27). Our expression-based classifiers mirrored these findings: predicted mTLS was significantly associated with improved PFS in univariate models (HR = 0.42, p = 0.007) and retained near-significance after adjusting for predicted TIL score (HR = 0.46, p = 0.057), while the TIL predictor was no longer significant (HR = 0.90, p = 0.75). In the I-SPY2 neoadjuvant trial (N = 987), predicted mTLS tumors had the highest pCR rates across TLS groups (39% overall, p = 0.055; 52% in HR− subset, p = 0.028). After adjustment for treatment arm, compared to TLS-low tumors, predicted mTLS remained associated with greater odds of pCR in the overall cohort (OR = 1.36, 95% CI: 0.99–1.86, p = 0.054) and the HR− subset (OR = 1.50, 95% CI: 0.96–2.33, p = 0.074), though these relationships did not reach statistical significance. These findings demonstrate that both histologically-defined and gene-predicted mTLS are independently associated with improved survival in ER− breast cancer and identify tumors more likely to respond to neoadjuvant therapy. These results support incorporating TLS assessment into future breast cancer classification and treatment frameworks.
Presentation numberPS3-12-08
Rac/cdc42 inhibitor mbq-167 modulates the breast tumor immune microenvironment
Suranganie Dharmawardhane, University of Puerto Rico Medical Sciences Campus, San Juan, PR
A. Torres-Sanchez1, A. Cruz-Collazo1, N. Grafals1, S. Dorta-Estremera2, S. Dharmawardhane1; 1Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, PR, 2Microbiology, University of Puerto Rico Medical Sciences Campus, San Juan, PR.
A uniform therapeutic approach for metastatic breast cancer has been challenging, especially for triple negative breast cancer (TNBC), due to the inherent heterogeneity and lack of targetable receptors. Therefore, we developed MBQ-167, as a specific inhibitor of the homologous Rho GTPases Rac/Cdc42 and demonstrated its efficacy in pre-clinical mouse models of HER2 (3+) and TNBC. MBQ-167 is currently in Phase 1 clinical trials in advanced BC patients, and so far, have not shown any adverse events, and is bioavailable at acceptable levels. Emerging data show the relevance of the tumor microenvironment (TME) to malignant cancer progression, where immunosuppressive immune cells such as macrophages, neutrophils, and Myeloid-Derived Suppressor Cells (MDSCs) are recruited to the TME to promote tumor cell invasion. Moreover, in the TME, cancer cells may hijack the infiltrating cytotoxic T cells by expressing programmed death ligand (PD-L1), which binds the PD-1 receptor on T cells to induce T cell death. The crosstalk signaling between the immune cells in the TME to instigate metastatic cancer cell migration into the circulatory system is regulated by Rac and Cdc42. Therefore, we investigated the effect of MBQ-167 on the TME and inflammatory responses to further demonstrate its potential as a TNBC therapeutic. By immunophenotyping cells from spleens and tumors following administration of vehicle or MBQ-167 to Balb/C mice bearing 4T-1 TNBC tumors, we found that MBQ-167 treatment significantly reduced macrophages (particularly M2 macrophages), MDSCs, and increased CD8+ T cells. Parallel to a decrease in lung metastases, MBQ-167 increased the CD8+ Granzyme B+ regulatory T cell ratio. Of the cytokines released into the TME to activate inflammatory and immunosuppressive immune cells, we found Interleukin-6 (IL-6) to be significantly decreased in mouse plasma following MBQ-167 treatment. As published by us, gene set enrichment analysis of the transcriptomic data from 4T-1 tumors in Balb/c mice treated with vehicle or MBQ167 demonstrated a statistically significant ~2-fold downregulation in the inflammatory mediators chitinase-3-like protein 1 (CHI3L1) and S100A9 that have also been associated with breast cancer metastasis. Therefore, ELISAs were performed following vehicle, or MBQ-167 using conditioned media from TNBC cells and macrophages in tissue culture, as well as serum from Balb/C mice with 4T-1 TNBC tumors. Results show that MBQ-167 treatment significantly reduced CHI3L1 and S100A8/A9 levels from HER2+ breast cancer cells, macrophages, and serum from Balb/C mice bearing 4T-1 TNBC. In addition to creating a favorable immune environment for inhibition of tumor growth and metastasis, MBQ-167 treatment also increased PDL1 levels in MDSCs and macrophages from spleens and tumors of mice with TNBC tumors following MBQ-167 treatment. Therefore, we tested the effect of MBQ-167 individually (50mg/kg 5X a week for 4 weeks) or in combination with anti-PD1 or anti-PDL1 (10mg/kg every 3 days for 2 weeks) in the 4T-1/Balb/C mouse model. Data show that MBQ-167 administration resulted in 80% reduction in tumor growth with no toxic effects. MBQ-167 was more effective than either anti-PD1 or anti-PDL1 in single or combined therapy in this TNBC mouse model. In conclusion, the Rac/Cdc42 inhibitor MBQ-167 targets metastatic BC cells and immunosuppressive inflammatory cells in the BC TME and is a viable TNBC therapeutic. Funded by DoD/BCRP HT94252310166, BC220526 and HT94252410094, BC230070.
Presentation numberPS3-12-09
Spatial immune profiling and pathological response after neoadjuvant therapy in early HER2-positive breast cancer
Carla Gullotta, Hospital Clinic Barcelona, Barcelona, Spain
C. Gullotta1, S. Cobo1, V. Albarràn Fernàndez1, C. Rubio-Perez1, E. Sanfeliu2, P. Galván1, A. Aguirre1, O. Castillo1, P. Blasco1, A. Martínez- Romero1, M. Marín-Anguilera3, B. Walbaum1, O. Martínez-Sáez1, M. Bergamino1, M. Vidal1, M. Muñoz1, T. Pascual1, B. Adamo1, F. Braso-Maristany1, A. Prat1, L. Angelats1; 1IDIBAPS, Hospital Clinic Barcelona, Barcelona, SPAIN, 2Pathology Department, Hospital Clinic Barcelona, Barcelona, SPAIN, 3IDIBAPS, Reveal Genomics, Barcelona, SPAIN.
Background:HER2-positive (HER2+) breast cancer is a heterogeneous disease, with variability in tumor architecture, immune infiltration, and protein expression within both the tumor (TU) and the surrounding tumor microenvironment (TME). A better understanding of how the TME influences treatment response is critical. This study investigates whether protein expression levels and intratumoral heterogeneity are associated with pathological complete response (pCR) in patients with HER2+ early breast cancer (EBC) treated with neoadjuvant therapy. Methods:A total of 54 HER2+ EBC core biopsies from patients treated with neoadjuvant HER2-targeted chemotherapy at Hospital Clinic of Barcelona (April 2022 – September 2023) were analyzed using GeoMx Digital Spatial Profiling. Protein expression was measured in the immune (CD45⁺) and tumor (PanCK⁺) compartments. For each sample, a minimum of three TU and TME regions of interest (ROIs) were selected to ensure adequate spatial coverage and capture intratumoral heterogeneity. Protein expression data were batch-corrected; for each ROI we computed the mean, standard deviation (SD), and coefficient of variation (CV). CV, as a relative metric, served as a proxy for intraregional heterogeneity, while the mean reflected protein expression. In parallel, the HER2DX genomic test was performed on bulk RNA from the same biopsy to evaluate transcriptomic correlates; PAM50 was also assessed. Stromal tumor-infiltrating lymphocytes (TILs) were assessed according to international guidelines. Associations between protein metrics, HER2DX scores, TILs, and pCR were tested using Wilcoxon, Kruskal–Wallis, and logistic regression in RStudio. Results:Among the 54 HER2+ cases, the median age was 58 years, and the pCR rate was 51.9%. Clinical and biological features showed general concordance with those reported in a HER2+ neoadjuvant cohort, including PAM50 subtypes (11.3% Basal-like, 43.4% HER2-Enriched, 18.9% Luminal A, 20.8% Luminal B, 5.7% Normal-like), menopausal status (66.7% postmenopausal), hormone receptor (HR) status (72.2% positive), and HER2DX pCR score (31.4% high, 27.5% medium, 41.2% low). Higher HER2 mean expression in TU ROIs (Odds ratio [OR] = 1.54; p = 0.008), but not HER2 CV (p = 0.327), was associated with pCR. In TME ROIs, increased CVs of T cell markers, CD3 (OR = 1.30; p = 0.020), CD4 (OR = 1.40; p = 0.010), CD8 (OR = 1.24; p = 0.040), and the plasma cell marker CD27 (OR = 1.24; p = 0.030), were also significantly associated with a higher probability of pCR, whereas their mean expression levels were not; while T cell exhaustion TIM3 marker mean expression in TME ROIs was inversely associated with pCR (OR = 0.23; p = 0.042). TILs levels, as quantitative and qualitative variable, were not significantly associated with pCR in this cohort (OR = 1.01, p = 0.300). Finally, the HER2DX pCR score was significantly associated with pCR status, both when analyzed as a continuous variable (OR 1.90 per 10-unit increase; 95% CI = 1.34–2.77; p<0.001) and when compared across predefined low and high score groups (OR = 88.7; p<0.001). These associations remained significant across multivariate models that included spatial protein expression metrics and HR status, whether the HER2DX pCR score was modeled as a continuous variable or as categorical risk groups. Conclusions:In HER2+ EBC, spatial heterogeneity of immune-related proteins, mainly the presence of T-cell markers, was associated with a higher pCR rate, while expression levels were not, suggesting a role for immune variability within the TME in modulating treatment efficacy. These results also highlight total HER2 abundance over regional heterogeneity. The HER2DX pCR score remained a robust predictor of response in all statistical analyses and spatial metrics did not outperform or override its predictive value in this cohort.
Presentation numberPS3-12-10
Enhancing responses to trastuzumab deruxtecan using tucatinib in HER2-negative breast cancer
Daniel S Peiffer, University of Chicago Hospitals, Chicago, IL
D. S. Peiffer, N. Thompson, L. Nguyen, N. Chen, F. M. Howard, H. Shah, K. Cole, R. Nanda, G. L. Greene; Medicine, University of Chicago Hospitals, Chicago, IL.
Background: HER2 status in breast cancer (BC) is determined by a combination of immunohistochemistry (IHC) and in situ hybridization. HER2-positive (HER2+) tumors overexpress the marker, whereas HER2-negative tumors do not. However, approximately ½ of all BCs express modest levels of HER2, defined as HER2-low BC, with the remaining tumors expressing no detectable levels of HER2, considered HER2-0 disease. The antibody drug conjugate (ADC) trastuzumab deruxtecan (T-DXd) has been shown to improve progressive-free and overall survival in metastatic HER2-low BC, leading to its FDA approval for the treatment of this population. Unfortunately, patients with HER2-0 BC are not eligible to receive T-DXd, despite trials demonstrating a subset of these patients respond to the ADC. Recent studies have shown that reversible tyrosine kinase inhibitors (TKIs) like tucatinib can increase membrane HER2 expression, potentially enhancing the efficacy of HER2-targeted ADCs. We hypothesize that tucatinib may increase HER2 expression in the HER2-0 BC, enhancing patient responses to T-DXd with low target expression. Methods: Baseline HER2 expression was assessed using IHC and flow cytometry (FC) across BC cell lines and patient-derived organoids (PDOs). CRISPR HER2-KO variants were created from HER2-low (MCF-7, T47D) and HER2-0 (MDA-MB-231, SUM149) cell lines. To assess T-DXd efficacy with or without tucatinib, growth assays were performed in vitro using Incucyte live-cell imaging and ATP quantification. Tucatinib’s effect on HER2 protein levels, stability, and downstream signaling were examined by FC, cycloheximide chase experiments, and western blot/qPCR analysis, respectively, to assess protein levels, stability, and downstream signaling. T-DXd uptake rates were measured using fluorescently tagged T-DXd by flow cytometry. Additionally, mass spectrometry quantified the deposition of the chemotherapy payload, deruxtecan (DXd). Results: T-DXd efficacy correlated with HER2 levels, as determined by FC and IHC, with membrane HER2 expression being required for response. Tucatinib increased membrane HER2 levels across all parental and CAS9-control cell lines, with no effect on the HER2-KO variants. Pulsing with tucatinib achieved similar results to continuous co-treatment in terms of membrane HER2 levels. The combination of T-DXd and tucatinib had little effect on cell growth in T-DXd-sensitive cell lines. However, it significantly enhanced efficacy in de novo resistant cell lines. These results correlated with increased T-DXd uptake and DXd deposition in vitro. Conclusions: The combination of T-DXd and tucatinib represents a novel treatment strategy that may expand the patient population that responds to T-DXd, particularly those with HER2-0 BC who tend to have highly variable responses to T-DXd at baseline. Given the increased side effect burden associated with tucatinib when used in combination with HER2-targeted ADCs, further studies evaluating tucatinib pulsing strategies are warranted and ongoing. This approach could optimize treatment efficacy while minimizing toxicity, improving outcomes for BC patients, particularly those with HER2-0 disease.
Presentation numberPS3-12-11
Neutrophil elastase promotes myoepithelial layer dissolution and breast cancer progression
Tarek Taifour, McGill University, Montreal, QC, Canada
T. Taifour1, Y. Gu2, A. Massé2, V. Sanguin-Gendreau3, D. Zuo3, B. Xiao3, S. Attalla3, W. Muller1; 1Goodman Cancer Institute and Department of Experimental Medicine, McGill University, Montreal, QC, CANADA, 2Goodman Cancer Institute and Department of Biochemistry, McGill University, Montreal, QC, CANADA, 3Goodman Cancer Institute, McGill University, Montreal, QC, CANADA.
Breast cancer progression from ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) is a critical transition that underlies the development of metastatic disease. DCIS now accounts for ~20% of all breast cancer diagnoses due to early screening; however, only about half of these cases will progress to IDC. Despite this, there are currently no biomarkers to identify high-risk DCIS, leading to widespread over-treatment. A defining hallmark that distinguishes DCIS from IDC is the presence of an intact myoepithelial cell layer, which physically restrains tumor invasion. Disrupting this layer is a requirement for breast cancer progression but remains a poorly understood process. Elucidating mechanisms of myoepithelial disruption is crucial to identify targetable biomarkers of DCIS progression and improve treatment of early breast cancer. Neutrophils are prominent pro-tumorigenic immune cells associated with poor outcomes and resistance to therapy. While elevated neutrophil levels have been reported in DCIS patient samples, their functional role in early progression remains unknown. In this study, we demonstrate that neutrophil depletion in breast cancer Genetically Engineered Mouse Models (GEMMs) results in striking retention of the myoepithelial layer and halts DCIS transition to IDC. Similarly, genetic ablation of the secreted protein Chitinase-3 like 1 (Chi3l1), a key neutrophil chemoattractant, phenocopies neutrophil depletion in two independent GEMMs and inhibits DCIS progression. In contrast, Chi3l1 overexpression leads to increased neutrophil infiltration, accelerated myoepithelial layer loss, and enhanced lung metastasis, a phenotype reversed by neutrophil depletion. Furthermore, elevated neutrophil levels in human DCIS samples also correlated with significant disruption of the myoepithelium. Mechanistically, we identify neutrophil elastase (NE) as a major effector: NE is secreted by tumor-infiltrating neutrophils and directly promotes myoepithelial layer dissolution. Pharmacological inhibition of NE preserves the myoepithelial barrier and significantly impairs DCIS progression. Despite the fact that only 50% of DCIS cases progress to IDC, the lack of predictive markers has led to widespread over-treatment. Our results reveal a novel immunological mechanism driving myoepithelial layer disruption and highlight neutrophils and NE as potential biomarkers and therapeutic targets for DCIS progression. These findings could help stratify DCIS patients for treatment based on neutrophil activity or NE expression, reducing over-treatment while targeting high-risk cases. Given the availability of NE inhibitors, our work offers a translationally actionable framework for preventing invasive breast cancer.
Presentation numberPS3-12-12
Association between Intratumoral Microbiota and Immune Microenvironment in Breast Cancer
Akimitsu Yamada, Yokohama City University Graduate School of Medicine, Yokohama, Japan
A. Yamada1, K. Kawashima1, M. Sasamoto1, H. Fukuoka1, M. Oshi1, K. Narui2, T. M. Dieter3, Y. Sekiguchi3, T. Ishikawa4, I. Endo1; 1Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, JAPAN, 2Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, JAPAN, 3Molecular Biosystems Research Institute, The National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, JAPAN, 4Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, JAPAN.
Background Cancer research has revealed intratumoral microbiomes as active modulators of tumor biology, distinct from gut microbiota. In breast cancer—the leading malignancy among Japanese women—these resident bacteria influence immune responses, inflammation, metabolism, and therapy resistance. Bacterial composition varies by tumor subtype and correlates with immunological activity and treatment outcomes. Microbes likely access breast tissue via hematogenous spread or ductal migration, then interact with immune cells, tumor cells, and stroma to shape the tumor microenvironment (TME). However, their clinical significance regarding immune infiltration, gene expression, and interactions with cancer stem cells—which drive tumor initiation, progression, and therapeutic resistance—remains unclear. Our study characterizes breast cancer’s intratumoral microbiota and investigates its role in immune landscape formation and transcriptomic profiles. By integrating microbiome and transcriptome analyses from paired tumor, normal, and fecal samples, we aim to elucidate how microbial ecosystems influence breast cancer heterogeneity, cancer stem cell (CSC) biology, and overall tumor behavior. Objective To investigate how intratumoral microbiota affects the TME and CSC distribution in breast cancer and to examine its relationship with clinical outcomes. Methods We collected fecal samples, tumor tissues, and adjacent normal tissues from 16 breast cancer patients who underwent surgical resection without neoadjuvant therapy. The median patient age was 65 years (range: 38-91 years). 16S rRNA gene amplicon sequencing was performed for both fecal and tissue samples. RNA sequencing of tumor tissues was conducted to evaluate the expression of cytokines, immune-related genes, and CSC markers. Microbiota profiles were analyzed in conjunction with transcriptomic data to identify potential correlations. Results Substantial interindividual variation in microbial profiles was observed within tumor tissues. Jaccard similarity analysis of microbial communities between tumor and matched normal tissues demonstrated a moderate positive correlation with intratumoral bacterial load (r = 0.67, p = 0.036). Several bacterial genera showed specific enrichment in tumor tissues compared to patient-matched normal tissues. Comparative analysis of fecal-tumor microbiota revealed shared amplicon sequence variants (ASVs) of 16SrRNA genes in 11 of 16 patients, suggesting multiple potential routes of bacterial translocation, including hematogenous and ductal pathways. Transcriptomic analysis revealed that elevated bacterial load was associated with altered immune signatures, including modifications in regulatory T cell and NK cell activity as assessed by xCell deconvolution analysis. However, CSC-specific signals were limited due to the low fraction of ALDH1-positive cells in our cohort. Gene Set Enrichment Analysis identified differences in hypoxia response, TNF-α signaling, and E2F target pathways that correlated with microbial abundance levels. Conclusion Our findings suggest that intratumoral microbiota may influence the immune microenvironment and key molecular pathways in breast cancer, potentially affecting disease progression. These results provide foundational evidence for the development of microbiota-targeted therapeutic strategies in breast cancer management. Further studies with larger cohorts and functional validation are warranted to establish causal relationships and clinical implications.
Presentation numberPS3-12-13
Adipocyte-tumor cell interactions drive aggressiveness in triple-negative breast cancer
Tiziana Triulzi, Fondazione IRCCS Istiututo Nazionale dei Tumori di Milano, Milano, Italy
T. Triulzi1, M. Francesconi1, S. Iannicelli1, V. Regondi1, L. De Cecco1, E. Tagliabue1, S. M. Pupa1, A. Vingiani2; 1Department of Experimental Oncology, Fondazione IRCCS Istiututo Nazionale dei Tumori di Milano, Milano, ITALY, 2Department of Diagnostic Innovation, Fondazione IRCCS Istiututo Nazionale dei Tumori di Milano, Milano, ITALY.
Triple-negative breast cancer (TNBC) is a particularly aggressive subtype of breast cancer, representing approximately 10-20% of all cases. It is distinguished by a unique tumor microenvironment in which immune components play a critical prognostic role. Beyond immune cells, various stromal elements contribute to cancer progression, with tumor cells actively shaping their microenvironment through bidirectional interactions that influence the recruitment and functional phenotype of these surrounding cells. Among stromal components, adipocytes—the predominant stromal cells in breast tissue—have gained increasing attention. This study aimed to determine whether interactions between TNBC cells and adipocytes promote cancer aggressiveness. At the cellular level, we found that the interaction between tumor cells (MDA-MD-231, SUM149PT, HCC1937, BT549 and MDA-MB-468) and mature adipocytes—derived from differentiation of ASC52 mesenchymal stem cells—induces adipocyte delipidation, marked by a reduction in both the number and size of lipid droplets and a significant decrease in overall lipid content compared to control adipocytes, as quantified by Oil red O staining. This phenotypic shift is accompanied by a marked downregulation of mature adipocyte markers, including PPARG, CEBPA, and ADIPOQ, as well as reduced PPARG transcriptional activity, evidenced by decreased expression of FABP4. Furthermore, these adipocytes exhibit a distinct transcriptional profile enriched in inflammatory pathways and genes involved in tumor microenvironment interactions. Tumor cells exposed to adipocytes acquire features of epithelial-mesenchymal transition and cancer stemness, as demonstrated by qPCR analysis of specific markers and flow cytometry. In patients, we defined an adipose tissue infiltration (ATi) score, defined as involvement of ≥10% of the tumor margin (ATi-positive), reflecting interaction between tumor cells and adipocytes. In a cohort of 92 consecutive patients with primary TNBC who underwent surgery and received adjuvant chemotherapy, the ATi score was associated with a higher relapse rate (p=0.0152), lower tumor-infiltrating lymphocytes (TILs), and older age. Notably, ATi-positive status was significantly associated with poorer disease-free survival (DFS) (HR=3.13; 95% CI: 1.16-8.50). This association was independent of other clinico-pathological factors including age, tumor size, lymph node involvement, and TILs in multivariate analysis, supporting the role of tumor-AT crosstalk in TNBC progression. Collectively, our findings strongly indicate that the interaction between tumor cells and adipocytes contributes to the aggressiveness of TNBC, underscoring the importance of further investigating the underlying mechanisms for potential therapeutic targeting. Supported by Fondazione AIRC
Presentation numberPS3-12-15
Mapping the imprint of taxane chemotherapy on the metastatic microenvironment in HR+ breast cancer with single cell and spatial profiling
Daniel L Abravanel, Dana-Farber Cancer Institute, Boston, MA
R. Strasser1, Y. Tam1, R. M. Oscanoa1, Å. Segerstolpe2, J. Nelson3, C. McCann3, L. DelloStritto4, A. Frangieh4, K. Helvie4, S. Vigneau4, A. R. Thorner4, K. Nguyen4, S. Strauss4, N. U. Lin4, S. M. Tolaney4, S. Farhi3, B. E. Johnson4, N. Wagle4, J. Klughammer1, D. L. Abravanel4; 1Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, GERMANY, 2Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 3Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA, 4Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.
Introduction: The tumor microenvironment (TME) plays a prominent role in critical elements of breast cancer progression including metastatic dissemination and therapeutic response and resistance. Differences between efficacy of checkpoint inhibitors as monotherapies as compared to combinations with conventional chemotherapies or antibody drug conjugates promotes the hypothesis that cancer-directed therapies not only impact tumor cells directly but also shape TME dynamics with potentially pivotal consequences on therapeutic response. Unfortunately, the impact of even common therapies on TME interactions as they occur in patients remains incompletely defined, particularly in HR+ metastatic breast cancer (MBC). Methods: Here, we leveraged recent advances in single-cell and spatial profiling techniques to address this shortcoming. Biopsy cores of liver metastases from 40 patients with HR+ MBC were profiled with both single-nucleus RNA sequencing (snRNA-seq) and Multiplexed Error-Robust Fluorescence In Situ Hybridization (MERFISH). Results: Integrating the snRNA-seq and MERFISH profiles yielded detailed description of the composition, gene expression profiles/programs and spatial organization of HR+ liver metastases. Of the 40 biopsies, 32 were from patients previously treated with taxanes: 16 in the adjuvant setting (treatment-biopsy interval 18-222 months; mean 84); 10 in both the adjuvant and metastatic settings (interval 0-61 months; mean 18); and 6 only in the metastatic setting (interval 1-48 months; mean 20). Multiple gene expression profiles were increased in biopsies from patients exposed in the metastatic setting only. The malignant cells from these were characterized by enriched oxidative phosphorylation (Wilcoxon rank sum p=0.022), glycolysis (p=0.029), and hypoxia (p=0.033) gene sets as well as increased MYC regulon activity. Correspondingly, the TME was enriched for HIF1A regulon activity as well as SPP1+ lipid-scavenging (p = 0.047) and angiogenic (p=0.035) macrophages, pericytes (p=0.031), lymphatic endothelial cells (p=0.009), and proliferating endothelial cells (p=0.003). Integrated spatial analyses revealed that this phenotype was elevated in regions of metastatic involvement compared to the adjacent normal liver (e.g. p=0.002 for lipid-scavenging macrophages), while interferon-induced inflammatory macrophages were relatively depleted (p=0.040). Conclusions: Taken together, these data not only furnish a detailed atlas of liver metastases in HR+ breast cancer, a common and clinically important site of metastatic spread with adverse prognostic implications in the most common, but seemingly least immunogenic, breast cancer subtype, but also map the imprint of taxane-based chemotherapy. We identify the setting of taxane exposure as a critical determinant of the impact of this key therapeutic class, with specific relevance in combination with immunotherapy, on the TME. Notably, taxane exposure limited to the metastatic setting was associated with a hypoxic/angiogenic and immunosuppressive/immune-excluded TME, even compared to cases with taxane treatment in both the adjuvant and metastatic setting, highlighting opportunities for future clinical translation.
Presentation numberPS3-12-16
Reprogramming the Tumor Microenvironment in TNBC: A Novel Strategy Combining Notch Inhibition with Radiotherapy and Checkpoint Blockade
Eileen P Connolly, Columbia University Medical Center, New York, NY
Q. Wang1, D. Banerjee2, E. C. Connolly3, D. Yamashiro2, E. P. Connolly1; 1Radiation Oncology, Columbia University Medical Center, New York, NY, 2Pediatric Oncology, Columbia University Medical Center, New York, NY, 3Radiation Oncology, Emory Unirsity, Atlanta, GA.
Background:High-dose radiotherapy (HDRT) is immunogenic, promoting tumor antigen release and enhancing immune checkpoint inhibitor (ICI) efficacy. However, HDRT also induces an immunosuppressive tumor microenvironment (TME), limiting the therapeutic response of combined RT + ICI. The Notch signaling pathway is a critical regulator of immune and stromal cell behavior. We hypothesized that inhibiting Notch signaling using a γ-secretase inhibitor (GSI) AL101 would overcome radiation-induced immunosuppression and synergize with HDRT and αPD-1 therapy in triple-negative breast cancer (TNBC).Methods:EO771 murine TNBC tumors were established in syngeneic C57BL/6 mice and athymic nude mice. Mice received GSI AL101 (6.5 mg/kg daily ×10), a single 12 Gy dose of HDRT (Day 3), and αPD-1 antibody (200 µg, Days 0, 3, and 6). A group of mice were sacrificed on Day 10 for mechanistic analyses; others were followed for survival and euthanized when tumors reached 1.5 cm³. Tumors were analyzed by spectral flow cytometry for immune cell subsets. Lung metastasis were evaluated by histology image analysis for metastasis/total lung area using HALO image analysis.Results:Neither GSI nor αPD-1 monotherapy inhibited EO771 tumor growth (median survival: control 14d vs GSI 18d and αPD-1 15d, p>0.05). GSI + HDRT modestly prolonged survival compared to RT alone (41d vs 35d, p < 0.05). Triple therapy (GSI + HDRT + αPD-1) resulted in durable tumor growth control and improved survival (98.5d, p <0.0001). Triple therapy also led to a marked reduction in lung metastases at day 10 (mean metastasis/lung area GSI + HDRT + αPD-1, 5.04±5.011%, vs Ctrl 26.85±18.91%, p<0.05) and at the survival endpoint (mean metastasis area/lung area GSI + HDRT + αPD-1 5.04±5.011%, vs Ctrl 26.85±18.91%, p0.05), confirming an immune-dependent mechanism. Spectral flowcytometry revealed that HDRT alone increased regulatory T cells (Tregs), exhausted T cells, and M2-like suppressive macrophages, while depleting M1-immunostimulatory populations. Triple therapy reversed these effects: Tregs and exhausted T cells were reduced, while CD8⁺ effector T cells and M1-like macrophages were increased. Importantly, CD103⁺ dendritic cells (DC), critical for antigen cross-presentation, were restored, suggesting that that GSI reshapes the TME toward an immunostimulatory state.Conclusions:Notch inhibition reprograms the radiation-altered TME to enhance responsiveness to checkpoint blockade in this TNBC model. Triple therapy prolongs survival and reduces metastasis by restoring CD103⁺ dendritic cells, activating CD8⁺ T cells, and suppressing immunosuppressive cell populations. These data support the translational potential of integrating Notch inhibition into breast cancer immuno-radiotherapy regimens.
Presentation numberPS3-12-17
Baicalein Reverses CAFs Activation in Triple-Negative Breast Cancer by Inhibiting the PI3K/Akt/mTOR Pathway
Wei Lv, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
W. Lv, F. Wu, C. Du, S. Zhang; The Breast Disease Diagnosis and Treatment Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, CHINA.
Background and purpose: Triple-negative breast cancer (TNBC) is the most aggressive subtype with high metastatic potential and the poorest prognosis. Cancer-associated fibroblasts (CAFs) promote tumor progression and treatment resistance. Baicalein (BAI), the primary active constituent of the traditional Chinese herb Scutellaria baicalensis, exhibits broad-spectrum antitumor activity and multi-target effects. Studies on baicalein-mediated regulation of CAFs remain limited, so this study aims to analyze the correlation between tumor-stroma ratio (TSR), CAFs activation markers, clinicopathological features, and prognosis in TNBC; explore novel mechanisms and key molecules underlying CAFs activation; elucidate the role and molecular mechanism of baicalein in reversing CAFs activation, thereby providing a scientific foundation for developing novel TNBC combination therapies.Methods: Clinicopathological data from TNBC patients and the TCGA database were analyzed to evaluate the association of TSR and CAFs activation markers with clinicopathological characteristics and prognosis. An ex vivo CAFs activation model was established to investigate the effects of activated CAFs on 4T1 cell migration, pathway alterations during CAFs activation, and underlying mechanisms. The reversal effect of baicalein on CAFs activation and its molecular mechanism were systematically evaluated. Molecular docking and dynamics simulations were performed to assess the binding affinity and stability of baicalein with key pathway proteins. A murine orthotopic TNBC co-implantation model was established to validate the in vivo effects of baicalein on CAFs activation reversal and TME remodeling.Results: Stroma-rich tumors were significantly associated with high histological grade, rapid proliferation, and PIK3CA mutations in TNBC patients. Both CAFs activation and high PI3K expression correlated with poor prognosis. Stroma-rich TNBC tissues exhibited higher CAFs activation and elevated stromal PI3K expression. Conditioned medium (CM) from 4T1 cells induced a CAFs-like morphology in 3T3 cells, with significantly upregulated protein levels of CAFs activation markers (α-SMA, FAP, S100A4, and Vimentin), upregulated CAFs activation-related genes (eg.Acta2) and extracellular matrix-related genes (eg.Col1a1). KEGG enrichment analysis highlighted significant alterations in the PI3K-Akt pathway. Activated CAFs showed increased ratios of p-PI3K/total PI3K, p-Akt/total Akt, and p-mTOR/total mTOR. Activated CAFs markedly promoted 4T1 cell migration. Baicalein downregulated the CAFs activation and extracellular matrix-related genes. KEGG analysis revealed the most pronounced enrichment in the PI3K-Akt pathway. The binding energy of baicalein and p110α is −8.4 kcal/mol, forming a thermodynamically stable complex. In the orthotopic TNBC model, the baicalein plus nab-paclitaxel exhibited the smallest tumor volume/weight, fewest lung metastases, and prolonged survival. Baicalein significantly reduced α-SMA, Vimentin, and Collagen I expression, decreased collagen deposition, and attenuated TME fibrosis. Tumor tissues from the baicalein group showed reduced p-PI3K/total PI3K, p-Akt/total Akt, and p-mTOR/total mTOR ratios. Conclusion: In TNBC patients, CAFs activation, stroma-rich TME, and aberrant PI3K activation are interrelated and associated with poor prognosis. PI3K/Akt/mTOR pathway activation is a novel mechanism driving CAFs activation in TNBC. Baicalein reverses CAFs activation by inhibiting this pathway, suppresses TNBC cell migration, and synergizes with taxanes to enhance antitumor efficacy.
Presentation numberPS3-12-18
Exploratory Analysis of Immune Cell Subsets in Luminal B HER2-negative Breast Cancer and Their Association with Pathologic Response
Alexandr Petrovskiy, N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
M. Khoroshilov1, E. Kovalenko1, Z. Kadagidze2, T. Zabotina2, E. Evdokimova1, Y. Zhulikov1, A. Petrovskiy3, I. Vorotnikov3, M. Kiselevskiy4, E. Artamonova1; 1Department of Chemotherapy №1, N.N. Blokhin National Medical Research Center of Oncology, Moscow, RUSSIAN FEDERATION, 2Laboratory of Clinical Immunology and Innovative Technologies, N.N. Blokhin National Medical Research Center of Oncology, Moscow, RUSSIAN FEDERATION, 3Department of Breast Tumors, N.N. Blokhin National Medical Research Center of Oncology, Moscow, RUSSIAN FEDERATION, 4Laboratory of Cellular Immunity, N.N. Blokhin National Medical Research Center of Oncology, Moscow, RUSSIAN FEDERATION.
Background: Although tumor-infiltrating lymphocytes (TILs) are associated with better outcomes in triple-negative and HER2-positive breast cancer, their prognostic value in luminal HER2-negative subtype remains unclear. This study aimed to evaluate TIL subpopulations in luminal breast cancer and their association with pathologic complete response.Materials and methods: This prospective trial included 72 pts. with ER+ (>10%), HER2 negative breast cancer II-III stages. Both stromal and intratumoral subpopulations of TILs were assessed with flow cytometry on a tumor sample obtained by core-biopsy directly before the start of neoadjuvant chemotherapy (NACT). We analyzed CD3-CD8+, CD3+HLA- DR+, CD3-HLA-DR+, HLA-DR+, CD8+CD28+, CD8+CD28-, CD8+CD28+/CD8+CD28-, CD4+/CD8+, 11b+28-, 11b+28+, 11b- 28-, 11b-28+, CD8+CD4+, CD4+CD152+ minor subpopulations and other linear subpopulations. All the pts received NACT with 4 cycles of dose-dense AC every 2 weeks then 4 cycles of docetaxel every 3 weeks in a N.N. Blokhin National Medical Research from 2018 to 2023.Results: The mean value of TIL is 2.58% (median 0.9%; range 0-30.6%). Stage II was diagnosed in 7 patients (9.7%), and stage III in 65 patients (90.3%). G2 tumors were found in 58 patients (80.6%), G3 in 14 (19.4%). The mean age was 46 ± 9 years (range 25-70), with most patients being premenopausal (72.2%). No significant differences in TIL levels were observed across age groups (median 0.9% in patients ≤50 years vs. 0.8% in >50 years; p=0.450). Nevertheless, older patients showed a numerical increase in CD16+ and CD8+CD279+(PD-1+) cells, along with a decrease in CD4+CD25+ lymphocytes, consistent with previous reports. There was no significant difference in mean value of total TIL levels between patients with good (RCB 0-1) and poor (RCB 2-3) pathologic response (3.92% vs. 2.09%, p=0.137). Most analyzed subpopulations (including CD3+CD4+, CD3+CD8+, CD4+CD25highCD127-/low, and others) showed no significant association with treatment response. However, CD4+CD25+ (11.2% vs. 7.5%; p=0.013) and CD8+PD-1+ lymphocytes (9.5% vs. 16.3%; p=0.041) significantly differed between the RCB 0-1 and RCB 2-3 groups. Higher levels of CD4+CD25+ (more than median) lymphocytes were significantly associated with favorable response (RCB 0-1 – 57.1% vs. 8.3%, p=0.001), while lower levels of CD8+PD-1+ lymphocytes were also associated with a better outcome (RCB0-1 46.2% vs. 12.0%, p=0.007).Univariate regression analysis identified the following as predictors of better pathologic response: absence of lymph node involvement (N0), G3 tumor grade, age ≤50 years, progesterone receptor expression <20%, low CD8+PD-1+, and high CD4+CD25+ levels. However, none of these factors retained independent predictive value in multivariate analysis.Conclusion: These findings suggest a potential prognostic relevance of specific TIL subpopulations, particularly CD4+CD25+ and CD8+PD-1+ lymphocytes, in luminal breast cancer. Further studies with larger cohorts are needed to validate these observations.
Presentation numberPS3-12-19
Spatial Immune Profiling in Inflammatory Breast Cancer Reveals a Central Role for Tumor-Associated Macrophages and a Spatial Context-Dependent Negative Prognostic Impact of B Cells
Christophe Van Berckelaer, University of Antwerp, Antwerpen, Belgium
C. Van Berckelaer1, S. Van Laere1, C. Vermeulen1, M. Kockx2, Y. Waumans2, K. Mariën2, F. Berditchevski3, F. Bertucci4, P. Vermeulen5, L. Dirix6, C. Colpaert7, G. R. Devi8, P. Van Dam1; 1Center for Oncological Research (CORE), University of Antwerp, Antwerpen, BELGIUM, 2CellCarta, CellCarta, Antwerpen, BELGIUM, 3Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UNITED KINGDOM, 4Predictive Oncology team, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, Marseille, FRANCE, 5Translational Cancer Research Unit (TCRU), ZAS Sint-Augustinus, Antwerpen, BELGIUM, 6Department of Oncology, ZAS Sint-Augustinus, Antwerpen, BELGIUM, 7Department of Pathology, AZ Turnhout, Turnhout, BELGIUM, 8Department of Surgery, Division of Surgical Sciences, Duke University School of Medicine, Durham, NC.
Background: Inflammatory breast cancer (IBC) is a rare, aggressive subtype of breast cancer with unfavorable prognosis. Despite its distinct clinical presentation and molecular features, the immune landscape of IBC and its role in driving the aggressive phenotype remain poorly understood. This study aimed to characterize the spatial immune landscape of IBC using digital image analysis (DIA), compare it with subtype-matched non-inflammatory breast cancer (nIBC), and evaluate prognostic implications of immune cell composition and localization. Methods: We examined pretreatment tumor samples from 161 IBC and 115 subtype-matched nIBC patients using an AI-assisted DIA pipeline. Immunostainings (H-DAB) were conducted using validated protocols for CD8 (cytotoxic T cells), FOXP3 (Tregs), CD79α (activated B cells), PDL1 (SP124), and CD163 (tumor-associated macrophages, TAMs). Slides were digitized and assessed using VISIOPHARM® software, enabling virtual multiplexing. We quantified DAB+ immune cells and their locations using XY coordinates to calculate density. Spatial colocalization was analyzed using ecological statistics, based on point pattern or quadrant analysis. In point pattern analysis, we counted immune cells within a 30 µm radius around CD8+ cells or tumor cells. For quadrant analysis, we applied the Morisita-Horn Index (MHI) after square tessellation (250×250 µm tiles). The MHI compares cell composition between tile pairs, ranging from 0 to 1, with higher values indicating greater dissimilarity. Associations with clinicopathological features and outcome were assessed using multivariate regression models. Transcriptomic validation was performed using Affymetrix gene expression data and consensusTME deconvolution. Results: IBC showed higher CD163+ tumor-associated macrophage (TAM) infiltration than nIBC. Gene expression data confirmed the IHC findings, and pathway analysis linked high TAM density with inflammatory and proliferative pathways. The spatial distribution of immune cells was prognostically relevant, with high CD8+ T-cell (OR: 0.41, 95% CI: 0.22-0.76, P = 0.004) and low CD79α+ B-cell infiltration (OR: 3.19, 95% CI: 1.68-6.03, P < 0.001) correlating with improved overall survival in IBC. The ratio of CD8+ T cells to FOXP3+ regulatory T cells within tumor area was a significant prognostic indicator (OR: 0.34, 95% CI: 0.14-0.83, P = 0.018), whereas absolute densities of CD8+ or FOXP3+ cells alone did not associate with outcome. Beyond density alone, spatial colocalization of B cells with tumor cells emerged as a key prognostic factor. A high tumor colocalization index for CD79α+ B cells was associated with significantly worse survival (OR: 3.59, 95% CI: 1.31-9.84, P = 0.013). Similarly, greater spatial similarity between CD79α+ B cells and tumor cells, assessed via the MHI, was linked to poor outcome (OR: 2.61, 95% CI: 1.19-5.74, P = 0.017). Incorporating spatial B-cell features improved the prognostic model (P = 0.0005). Conclusions: This comprehensive spatial immune profiling effort in a large IBC cohort revealed the immunosuppressive nature of the IBC microenvironment and underscored the central role of TAMs in promoting its aggressive behavior. Spatial context, not immune cell abundance, was most predictive of outcome. High colocalization of CD79α+ B cells with tumor cells associated with poor survival, suggesting a tumor-promoting role. These findings demonstrate the value of spatial immunophenotyping beyond conventional quantification and support exploring TAM- and B cell-targeted therapies in IBC.
Presentation numberPS3-12-20
Activated PMN-MDSC phenotype associates with immune-related adverse events to immunotherapy in triple negative breast cancer
Janita Basit, Cleveland Clinic, Cleveland, OH
J. Basit1, P. Parthasarathy1, I. Gautam1, N. Stabellini2, P. Rayman1, M. Patel1, P. Pavicic1, J. Powers1, A. Moen1, B. Race1, E. Mundell1, A. Trevino1, T. Chan1, B. Moftakhar2, T. Mizukami2, C. Owusu3, T. Alban1, V. Makarov1, A. Montero3, M. Diaz-Montero1; 1Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, 2University Hospitals/Seidman Cancer Center, University Hospitals, Cleveland, OH, 3University Hospitals/Seidman Cancer Center, Univeristy Hospitals, Cleveland, OH.
Introduction/Background: Triple-negative breast cancer (TNBC) is an aggressive subtype, that comprises approximately 15-20% of all breast cancers. Currently, the standard for care in early-stage TNBC, as established the KEYNOTE-522 (KN-522) trial is combined neoadjuvant cytotoxic chemotherapy plus pembrolizumab. The trial demonstrated that the addition of pembrolizumab, significantly improved pathologic complete response rates and survival outcomes. However, pembrolizumab is also associated with immune-related adverse events (irAEs), leading to early treatment discontinuation. In the KN-522 trial, irAEs occurred in 34% of patients. Identifying predicative biomarkers may inform new strategies for intervention and management in patients likely to develop irAEs. Therefore, this study aimed to identify circulating immune determinants of irAEs to immunotherapy in TNBC. Methods: Peripheral blood was collected at multiple time points from 41 patients with TNBC undergoing neoadjuvant therapy with the KN-522 regimen. Blood was collectedat: baseline (BL), on cycle 3 day 1, and after surgery. Peripheral blood mononuclear cells (PBMCs) were isolated via density gradient centrifugation and profiled using high-dimensional flow cytometry to assess immune cell composition and phenotypes. For transcriptomic analysis, single-cell RNA sequencing was performed using the Parse Biosciences platform (pipeline split-pipe v1.2.0). The data was then analyzed with the Seurat pipeline to identify differentially expressed genes and investigate cell-state dynamics across samples. In parallel, plasma cytokine levels were quantified using the Olink platform to assess circulating biomarkers associated with irAE development. Results: Twelve of the 40 patients enrolled in the study (30%) experienced a documented irAE, including hypothyroidism (n = 4), hepatitis (n = 3), adrenal insufficiency (n = 2), colitis (n = 1), myocarditis (n = 1), and hemophagocytic lymphohistiocytosis (n = 1). At baseline (BL), flow cytometry revealed an accumulation of PMN-MDSCs with an activated/platelet-associated phenotype (CD16⁻ CD62p⁺ CD42b⁺; 42.1% vs. 1.24%) among irAR+ patients. In contrast irAE⁻ patients had significantly higher proportions of immature non-platelet-associated APC like traits PMN-MDSC (CD80+, CD86+, HLDR+; 90.4% vs. 40.7%). Pathway analysis showed irAE+ patients at BL have enriched interferon-alpha/gamma response, TNFα signaling via NF-κB, IL2–STAT5 signaling. On-treatment, irAE⁺ patients exhibited either sustained or amplified enrichment of these pathways. Proteomics analysis showed a correlation between irAE and the presence of the following pro-inflammatory cytokines: IL-10, CLEC7A, IL-1 alpha and CXCL1. Conclusion(s): Together these multi-omic analysis indicates that patients who develop irAEs may have a distinctive immune signature characterized by increased frequencies of activated PMN-MDSCs, and an inflammatory milieu conductive to overly activated CD4⁺ T cells, and B cells.
Presentation numberPS3-12-21
A Novel Spatial Profiling Approach Reveals Distinct Fibroblast and Immune Architectures in Invasive Pleomorphic versus Classic Lobular Carcinomas
Elizve N Barrientos-Toro, The University of Texas MD Anderson Cancer Center, Houston, TX
E. N. Barrientos-Toro1, R. Pandurengan1, H. Batra1, Q. Ding2, A. Contreras2, A. A. Sahin2, C. Haymaker1, M. G. Raso1; 1Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 2Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
A Novel Spatial Profiling Approach Reveals Distinct Fibroblast and Immune Architectures in Invasive Pleomorphic versus Classic Lobular CarcinomasElizve Barrientos-Toro1, Renganayaki Pandurengan 1 , Harsh Batra1, Qingqing Ding2 , Alejandro Contreras2, Cara Haymaker1, Aysegul Sahin2, Maria Gabriela Raso1.1Department of Translational Molecular Pathology, 2Department of Anatomical Pathology, UT MD Anderson Cancer Center, Houston, TX, USABackgroundInvasive Lobular carcinomas (ILC) comprise a histologically and biologically diverse group of breast cancers. While classic invasive lobular carcinomas (ICLCs) are typically characterized by cohesive, infiltrative growth and immune-excluded microenvironments, pleomorphic invasive lobular carcinomas (IPLCs) display more aggressive behavior. Based on our previous findings of differential CAF cell densities in five phenotypes between CLCs and PLCs, we aimed to investigate the spatial relationships and distribution patterns within these phenotypes, as the spatial organization of fibroblasts and immune cells in these histologic subtypes remains underexplored.MethodsWe performed high-dimensional multiplex immunofluorescence (mIF) spatial analysis on 12 cases of ILC (IPLCs: cases 7–12; ICLCs: cases 13–18). A comprehensive fibroblast-focused panel (including FAP, S100A4, Thy1, A-SMA) and immune markers (CD45) were used to characterize tumor (CK+), fibroblast, and immune compartments. Spatial clustering and neighborhood analysis were conducted using the Spatial Image Analysis of Tissues (SPIAT) R package v 4.5.1, to define the microenvironmental architecture.ResultsIPLCs demonstrated marked spatial heterogeneity, with fragmented CK+ tumor regions interspersed among stromal and immune elements. Fibroblast landscapes in IPLCs were notably diverse, featuring abundant hybrid subtypes (e.g., S100+Thy1+, A-SMA+FAP+) and frequent tumor-stroma integration within shared neighborhoods. These dynamic fibroblast populations supported active extracellular matrix (ECM) remodeling, potentially facilitating invasion and contributing to partial immune engagement. CD45+ immune cells were consistently more prominent in IPLCs, forming discrete immune-enriched clusters and mixed neighborhoods, suggesting an immune-intermediate or partially inflamed phenotype. In contrast, ICLCs displayed large, cohesive CK+ tumor regions with minimal fragmentation and low spatial heterogeneity. Fibroblast subtypes were simpler and predominantly peripherally distributed, and they rarely formed hybrid phenotypes. The stroma primarily acted as structural support, with limited ECM remodeling and minimal immune modulation. CD45+ cells were sparse and diffusely scattered, consistent with an immune-excluded (“cold”) microenvironment.ConclusionsWhile descriptive, our data reveal profound differences in the spatial microenvironments of IPLC and ICLCs. IPLCs exhibit a dynamic, heterogeneous stroma characterized by integrated fibroblast-tumor interactions and increased immune infiltration, aligning with their aggressive clinical behavior. In contrast, ICLCs maintain a cohesive tumor architecture with simplified, peripheral stroma and sparse immune presence. These insights highlight the significance of spatial context as an innovative and informative strategy in comprehending ILC biology and suggest that stromal and immune features may inform therapeutic strategies, including potential vulnerability to stroma- or immune-targeted interventions in IPLCs
Presentation numberPS3-12-22
Tumor cell AADAT suppresses CD8⁺T cell function by depleting malate, driving progression in Triple-Negative Breast Cancer
Megha Chatterjee, Baylor College of Medicine, Houston, TX
M. Chatterjee1, F. Gu1, L. Yu2, D. Varghese2, J. Park3, B. Piyarathna1, S. Thota1, W. Zhang2, Y. Gao2, U. Rasaily1, Y. Qiu4, V. Putluri5, B. Karanam6, N. Chandandeep2, B. Simons7, J. Asirvatham8, A. Rao8, C. Morrison9, G. Das10, C. Ambrosone11, E. Seeley12, B. Kaipparettu2, I. Kurland4, N. Putluri5, Q. Zhu2, X. Zhang2, A. Sreekumar5; 1Molecular and cellular Biology, Baylor College of Medicine, Houston, TX, 2Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 3Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 4Stable isotope & metabolomics core facility, Albert Einstein College of Medicine, Bronx, NY, 5Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 6Department of Biology and Cancer Research, Tuskegee University, Tuskegee, AL, 7Center for Comparative Medicine, Baylor College of Medicine, Houston, TX, 8Division of Anatomic Pathology, Baylor Scott & White Health, Temple, TX, 9Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center,, Buffalo, NY, 10Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center,, Buffalo, NY, 11Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center,, Buffalo, NY, 12Department of Chemistry, College of Natural Sciences, The University of Texas at Austin, Austin, TX.
Triple-negative breast cancer (TNBC) remains refractory to most targeted therapies and is characterized by a profound immunosuppressive tumor microenvironment. Leveraging integrated transcriptomic, metabolomic, immunohistochemical, and clinical datasets, we identify the mitochondrial kynurenine-pathway enzyme α-aminoadipate aminotransferase (AADAT) as a previously unrecognized metabolic immune checkpoint in TNBC. AADAT transcripts and protein were significantly up-regulated in human TNBC specimens and showed a strong inverse correlation with intratumoral CD8⁺ T-cell abundance and patient survival. shRNA-mediated silencing of AADAT in orthotopic murine TNBC models curtailed primary tumor growth and distant metastasis in a CD8⁺ T-cell-dependent fashion, augmented effector T-cell activation, and rendered tumors exquisitely sensitive to combined PD-1/CTLA-4 blockade. Mechanistically, AADAT loss decreased mitochondrial CoQ₁₀ pools, triggering a compensatory rise in secreted malate that directly boosted CD8⁺ T-cell oxidative metabolism, reactive oxygen species generation, and production of TNF-α and IFN-γ via an increased NAD⁺/NADH ratio. Exogenous malate recapitulated the antitumor effects of AADAT knockdown and synergized with paclitaxel plus anti-PD-1 therapy to suppress tumor burden and prolong overall survival. Consistent with these preclinical findings, higher intratumoral malate levels in TNBC patient cohorts associated with enhanced functional CD8⁺ T-cell infiltration, reduced T-cell exhaustion signatures, and superior post-chemotherapy outcomes. Collectively, these data position AADAT as a critical metabolic orchestrator of immune escape in TNBC and nominate malate supplementation as a readily translatable adjuvant strategy to amplify the efficacy of existing chemo-immunotherapy regimens.
Presentation numberPS3-12-24
Immune Landscape in Tumor Draining Lymph Nodes after Neoadjuvant Chemoimmunotherapy in Early Triple Negative Breast Cancer
Anton Oseledchyk, University Hospital Basel, Basel, Switzerland
A. Oseledchyk1, P. Caloba2, S. Uzun3, K. Schaeuble4, S. Münst3, W. P. Weber1, C. Kurzeder5, M. Vetter6, H. Läubli2, M. Binder1, A. Zippelius4; 1Medical Oncology, University Hospital Basel, Basel, SWITZERLAND, 2Laboratory of Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel, Basel, SWITZERLAND, 3Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, SWITZERLAND, 4Laboratory of Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, SWITZERLAND, 5Department of Gynecology and Gynecologic Oncology, University Hospital Basel, Basel, SWITZERLAND, 6Department of Hematology and Oncology, Cantonal Hospital Basel-Land, Liestal, SWITZERLAND.
Background: The integration of immune checkpoint inhibitors (ICIs) with neoadjuvant chemotherapy has significantly improved clinical outcomes for patients with early triple-negative breast cancer (TNBC). However, approximately one-third of patients still fail to achieve a major pathological response, facing a substantially increased risk of recurrence and death. While immune correlates within the tumor microenvironment and peripheral blood have been extensively studied, the immunological dynamics within tumor-draining lymph nodes (TDLNs) – key sites for the initiation and coordination of anti-tumor immunity – remain poorly understood. Here we present our ongoing investigation into the immune architecture of TDLNs following chemoimmunotherapy to uncover predictive signatures of response and mechanisms of resistance. Methods: Sentinel lymph nodes from early TNBC patients undergoing neoadjuvant chemoimmunotherapy are analyzed using the PhenoCycler multiplex imaging platform, deploying a comprehensive 50-marker panel. This approach enables deep spatial and phenotypic profiling of adaptive immune response, including T-cell priming pathways. We quantify immune cell density, diversity, and spatial organization, with a focus on cellular neighborhood analysis. In addition, we employ an innovative tracking method to identify anti-PD1-bound cells, providing direct insights into the pharmacodynamic effects of PD-1 blockade within TLDNs. Results: To date, we have analyzed TDLNs from four stage II TNBC patients – two with complete pathologic response (pCR, residual cancer burden (RCB)=0) and two with residual disease (non-pCR, RCB=1). Preliminary results reveal a trend toward higher frequency of CD8⁺ (2.7% vs. 1.4% of CD45⁺ cells) and CD4⁺ T cells (19.9% vs. 8.5% of CD45⁺ cells) within the paracortical regions of pCR patients. Furthermore, the composition of CD8⁺ T cells differed, with increased proportions of precursor-exhausted (Tpex) (16.0% vs. 2.7% of CD8⁺ cells), and terminally exhausted (Tex) (10.2% vs. 1.1% of CD8⁺ cells) subsets observed in pCR cases. Ongoing analyses include mapping of key immune populations, such as plasmacytoid dendritic cells (pDC), conventional dendritic cells (cDC1 and cDC2), Th1, Th2, and T follicular helper (Tfh) cells, allowing us to compute cell-to-cell distances and generate individualized neighborhood profiles to identify spatial signatures associated with treatment response. Conclusion: Our high dimensionality spatially resolved analysis provides a powerful window into the immunological landscape of TDLNs in TNBC. By preserving tissue architecture and capturing cellular interactions, we aim to unravel how TDLNs contribute to effective anti-tumor immunity or harbor resistance in the context of chemoimmunotherapy. Preliminary findings suggest distinct immune patterns in patients achieving pCR vs. those with residual disease. A larger cohort, including comprehensive spatial and activation marker analysis, will be presented at the conference.
Presentation numberPS3-12-25
First Preclinical Evaluation of Inavolisib-Based Combinatorial Strategies in PIK3CA-Mutant Breast Cancer Brain Metastasis Models
Zhao Jianli, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
Z. Jianli1, B. Gao2, L. Ding2, D. Bi2, T. Chen2, Z. Wu2, D. Huang2, Y. Wang2; 1Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA, 2Sun Yat-sen Memorial Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, CHINA.
First Preclinical Evaluation of Inavolisib-Based Combinatorial Strategies in PIK3CA-Mutant Breast Cancer Brain Metastasis Models Jianli Zhao1,2#, Boying Gao1,2#, Linxiaoxiao Ding1,2#, Dexi Bi1,2#, Tao Chen1,2#, Zhenghao Wu1,2,3*, Di Huang1,2*, Ying Wang1,2*.1Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; 2Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; 3Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; # These authors contributed equally and should be considered co-first authors. Introduction:Brain metastases are a major cause of mortality in breast cancer, particularly in HER2-positive and triple-negative breast cancer (TNBC) subtypes. Due to the restrictive nature of the blood–brain barrier (BBB), conventional anti-tumor agents often show limited efficacy in the central nervous system. Moreover, PIK3CA mutations, common in breast cancer, activate the PI3K–AKT–mTOR pathway, promoting tumor proliferation, invasion, and therapeutic resistance. However, how to effectively treat PIK3CA-mutant breast cancer brain metastases (BCBM) remains a critical clinical challenge. Here, we report the first preclinical evaluation of Inavolisib, a selective PI3Kα inhibitor, in intracranial animal models and patient-derived xenografts (PDX) of BCBM harboring PIK3CA mutations across HER2-positive and TNBC subtypes. Methods:We developed the world’s first intracranial BCBM mouse models and PDX models with confirmed PIK3CA mutations, using direct intracranial injection of tumor cells derived from HER2+ and TNBC subtypes. These models were used to assess the anti-tumor efficacy of Inavolisib monotherapy and its combinations with clinically relevant agents. For TNBC models, combinations included PD-1 antibody and albumin-bound paclitaxel. For HER2-positive models, Inavolisib was combined with Trastuzumab + Pertuzumab, T-DXd (Trastuzumab deruxtecan), or Tucatinib. Tumor size and survival were measured, and all findings were validated in corresponding PIK3CA-mutant PDX models. Dose-response and toxicity profiles were also evaluated. Results:In TNBC brain metastasis models, Inavolisib monotherapy had limited efficacy. However, the combination of Inavolisib with anti–PD-1 antibody led to significant tumor volume reduction and prolonged survival, demonstrating synergistic intracranial activity. In HER2-positive BCBM models, Inavolisib + Trastuzumab + Pertuzumab and Inavolisib + T-DXd both resulted in marked suppression of brain tumor growth and significant extension of survival. Conversely, combination with HER2-TKI Tucatinib showed minimal additional benefit. These results were consistently reproduced in PIK3CA-mutant PDX models. Notably, T-DXd-based regimens demonstrated a clear dose–response relationship and maintained a favorable safety profile. Conclusion:This study provides the first preclinical evidence of Inavolisib-based combination strategies in PIK3CA-mutant breast cancer brain metastases, using both novel intracranial animal models and PDX systems. Our results suggest that: Inavolisib combined with PD-1 antibody is a promising regimen for TNBC with brain metastases; Inavolisib + T-DXd or Trastuzumab + Pertuzumab shows potent activity in HER2-positive BCBM. These findings highlight the potential of PI3K-targeted combinatorial strategies for overcoming the BBB challenge in breast cancer brain metastases and provide a strong rationale for clinical translation.
Presentation numberPS3-12-26
Interplay Between Therapy-Induced Senescence and Macrophage Polarization in the Tumor Microenvironment of Triple-Negative Breast Cancer
Homood Moqbel As Sobeai, King Saud University, Riyadh, Saudi Arabia
H. M. As Sobeai, S. Almufadhili, S. Alhudaithi, M. Alotaibi; College of Pharmacy, King Saud University, Riyadh, SAUDI ARABIA.
Tumor microenvironment (TME) plays a critical role in cancer progression, with tumor-associated macrophages (TAMs) acting as key modulators of immune suppression and metastasis. Emerging evidence suggests that cellular senescence may influence the function of TAMs; however, the underlying mechanisms remain poorly understood. In this study, we investigated the interplay between therapy-induced senescence (TIS) and TAMs, focusing on the impact of senescent cancer cells on macrophage phenotype and the therapeutic modulation of TAMs using Pexidartinib (PLX3397), an inhibitor of M2-like polarization. Using the 4T1 triple-negative breast cancer cell line and RAW264.7, the established macrophage cell line, we found that conditioned media from senescent cancer cells did not induce macrophage senescence. In contrast, direct exposure of RAW cells to doxorubicin significantly induced senescence and promoted an M2-like, pro-tumorigenic phenotype characterized by elevated CD206 expression. At the molecular level, senescent macrophages showed increased expression of p53 and p21, confirming senescence induction. Moreover, a reduction in IL-6 mRNA levels supported the shift toward M2 polarization. Notably, pretreatment with PLX3397 led to a marked reduction in macrophage numbers in senescent cultures, suggesting impaired TAM survival. These findings reveal a role for senescence in promoting M2-like macrophage polarization and highlight the potential of PLX3397 as a therapeutic strategy to target senescent TAMs and reshape the immune landscape within the TME.
Presentation numberPS3-12-27
Comparative analysis of the tumor immune microenvironment (TIME) and primary and metastatic tissue in HR+/HER2- and triple-negative breast cancer (TNBC)
Alexis LeVee, UCLA David Geffen School of Medicine, Los Angeles, CA
A. LeVee1, J. Bitar2, E. B. Jaeger3, U. Jariwala3, J. Mercer3, C. Egelston4, H. McArthur5, Y. Yuan2, I. Kang6; 1Department of Medicine, Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA, 2Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 3Medical Affairs, Tempus AI, Inc, Chicago, IL, 4Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, 5Department of Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX, 6Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA.
Background: Immune checkpoint inhibitor (ICI) based regimens are standard of care for patients with TNBC and clinical trials are investigating ICI in HR+/HER2- breast cancer, with encouraging results in a subset of patients. The tumor immune microenvironment (TIME) plays a critical role in ICI response and can differ significantly by metastatic site. Given the expanding integration of ICIs in HR+/HER2- breast cancer trials, we examined the TIME across sites of disease and subtypes to illustrate how these anatomical sites are molecularly different and may impact ICI benefit. Methods: We used Tempus Lens (Tempus AI, Inc., Chicago, IL) to retrospectively analyze de-identified next-generation sequencing data from patients with HR+/HER2- breast cancer (n=6,818) and TNBC (n=2,715) in the Tempus Database. Metaplastic histology was excluded. Among HR+/HER2- breast cancer and TNBC, tumors from primary breast (PB, n=4451), liver (n=1117), bone (n=845), lymph node (LN, n=425), lung (n=388), and brain/CNS (n=234) metastatic sites were sequenced with Tempus xT DNA (648-gene panel) and xR RNA assays. TMB, MSI, PD-L1 (CPS, clone 22c3), and proportions of B, T (CD4+, CD8+), NK cells, and macrophages of immune cells via quanTIseq deconvolution were compared across metastatic sites in HR+/HER2- breast cancer and compared to TNBC. Chi-squared/Fisher’s exact or Kruskal-Wallis tests were used to assess statistical significance. Results: In this study, the median age at diagnosis in the HR+/HER2- and TNBC cohort was 58 and 57, respectively and both cohorts were diverse: White (50%/44%), African American (8%/14%), Hispanic (6.7%/7%), Asian (3.1%/2.3%), other race (5.5%/5.5%) and unknown (33%/34%). Overall, 67% of patients were stage IV. Among patients with HR+/HER2- breast cancer, brain/CNS (14%) was most commonly TMB high (≥10 mut/mB) followed by bone (9.4%) and liver (8.9%) (p=10% (positive) proportions were highest in PB (4.1%), lung (3.6%) and LN (4.9%) (p<0.001). LN had the highest proportions of B cells and T cells (CD4+ and CD8+) and the lowest proportion of macrophages (M1 and M2) and NK cells (p<0.001). Besides LN, CD8+ T cells were in higher proportions in lung and PB compared to liver, bone, and brain/CNS metastases (p<0.001). In TNBC, PB, lung, and LN metastases had the highest proportions of PD-L1 positive status as well as CD8+ T cell proportions compared to liver, bone, and brain/CNS (both, p<0.001). Compared to TNBC, HR+/HER2- breast cancer had different proportions for all immune cell populations in PB and all non-breast sites (exception NK cells, all else p<0.05). Notably, HR+/HER2- breast cancer had lower proportions of CD8+ T cells and M1 macrophages and higher proportions of M2 macrophages across PB and all non-breast sites compared to TNBC (all, p<0.001). Liver metastases had the highest proportions of M1 macrophages, NK cells, and neutrophils compared to all other sites in HR+/HER2- breast cancer, with a similar pattern seen in TNBC (all, p<0.001). Conclusions: In HR+/HER2- and TNBC, the TIME significantly differed according to site of disease. Although more commonly TMB high, liver, bone, and brain/CNS metastases were composed of lower proportions of CD8+ T cells in HR+/HER2- breast cancer. HR+/HER2- breast cancer was less immunogenic across all disease sites compared to TNBC. However, patients with breast, lung, and LN metastases had the highest proportions of CD8+ T cells in both subtypes, indicating a subset of patients with HR+/HER2- breast cancer that could be enriched for ICI response. These analyses may support TIME biomarkers for patient selection in ICI focused clinical trials for patients with breast cancer.
Presentation numberPS3-12-28
Hydrogel-mediated local anti-CTLA-4 delivery potentiates immune activation after breast tumor cryoablation
Flávia Sardela de Miranda, TTUHSC, Lubbock, TX
F. Sardela de Miranda1, Y. Dauletov2, R. L. Babcock3, M. F. Mahecha1, P. J. Gukhool1, S. P. Singh4, H. S. Gill5, R. Layeequr Rahman1, M. W. Melkus1; 1Department of Surgery and Breast Center of Excellence, TTUHSC, Lubbock, TX, 2Department of Chemical Engineering, TTU, Lubbock, TX, 3Department of Cell Biology and Biochemistry and Breast Center of Excellence, TTUHSC, Lubbock, TX, 4Department of Internal Medicine and Breast Center of Excellence, TTUHSC, Lubbock, TX, 5Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC.
Background: Triple-negative breast cancer (TNBC) is highly aggressive with few treatment options and a significant risk of recurrence and metastasis owing to immune suppressive signals. Cryoablation offers dual advantage of local tumor necrosis while simultaneously stimulating anti-tumor immune responses via releasing tumor-associated antigens offering systemic control. Combining cryoablation with immune checkpoint inhibitors (ICIs) fosters stronger anti-tumor immunity, including abscopal effects. Nevertheless, systemic administration of ICIs like anti-CTLA-4 lead to widespread immune activation and immune-related adverse events (irAEs). To enhance the efficacy of immunotherapy while reducing systemic toxicity, we developed an injectable crosslinked chitosan-alginate hydrogel for localized and sustained delivery of anti-CTLA-4 – vinylated Poly(vinyl alcohol) conjugate in the tumor bed. Methods: Pharmacokinetic studies were conducted in naïve BALB/c mice to compare local (mammary fat pad, MFP; injection) and systemic (intraperitoneal; IP) administration of an anti-CTLA-4 hydrogel formulation (200 µg for direct comparison). Mice were bled on days 1, 4, and 8, and were sacrificed on day 14 for necropsy, during which plasma and mammary fat pad tissues were analyzed for anti-CTLA-4 concentrations using ELISA. The peripheral blood was further examined for T cell (CD4⁺, CD8⁺) activation by ICOS expression. Therapeutic efficacy was evaluated in our bilateral 4T1 tumor-bearing cryoablation mouse model; the left tumor was cryoablated and mice were treated with 200 µg of anti-CTLA-4 either systemically (IP) or locally via the hydrogel. Tumor size, hydrogel retention, T cell activation, and systemic anti-CTLA-4 levels were assessed post-treatment on days 7 and 14. Results: Non-tumor bearing mice treated with gel-formulated anti-CTLA-4 through local injection into the MFP exhibited significantly lower circulating antibody levels compared to those receiving systemic (IP) administration, as measured by ELISA (p < 0.05). At day 14, locally treated mice showed an approximately 10-fold increase in anti-CTLA-4 concentration within the mammary fat pad tissue and greater tissue mass relative to systemically treated controls (p < 0.05). Flow cytometry analysis revealed elevated ICOS expression on CD4⁺ and CD8⁺ T cells in the blood at day 8 in both groups, indicating intact immunostimulatory activity of the antibody; by day 14, no significant differences in ICOS expression were observed. In a bilateral 4T1 breast cancer cryoablation mouse model, local administration of anti-CTLA-4 into the cryoablated tumor site resulted in significantly higher antibody retention at the tumor and lower systemic exposure compared to IP delivery. At day 7, locally treated tumors were larger and contained higher anti-CTLA-4 concentrations reflecting gel retention, while by day 14, tumors in both groups were nearly fully reabsorbed. Peripheral blood analysis confirmed lower plasma levels of anti-CTLA-4 in the locally treated group. Flow cytometry showed comparable ICOS expression on T cells from the PB across both groups at days 7 and 14, indicating equivalent T cell activation. Conclusions: Local delivery of anti-CTLA-4-vinylated PVA conjugates using a chitosan-alginate hydrogel after cryoablation enhanced retention of anti-CTLA-4 in the tumor and provided equivalent activation of T cells in the blood, reducing systemic antibody exposure. This strategy presents a promising approach to minimize irAEs without compromising therapeutic efficacy. Moreover, this hydrogel-based method requires additional validation with lower doses of anti-CTLA-4 to further de-escalate treatment-associated toxicity. It supports its future application in clinical settings to enhance combinatorial cryoablation and ICI therapy.
Presentation numberPS3-12-29
Trop2 remodeling of the immune microenvironment in triple negative breast cancer
Bogang Wu, Massachusetts General Hospital, Harvard Medical School, Boston, MA
B. Wu1, W. Thant1, E. Bitman1, T. Liu1, J. Liu2, E. Paschalis2, B. Patel1, C. Nawrocki1, K. Xu1, L. Nieman1, D. Ting1, N. Thimmiah1, S. Sun1, R. Abelman1, V. Bossuyt1, S. Isakoff1, L. Spring1, A. Bardia1, L. Ellisen1; 1Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 2Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA.
Immune exclusion inhibits anti-tumor immunity and response to immunotherapy, but its mechanisms remain poorly defined. In triple-negative breast cancer (TNBC), an aggressive and generally immune-rich subtype, an immune-cold microenvironment predicts poor prognosis due to a limited response to chemotherapy and immune checkpoint inhibitors. To identify mechanisms regulating immune infiltration in TNBC, we performed spatial transcriptomic analysis comparing immune-enriched versus immune-cold tumors. We reveal that Trophoblast Cell-Surface Antigen 2 (TROP2), a key target of anti-cancer Antibody Drug Conjugates (ADCs), controls barrier-mediated immune exclusion in TNBC through Claudin 7 association and tight junction regulation. TROP2 expression is inversely correlated with T cell infiltration and predicts poor outcomes in TNBC. Loss-of-function and reconstitution experiments demonstrate TROP2 is sufficient to drive tumor progression in vivo in a CD8 T cell-dependent manner, while its loss deregulates expression and localization of multiple tight junction proteins, enabling T cell infiltration. Employing a humanized TROP2 syngeneic TNBC model, we show that TROP2 targeting via hRS7, the antibody component of the ADC Sacituzumab govitecan (SG), enhances the anti-PD1 response and improves T cell accessibility and effector function. Correspondingly, TROP2 expression is highly associated with lack of response to anti-PD1 therapy in human breast cancer. Thus, TROP2 controls an immune exclusion program that can be targeted to enhance immunotherapy response.
Presentation numberPS3-12-30
Targeting dendritic cells with CD40 agonist-based triplet immunotherapy induces IL12B-driven tumor control in triple-negative breast cancers
Sangeetha M Reddy, University of Texas Southwestern Medical Center, Dallas, TX
P. L. Mylabathula, R. Ali, A. Alkhani, X. Qi, S. Hebbale, J. Zhu, I. Chan, S. M. Reddy; Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX.
Background: Although immunotherapies deliver lasting remissions in many solid cancers, triple-negative breast cancers demonstrate limited immunity. One potential reason for this is functional impairment within the antigen-presenting cell (APC) compartment. Cross-presenting conventional dendritic cells (cDC1s) are essential for initiating effective CD8+ T cell responses but are frequently scarce or suppressed in breast tumors. We evaluated a rationally designed triplet regimen combining immunogenic anthracycline chemotherapy, Flt3 ligand to expand dendritic cell populations, and a CD40 agonist to activate them, aiming to reprogram the tumor microenvironment and promote durable anti-tumor T cell responses. Methods: EO771, AT3, or 4T1 tumors were implanted orthotopically in female mice. Once tumors reached ~50 mm³, mice received pegylated liposomal doxorubicin (Doxil, day 1), recombinant Flt3 ligand (Flt3L, days 1-5), and a CD40 agonist antibody (CD40a, days 11, 14, 17) or monotherapy or doublet combinations. Tumor burden and survival were monitored longitudinally. Tumors and draining lymph nodes were analyzed using flow cytometry, single-cell RNA sequencing (scRNA-seq), and TCR sequencing (TCR-seq). Mechanistic dependencies were assessed through genetic knockout models, antibody-mediated depletion, cytokine or chemokine blockade, and lymphocyte trafficking inhibition. Results: Triplet treatment significantly delayed tumor progression and prolonged survival compared to monotherapy or doublet combinations. To elucidate the underlying immune mechanisms, flow cytometry and scRNA-seq revealed robust activation across all three major APC lineages: dendritic cells (DCs), macrophages, and B cells. Specifically, Flt3L expanded cDC1s, cDC2s, and mature dendritic cells enriched in immunoregulatory features (mregDCs), while CD40a upregulated CD80, CD86, β2-microglobulin, and Il12b. Inflammatory macrophages expressing high levels of Cxcl9 were enriched, and B cells exhibited increased markers of activation and memory differentiation. Consistent with APC activation, triplet therapy increased intra-tumoral CD8+ T cell infiltration, reduced expression of exhaustion markers (LAG-3, TIM-3), and promoted stem-like features. TCR-seq demonstrated clonal expansion of T cells without changes in Shannon diversity, indicating predominant expansion of pre-existing T cell clones. Mechanistic studies demonstrated that cDC1s are essential for therapeutic efficacy, as tumor control was lost in Batf3-/- mice but remained intact following depletion of CSF1R+ macrophages or CD20+ B cells. CD8+ T cell depletion partially reduced efficacy, while co-depletion of CD8+ and CD4+ T cells resulted in complete loss of tumor control, indicating a cooperative role for both subsets. Removal of regulatory T cells further enhanced treatment response, suggesting they act as a barrier to optimal efficacy. Importantly, therapy remained effective despite FTY720-mediated inhibition of lymphocyte trafficking from the tumor draining lymph node, pointing to a role for pre-existing intra-tumoral T cells. Finally, neutralization of IL-12 completely abolished tumor control, whereas CXCL9 or CXCR3 blockade had little impact, underscoring the central role of IL-12 in mediating therapeutic response. Conclusion: This CD40 agonist-based triplet immunotherapy reprograms the tumor microenvironment through coordinated APC activation and IL-12-driven CD4+ and CD8+ T cell responses. cDC1s are essential mediators of therapeutic efficacy, while macrophages and B cells adopt inflammatory and activated states but are dispensable for treatment effect. Tregs constrain treatment potency and are a rational co-target to improve responses in breast cancer.
Presentation numberPS3-13-01
Cik cell therapy remodels the immune microenvironment and suppresses cutaneous metastatic breast cancer in preclinical models
Chi-Wen Luo, Kaohsiung Medical University Hospital, Kaohsiung city, Taiwan
M. Hou1, S. Chang2, F. Chen1, M. Pan2, 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: Cutaneous metastases in breast cancer (CMBC) are clinically challenging and associated with poor prognosis and limited therapeutic options. Conventional treatments offer minimal survival benefit and often fail to control skin lesions. Emerging data suggest that immunotherapy, particularly adoptive cell therapy, may offer therapeutic promise. This study evaluates the feasibility and immunologic impact of cytokine-induced killer (CIK) cells in preclinical models of CMBC. Methods: We established in vitro and in vivo models using murine breast cancer cells with cutaneous metastatic potential. CIK cells were generated from peripheral blood mononuclear cells of murine splenocytes. Cytotoxicity assays, flow cytometry, transcriptomic profiling, and immunohistochemistry were used to assess anti-tumor effects and immune-related changes. The impact of CIK therapy alone or in combination with a TLR7 agonist was also examined. Results: Transcriptomic analysis of cutaneous lesions revealed upregulation of immune effector molecules (CXCL9/10/11, B2M, CD27) and checkpoint pathways (PD-L1). CMBC cells showed heightened sensitivity to CIK-mediated cytotoxicity compared to non-cutaneous metastatic cells. In vivo, CIK therapy significantly reduced cutaneous tumor burden and prolonged survival. Co-administration of a TLR7 agonist enhanced tumor cell apoptosis, reduced PD-L1 expression, and increased T-cell infiltration, suggesting a synergistic immunomodulatory effect.Conclusions: CIK cell therapy demonstrates strong preclinical efficacy against breast cancer cutaneous metastases and reshapes the tumor immune microenvironment. These findings support further development of CIK-based immunotherapy, potentially in combination with innate immune modulators, for patients with CMBC—a clinical subgroup with high unmet need.
Presentation numberPS3-13-02
Tumor Microenvironment in Asian-American and Native Hawaiian/Pacific Islander Women With Triple Negative Breast Cancer
Edward Tri Nguyen, University of Hawaii Internal Medicine Residency Program, HONOLULU, HI
E. T. Nguyen1, S. Cheng2, M. William3, C. Chang4, B. Nguyen5, V. Khadka5, Y. Deng5, L. W. Loo3, X. Wang3, J. A. Fukui3; 1Internal Medicine, University of Hawaii Internal Medicine Residency Program, HONOLULU, HI, 2General Surgery, University of Hawai’i Surgical Residency Program, HONOLULU, HI, 3Cancer Biology, University of Hawaii Cancer Center, HONOLULU, HI, 4Pathology, University of California Davis, Sacramento, CA, 5Genomics and Bioinformatics Shared Resource, University of Hawaii Cancer Center, HONOLULU, HI.
Background: Triple negative breast cancer (TNBC) accounts for approximately 10-20% of breast cancer cases and has the worst overall survival and prognosis compared to other breast cancer subtypes. A higher percent of stromal tumor infiltrating lymphocytes (sTILs) in the tumor microenvironment (TME) of TNBC correlates with an improved prognosis. Also, high levels of TILs is associated with greater likelihood of pathologic complete response (pCR) in response to neoadjuvant treatment (NAT). Previous studies showed that all breast tumors from Asian Americans (AA) and Native Hawaiians and Pacific Islanders (NHPI) patients have a higher percent of sTILs compared to tumors from White patients. Although AAs have a lower cancer death ratio at 0.58, NHPIs have a higher cancer death rate ratio at 1.21 compared to White patients. Our study aims to molecularly characterize the tumor microenvironment in the AA and NHPI populations of patients with TNBC who received NAT. Methods: A retrospective chart review was conducted on 12 AA and 12 NHPI patient cases with early stage TNBC from 2015 to 2022 from a medical oncology clinic within the Hawai’i Pacific Health system. Breast cancer patient data collected included: age at diagnosis, histology, clinical stage, body mass index (BMI), sTILs, pCR, residual cancer burden, and NAT. Pre-treatment core biopsy samples were processed and gene expression profiles analyzed using the NanoString nCounter® Breast Cancer 360™ Panel and the nSolver Advanced Analysis Module. Differentially expressed genes (DEGs) and pathway analyses were generated using the ROSALIND® bioinformatics software. Results: Both AAs (66.7%) and NHPIs (58.3%) had a majority of the basal immunosuppressed (BLIS) TNBC subtype. When we compared TNBC tumors from AA and NHPI patients, 29 DEGs were statistically significant (p<0.05, adjusting for age) and the immune cell profiles revealed that NHPI tumors were observed to have fewer monocytes compared to AA tumors (p<0.05). When we compared the AA responders and nonresponders to NAT, 68 DEGs were statistically significant and the pathways involved in interleukin-2 signaling and B-cell activation were identified as significant (p<0.05, adjusting for age). In the immune cell profile analysis, AA responders expressed more CD8+ T-cells, memory B cells, and resting dendritic cells compared to AA nonresponders (p<0.05). When the NHPI responders and nonresponders to NAT were compared, 105 DEGs were statistically significant and the pathways associated with allograft rejection, PD-1 signaling, and antigen processing and presentation were identified as significant (p<0.05, adjusting for age). The immune profile analysis revealed no differences. When all responders and nonresponders to NAT were compared, 140 DEGs were statistically significant, and the pathways associated with allograft rejection and PD-1 signaling were identified as significant (p<0.05, adjusting for age). The immune profile analysis revealed no differences. Conclusions: In this pilot study, we characterized the gene expression profiles of tumors from AA and NHPI patients with TNBC. Among the two races, there were differences with DEGs but not in pathways. Meanwhile, AA and NHPI responders and nonresponders to NAT had differences in DEGs and biological pathways among the two population groups, suggesting that there may be differences in the biological response to NAT among AA and NHPI. In addition, all responders to NAT had pathways associated with differences in immune response compared to all nonresponders, suggesting that the immune response plays a role in the TME and in different biological mechanisms for the patient’s response to NAT. Future analysis will involve a larger cohort of samples with additional analysis of the TME. We hope to integrate our findings to guide NAT for multiethnic patients.
Presentation numberPS3-13-03
Neuron Projection Guidance signature associates with tumor progression and changes in the tumor microenvironment in triple-negative breast cancer
Kei Kawashima, Roswellpark Comprehensive Cancer Center, Buffalo, NY
K. Kawashima1, K. Chida1, T. AlRommah1, M. Oshi2, A. Yamada2, I. Endo2, K. Takabe1; 1Surgical Oncology, Roswellpark Comprehensive Cancer Center, Buffalo, NY, 2Gastroenterological surgery, Yokohama City University, Yokohama, JAPAN.
Background Tumor-associated neurogenesis has emerged as poor prognostic indicators in various solid tumors. Recent studies have suggested that neurogenesis related molecules, particularly those involved in the neuron projection guidance pathway, may promote tumor growth, metastasis, and change the tumor microenvironment of triple-negative breast cancer (TNBC). However, their clinical relevance remains to be fully understood. We hypothesized that the “neuron projection guidance score” may be associated with prognosis of TNBC patients. Methods Transcriptomic and clinical data were obtained from the TCGA-BRCA (n = 1084) cohort, and METABRIC (n = 1980) as a validation cohort. For treatment response analysis, GSE194040 was utilized. Gene sets corresponding to the GO term neuron projection guidance were extracted from MSigDB and used to calculate a GSVA-based Neuron Projection Guidance (NPG) score. Patients were grouped into neuron projection guidance score -high and -low groups by the median score. Associations with clinicopathologic variables, prognosis, immune landscape, and treatment response were evaluated. Results Using the xCell algorism, neuron projection guidance (NPG) score was associated with high neuron cell infiltration in TCGA. Gene set enrichment analysis (GSEA) revealed that NPG score-high TNBC was associated with pathways related to cell proliferation and migration, such as epithelial mesenchymal transition, angiogenesis, and TGF-beta signaling. Immune deconvolution using xCell revealed lower enrichment of Th1 and Th2 cells in the NPG score -high group. Consistently, CIBERSORTx showed reduced proportions of CD8⁺ T cells and M1 macrophages in the NPG-high group, suggesting that tumors with elevated NPG score are characterized by an immunosuppressive tumor microenvironment. Furthermore, analysis using pre-calculated scores from Thorsson et al. revealed that the NPG score-high group had a significantly lower tumor mutation burden compared to the score-low group. This finding suggests that the score-high group is associated with poor response to immune checkpoint inhibitors (ICI). To further investigate this, we extracted a subset of TNBC patients from the GSE194040 cohort who received neoadjuvant chemotherapy with ICI and evaluated the association between the NPG score and pathological complete response (pCR) to ICI therapy. pCR rates did not differ significantly between the two groups, but pCR cases tended to show lower NPG scores. In survival analysis, high NPG-scores were significantly associated with poorer overall survival in TNBC consistently in TCGA and METABRIC cohorts, while no such trend was observed in any other breast cancer subtypes. Conclusions This study highlights the prognostic significance of neuron projection guidance score in TNBC. High scores were associated with cell proliferation-related gene sets, and an immunosuppressive tumor microenvironment. These results indicate that intratumoral neurogenesis may contribute to changes in tumor behavior and therapeutic response.
Presentation numberPS3-13-04
Heterogeneous dormant breast cancer cells leverage bone marrow cell plasticity at the single-cell level to drive metastasis initiation
Noriko Gotoh, Cancer Research Institute, Kanazawa University, ??????, Japan
N. Gotoh; Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, ??????, JAPAN.
Breast cancer remains a leading cause of cancer-related deaths among women worldwide, largely due to the high risk of recurrence and metastasis in HER2-negative and hormone receptor-negative triple-negative breast cancer (TNBC) patients. While bone metastasis is common, the molecular mechanisms driving its initiation remain poorly understood. We recently identified a subpopulation of cancer stem cells (CSCs) with ancestral features, marked by the FXYD domain-containing ion transport regulator 3 (FXYD3), a Na+/K+ pump component. These “ancestor-like CSCs” persist in breast tissues during neoadjuvant chemotherapy (NAC), linking them to drug-tolerant persisters (DTPs) and positioning them as critical therapeutic targets for preventing local recurrence (Li M, Gotoh N et al., J Clin Invest, 2023). Despite this, the mechanisms underlying DTP dormancy in the bone marrow and their transformation into bone metastasis-initiating cells remain unclear. Given the stromal richness of TNBC tissues, we explored interactions between cancer-associated fibroblasts (CAFs) and breast cancer cells using a co-culture system. RNA-sequence analysis revealed a strong upregulation of granulocyte colony-stimulating factor (GCSF) in co-cultured CAFs. Notably, we identified FXYD3+ and GCSF receptor-positive cancer cells as a distinct subpopulation of CSCs with bone metastasis initiation potential. To investigate the temporal-spatial dynamics of bone metastasis initiation, we performed single-cell RNA sequencing (10x Genomics) and immunofluorescent imaging using metastatic bone marrow from the patient-derived xenograft (PDX) models. These analyses revealed how bone metastasis-initiating CSCs exploit the plasticity of bone marrow cells to establish a CSC niche at the single-cell level. Our findings provide critical insights into the mechanisms of recurrence and metastasis in breast cancer, offering novel therapeutic opportunities to target ancestor-like CSCs and prevent disease progression.
Presentation numberPS3-13-05
Epigenetic silencing of DNA sensing pathway by FOXM1 blocks stress ligand-dependent anti-tumor immunity and immune memory in triple negative breast cancer.
Jian Huang, UT Health San Antonio, San Antonio, TX
J. Huang, D. Medina, D. Singh, S. Abdulsahib, P. Subbarayalu, T. Do, P. Prabhakar, J. Prochnau, M. Rao; Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX.
Objective: This study investigates the role of transcription factor FOXM1 in creating an immune-suppressive tumor microenvironment (TME) by regulating stress ligands and the STING pathway in triple-negative breast cancer (TNBC).Methods: We performed in vitro and in vivo assays using CRISPR/Cas9-mediated knockout of FOXM1 in TNBC cell lines and syngeneic mouse models. Using single-cell RNA sequencing, we analyzed the impact of tumor-intrinsic FOXM1 on stress ligand expression and its downstream effects on immune cell interactions. FOXM1’s epigenetic regulation on STING pathway was further validated through chromatin immunoprecipitation.Results: We found tumor-intrinsic FOXM1 establishes an immune-suppressive TME by downregulating stress ligands like ULBP1 on cancer cells. This inhibition disrupts NKG2D-NKG2DL signaling, a key pathway for initiating natural killer and T cell cytotoxicity against cancer cells. FOXM1 achieves this by epigenetically silencing the DNA-sensor protein STING via a DNMT1-UHRF1 complex. Notably, patients with elevated FOXM1 and DNMT1 levels and reduced STING and ULBP1 levels show poorer survival rates and reduced responsiveness to immunotherapy.Conclusions: Our findings identify FOXM1 as a key regulator of immune evasion in TNBC, revealing a new pathway by which it shapes the TME through epigenetic repression. Targeting FOXM1 or its downstream effectors could enhance immunotherapy efficacy, offering new therapeutic strategies for patients with TNBC.
Presentation numberPS3-13-06
Antitumor activity of the oncolytic virus JX-594 in patient-derived xenograft models of triple-negative breast cancer
Jihye Lee, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea, Republic of
J. Lee1, Y. Kim2, Y. Cha3, N. Lee4, K. Oh4, J. Jeong5, J. Kim*2; 1Institute of Breast Cancer Precision Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 2Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 3Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF, 4Research center, SillaJen, Inc., Yongin-si, Gyeonggi-do, KOREA, REPUBLIC OF, 5Department of Surgery, Institute of Breast Cancer Precision Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, KOREA, REPUBLIC OF.
Introduction: Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). This profile limits treatment options primarily to chemotherapy, which is often associated with drug resistance, poor clinical outcomes, and is further challenged by the intrinsic heterogeneity and immunosuppressive tumor microenvironment of TNBC. JX-594 (pexastimogene devacirepvec; Pexa-Vec®), an oncolytic vaccinia virus derived from the Wyeth vaccine strain, has demonstrated multiple antitumor mechanisms, including direct oncolysis, immune activation, and disruption of tumor vasculature. Patient-derived xenograft (PDX) models closely replicate the original human tumors, providing a clinically relevant platform for evaluating novel therapeutics such as JX-594. In this study, we investigated the antitumor efficacy of intratumoral JX-594 administration in TNBC PDX models and assessed its effects on tumor cell proliferation, angiogenesis, and viral replication. Methods: Patient-derived xenograft (PDX) models were established by implanting fresh tumor tissues from two TNBC patients (ages 54 and 55) into the mammary fat pads of female NOD/SCID mice. Tumors were serially passaged and confirmed to retain human origin via Short Tandem Repeat (STR) profiling. Once tumors reached approximately 30 mm³ in volume, mice were randomized into two groups (n=6 per group per model): control or JX-594 treatment. JX-594 (1×10⁷ PFU in 50 µL saline) was administered intratumorally once weekly for four weeks. Tumor size, body weight, and general health were monitored. On day 28, mice were sacrificed, and tumors were collected, formalin-fixed, and paraffin-embedded for hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) analysis of Ki-67, CD31, and vaccinia antigen. Results: Administration of JX-594 significantly inhibited tumor growth in both TNBC PDX models compared to controls over a 28-day observation period. By the end of the study, mean tumor volumes in JX-594-treated groups were reduced by 91.7% relative to controls, with significant differences observed by four weeks after treatment initiation (p < 0.01). Importantly, oncolytic virotherapy was well tolerated, with no evidence of systemic toxicity or weight loss in treated mice. IHC analysis revealed extensive tumor cell death in JX-594-treated TNBC PDX models, as indicated by markedly reduced Ki-67 proliferation index and CD31 vascular marker expression levels. The presence of vaccinia antigen confirmed active viral replication within tumors. Conversely, control groups showed higher Ki-67 and CD31 expressions. Conclusion: This study demonstrates that intratumoral administration of the oncolytic vaccinia virus JX-594 effectively suppresses tumor growth in PDX models of TNBC. JX-594 induces tumor cell death, inhibits proliferation, and disrupts tumor vasculature, collectively contributing to its potential antitumor activity. These findings support the potential of JX-594 as a promising therapeutic strategy for treatment of TNBC.
Presentation numberPS3-13-08
The impact of immediate lipofilling on oncological outcomes, complication rates and patient reported outcomes in breast conserving surgery: a systematic review and meta-analysis
James Lucocq, Western General Hospital, Edinburgh, United Kingdom
J. Lucocq, H. Baig, J. Dixon; Edinburgh Breast Unit, Western General Hospital, Edinburgh, UNITED KINGDOM.
Background: There is concern from in-vitro studies that adipose-derived stem cells could promote cancer recurrence through their proliferative properties. The impact of immediate lipofilling (ILF) in the setting of breast conserving surgery on oncological outcomes, complications and patient-reported outcomes are unknown. Methods: A systematic search of Medline, Embase and Cochrane Central was conducted for studies investigating the impact of ILF in patients undergoing BCS. Random-effects meta-analysis were conducted for oncological outcomes (local [LR], regional [RR], distant [DR] and overall recurrence). Results: Six studies fulfilled the inclusion criteria including 252 patients. The pooled LR, RR, DR and overall recurrence rates were 2.38%, 1.52%, 3.03% and 5.95%, respectively (median follow-up, 39 months). A meta-analysis of studies comparing ILF (n=170) with no-ILF (n=362) found no difference in LR (OR, 0.77; 95%CI, 0.19-3.17; p=0.714), RR (OR 1.73, 95%CI 0.36-8.21, p=0.686), DR (OR 1.37, 95%CI, 0.51-3.63; p=0.627) or overall recurrence (OR 1.23, 95%CI, 0.60-2.52; p=0.569). Cancer-specific survival in the ILF group was 100% compared to 98.7% with no ILF. Post-operative calcifications (12.9% [21/163] vs. 0%; [0/72], p=0.002) and fat necrosis (7.8% (17/217] vs. 2.8% [8/283], p=0.011) were significantly more common with ILF, but early complications (e.g haematoma and infection) showed no difference (p>0.05). Three studies reported superior Breast-Q scores in ILF compared to no-ILF. No randomised controlled trials have been conducted. Conclusion: Immediate lipofilling following BCS is oncologically safe, enhances aesthetic outcomes and causes minimal morbidity.
Presentation numberPS3-13-09
Release of tumor antigens from breast cancer cells by a novel ultrasound ablation method
Suhe Wang, University of Michigan, Ann Arbor, MI
S. Wang, S. Tang, R. McGinnis, Z. Cao, Z. Xu; Internal Medicine, University of Michigan, Ann Arbor, MI.
Release of tumor antigens from breast cancer cells by a novel ultrasound ablation method Abstract Background: HER2-positive breast cancer accounts for about 20% of all breast cancer cases and is one of the most aggressive subtypes of breast cancer (1,2). These patients are often resistant to current antitumor treatments including immunotherapy. Even with new therapeutic strategies currently available, around 30% of patients relapse, often develop metastatic disease, and the 5-year survival rate for this patient cohort is only about 22%. Histotripsy (HT) is a non-invasive, non-thermal, non-ionizing ultrasound ablation method that utilizes high-pressure, microsecond ultrasound pulses at low-duty cycle to create cavitation at a focal point (3). It has been reported that HT can effectively inhibit the tumor at its target site and also arrest tumor growth at the remote site (3,4). The mechanism of the abscopal effect remains unclear but is thought to be related to HT-triggered systemic anti-tumor immune responses. The release of intact or immunogenic tumor antigens is critical in the induction of systemic and specific anti-tumor immune responses. Methods: HER2-positive murine mammary tumor cells were injected into HER2 transgenic mouse’s mammary fat pads. Tumor-bearing mice were treated with histotripsy with acute tumor resection. Treated tumors were respected and separated into cell-free and residual cell pellet fractions using centrifugation. Total protein concentration in both fractions was quantified using BCA protein assays. Ultra-performance liquid chromatography (UPLC) and Western blotting were used to evaluate and characterize HER2 protein. Histotripsy was performed using a custom built 8-element, 1 MHz transducer with ultrasound-guided targeting. The tested treatment dose was defined as the number of pulse repetitions per focal location (ppl). Then, 100 ppl was chosen for this study. Other acoustic parameters such as pulse repetition frequency remained fixed at 100 Hz PRF. Results: Our data showed that the HT cavitation can accurately target and disrupt tumor tissues and release more total proteins into the cell free fraction than untreated tumor by BCA assay. HT treatment significantly increases the level of released HER2 protein as measured by UPLC and Western blot analysis. UPLC analysis further showed that the cell-free fractions exhibited a protein peak with a retention time at around 9.3 minutes. This released antigen appeared identical to recombinant HER2 protein, suggesting that the HER2 released by HT is in line with intact protein. Conclusions: Our findings have demonstrated that HT-treated orthotopic breast tumor could trigger the release of intact tumor antigens, which in some manner can stimulate the host immune system. The release of such intact and concealed tumor antigens can create the basis for HT-induced systemic anti-tumor immune responses and a favorable environment for immunotherapies. Finally, this increase in immune activity potential enhances the host’s ability to prevent tumor recurrence and metastasis. References:1.Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther. 2025 Feb 19;10(1):49.2.Khan MM, Yalamarty SSK, Rajmalani BA, Filipczak N, Torchilin VP. Recent strategies to overcome breast cancer resistance. Crit Rev Oncol Hematol. 2024 May;197:104351.3.Xu Z, Hall TL, Vlaisavljevich E, Lee FT Jr. Histotripsy: the first noninvasive, non-ionizing, non-thermal ablation technique based on ultrasound. Int J Hyperthermia. 2021;38(1):561-575.4.Pepple AL, Guy JL, McGinnis R, Felsted AE, Song B, Hubbard R, Worlikar T, Garavaglia H, Dib J, Chao H, Boyle N, Olszewski M, Xu Z, Ganguly A, Cho CS.Spatiotemporal local and abscopal cell death and immune responses to histotripsy focused ultrasound tumor ablation. Front Immunol. 2023 Jan 23;14:1012799.
Presentation numberPS3-13-10
Immunomodulation of Tumor Microenvironment by Reversing Hypoxia
Pavani Chalasani, The George Washington Cancer Center, Washington, DC
P. Egwon1, A. Nanduru2, A. Kiss3, G. Cresswell3, D. Roe4, E. Unger5, J. Johnson6, P. Chalasani3; 1Oncology, NCI, Bethesda, MD, 2Medicine, The George Washington University, Washington, DC, 3Medicine, The George Washington Cancer Center, Washington, DC, 4Biostatistics, The University of Arizona Cancer Center, Tucson, AZ, 5N/A, Nuvox Therapeutics, Tucson, AZ, 6NA, Nuvox Therapeutics, Tucson, AZ.
Immunomodulation of Tumor Microenvironment by Reversing Hypoxia Background:Triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and higher mortality than other breast cancer subtypes. Tumor hypoxia due to rapid proliferation of cancer cells impairs T cell infiltration and function in TNBC. Although reversing tumor hypoxia has shown promise in pre-clinical tumor models, existing oxygenation strategies remain limited for clinical translation. NanO₂ (NuvOx Pharma), a novel perfluorocarbon-based oxygen therapeutic composed of 2% w/v dodecafluoropentane, improves tissue oxygenation without receptor binding. It has been shown to reverse tumor hypoxia in preclinical models and in patients with Glioblastoma Multiforme. We hypothesized, that reversing hypoxia with NanO₂ will change tumor microenvironment (TME) to enhance response to immunotherapy and chemotherapy. Methods:We tested NanO₂ on hypoxia reversal, changes in TME in a syngeneic TNBC model (4T1 cells in BALB/c mice). Flow cytometry and IHC (CD4, CD8a, CD19, CD206, F4/80, PD-1) were used to characterize immune cell populations. In our first series of experiments we tested the effect of NanO₂ with/without oxygen in 2 dosing regimens: single dose or five daily doses. Seventy mice received either saline or NanO₂ intravenously, with air or O₂ exposure for 17 days starting on day 5 post-tumor implantation. In the second series of experiments, we tested the effect of NanO₂ in combination with immunotherapy (IT), chemotherapy (CTX, paclitaxel) or both IT and CTX. BALB/c mice were treated for three weeks starting on day 3 post-implantation with IV NanO₂ (every 3 days), anti-PD-1 (200 µg IV every 5 days), Paclitaxel (2 µg/g IP every 3 days), or combinations. Due to unexpected mortality, only 26 mice reached the endpoint.Results:Treatment with NanO₂ decreased hypoxic cells when combined with oxygen. In addition, it decreased PD-1 positive cells, increased CD8 cell infiltration. Tumor associated macrophages (TAMs) were also increased. PD-1 expression decreased supporting that reversing hypoxia can enhance response to IT. In addition, in the second series of experiments we demonstrated that the TME changes by NanO₂ +oxygen were enhanced by IT, CTX and combination IT+CTX. Conclusions:NanO₂ synergizes with immunotherapy and chemotherapy to remodel the TME to enhance anti-tumor immune responses in in vivo TNBC models.Figures to Upload:CD8 IHC
Decreased Hypoxic Cells![[{7cd5b340-c7e2-4a45-8748-e775b5650e6f}sad3ad.html_g�0004.gif]]({7cd5b340-c7e2-4a45-8748-e775b5650e6f}sad3ad.html_g4.gif)
Presentation numberPS3-13-11
Expression of Brutons tyrosine kinase (BTK) in breast cancer associates with tumor immune microenvironment but lacks predictive value for clinical outcome
Tamrah ALRammah, Roswell park comprehensive cancer center, BUFFALO, NY
T. ALRammah; surgical oncology, Roswell park comprehensive cancer center, BUFFALO, NY.
Background: Bruton’s tyrosine kinase (BTK), a downstream mediator of the B-cell receptor (BCR) signaling pathway, is known to be essential in differentiation and proliferation of B-cell. Thus, BTK inhibitors are used to treat refractory mantle cell lymphoma and chronic lymphocytic leukemia. Recent discoveries have revealed the expression of BTK in myeloid-derived suppressor cells, known to worsen breast cancer (BC) outcomes. To this end, it was of interest to investigate the clinical relevance of BTK expression in BC. Methods: Total of 9297 BC patients from 23 independent cohorts with tumor transcriptome and clinical data were analyzed. Results: Based on two independent single-cell sequencing cohorts, approximately 15% of B-cells and 30% of myeloid cells within the tumor microenvironment expressed BTK, while no other cells exhibited BTK expression, suggesting that the BTK expression signal from bulk tumor samples primarily originates from these cells. Correlation with BTK expression was highest in macrophage regulation (r=0.94), and leukocyte fraction, lymphocyte infiltration, TCR Shannon and TCR Richness (all r>0.74), but not with BCR Shannon nor BCR Richness. Total macrophage, M1 and M2 macrophage, dendritic cells, total B-cells, memory B-cells, as well as CD8 and CD4 cells were all highly infiltrated in BTK high BCs. Moreover, BTK expression correlated with both PD1 & PD-L1 in all BC subtypes. Biologically, BTK high BC expression was linked to immune-related gene sets, with gene set enrichment analysis (GSEA) showing enrichment in pathways related to immune response such as Complement, Inflammatory response, Allograft rejection, IFN-γ, IFN-α, IL2, IL6, and TNFα signaling. At the same time, BTK high BC enriched tumor-aggravating signaling pathways such as PI3k/AKT/mTOR signaling and mTORC1 signaling. The cytolytic activity score, reflecting the global immune killing, was significantly higher in the BTK high BC. Given that BC with abundant tumor infiltrating lymphocytes respond better to chemotherapy, the relationship between BTK expression and response to neoadjuvant chemotherapy was assessed. Somewhat unexpectedly, BTK expression was associated with better response in only one out of ten neoadjuvant cohorts. Further, no change in BTK expression were observed before and after neoadjuvant chemotherapy in six cohorts. While BTK expression was higher in primary sites compared to metastatic sites, it was not associated with distant recurrence. Across the TCGA, METABRIC, and SCAN-B cohorts, no significant overall survival differences were observed between the BTK high and low expression groups. All findings were consistent regardless of the BC subtypes. Conclusions: Myeloid cells and B-cells were the source of BTK expression, and it was associated with macrophage regulation and infiltration of various types of immune cells and proteins including PD1 & PD-L1 and enriched immune related gene sets. However, BTK expression was not associated with response to neoadjuvant chemotherapy or Impact on survival, regardless of BC subtypes.
Presentation numberPS3-13-12
Parp inhibition augments response to anti-pd1 therapy in hrd mutated breast cancer
Sibapriya Chaudhuri, University of California San Francisco, San Francisco, CA
S. Chaudhuri, A. Emata, J. Camara Serrano, V. Steri, S. Thomas, P. Munster; Medicine, University of California San Francisco, San Francisco, CA.
Introduction: Over 2 million women develop breast cancer globally every year despite immense advances in disease biology, genomic assessment and novel treatment options. Many preclinical and clinical studies suggest a benefit of combining PARP inhibitors (PARPi) and immune checkpoint inhibitors (ICi) in patients with homologous recombination intact and mutated cancers. However, similar to the varied benefits of either drug class as single agent, the benefits of such combinations are strongly dependent on tumor type, tissue and mutational context. In this study, we explored the association of immune cell composition in the tumor tissues for driving the responses. Methods: To study how select homologous recombination deficiency (HRD) mutations respond to a combination of a PARPi with an ICi, we used CRISPR engineering to repress BRCA1, BRCA2, ATM, CHEK2, PALB2 in different murine and human breast cancer cell lines. The CRISPR engineered cells were implanted in mice and treated with PARPi and ICi to determine whether the ICi, PARPi and their combination induced response. Using spectral flow-cytometry, immunohistochemistry and RNA sequencing, the different immune cell population in the tumor microenvironment were analyzed.Results: In vitro, induced suppression of BRCA1 and PALB2 in the EMT6 cell lines showed higher sensitivity to PARPi (IC50 0.01 uM and 0.098 uM) than suppression of BRCA2 and ATM (IC50 0.16 uM and IC50 0.28 uM). In contrast, induced suppression of CHEK2 showed resistance to PARPi compared to their parental EMT6 cells (IC50 13.9 uM versus 5.3 uM). We further validated these findings in MDA-MB231 and observed similar trends. Expression levels of PDL1 and select chemokines (CCL2, CCL5, CXCL9, CXCL10 and CXCL11) were associated with sensitivity to PARPi. We investigated the efficacy of combining PARPi with ICi in our in vivo models. In EMT6 background, presence of BRCA1 or BRCA2 mutations showed sensitivity to either single agent or the combination. The response correlates with higher CD8+ T cells infiltration. Presence of ATM mutation has shown benefit to either single agent but not enhanced by the combination. This mirrors the findings in the clinic.Conclusion: Using inducible suppression of HRD function in an immune competent mouse breast cancer model helps to understand the differential role of individual HRD mutations (BRCA1, BRCA2, ATM, CHEK2, PALB2) in altering inflammatory signals in response to PARPi and ICi. This data may guide the clinical development of such combination in breast and other cancers.
Presentation numberPS3-13-13
Cryoablation Combined with Immune Checkpoint Inhibitors Enhances Conventional Dendritic Cell Activation in Abscopal Tumors and Tumor-Draining Lymph Nodes.
Flávia Sardela de Miranda, Texas Tech University Health Sciences Center, Lubbock, TX
F. Sardela de Miranda1, R. L. Babcock2, G. P. Boligala2, M. F. Mahecha1, P. J. Gukhool1, A. K. Garcia3, A. G. Oliver3, C. Bose4, S. Almodovar3, K. Pruitt5, S. P. Singh6, M. W. Melkus1, R. Layeequr Rahman1; 1Department of Surgery and Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, 2Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, 3Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, 4Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, 5Department of Pharmacology, University of North Carolina, Chapel Hill, NC, 6Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX.
Background: Immune checkpoint inhibitors (ICIs), such as anti-CTLA-4 which acts primarily during T cell priming and anti-PD-1 which targets exhausted T cells, have shown clinical efficacy in triple-negative breast cancer (TNBC) but are associated with immune-related adverse events. These limitations underscore the need for combination strategies that enhance therapeutic outcomes and reduce ICI dosing requirements. Cryoablation, a non-surgical procedure, “kills” the tumor through freeze-thaw cycles while preserving tumor-associated antigens (TAAs). This procedure induces tumor necrosis and release of damage-associated molecular patterns that trigger antitumor immune responses and may enhance the abscopal effect – systemic immune response against distant tumors. The immune activation is mediated by immune cells recruited to the ablation site and tumor-draining lymph nodes (TdLNs). Conventional dendritic cells (cDC) play a central role in antitumor immunity by capturing TAAs and presenting them to T cells. Among cDC subsets, cDC1 – including lymphoid resident (CD8α⁺) and migratory (CD103⁺) populations – specialize in cross-presenting antigens to CD8⁺ T cells, while cDC2 primarily present antigens to CD4⁺ T cells. We hypothesized that combining cryoablation with ICIs enhances antitumor immunity by promoting antigen presentation and T cell activation. Methods: In a murine TNBC cryoablation model, 4T1-12B cells were bilaterally injected into BALB/c mice. The left (primary) tumors were cryoablated at 2 weeks, while the right tumors (abscopal tumors) were left intact to represent distant metastatic tumors to examine the immune abscopal effect. PBS or 100 µg ICI (anti-CTLA-4, PD-1, or PD-L1) were administered by I.P. injection 24h pre- and post-cryoablation. Mice were sacrificed a week later and evaluated for early immune activation. The primary and abscopal tumors and TdLNs, peripheral blood, and spleen were collected, processed and analyzed for immune system activation by flow cytometry. Results: Preliminary analyses demonstrated that combining cryoablation with anti-CTLA-4 therapy enhanced CD4+ and CD8+ T cell activation in both the spleen and peripheral blood compared to cryoablation alone or in combination with PD-1/PD-L1 blockade. Notably, relative to cryoablation monotherapy, the addition of anti-CTLA-4 increased the frequency and activation of both cDC1 and cDC2 subsets in the abscopal TdLNs. Among cDC1 populations, the lymphoid resident subset was significantly elevated in the cryoablation plus anti-CTLA-4 group. Furthermore, abscopal tumors from this group showed a higher frequency of activated CD4⁺ and CD8⁺ T cells, as well as NKT cells. Conclusions: These results suggest combination of cryoablation with anti-CTLA-4 therapy enhances systemic antitumor immunity more effectively than cryoablation alone or with PD-1/PD-L1 inhibition. One possible explanation is that cryoablation destroys the entire tumor immune microenvironment, resulting in fewer effector T cells for PD-1/PD-L1 inhibitors to act upon, which limits efficacy in this context. In contrast, anti-CTLA-4 may act earlier in the immune response by promoting de novo T cell priming. We observed increased frequencies and activation of cDC1 and cDC2 in the TdLNs of abscopal tumors, which likely contributed to the elevated numbers of activated CD4⁺ and CD8⁺ T cells in the abscopal tumor microenvironment. Altogether, these findings highlight the potential of cryoablation combined with anti-CTLA-4 to enhance antitumor immunity. Our results support further investigation into this combination strategy to assess its long-term potential to prevent tumor recurrence and metastasis.
Presentation numberPS3-13-14
Transcriptomic Analysis of Differentially Expressed Genes in Triple-Negative Breast Cancer: Insights into Imune Cell Infiltration and the Tumor Microenvironment
Dyego Lucas Pereira Nunes, Instituto D’Or de Pesquisa e Ensino (IDOR), Salvador, Brazil
D. L. Nunes1, R. V. Gisele2, V. D. Bertoni3, C. A. Rocha4; 1Oncologia, Instituto D’Or de Pesquisa e Ensino (IDOR), Salvador, BRAZIL, 2Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BRAZIL, 3Oncologia, Clínica AMO, Salvador, BRAZIL, 4Centro de Pesquisa Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BRAZIL.
Transcriptomic Analysis of Differentially Expressed Genes in Triple-Negative Breast Cancer: Insights into Immune Cell Infiltration and the Tumor MicroenvironmentBackground:Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive subtype associated with poor prognosis and limited therapeutic options. Molecular and immunological characterization of the tumor microenvironment (TME) may support the identification of prognostic biomarkers and new therapeutic targets.Objective:To evaluate, through an in silico approach, the gene expression profile and immune composition of the TNBC tumor microenvironment compared to hormone receptor-positive (HR+) and HER2-positive subtypes.Methods:We used clinical and RNA-seq data from the TCGA-BRCA dataset, comprising 1,101 breast cancer patients. Tumor samples were classified into TNBC, HR+, and HER2+ groups. Immune and stromal cell composition was estimated using the CIBERSORT and ESTIMATE algorithms. Differentially expressed genes (DEGs) were identified using the DESeq2 package. Gene co-expression network analysis was conducted using WGCNA, and overall survival was analyzed using Kaplan–Meier curves.Results:A total of 226 DEGs were identified: 206 in the TNBC vs HR+ comparison and 96 in the TNBC vs HER2+ comparison, with 76 genes shared between both. TNBC samples showed higher levels of CD8⁺ T cells (p < 0.01), activated dendritic cells, and immune scores (p < 0.001), but also increased T regulatory cells (p < 0.05) and M2 macrophages (p < 0.0001), suggesting a complex immune landscape with both cytotoxic and immunosuppressive elements. WGCNA identified two relevant modules: the blue module, correlated with stromal score, included DSG1, ACAN, RHCG, and KRT6C, genes linked to extracellular matrix remodeling; and the turquoise module, correlated with immune score, included CD274 (PD-L1), LAG3, IDO1, CXCL10, and GZMB, markers of immune activation and evasion. Survival analysis showed that CXCL13, CD3G, and GZMB were significantly associated with worse overall survival (p < 0.05).Conclusion:This integrative analysis revealed distinct gene signatures and immune patterns in TNBC, with enrichment of pathways related to inflammation, immunosuppression, and stromal remodeling. These findings suggest potential prognostic and predictive biomarkers and reinforce the TME as a relevant target for future therapeutic strategies. These results highlight promising targets for translational research and contribute to a better understanding of TNBC biological heterogeneity.
Presentation numberPS3-13-15
Dasatinib-induced Src inhibition blocks nonangiogenic vascularization and potentiates antibody-drug conjugates in high-grade breast cancer
Masafumi Shimoda, Hyogo Medical University, Nishinomiya, Japan
M. Shimoda1, I. Seto2, M. Masuyama2, K. Kikumori2, K. Shimazu2; 1Department of Breast and Endocrine Surgery, Hyogo Medical University, Nishinomiya, JAPAN, 2Department of Breast and Endocrine Surgery, University of Osaka, Suita, JAPAN.
Background: Nonangiogenic vascularization (NAV)—encompassing vascular mimicry (VM) and vessel cooption (VCO)—allows high-grade breast cancers to secure a blood supply and evade antiangiogenic therapy. We aimed to identify a small-molecule inhibitor that disrupts NAV. Methods: A library of 259 clinically relevant compounds was screened using VM tube formation assays in trastuzumab-resistant, HER2-positive JIMT-1 cells and triple-negative MDA-MB-231 cells. VCO was evaluated by the adhesion of cancer cells to preformed human umbilical vein endothelial cell tubes. Mechanistic studies involved Src pathway immunoblotting and siRNA knockdown. Antitumor efficacy was evaluated in orthotopic xenografts, with vasculature characterized by mouse CD31, mouse CD105, and human SERPINE2 immunostaining and apoptosis evaluated by TUNEL assay. Combination studies with trastuzumab-emtansine (T-DM1) were conducted in the JIMT-1 model. Results: Three Src inhibitors—dasatinib, bosutinib, and ponatinib—emerged as potent VM inhibitors, with dasatinib being the most active (IC₅₀ = 1.4 nM in JIMT-1 cells; 7.3 nM in MDA-MB-231 cells). Dasatinib also abrogated VCO, and Src knockdown phenocopied its effects, confirming pathway dependence. In vivo, dasatinib significantly reduced tumor growth without increasing the number of TUNEL-positive cells; however, it markedly depleted the number of CD31-positive mature vessels and SerpinE2-positive tumor cells but spared CD105-positive immature vessels. Dasatinib improved the antitumor activity of T-DM1; specifically, the combination further suppressed CD31-positive vasculature. Conclusions: Dasatinib disrupts NAV by targeting Src, translating into tumor growth inhibition and improved efficacy of T-DM1 therapy. These results support repurposing dasatinib to overcome NAV-mediated therapeutic resistance in high-grade breast cancer.
Presentation numberPS3-01-02
The Role of a Low-Fat Dietary Intervention on Intake of Dietary Advanced Glycation End Products in Postmenopausal Women
Margaret S Pichardo, University of Pennsylvania, Philadelphia, PA
M. S. Pichardo1, R. P. Hunt2, L. L. Peterson3, K. Pan4, M. Coday5, S. Jung6, L. Snetselaar7, M. L. Neuhouser2, S. E. Steck8, F. K. Tabung9, N. Saquib10, J. E. Manson11, R. T. Chlebowski12; 1University of Pennsylvania, Philadelphia, PA, 2Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 3Division of Medical Oncology, Department of Medicine, Siteman Cancer Center,, Seattle, WA, WA, 4Kaiser Permanente Southern California, Downey, CA, 5Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, 6Translational Sciences Section, School of Nursing, Department of Epidemiology, Fielding School of Public Health, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 7Epidemiology and Endocrinology/Metabolism, University of Iowa, Iowa City, IA, 8Department of Epidemiology & Biostatistics, Arnold School of Public Health, University of South Carolina,, Columbia, SC, 9Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine and Comprehensive Cancer Center, Columbus, OH, 10Clinical Sciences Department, College of Medicine, Sulaiman AlRajhi University, Al-Bukairyah, Al-Qassim, Saudi Arabia, 11Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School,, Boston, MA, 12The Lundquist Institute, Torrance, CA
Background: Advanced glycation end-products (AGEs) are compounds produced in the body during the metabolism of protein, lipids or nucleic acids, and have been implicated in breast cancer risk and prognosis. In the Women’s Health Initiative (WHI) Dietary Modification (DM) randomized trial, the low-fat dietary intervention significantly reduced breast cancer mortality (P = 0.02) over 20 years of follow up. As dietary AGEs (dAGEs) are associated with westernized diets and adversely impact breast cancer prognosis, we examined whether the WHI low-fat dietary pattern influenced consumption of dAGEs. Methods: A total of 48,835 postmenopausal women, ages 50 to 79, were randomized to a low-fat dietary intervention (n = 19,541) versus usual diet comparison (n=29,294). Food frequency questionnaires were administered at baseline, year 1, and yearly thereafter from a rotating subgroup and were used to estimate dAGE scores (kilo Units/100 kilocalories [kU/1000 kcal]) using a commonly referenced database. Multivariate regressions with repeated measures for dAGEs were examined by DM randomization group and follow-up duration (baseline, years 1, 2-4 and 5-7). Results: Baseline mean dAGE scores were similar for intervention (7,542 kU/1000 kcal, SD = 912) and comparison (7,514 kU/1000 kcal, SD=917) groups. In multivariate regression, the largest difference in dAGE was observed after 1-year follow-up (intervention: 5,362 kU/1000 kcal versus comparison: 7,159 kU/1000 kcal). Subsequently, persistently lower dAGE levels were found in the intervention versus comparison group, with some attenuation of the difference: visit years 2-4 (intervention: 5,818 kU/1000 kcal v. comparison: 7,458 kU/1000 kcal) and visit years 5-7 (intervention: 6,236 kU/1000 kcal v. comparison: 7,669 kU/1000 kcal; P <0.0001). Conclusions: Among postmenopausal women, a low-fat dietary intervention substantially reduced dAGE intake. Future studies will explore whether reduced dAGE intake moderated the favorable breast cancer outcomes previously reported in the WHI DM trial.
Presentation numberPS3-01-17
Brca1/2 mutation status and its prognostic implications in breast cancer: a 10-year survival analysis in peru
Natalia Valdiviezo, Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, Lima, Peru
N. Valdiviezo1, P. Mora2, I. Otoya1, M. Acosta2, J. Herzog3, S. Neciosup1, T. Vidaurre1, Z. Morante1, C. Castañeda1, H. Gómez4, C. Rabanal1, L. Reynaga3, Y. Sullcahuaman2, H. Guerra5, M. Velarde6, J. Cotrina7, V. Acuña8, S. Gruber3; 1Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, Lima, Peru, 2Unidad Funcional de genética y biología molecular, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, Lima, Peru, 3City of Hope National Medical Center, Duarte, CA, 4OncoSalud AUNA, Lima, Peru, 5Anatomía Patológica y Patología Oncológica, Patólogos Especializados SAC, Lima, PERU, Lima, Peru, 6Cirugía de Mamas y Tejidos Blandos, Instituto Nacional de Enfermedades Neoplásicas, Lima, PERU, Lima, Peru, 7Cirugía de Mamas y Tejidos Blandos, Instituto Nacional de Enfermedades Neoplásicas, Lima, Peru, 8Facultad de Farmacia y Bioquímica, Universidad Nacional Mayor de San Marcos, Lima, PERU, Lima, Peru
Breast cancer is a heterogeneous disease with genetic, environmental, and lifestyle causes. Among the hereditary forms, mutations in BRCA1 and BRCA2 represent the main risk factors, accounting for approximately 20-25% of hereditary cases and 5-10% of all cases. These tumor suppressor genes maintain genomic stability through DNA repair; their mutations increase the predisposition to early-onset breast cancer, bilateral tumors, and ovarian cancer. The objective of this study was to characterize and compare the clinical, pathological, and survival profiles of Peruvian breast cancer patients according to BRCA1/2 mutation status, and to evaluate their prognostic impact on overall survival. METHODS: We included 1,446 breast cancer patients evaluated at INEN (2013-2020) under protocol INEN13-48. Genetic testing for BRCA1/2 was performed at City of Hope. Group comparisons by BRCA status were conducted using standard statistical tests. Propensity score matching (1:1) by age and clinical stage resulted in 242 matched patients. Ten-year overall survival was analyzed with Cox regression models, accounting for delayed entry. RESULTS: A total of 1,441 breast cancer patients were analyzed based on BRCA mutation status (BRCA-negative, BRCA1+, and BRCA2+). Significant clinicopathological differences were observed between the groups. The median age at diagnosis was 41 years (range: 18-84). BRCA1 carriers were diagnosed at a significantly younger age (median: 39 years) than BRCA-negative and BRCA2 patients (p = 0.042). Clinical stage II was the most frequent in all groups (~43%). Although no statistically significant differences were observed in stage distribution, stage IV was slightly more prevalent in BRCA2 (13.2%). Molecular subtypes differed significantly between groups (p < 0.001): BRCA1: Predominantly triple-negative tumors (85.4%) and BRCA2: Higher proportion of HR+/HER2- tumors (47.6%). Regarding tumor proliferation, a Ki-67 index ≥ 20% was observed in 100% of BRCA1 and BRCA2 patients, significantly higher than in BRCA-negative patients (80.6%, p < 0.001). The presence of a second cancer was more frequent in BRCA1+ (37.9%) and BRCA2+ (25.5%) patients compared to BRCA-negative individuals (13.9%, p < 0.001). The most common type was a second breast cancer (69.4% in BRCA1; 66.7% in BRCA2), and a notable proportion of ovarian cancer was found in BRCA1+ patients (25%). Survival analysis revealed that BRCA1+ patients had a significantly higher risk of death compared to BRCA-negative patients (HR = 1.83; 95% CI: 1.12-2.98; p = 0.016). Advanced clinical stage (III/IV vs. I/II) was associated with nearly a threefold increased risk of death (HR = 2.70; 95% CI: 1.65-4.41; p < 0.001), and the triple-negative subtype was associated with a significantly higher mortality risk (HR = 2.10; 95% CI: 1.19-3.68; p = 0.01). CONCLUSIONS: BRCA1 mutation carriers were diagnosed at a younger age and exhibited a more aggressive clinical and pathological profile, characterized by a predominance of the triple-negative subtype, high Ki-67 index, and G3 histological grade. Additionally, they presented a higher frequency of second cancers (37.9%), particularly breast and ovarian cancer. These findings highlight the importance of considering BRCA status in prognosis assessment and treatment planning for breast cancer patients.
Presentation numberPS3-02-14
Incidental Histopathological Findings in Risk Reducing Breast Surgery: A Cross Sectional Study
Cesar Cabello, State University of campinas – UNICAMP, campinas, Brazil
T. Fernandes, D. Silva, A. O. Magalhaes, B. Duarte, A. cabello, S. Ramalho, L. R. da Silva, G. Paiva, r. torresan, F. Brenelli, M. Minari, T. Gaspar, A. Viaro, S. Teixeira, M. Faucz, C. Alem, C. Cabello; State University of campinas – UNICAMP, campinas, Brazil
Abstract: Purpose: Risk-reducing breast surgery (RRBS) is an established preventive strategy for women at high genetic or familial risk of breast cancer. Histopathological evaluation of RRBS specimens may reveal incidental findings (IFs), including occult malignancies and precursor lesions, but their prevalence and predictors remain insufficiently described. This study aimed to estimate the prevalence of IFs and identify associated risk factors in a Brazilian cohort. Methods: We conducted a cross-sectional analysis of 154 women who underwent RRBS at a tertiary public cancer center in Brazil between 2013 and 2025. Eligible patients carried pathogenic/likely pathogenic germline variants and/or fulfilled NCCN high-risk criteria. Histopathological assessment followed an institutional protocol with extensive tissue sampling. Logistic regression models evaluated demographic, clinical, and surgical predictors of IFs. Results: IFs were identified in 35% (54/154) of patients, including atypical proliferations (70%), DCIS (22%), and invasive neoplasia (8%). On multivariate analysis, older age (OR 1.071) and bilateral mastectomy (OR 3.27) were significant independent predictors of IFs. For invasive and DCIS findings, age was the only significant factor (OR 1.069). Genetic status showed no significant association. Conclusion: This study reports the highest prevalence of IFs in RRBS specimens to date, largely attributable to the comprehensive sampling protocol. Associations with clinical characteristics, particularly age, contrast with the absence of correlation between negative genetic testing and reduced risk. These findings highlight the importance of integrating detailed clinical and familial risk assessment into surgical decision-making, especially in resource-limited systems with restricted access to genetic testing.
Presentation numberPS3-02-18
Association of epidemiological and reproductive factors with risk of hormone receptor-positive/negative breast cancer: a case-control study in Beijing, China (CABC011)
Huiping Li, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China, Beijing, China
H. Li1, Y. Lei2, F. Zhao3, H. Liao1, Y. Zhang4, Y. Liu1, R. Zhang1, H. Zhao2, R. Ran1, H. Chen5, Q. Zhan3; 1Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China, Beijing, China, 2Department of General Surgery, Peking University Third Hospital, Beijing, 100191, China, Beijing, China, 3Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China, Beijing, China, 4Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital and Institute, Beijing, 100142, China, Beijing, China, 5Jiangsu Hengrui Medicine CO., LTD., Lianyungang, 222047, China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China., Hangzhou, China
Background: The reason of rapidly increasing of breast cancer (BC) incidence in China has invested in recent years. However, the pathogenesis of BC, especially hormone receptor-positive/negative (HR+/−) BC among the Chinese population, remains unclear.Methods: This was a case-control study conducted in Beijing, China. BC patients and healthy women were enrolled as cases and controls, respectively. Information on demographic factors [(age, body mass index (BMI), education level, family history of cancer)] and reproductive factors (menopausal status, age at menarche, number of pregnancies, number of children, and weather performed breast feeding) were collected using a questionnaire of 28 items. We also collected data on the patients’ estrogen receptor (ER) and progesterone receptor (PR) statuses from their medical records. Logistic regression were used to estimate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs) for the risk factors of interest.Results: We analyzed 471 BC patients and 473 healthy controls. ER and PR expression data were available for 439 BC patients; 355 (75.4%) had HR+ (ER+ and/or PR+) BC, while 84 (17.8%) had HR− (ER− and PR−) BC. Multiple logistic regression analysis indicated that higher education level (OR: 0.619; 95% CI: 0.513~0.747; P < 0.001), was a protective factor against in all cohort both in HR+ and HR- BC; but in HR+ group showed that full pregnancy (OR: 0.622, 95% CI: 0.425~0.911, P = 0.015), bearing more children (OR: 0.622, 95% CI: 0.425~0.911, P= 0.013), less abortion and miscarry (OR: 1.358, 95% CI:1.114 ~1.655,P = 0.002); took longer period of breast feeding (OR: 0.798, 95% CI: 0.691~0.921, P = 0.002) , and without family history of BC (OR:9.691, 95% CI: 2.088~44.983, P = 0.004) were protective factors against HR+ BC; while family history of BC in first degree relatives (OR: 3.076; 95% CI: 0.988~9.484; P = 0.05) was a risk factor for HR- BC.Conclusions: Higher education level protected against BC, while a family history of BC was a risk factor for BC. Reproductive factors play a more significant role in affecting HR+ BC risk; while genetic factors like family history of BC in first degree relatives are more relevant in the development of HR− BC.
Presentation numberPS3-03-08
Initial results from RECAST DCIS: Multicenter platform trial testing active surveillance and novel endocrine therapy agents for DCIS management
Kelly C Hewitt, University of Utah, Salt Lake City, UT
K. C. Hewitt1, C. Yau2, R. Mukhtar2, C. Thompson3, J. Hui4, K. Westbrook5, J. Jackson2, C. Carruthers6, C. Farley7, S. Bommakanti8, K. Brownson1, B. Fan9, I. Meszoely10, T. Balija11, M. H. Mcnatt12, P. Gilman6, J. Marti13, T. Li2, S. Horton14, A. Borowsky2, J. Rosenbluth2, L. Esserman2; 1University of Utah, Salt Lake City, UT, 2University of California, San Francisco, San Francisco, CA, 3University of California, Los Angeles, Los Angeles, CA, 4University of Minnesota, Minneapolis, MN, 5Duke University, Durham, NC, 6Mainline Health, Philadelphia, PA, 7Emory University, Atlanta, GA, 8Hennepin Healthcare, Minneapolis, MN, 9University of Chicago, Chicago, IL, 10Vanderbilt University, Nashville, TN, 11Mt Sinai, New York, NY, 12Wake Forest, Winston Salem, NC, 13Englewood Health, Englewood, NJ, 14Quantum Leap Healthcare Collaborative, San Francisco, CA
Ductal carcinoma in situ (DCIS) of the breast is a heterogeneous, non-invasive condition characterized by the proliferation of malignant epithelial cells within the duct, with variable risk of progression to invasive disease. Current standard treatment involves surgical resection followed by possible adjuvant endocrine therapy and/or adjuvant radiation. However, a substantial proportion of DCIS may never progress to invasive disease and thus may be overtreated with the current standard of care. Consequently, active surveillance (AS) is emerging as a treatment option for patients with DCIS. DCIS is definitely a risk factor for breast cancer, and should be reframed as an opportunity for preventive intervention. RECAST (Re-evaluating Conditions for Active Surveillance Suitability as Treatment) is a Phase II adaptive platform trial studying the efficacy of up front endocrine therapy to identify appropriate candidates for long-term active surveillance versus those who would benefit from standard surgical treatment. Patients with hormone receptor positive DCIS of any grade undergo baseline breast MRI and are randomized to one of four hormonal therapy regimens, including standard endocrine therapy, z-endoxifen, elacestrant or HAV-008 with the potential to offer equivalent or improved benefit with less adverse effects. Serial MRI at 3 and 6 months is then used to evaluate change in background parenchymal enhancement (BPE) and DCIS lesion. Patients deemed low risk based on imaging response have the option to continue AS and avoid surgery and radiation and continue their endocrine therapy for 3 years. The primary endpoint of the trial is to determine whether novel endocrine therapy increases the fraction of patients who will be suitable for long-term AS as measured by the fraction of patients who remain on active surveillance at 7 months compared to control. To date, over 50 patients are enrolled in the trial. Our target accrual of 400 patients. A total of 17 (34%) patients have reported an AE, all grade 1 or 2, with no serious adverse events reported and no safety signals identified. Of the 50 patients currently on treatment, 25 have reached or gone beyond the 6-month mark and 22 (88%) of those patients remain on active surveillance with a median follow up of 351 days. At the 3 month point, one patient progressed on imaging and two chose to come off surveillance due to other reasons. At 6 months, 8% (2/25) progressed on imaging and one additional patient has proceeded to surgery at the 1 year point. One patient who went on to have surgery was found to have invasive disease (pT1bN0). Robust accrual continues at sixteen currently active sites with five more sites anticipated to open in the next few months. The shift to using hormonal therapy in the neoadjuvant setting allows investigators to assess response to endocrine therapy, characterize risk of progression to invasive cancer and helps to better inform treatment-making decisions before continuing AS or proceeding with surgery, and appropriately assesses the value of endocrine risk reduction prior to surgery to avoid it. The trial uses a patient-centered approach and gives all patients a window of opportunity to assess their response as well as medication tolerability to make an informed decision about treatment approach.
Presentation numberPS3-03-21
Incidence and Survival of Inflammatory Breast Cancer: A 10-Year Single-InstitutionRetrospective Analysis in Riyadh Saudi Arabia.
Sadal REFAE, King Abdulaziz Medical city – Minsistry of national Guard, Riyadh, Saudi Arabia
S. REFAE, mohammed AlKAIYAT, Haitham ARABI, Joud ALQAHTANI, Mohammed ALGARNI, Adel AlRashdi, Kanaan Al-Shammari, Alaa Al Zare, Nada ALSUHAIBANI, Nafisa Abdulhafiz; King Abdulaziz Medical city – Minsistry of national Guard, Riyadh, Saudi Arabia
Backgound: Inflammatory breast cancer (IBC) is a rare but aggressive primary breast carcinoma that accounts for approximately 5% of all breast carcinomas and is characterized by erythema, edema, and “peau d’orange“. We assessed the incidence of IBC in younger age groups and its association with worse outcomes than non-inflammatory breast cancer. Despite the availability of multimodal treatment options for IBC, there is an unmet need for information on the incidence and outcomes of IBC in a sufficiently large and diverse population, specifically in the Gulf region of the Middle East. Herein, we retrospectively explored the incidence and survival of IBC at a single institution in Saudi Arabia over 10 years Method: We conducted a retrospective analysis of patients diagnosed with breast cancer between 2015 and 2025 at our institution. Initial screening was performed to identify patients with inflammatory breast cancer (IBC). For patients diagnosed with IBC, we collected variables related to patient demographics, disease characteristics, treatment modalities, and survival. Results: A total of 1,806 cases were screened, of which 43 were diagnosed with IBC, yielding an incidence of 2.4%. The median age of the patients with IBC was 48.9 years (SD = 13.1). The majority were female (n = 42, 97.7%) and Saudi nationals (n = 41, 95.3%). Approximately 60% of the patients were premenopausal. The most prevalent pathological diagnosis was invasive ductal carcinoma (IDC), which accounted for 88.4% of cases. Nearly half of the patients (46.5%) presented with metastatic disease. Hormone receptor-positive tumors were observed in 46.5% of cases, HER2-positive tumors in 30% (n = 13), and HER2-enriched tumors in 16.3% (n = 7). Triple-negative breast cancer (TNBC) was identified in 37.2% (n = 16) of the patients. Only two patients carried BRCA1 mutations. Most patients received chemotherapy (86%), 51.2% underwent surgery, and 41.9% received radiotherapy. All patients with HER2-positive disease (n = 13, 100%) received anti-HER2 therapy. Immunotherapy and CDK4/6 inhibitors were administered to 11.6% and 13.6% of the patients, respectively. At the time of data analysis, 65.1% (n = 28) of the patients were alive. The overall median survival was 49.2 months (95% CI [18.3, 80.2]). The median survival for TNBC cases was 20 months (95% CI [1.0, 73.9]) compared to 49.2 months (95% CI [26.4, 72.0]) for non-TNBC cases; however, this difference was not statistically significant (p =.43).Conclusion: inflammatory breast cancer IBC incidence is 2.4 % over 10 years compared to 2-4% of all breast cancer subtype in the United States. IBC is an aggressive form of breast cancer with a poor prognosis. This may be potentially worsened by the triple-negative subtype. Multicenter studies are needed to better assess IBC’s prevalence and prognostic factors of IBC to improve patient outcomes.
Presentation numberPS3-03-29
Modeling of BRCA1/2 Breast Cancers Using iPSC Derived 3D Human Organoids Platform
Nur Yucer, University of Texas Health Science Center San Antonio, san antonio, TX
N. Yucer1, A. Okim1, S. Dhungana1, D. Bacich1, K. Lawrenson1, X. Cui2, S. Gayther1; 1University of Texas Health Science Center San Antonio, san antonio, TX, 2Cedars-Sinai Medical Center, Los Angeles, CA
BRCA1 and BRCA2 mutations (BRCA1mut, BRCA2mut) increase the risk of breast cancer (BC) in women. The cumulative risk of developing breast cancer is estimated to be between 45-75% for carriers of BRCA1mut and 41-70% for carriers of BRCA2mut, compared to a population risk of about 13% in the USA. Although BRCA1 and BRCA2 were identified as breast cancer susceptibility genes nearly 30 years ago, many aspects of their functional roles in the development of specific types of breast cancer remain poorly understood. Both genes play a crucial role in repairing DNA double-strand breaks, but the types and characteristics of breast cancer associated with each gene differ. Patients with BRCA1muttypically develop basal-like triple-negative breast cancer (TNBC), which is associated with poor survival rates. In contrast, patients with BRCA2mut are more likely to develop luminal-like estrogen receptor-positive (ER+) breast cancer. To date, no applicable human breast tissue model systems are available for studying preneoplastic changes and neoplastic transformation elicited by variations in the underlying genetic and/or microenvironment risk factors, particularly for BRCA carriers. In this study, we aimed to elucidate the effects of germline BRCA1mut and BRCA2mut on breast cancer development. We also sought to identify the hormonal and somatic genetic factors that influence the initiation and progression of breast cancer in individuals with these mutations. To achieve this, we utilized a patient-specific, induced pluripotent stem cell (iPSC)-derived 3D mammary organoid model developed in our lab. iPSC technology and 3-dimensional (3D)-tissue engineering have provided opportunities to model human disease in vitro. This model system can self-renew and differentiate into multiple lineages and intrinsically self-organize to form 3D tissue architecture. Most importantly, tissue-derived iPSC from a specific individual harbors/retains both the genetic mutation and the whole genetic background of the patient. Several iPSC-derived, inherited disease models have been used to reproduce genetic mutation-associated high-risk cancers. These studies have revealed disease pathogenesis and carcinogenesis-initiating events in relevant human cell types. To model the risk of developing breast cancer associated with BRCA1/2 pathogenic variants, we generated three different BRCA1mut and BRCA2mut female iPSC cell lines and developed an iPSC-derived 3D mammary gland epithelium organoid model. Our initial study found that following differentiation into mammary gland organoids, both BRCA1mutand BRCA2mut heterozygous mutation carriers conferred a neoplastic phenotype reminiscent of a ductal carcinoma in situ(DCIS), a proposed precursor of BC (i.e., organoid models retained the wildtype copy of BRCA1 or BRCA2) compared to BRCA wildtype (BRCA1/2WT) controls, which retained both copies of both genes (confirmed by whole genome sequencing). Importantly, the development of DCIS in BRCA2mut carriers is dependent on estrogen (E2) exposure, while in BRCA1mut carriers, it is independent of hormonal influences. This indicates that BRCA haploinsufficiency contributes to the observed phenotype in both BRCA1mut and BRCA2mut carriers. Moreover, BRCA2mut are more likely to lead to estrogen receptor-positive (ER+) breast cancer, while BRCA1mut are associated with a higher risk of developing TNBC. Currently, we are expanding the BRCA1/2 iPSC lines cohort to generate mammary gland organoids with different BRCApathogenic variants and additional somatic alterations, aiming to estimate risk scores in developing BRCA1mut versus BRCA2mut breast cancer subtypes.
Presentation numberPS3-05-24
Sex Hormone Responses to Alternate-Day Fasting-Low-Carbohydrate in Premenopausal and Postmenopausal Women
Faiza Kalam, The Ohio State University Comprehensive Cancer Center – James, Division of Cancer Prevention & Control, Department of Internal Medicine, Columbus, OH
F. Kalam1, R. Akasheh1, F. Tounkara2, M. Hafez3, K. Varady4, T.-Y. Cheng1; 1The Ohio State University Comprehensive Cancer Center – James, Division of Cancer Prevention & Control, Department of Internal Medicine, Columbus, OH, 2Department of Biomedical Informatics (Biostatistics & Population Health), College of Medicine, The Ohio State University, Columbus, OH, 3St. Luke’s University Health Network, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 4Department of Kinesiology & Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL
Background: Intermittent fasting is increasingly used to manage weight and metabolic disease. Alternate-day fasting (ADF) combined with low carbohydrate (ADF-LC) lowers body weight and insulin. Because insulin suppresses hepatic sex hormone-binding globulin (SHBG), ADF-LC is expected to increase SHBG without increasing free testosterone (free T); the adrenal androgen dehydroepiandrosterone (DHEA) may also shift with metabolic changes. We tested the impact of an ADF-LC intervention on insulin and androgen-related hormones and evaluated whether changes in these markers (SHBG, free T, DHEA) tracked with body weight change, and whether hormone levels and menopausal status were associated with body weight over 24 weeks. Methods: Women with obesity completed a single-arm, 24-week ADF-LC intervention (12-week loss; 12-week maintenance). Fasting morning body weight and SHBG, free T, DHEA, and insulin were assessed at weeks 0, 12, and 24. Analyses included women (n=28; premenopausal n=13; postmenopausal n=15). We ran change-on-change regressions of Δweight on ΔSHBG, ΔDHEA, Δfree T, and Δinsulin (univariable; multivariable with age and height; plus models adding menopausal status and the interactions ΔSHBG×menopause and Δinsulin×menopause). We also fit a random-intercept mixed model for weight over time including time, SHBG, DHEA, free T, insulin, menopausal status, age, height, and time×hormone interactions. Estimates are reported as β (95% CI, p). Results: Participants lost weight over 24 weeks intervention (p<0.01). In the mixed-effects model, body weight decreased over time (β=−4.7; 95% CI -6.3, -3.0). Higher SHBG was associated with lower body weight (β=-5.8; -11.0, -0.99), and higher insulin with higher body weight (β=7.8; 3.7, 12.0). DHEA, age, and menopausal status were not significant (e.g., DHEA β=0.02; −2.6, 2.7). A time×free T interaction was significant (β=-4.7; -9.1, -0.30), indicating greater weight loss over time at higher testosterone. In change-on-change analyses, insulin change was positively associated with weight change (univariable β=0.76; 0.13, 1.4; p=0.019; multivariable β=0.80; 0.15, 1.5; p=0.018; model including menopause β=0.97; 0.09, 1.9; p=0.033), whereas changes in SHBG, DHEA, and free T were not significant; ΔSHBG×menopause and Δinsulin×menopause were non-significant. Conclusions: Over 24 weeks, ADF-LC was associated with weight loss. In the models, higher SHBG related to lower body weight, higher insulin to higher body weight, and larger insulin reductions tracked with greater weight loss; DHEA, age, and menopausal status were not determinants. A time×testosterone interaction indicated greater weight loss at higher testosterone. Overall, no adverse androgenic signal was observed. These findings support testing ADF-LC in larger, controlled trials and mechanistic mediation along insulin-SHBG pathways relevant to breast-cancer prevention.
Presentation numberPS3-05-25
A blood test for personalized breast and lung cancer risks: findings from the MD Anderson MERIT study
Johannes Fahrmann, UT MD Anderson Cancer Center, Houston, TX
J. Dennison, E. Irajizad, O. Weaver, E. Ostrin, J. Fahrmann, J. Vykoukal, J. Leung, H. Khoshfekr Rudsari, S. Khoramisarvestani, N. Kettner, S. Hanash; UT MD Anderson Cancer Center, Houston, TX.
Background: The MERIT cohort (Mammography, Early Detection, Risk Assessment, and Imaging Technologies) has enrolled 8,000 participants undergoing annual screening mammography at MD Anderson Cancer Center since 2017. The goal is to integrate clinical and imaging data with blood-based biomarker profiles to estimate risk of developing breast and other cancers. Annual blood collection enables evaluation of longitudinal algorithms for changes in biomarker levels. Here we report the performance of prespecified blood biomarker algorithms for assessing the risk of harboring or developing breast and lung cancers within one year of blood draw. Methods: Biomarker combination rules and decision thresholds were prespecified and locked. For longitudinal modeling, a parametric empirical Bayes (PEB) approach was applied to serial biomarker data to refine risk estimates. Results: Biomarker levels were assayed in 1,762 non-cancer controls, 221 breast cancer cases, and 21 lung cancer cases. Limiting the analyses to samples collected within one year of diagnosis included 104 breast cancer cases and 12 lung cancer cases. For breast cancer, using a 1.67% 5-year risk threshold (the Gail model “elevated-risk” cut-point), the biomarker panel classified an additional 13% of women with breast cancer at elevated risk who were missed by the Gail model. Incorporating 120 serial samples and applying the longitudinal algorithm further improved risk classification of breast cancer by an additional 9% without increasing the false-positive rate. For lung cancer, the biomarker panel achieved 93.0% specificity (95% CI: 89.0 to 96.0) and 42.0% sensitivity (95% CI: 33.0 to 50.0) at a prespecified 1% 6-year risk threshold, comparable to the U.S. Preventive Services Task Force eligibility criteria for low-dose CT (LDCT) screening. Among never-smokers, the panel classified 40% of women who developed lung cancer as elevated risk at 90% specificity. Incorporating 59 serial samples and applying the longitudinal algorithm resulted in a further 7% improvement in classification at fixed specificity. Conclusions: A blood biomarker panel for breast cancer risk assessment identified women at increased risk who were missed by the Gail model. A blood biomarker panel for lung cancer identified women at increased risk for lung cancer, including never-smokers who may benefit from LDCT screening. Longitudinal blood collection and application of PEB further improved risk assessment without increasing false positives, supporting the utility of blood testing for breast and lung cancer risks in the context of mammography screening.
Presentation numberPS3-05-26
Triple incretin receptor agonist induced weight loss delays tumor latency in obesity-associated breast cancer
Naveed Pervaiz, The University of Tennessee Health Sceience Center, Memphis, TN
N. Pervaiz1, S. J. Marathe1, Z. Powell1, L. G. McGrath1, Z. T. Mustafa1, L. Wang2, M. C. Playdon2, S. A. Summers2, J. Hao3, B. Li3, J. F. Pierre4, L. Makowski1; 1The University of Tennessee Health Sceience Center, Memphis, TN, 2The University of Utah, Salt Lake City, UT, 3The University of Iowa, Iowa City, IA, 4The University of Wisconsin-Madison, Madison, WI
Objective: To evaluate the impact of GLP-1R/GIPR/GCGR agonist-associated weight loss on breast cancer (BC) risk and tumor progression. Background: BC is the second leading cause of cancer death among women in the US. Obesity increases the risk of multiple cancers, including BC. Previous pre-clinical studies demonstrated that GLP-1 receptor agonists promote weight loss, improve metabolic dysfunction, reduce cancer risk and blunt progression in pancreatic and lung cancer, but effects on spontaneous BC were unknown. Hypothesis: To assess the impact of a novel triple incretin receptor agonist (TIRA) on BC risk and tumor progression in mice with obesity-associated metabolic dysfunction. Methods and Approach: Female FVB C3(1)-T antigen (“C3Tag”) mice, a genetically engineered mouse model (GEMM), which develop BC spontaneously, were utilized to evaluate effects of medical weight loss on tumor risk and outcomes. Overweight mice were subjected to: 1) vehicle (VEH) control to maintain body weight; 2) TIRA as weight loss intervention; or 3) caloric restriction to allow for weight matched (WM) controls. Mice were fed a high-fat diet from 8 weeks of age. After gaining weight, they received subcutaneous TIRA (15 nmol/kg) or vehicle every other day. One group was sacrificed at 15 weeks to investigate preneoplastic lesions in the mammary gland. The second group was allowed to proceed to endpoint to examine tumor latency, progression, and survival. N=10-12 per group. Gut microbiome and circulating mediators were quantified by proteomics and metabolomics. One way ANOVA was used. Results: TIRA and WM induced significant body weight loss (~15-20%) within 2-3 weeks, followed by weight stabilization, with reduced circulating leptin concentration compared to VEH. TIRA and WM also reduced 4-hour fasting blood glucose, decreased gonadal white adipose tissue, and mammary fat pad mass. Moderately reduced muscle mass was observed in TIRA and WM mice in the preneoplasia group but not in the long-term survival group. TIRA delayed gastric emptying revealed by higher cecal contents compared to VEH and WM in the preneoplasia group. TIRA-induced ceramide, metabolite, and microbiome changes were evident. In the survival group, TIRA significantly delayed tumor latency compared to VEH. Conclusion: TIRA and WM effectively induced weight loss and improved metabolic dysfunction with both interventions demonstrating significantly reduced adiposity, hyperglycemia, and tumor-promoting adipokines. TIRA-associated weight loss significantly delayed tumor onset whereas WM did not, suggesting a TIRA-specific anti-tumor effect. Results demonstrate that pharmacological weight loss suppressed early tumor progression – despite being in a transgenically driven GEMM – and improved outcomes in obesity-associated BC.
Presentation numberPS3-05-28
Landscape of homologous recombination repair-related gene mutation and homologous recombination deficiency in Chinese patients with HER2-low early breast cancer
Chang Gong, Sun Yat-sen Memorial Hospital, Guangzhou, China
R. Hu, Y. Xia, Q. Lin, W. Zhang, J. Chen, Y. Zhu, Y. Yang, Q. Lin, Y. Quan, H. Liu, C. Gong; Sun Yat-sen Memorial Hospital, Guangzhou, CHINA.
Background: Human epidermal growth factor receptor 2 (HER2)-low (immunohistochemistry [IHC] 1+ or IHC 2+ with fluorescent in situ hybridization [FISH]-negative) early breast cancer (EBC) constitutes a significant proportion of breast malignancies. Genomic instability, particularly defective DNA repair, is a recognized driver of tumorigenesis. Homologous recombination repair (HRR)-related gene mutation leading to homologous recombination deficiency (HRD) increases risks for tumorigenesis and has been proved as a viable therapeutic target in ovarian cancer and triple-negative breast cancer, with therapies such as poly (adenosine diphosphate-ribose) polymerase inhibitors and carboplatin. However, current evidence is largely derived from studies in European and American populations. Therefore, clinical research focusing on Chinese patients with HER2-low EBC is warranted.Methods: This prospective, single-center, observational study (NCT05466786) aimed to enroll 255 treatment-naïve patients with operable primary HER2-low (IHC 1+ of IHC 2+/FISH-negative) invasive EBC, who had completed HRR-related gene mutation and HRD assessments using a customized next-generation sequencing panel targeting 520 cancer-related genes. The HRD score was derived from the combined assessment of three genomic scar signatures: loss of heterozygosity, telomeric allelic imbalance, and large-scale state transitions. HRD positivity was defined as an HRD score ≥42 and/or the presence of a BRCA1/2 mutation. All enrolled patients received standard therapies according to the National Comprehensive Cancer Network guidelines. The primary outcome was to characterize the landscape of HRR-related gene mutations and HRD.Results: From August 2022 to September 2025, 129 of 255 planned patients were enrolled. The median age of the patients was 43 years (range 27-68), and 38 (30%) had family history of malignant tumor. Tumors were predominantly T1-2 (112 [87%]), node-negative (74 [67%]), and stage II-III (95 [74%]); 103 (80%) were hormone receptor (HR)-positive (estrogen receptor ≥10%), and 102 (79%) had Ki67 expression ≥20%. Among HRR-related gene mutations, BRIP1 (17 [13%]), NBN (11 [9%]), PALB2 (8 [6%]), BRCA 2 (8 [6%]), BRCA 1 (6 [5%]), ATM (4 [3%]), and ATR (4 [3%]) were most common. The median HRD score was 20 (Interquartile range 10-35), and 26 (20%) patients were HRD-positive (including 14 had gBRCA1/2 mutation). HRD-positive status was significantly more common in tumors with histological grade-III, HR-negative, or Ki67 expression ≥20%. Among the 520 cancer-related genes, somatic alterations were frequently observed in PIK3CA (58 [45%]), TP53 (48 [37%]), GATA3 (34 [26%]), MYC (22 [17%]), CCND1 (21 [16%]), FGF3 (20 [16%]), FGF4 (20 [16%]), and FGF19 (20 [16%]). Recurrent germline alterations included BRCA2 (8 [6%]), BRCA1 (6 [5%]), PALB2 (5 [4%]), ATM (2 [2%]), BLM (1 [1%]), and MSH3 (1 [16%]).Conclusions: This study delineates the landscape of HRR-related gene mutation and HRD in Chinese patients with HER2-low EBC. HRD-positivity was significantly associated with histological grade III, HR-negative status, and Ki67 expression ≥20%. Further enrollment is warranted to provide a more comprehensive representation of HRR-related genomic alterations and HRD in this patient population.
Presentation numberPS3-07-23
Survivor plots for quantitated adjunctive statistically standardized ER, PgR, and HER2 in adjuvant postmenopausal breast cancer: Canadian Cancer Trials Group MA.27 trial of exemestane versus anastrozole
Judith-Anne W Chapman, Queen’s University, Kitchener, ON, Canada
J. W. Chapman1, J. Bayani2, S. SenGupta2, J. M. S. Bartlett3, T. Piper3, M. A. Quintayo2, S. Virk4, P. E. Goss5, J. N. Ingle6, M. J. Ellis7, G. W. Sledge8, G. T. Budd9, M. Rabaglio10, R. H. Ansari11, R. Tozer12, D. P. D’Souza13, H. Chalchal14, S. Spadafora15, V. Stearns16, E. A. Perez17, K. A. Gelmon18, T. J. Whelan12, C. Elliott4, L. E. Shepherd4, B. E. Chen4, K. J. Taylor3; 1Queen’s University, Kitchener, ON, CANADA, 2University of Toronto, Toronto, ON, CANADA, 3University of Edinburgh, Edinburgh, UNITED KINGDOM, 4Queen’s University, Kingston, ON, CANADA, 5Harvard, Boston, MA, 6Mayo Clinic, Rochester, MN, 7Guardant Health, Palo Alta, CA, 8Caris Life Sciences, Irving, TX, 9Cleveland Clinic, Cleveland, OH, 10Inselspital Bern, Bern, SWITZERLAND, 11Indiana School of Medicine, South Bend, IN, 12McMaster University, Hamilton, ON, CANADA, 13London Regional Health Science Centre, London, ON, CANADA, 14Alan Blair Cancer Center, Regina, SK, CANADA, 15Algoma Regional Cancer Centre, Sault Ste Marie, ON, CANADA, 16Weill Cornell Medicine, New York, NY, 17Mayo Clinic, Redwood City, CA, 18University of British Columbia, Vancouver, BC, CANADA.
Background: Survivor plots permit adjustment by baseline demographics and clinical characteristics; log-normal survival analysis may be more robust than a Cox model under departures from model assumptions. We depicted distant disease-free survival (DDFS) for statistically standardized quantitated ER, PgR, and HER2. Methods: We utilized CCTG MA.27 (NCT00066573), an adjuvant phase III trial of exemestane versus anastrozole in postmenopausal women with ER+ and/or PgR+ tumours. IHC ER, PgR, and HER2 HSCORE were centrally assessed by machine image quantitation, and statistically standardized to mean of 0, standard deviation (SD) of 1 following Box-Cox variance stabilization transformations of square for ER, natural logarithm (ln) and square root for PgR, and ln for HER2. For the ln transformation, 0.1 was added to 0 HSCOREs for a stable transformation. The primary endpoint was DDFS at the longest trial follow-up of 4.1 years. For log-normal survival analysis, ln(time) is a linear function of covariates and had a normal distribution; the Cox models were log-linear for covariates. Survival was depicted with log-normal and Cox survivor plots adjusted at the means of age, T and N stage, grade, lymphovascular invasion, treatment, and baseline patient demographics. We examined ER and PgR ASCO/CAP cut-points at (0; >0), HER2 cut-points at (IHC 0, 0 stain; IHC 0, (0,10%] stain; IHC 1+; IHC 2+; IHC 3+), and ER/PgR/HER2 standardized cut-points at standard deviations about mean of 0 (<-1; (-1,0]; (0,1]; >1). P was based on the likelihood ratio criterion (~Χ2(1)) for the addition of ER/PgR/HER2 to the model of baseline covariates, and had nominal significance if P<0.05. Log-normal and Cox multivariable regressions with unstandardized and standardized categorical, and continuous, ER, PgR, and HER2 were adjusted by baseline covariates; 2-sided Wald tests had nominal significance if P<0.05. Results: Of 7576 MA.27 patients, 2325 patients had quantitated image analysis assessments for ER, PgR, and HER2; 113 of 2325 (4.9%) patients experienced a DDFS event during follow-up. Multivariable log-normal survival analysis indicated DDFS was significantly longer for patients with T1 (p=0.01), N0 (p=.002), and grade 1/2 tumors (p=0.01); similar significance levels were found with a Cox model. Log-normal survivor plots were smooth, while Cox survivor plots were step-wise discontinuous at events. Adjusted log-normal survivor plots indicated similar 5-year DDFS for patients by standardized ER categories (p=0.43): 95.4% for those with ER <-1.0 (n=401); 95.8%, for (-1.0,0.0] (n=737); 96.1%, for (0.0,1.0] (n=737); and 96.4%, for >1.0 (n=450). The log-normal standardized PgR survivor plots indicated patients had significantly different 5-year DDFS (p<.001) by categories, with both ln and square root transformations of HSCORE: with a ln transformation, 93.7% for <-1.0 (n=607); 95.4%, for (-1.0,0.0] (n=390); 96.8%, for (0.0,1.0] (n=826); and 97.8%, for >1.0 (n=502). Patients had similar 5-year DDFS by standardized HER2 categories (p=0.71): 96.2% for <-1.0 (n=83); 96.0%, for (-1.0,0.0] (n=1201); 95.9%, for (0.0,1.0] (n=527); and 95.7%, for >1.0 (n=514). In full fit multivariable log-normal survival analyses, DDFS was not significantly different for ER (p=0.51-0.89) and HER2 (p=0.12-0.98); higher PgR was associated with significantly better DDFS (p=0.01 to <.001). Adjusted Cox survival analyses had similar results. Conclusions: Survivor plots adjusted for baseline demographic and clinical characteristics provided similar indications of significance for standardized ER, PgR, and HER2 as those seen in multivariable models with both log-normal and Cox survival analyses. Higher PgR was associated with significantly better DDFS.
Presentation numberPS3-06-12
Pre-existing TOP2A-High Cells Drive Resistance to Trastuzumab Deruxtecan in HER2+ Breast Cancer
Se-eun Han, Wittgen Biotechnologies, Berkeley, CA
S. Han1, M. Jung1, J. Lee2, S. Rhie1; 1Wittgen Biotechnologies, Berkeley, CA, 2Preclinical Core, Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, HI.
Background: Trastuzumab deruxtecan (T-DXd) has significantly improved outcomes for patients with HER2-positive (HER2+) breast cancer. However, acquired resistance remains a critical clinical challenge. The underlying mechanisms driving this resistance are not fully understood. Emerging evidence suggests that resistance may arise from pre-existing tumor cell heterogeneity rather than being acquired de novo. Among candidate pathways, dysregulation of DNA replication and repair has been implicated in therapeutic resistance. Topoisomerase II alpha (TOP2A), a key enzyme in DNA topology and replication, is frequently overexpressed in aggressive breast cancers and associated with poor prognosis and treatment resistance. Building on this concept, we hypothesized that a subpopulation of resistant-like cells exists within tumors prior to treatment with T-DXd and is characterized by a distinct molecular signature involving TOP2A. Methods: We performed whole transcriptome analysis to compare gene expression profiles of T-DXd-resistant (TDXd-R) cells against their parental counterparts. We then analyzed public single-cell RNA sequencing (scRNA-seq) datasets from 17 HER2+ breast cancer cell lines and 8 HER2+ patient tumors to identify pre-existing cellular populations. Bioinformatics analyses included differential gene expression, gene set enrichment analysis (GSEA), gene regulatory network (GRN) analysis, and drug-gene database (DsigDB) screening to predict synergistic therapies. Results: Transcriptomic analysis revealed that HER2+ TDXd-R cells consistently exhibited significant upregulation of TOP2A expression and E2F signaling. GSEA confirmed activation of pathways related to the cell cycle, DNA replication, and genomic stability in the TDXd-R cell lines. Notably, scRNA-seq analysis demonstrated that a distinct TOP2A+ HER2+ cell cluster was present in all examined HER2+ cell lines and patient tumors prior to T-DXd exposure. With GRN analysis, we found these pre-existing putative resistant cells were characterized by high activity of E2F signaling pathways, involved in S/G2-M cell cycle progression and DNA synthesis. Based on this molecular signature, drug prediction analysis identified potential synergistic agents, including the topoisomerase inhibitor lucanthone, as candidates to target T-DXd resistant population. Conclusion: Our findings suggest that resistance to T-DXd in HER2+ breast cancer is driven by the selection and expansion of a pre-existing cancer cell subpopulation defined by high TOP2A expression and its elevated E2F-associated regulon activity. This molecular signature may serve as a biomarker for innate resistance. Furthermore, targeting this population with T-DXd and synergistic agents such as lucanthone could be a promising therapeutic strategy to overcome or prevent resistance to T-DXd and improve patient outcomes.
Presentation numberPS3-12-14
Targeting the Tumor Secretome to Enhance Immune Responses in Breast Cancer Liver Metastasis
Michelle M. Williams, University of Pittsburgh, Pittsburgh, PA
A. Phan, L. McCue, T. Hintelmann, M. M. Williams; University of Pittsburgh, Pittsburgh, PA.
Background: Nearly half of metastatic breast cancers spread to the liver, a metastatic site that responds poorly to immune checkpoint blockade. Although liver metastases are considered immunologically cold and this is thought to drive resistance, few studies investigate heterogeneity in the liver metastatic microenvironment. This gap in knowledge limits our understanding of the mechanisms driving therapy resistance. Objective: We aimed to characterize the heterogeneity of immune phenotypes in breast cancer liver metastases (BCLM) and postulated that comparison of immune hot and cold BCLM would reveal factors produced and secreted by the tumor to support an immune cold phenotype. Methods: We profiled a cohort of 20 human BCLM specimens using the NanoString nCounter system. Abundance of immune cells and secreted factors of interest were confirmed using multispectral fluorescence (mIF markers: PanCK+/tumor, CD4+ or CD8+/T cell, CD68+/macrophage, CD66b+/neutrophil) and immunohistochemistry (IHC). Similar approaches were conducted in a validation cohort of 19 BCLM. Single cell mRNA sequencing (scRNAseq) from 6 BCLM from GSE249361 were accessed and analyzed using CellChat to reveal differential communication between immune cells and AGR2-negative versus AGR2-expressing tumor cells. 4T1 or 66Cl-4 mouse mammary carcinoma spheroids were co-cultured with activated CD8+ splenocytes in the presence and absence of a secreted factor of interest, recombinant AGR2 (rAGR2). T cell distance from the center of spheroids was quantified using ImageJ. Three BCLM patient derived organoids (PDOs) were developed by the Hope for OTHERS rapid autopsy program at the University of Pittsburgh/UPMC Hillman Cancer Center. AGR2 levels were assessed in PDOs via IF. Results: nCounter analysis of 20 BCLM revealed that liver metastases lie across a spectrum of immune infiltration. Confirmatory multispectral fluorescence demonstrated that nearly half of the tumors (n=11) had abundant expression of macrophages and T cells, suggesting that they could be primed for response to checkpoint inhibitors. We compared differentially expressed genes between the most immune hot (n=5) and cold (n=5) metastases to reveal essential factors preventing immune infiltration in cold tumors. The most highly expressed gene in immune cold compared to immune hot BCLM was anterior gradient-2 (AGR2), a tumor secreted factor that correlated negatively with immune cell abundance in breast tumors. In a validation cohort of 19 BCLM, we confirmed that low AGR2 expression predicted a 2-fold increase in CD8+ T cell density. Datamining a scRNAseq dataset revealed that AGR2 is heterogeneously expressed in the tumor compartment. Using CellChat we showed that tumor cells lacking AGR2 have enhanced communication with CD8+ T cells through the MHC-I pathway, suggesting that AGR2 may dampen tumor antigen presentation. Alternatively, tumor cells expressing AGR2 have increased communication with several members of the tumor microenvironment through fibronectin signaling, revealing that tumor AGR2 may also enhance extracellular matrix dynamics. Preliminary mechanistic studies revealed that treatment with recombinant AGR2 decreases CD8+ splenocyte penetration into spheroids by 30%. Finally, AGR2 levels were heterogeneous in BCLM PDOs and they corresponded with the expression of AGR2 and the immune status in the original tumor. Conclusions: These findings suggest that BCLM produce and secrete AGR2 to limit T cell infiltration and activation. We are currently developing a platform with the BCLM PDOs to assess the impact of AGR2, and additional secreted factors, on immune cell responses using matching peripheral blood. We expect that these findings will support further development of AGR2 blocking antibodies as a companion or alternative immunotherapy approach in metastatic breast cancer.
Presentation numberPS3-06-14
Interpretable Graph Learning on Preoperative Biopsy Predicts Pathological Complete Response to Neoadjuvant Therapy in Breast Cancer
Guojun Zhang, The Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital & Peking University Cancer Hospital Yunnan, Kunming, China
W. Hou1, Z. Pu2, Z. Xu3, A. Wu1, Z. Liu1, K. Zhao4, C. Duan1, J. Guo5, K. Chen6, S. Qiu7, Z. Du8, X. Zhao8, J. Bai1, H. Zeng9, G. Zhang1; 1The Third Affiliated Hospital of Kunming Medical University & Yunnan Cancer Hospital & Peking University Cancer Hospital Yunnan, Kunming, CHINA, 2School of Intelligent System Engineering, Sun Yat-sen University, Guangzhou, CHINA, 3Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Peking University Cancer Hospital Yunnan, Kunming, CHINA, 4Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, CHINA, 5Department of Radiology, First Affiliated Hospital of Kunming Medical University, Kunming, CHINA, 6Sun Yat-Sen Memorial Hospital, Guangzhou, CHINA, 7Clinical Research Center, Shantou Central Hospital, Shantou, CHINA, 8Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, CHINA, 9The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, CHINA.
Predicting neoadjuvant therapy (NAT) response in breast cancer patients still remains challenging due to the high heterogeneity of tumors and modality differences in medical data. In this paper, we proposed a novel graph learning framework, a clinicopathological-guided spatial-variant network (CSV-Net), which integrates whole-slide images (WSIs) from biopsy samples with clinicopathological (CP) variables to predict pathological complete response (pCR) of patients receiving NAT. CSV-Net incorporates two novel components: (1) a spatial-variant graph convolution (SV-GraphConv) layer, which models the spatial-semantic heterogeneity inherent in whole-slide images (WSIs), and (2) a clinicopathological-guided graph pooling (CG-GraphPool) module that dynamically integrates clinicopathological (CP) variables with WSI features at a fine-grained level, enhancing both performance and interpretability. We evaluated CSV-Net on a retrospective dataset of 950 breast cancer patients from 5 medical centers. CSV-Net achieved ROC-AUCs of 0.845 (95% CI: 0.801-0.886) and 0.815 (95% CI: 0.755-0.873) in predicting pCR on the internal and external validation sets, respectively, outperforming state-of-the-art methods. Notably, subgroup analysis highlighted CSV-Net’s robust performance in patients with different molecular subtypes (e.g. Luminal, HER2 overexpression and TNBC), underscoring its clinical utility. Furthermore, disease-free survival relevance and transcriptomic profiling experiments confirmed the model’s prognostic value and biological interpretability, suggesting potential applications in personalized treatment planning and biomarker discovery.
Presentation numberPS3-09-08
Five-year analysis of distant disease-free survival (DDFS) across key subgroups from the phase 3 NATALEE trial of ribociclib (RIB) plus a nonsteroidal aromatase inhibitor (NSAI) in patients with HR+/HER2− early breast cancer (EBC)
Sara Hurvitz, Univ Washington, Seattle, WA
S. Hurvitz1, M. Jarzab2, A. Ring3, P. Sharma4, I. Temciuc5, H. Hu6, M. Akdere7, J. Pablo Zarate6, D. Yardley8; 1Univ Washington, Seattle, WA, 2Maria Sklodowska-Curie National Research Institute of Oncolog, Gliwice, Poland, 3Royal Marsden Hospital NHS Foundation Trust, Surry, United Kingdom, 4University of Kansas Medical Center, Westwood, KS, 5Translational Research in Oncology, Edmonton, AB, Canada, 6Novartis Pharmaceuticals Corporation, East Hanover, NJ, 7Novartis Pharma AG, Basel, Switzerland, 8Sarah Cannon Research Institute, Nashville, TN
Background: Despite improvements in outcomes in patients with HR+ EBC, distant recurrence remains a major concern given that there is no cure for metastatic breast cancer. The NATALEE trial has shown invasive disease-free survival and DDFS benefits with RIB + NSAI in patients with stages II and III HR+/HER2− EBC. We present updated DDFS data from the prespecified 5-y landmark analysis of NATALEE across clinically relevant subgroups. Methods: In NATALEE, patients were randomized 1:1 to receive either RIB (400 mg/d, 3 wk on/1 wk off for 3 y) + NSAI (anastrozole 1 mg/d or letrozole 2.5 mg/d for ≥5 y) or NSAI alone; men and premenopausal women also received goserelin. Patients with anatomic stage IIA (either node-negative [N0] with additional risk factors or N1 [1-3 axillary lymph nodes]), IIB, or III disease per AJCC (8th edition) were included in the trial. DDFS, a secondary endpoint, was defined as the time from randomization to the first event of distant recurrence, second primary non-breast invasive cancer (except for basal and squamous cell skin carcinomas), or death from any cause. Kaplan-Meier analysis was used to estimate survival rates at each time point, and a Cox proportional hazards model was applied to calculate hazard ratios (HRs) for RIB + NSAI vs NSAI alone, stratified based on study stratification factors. DDFS was analyzed across clinically relevant subgroups, including anatomic stage and nodal status Results: At the updated data cutoff of May 28, 2025, with a median duration of follow-up for DDFS of 55.5 months, RIB + NSAI continued to show a DDFS benefit (HR, 0.709 [95% CI: 0.608-0.827]; nominal 1-sided P<.0001) and a distant recurrence-free survival benefit (HR, 0.699 [95% CI: 0.594-0.824]; nominal 1-sided P<.0001) over NSAI alone in the intent-to-treat population. The most common sites of distant recurrence were bone, liver, lung/pleura, and lymph nodes with fewer events observed with RIB + NSAI vs NSAI alone across all sites. The DDFS benefit was observed irrespective of anatomic stage (stage IIA [n=1001]: HR, 0.374 [95% CI: 0.218-0.644]; stage IIB [n=1045]: HR, 0.863 [95% CI: 0.580-1.285]; stage IIIA [n=1830]: HR, 0.735 [95% CI: 0.569-0.951]; stage IIIB [n=317]: HR, 0.644 [95% CI: 0.390-1.063]; stage IIIC [n=892]: HR, 0.785 [95% CI: 0.594-1.037]). Likewise, a consistent improvement in DDFS with RIB + NSAI vs NSAI alone was observed irrespective of nodal status (N0 [n=614]: HR, 0.539 [95% CI: 0.318-0.913]; node-positive [N+] [n=4479]: HR, 0.723 [95% CI: 0.615-0.849]). The DDFS benefit with RIB + NSAI vs NSAI alone was similar across other clinically relevant subgroups, which included subgroups based on menopausal status, age, prior endocrine therapy duration, and Ki-67 status. Conclusions: In this prespecified 5-y analysis, with all patients off RIB treatment for a median of 2 y, the DDFS benefit with RIB + NSAI was sustained or improved compared with prior NATALEE analysis. This benefit was observed across all key subgroups, including N0 disease. These findings support the use of adjuvant RIB + NSAI to reduce distant recurrence risk in the NATALEE-eligible high-risk HR+/HER2− EBC population.
Presentation numberPS3-06-15
A Retrospective Study on a Predictive Model to Probe the Impact of Atmospheric Environment on Breast Cancer Prognosis: A Cohort of 17,438 Patients
Yunqin Yuan, School of artificial intelligence, Taizhou University, Taizhou, China, Taizhou, China
Y. Yuan1, A. Yuan2, W. Wang1, Z. Hu2, C. Zheng2, Z. Zheng2, W. Liang2, Y. Zhang2, Y. Pan2, C. Zhang1; 1School of artificial intelligence, Taizhou University, Taizhou, China, Taizhou, CHINA, 2Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), School of Medicine, Taizhou University, Taizhou, CHINA.
A Retrospective Study on a Predictive Model to Probe the Impact of Atmospheric Environment on Breast Cancer Prognosis: A Cohort of 17,438 PatientsAbstractBackground: In survival analysis tasks, DeepSurv, DeepHit, etc., are deep learning-based models designed to handle one-dimensional time series data. Their primary objective is to predict an individual’s survival probability and its relationship with time. However, traditional one-dimensional data processing methods may overlook certain spatial structural information inherent in the data—a limitation that is particularly significant for specific types of biomedical or clinical data. Therefore, we propose an innovative approach: converting one-dimensional data into grayscale images. This conversion introduces additional spatial features, enabling the model to better capture complex patterns in the data. Our aim is to develop a predictive analysis model for the impact of atmospheric environment on breast cancer prognosis.Methods: This study is a multi-center retrospective cohort study on breast cancer, involving 17,438 breast cancer patients. The endpoint outcome is disease-free survival (DFS). The features include age, gender, HDI, history of malignant tumors, family history of breast cancer, ki67, pathological type, molecular subtype, TNM staging, O3, PM2.5, monthly average precipitation, monthly average wind speed, and monthly average temperature. Firstly, we expand the dimensionality of one-dimensional survival data by mapping it to grayscale images, making it compatible with the input format of convolutional neural networks (CNNs) for processing image data. Through this conversion, we can effectively extract spatial features using convolutional operations, thereby enhancing the model’s predictive capability. Subsequently, we develop a CNN-based deep neural network model that incorporates convolutional layers to process these grayscale image data while retaining the core concepts of survival analysis. This model aims to predict an individual’s relative risk value by learning deep patterns in the data and can handle prognostic data with high complexity and non-linear relationships.
Presentation numberPS3-10-12
Adjuvant chemotherapy and anthracycline use in older adults with high oncotype DX scores: A SEER-Medicare analysis
Anna R. Schreiber, University of Colorado Anschutz Medical Campus, Aurora, CO
A. R. Schreiber, E. Molina Kuna, E. Soto Pérez de Celis, J. R. Diamond; University of Colorado Anschutz Medical Campus, Aurora, CO
Background: Determining what adjuvant chemotherapy regimen to select in older patients with higher-risk, early-stage hormone receptor (HR)+, human epidermal growth factor receptor 2 (HER2)- breast cancers can be challenging. Patients with early-stage HR+/HER2-, node-negative breast cancer with high Oncotype DX recurrence scores (>31) may benefit from the addition of anthracyclines (1). However, it is not known whether this is true for older patients. The purpose of this study was to investigate outcomes based on Oncotype DX score and type of adjuvant treatment among older patients with early-stage, HR+/HER2- breast cancer using the Surveillance, Epidemiology, and End Results (SEER)-Medicare database. Methods: Using the SEER-Medicare database, we identified 5,439 women aged 66 years and older with a diagnosis of Stage I-III HR+/HER2- breast cancer between 2000 and 2019, and who had a recorded Oncotype DX score. We assessed patient characteristics, type of therapy received (anthracycline+taxane-based chemotherapy, taxane-based chemotherapy, or endocrine therapy), and Oncotype DX recurrence score (low [<11], intermediate [11-25], high [≥26]). Kaplan Meier survival curves were generated to assess overall survival (OS) and cancer-specific survival (CSS) based on Oncotype DX scores and therapy received. Results: Of the 5,439 patients in the cohort, most had an intermediate Oncotype DX score (60%), followed by a low Oncotype DX score (27%), and a high Oncotype DX score (13%). Most patients were aged 66-74 years (76%), followed by those aged 75-84 (22%). Only 1% were ≥85 years of age. Patients with a high Oncotype DX score had mostly node-negative disease (80%), with 17% having 1-3 positive nodes. Most patients in the high Oncotype DX score group received endocrine therapy alone (63%), followed by taxane-based chemotherapy (21%), and anthracycline+taxane-based chemotherapy (5%). Anthracyclines were used more often in patients aged 66-74 years. Most patients who received anthracyclines had a Charlson Comorbidity Index (CCI) of 0-1 (82%). In patients with a high Oncotype DX score and node-negative disease, anthracycline+taxane-based regimens did not improve OS or CSS when compared to taxane-based regimens and endocrine therapy. Importantly, the use of any chemotherapy did not improve OS and CSS in this group compared with endocrine therapy alone. In patients with a high Oncotype DX score and 1-3 positive nodes, anthracycline+taxane-based regimens did not improve OS and CSS when compared to taxane-based regimens and endocrine therapy. Likewise, the use of any chemotherapy did not improve OS and CSS in this group compared with endocrine therapy alone. Conclusion: The use of adjuvant chemotherapy, as well as the addition of anthracyclines, did not improve outcomes in patients aged ≥66 years with early-stage HR+/HER2- breast cancer and a high Oncotype DX score (RS≥26), regardless of nodal positivity. It is important to note that the overall number of patients who received an anthracycline+taxane-based regimen was small, so results should be interpreted with caution. This lack of benefit was seen even in a population with lower comorbidity scores, which suggests its use in highly selected individuals. The decision to treat patients with high-risk, early-stage HR+/HER2- breast cancer with adjuvant chemotherapy should be individualized, taking into account the limited benefits in OS compared with reports in younger individuals. 1. Chen, Nan. “San Antonio Breast Cancer Symposium .” GS3-03: Impact of Anthracyclines in High Genomic Risk Node-Negative HR+/HER2- Breast Cancer, 2024, https://sabcs.org/Portals/0/Documents/Embargoed/GS3-03%20Embargoed.pdf?ver=i2jS4VEzTIiDPEWdqDHRwA%3d%3d. Accessed 2025.
Presentation numberPS3-11-06
Long-term prognostic and predictive value of lobular histology in the PALLAS trial
Guilherme Nader-Marta, Dana-Farber Cancer Institute, Boston, MA
G. Nader-Marta1, C. Desmedt2, D. Hlauschek3, A. DeMichele4, G. Zoppoli5, E. Blondeaux6, E. de Azambuja7, M. Bellet-Ezquerra8, O. Metzger-Filho1, J. M. Suga9, G. Pfeiler10, M. P. Goetz11, M. Ruiz-Borrego12, S. Loibl13, J. Meisel14, A. Ring15, K. Van Baelen2, E. P. Mamounas16, N. Zdenkowski17, E. Agostinetto7, M. Lambertini18, E. Gauthier19, L. Soelkner3, A. C. Dueck20, M. Gnant3, E. L. Mayer1; 1Dana-Farber Cancer Institute, Boston, MA, 2Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, BELGIUM, 3Austrian Breast and Colorectal Cancer Study Group (ABCSG), Vienna, AUSTRIA, 4University of Pennsylvania, Philadelphia, PA, 5Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, ITALY, 6Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 7Clinical Trials Support Unit, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, BELGIUM, 8Vall d’Hebron Institute of Oncology (VHIO), Barcelona, SPAIN, 9Kaiser Permanente Vallejo Medical Center, Vallejo, CA, 10Department of Gynecology and Gynecological Oncology, Medical University of Vienna, Comprehensive Cancer Center, Vienna, AUSTRIA, 11Mayo Clinic, Rochester, MN, 12Hospital Universitario Virgen del Rocío, GEICAM Spanish Breast Cancer Group, Sevilla, SPAIN, 13German Breast Group, Neu-Isenburg, GERMANY, 14Winship Cancer Institute, Emory University, Emory, GA, 15The Royal Marsden NHS Foundation Trust, London, UNITED KINGDOM, 16AdventHealth Cancer Institute, Orlando, FL, 17Lake Macquarie Private Hospital, Gateshead, AUSTRALIA, 18Department of Medical Oncology, U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, ITALY, 19Pfizer, Inc., San Francisco, CA, 20Alliance Statistics and Data Management Center and Mayo Clinic Rochester, Rochester, MN.
Background: Invasive lobular carcinoma (ILC) accounts for up to 15% of breast cancer (BC) cases and has distinct clinical and biological features compared to invasive carcinoma of no special type (IBC-NST). Prior studies, including PENELOPE-B, suggested greater benefit from adjuvant palbociclib (palbo) in ILC. We assessed the prognostic and predictive value of histology in patients (pts) treated with endocrine therapy (ET) ± palbo. Methods: PALLAS is a global, phase III trial in which pts with stage II-III, HR+/HER2− BC were randomized to 2 years (y) of adjuvant palbo + ET or ET alone. Pts with IBC-NST or pure ILC per local pathology assessment were included. Endpoints included invasive disease-free (IDFS; primary), distant recurrence-free (DRFS), locoregional recurrence-free (LRRFS), and overall survival (OS). Prognostic value was assessed using univariable and multivariate Cox models adjusted for age, T and N stage, grade, and progesterone receptor. Predictive value was assessed using histology-by-treatment interactions. Results: Among 5,748 pts in the ITT population, 4,982 (87%) were included (3,838 IBC-NST [67%], 1,144 ILC [20%]); median follow-up 6.9y. Compared with IBC-NST, ILC was associated with older age (median 54 vs 51 y), higher T stage (T3/T4: 41.7% vs 20.4%), fewer grade 3 tumors (12.7% vs 33.5%), higher mastectomy rates (74.5% vs 58.4%), less (neo)adjuvant chemotherapy (72.2% vs 86.2%), and less tamoxifen as initial ET (28.8% vs 34.5%) (all p<0.001). ILC prevalence varied by country: lower in Asia (Japan 10.5%, Korea 0%, Taiwan 2.8%) and higher in Europe (23.1%) and North America (24.0%). When evaluating the predictive value, palbo appeared to confer greater benefit in ILC for all endpoints (HRs ranging from 0.41 to 0.86) and not in IBC-NST (HRs ranging from 0.97 to 1.01), although the histology-by-treatment interaction was significant only for LRRFS (Table). In the overall population, 7-year IDFS was 79.3% for IBC-NST and 75.8% for ILC. While the difference was significant in univariable analysis (HR 1.17, 95% CI 1.01-1.35), it was not in multivariable analysis (HR 1.06, 0.91-1.24). Notably, the proportional hazards assumption was violated, and time-dependent modeling demonstrated that worse outcomes for ILC on IDFS, DRFS, and other endpoints emerged predominantly beyond 3 years. Conclusions: In this large, randomized dataset of pts with early HR+/HER2− breast cancer with long-term follow-up, ILC demonstrated distinct clinicopathologic features and geographic distribution. While ILC was not an independent prognostic factor over the entire follow-up, time-dependent modeling revealed worse long-term outcomes which only emerged after 3 years. Palbo appeared to be associated with more favorable outcomes in ILC than in IBC-NST across endpoints. These findings suggest a potential benefit of palbo in ILC that warrants further investigation in other adjuvant trials evaluating CDK4/6 inhibitors.
| Predictive value of histology (palbo+ET vs ET only) | |||
| Unadjusted HR (95% CI) | Interaction P-value | ||
| IDFS | IBC-NST | 0.97 (0.83–1.12) | 0.11 |
| ILC | 0.76 (0.59–0.98) | ||
| LRRFS | IBC-NST | 0.99 (0.73–1.35) | 0.02 |
| ILC | 0.41 (0.21–0.80) | ||
| DRFS | IBC-NST | 0.98 (0.83–1.16) | 0.17 |
| ILC | 0.78 (0.59–1.03) | ||
| OS | IBC-NST | 1.01 (0.82–1.24) | 0.44 |
| ILC | 0.86 (0.61–1.22) | ||
| Prognostic value of histology (ILC vs IBC-NST) | |||
| Unadjusted HR (95% CI) | Adjusted HR (95% CI) | ||
| IDFS | 1.17 (1.01–1.35) | 1.06 (0.91–1.24) | |
| LRRFS | 0.85 (0.61–1.20) | 0.83 (0.58–1.19) | |
| DRFS | 1.22 (1.04–1.44) | 1.09 (0.92–1.30) | |
| OS | 1.18 (0.96–1.44) | 1.01 (0.81–1.26) | |
| Time-dependent prognostic value (ILC vs IBC-NST) | |||
| Adjusted HR (95% CI), 0–3 years | Adjusted HR (95% CI), >3 years | ||
| IDFS | 0.87 (0.69–1.09) | 1.27 (1.04–1.57) | |
| LRRFS | 0.76 (0.44–1.30) | 0.89 (0.56–1.42) | |
| DRFS | 0.80 (0.62–1.04) | 1.41 (1.13–1.75) | |
| OS | 0.70 (0.47–1.06) | 1.17 (0.91–1.50) |
Presentation numberPS3-11-10
Neoadjuvant HSP90 inhibition with ganetespib significantly improves distant relapse-free survival in I-SPY2 patients with HER2-negative breast cancer with low expression of immune genes and no DNA repair deficiency
Tam B Bui, University of California San Francisco, San Francisco, CA
T. B. V. Bui1, D. M. Wolf1, C. Yau1, R. W. Sayaman1, K. Chow1, S. W. Choi1, M. C. Bruck1, K. Santos-Parker1, A. M. Glas2, J. E. Lang3, A. Forero-Torres4, L. Brown-Swigart1, G. L. Hirst1, E. F. Petricoin5, J. Perlmutter6, W. F. Symmans7, P. R. Pohlmann7, D. Yee8, L. Pusztai9, I-SPY investigators, N. M. Hylton1, A. M. DeMichele10, J. M. Rosenbluth1, L. J. Esserman1, L. J. Van ‘T Veer1; 1University of California San Francisco, San Francisco, CA, 2Quantum Leap Healthcare Collaborative, San Francisco, CA, 3Cleveland Clinic, Cleveland, OH, 4Kirklin UAB Hematology Oncology, Birmingham, AL, 5George Mason University, Manassas, VA, 6Gemini Group, Ann Arbor, MI, 7The University of Texas MD Anderson Cancer Center, Houston, TX, 8University of Minnesota, Minneapolis, MN, 9Yale School of Medicine, New Haven, CT, 10University of Pennsylvania, Philadelphia, PA
Introduction: The I-SPY2 neoadjuvant platform trial (NCT01042379) evaluates new therapies in high molecular risk, stage 2-3 breast cancer patients. Between 2010 and 2022, 24 therapies were tested in 2,117 patients. One-third of these patients had HER2-negative tumors with low expression of immune genes and no DNA repair deficiency (DRD) (HER2-/Immune-/DRD- subtype). These patients had very low pathologic complete response (pCR) rates (~10% on average) across all treatment and control arms. We performed a high-throughput drug screen (n=386 compounds) in triple-negative (TN), immune-low, DRD-low breast cancer patient-derived organoids with confirmed in vitro resistance to platinum chemotherapy to identify effective treatments for this tumor type. HSP90 inhibitors, including ganetespib, were one of the most potent drug classes in the screen. HSP90 is an abundant chaperone protein involved in the folding of oncoproteins under basal conditions and during stress response. Neoadjuvant ganetespib was tested in I-SPY2 in combination with standard chemotherapy in HER2-negative breast cancer but did not meaningfully improve pCR rates (PMID 36456573). Here, we re-evaluate the ganetespib arm by Response Predictive Subtypes with 5-year median follow-up data, focused on patients with breast tumors of the HER2-/Immune-/DRD- subtype. Methods: HER2-/Immune-/DRD- patients (N=221; 79% HR+, 21% TN) and HER2-/Immune+ and/or DRD+ patients (N=219; 32% HR+, 68% TN) were randomized to neoadjuvant ganetespib (150 mg/m2 ganetespib every 3 weeks with paclitaxel, then doxorubicin plus cyclophosphamide (AC)) (n=38 HER2-/Immune-/DRD- and n=55 HER2-/Immune+ and/or DRD+), or standard neoadjuvant chemotherapy control (paclitaxel-AC) (n=183 HER2-/Immune-/DRD- and n=164 HER2-/Immune+ and/or DRD+). The primary endpoint, pCR, was defined as the absence of invasive disease in the breast and regional nodes at surgery. Distant relapse-free survival (DRFS) was assessed using Kaplan-Meier curves, with hazard ratios (HR) estimated by Cox regression. Residual cancer burden (RCB) and serial MRI-based functional tumor volume (FTV) were collected as investigational early measures of treatment efficacy in the neoadjuvant setting. Results: In HER2-/Immune-/DRD- patients, pCR rates were low in the ganetespib and control arms (13% vs. 9%), compared with 38% vs. 31%, respectively, in HER2-/Immune+ and/or DRD+ patients. In both subtypes, patients who achieved a pCR with either treatment had excellent DRFS (100% at 5 years). Interestingly, in HER2-/Immune-/DRD- patients treated with ganetespib with no pCR (n=32), we observed only 1 DRFS event, compared with 35 DRFS events in HER2-/Immune-/DRD- control-treated patients with no pCR (n=166); HR 0.12 (95% CI 0.02-0.90; p-value: 0.039). The respective DRFS at 5 years was 96% with ganetespib vs. 77% with control, showing a significantly improved DRFS with neoadjuvant ganetespib for HER2-/Immune-/DRD- patients, despite their non-pCR. This treatment effect remained significant when adjusted for hormone receptor status. Among non-pCR patients, RCB measure and FTV decrease explained a portion of ganetespib’s significant DRFS benefit. In contrast, ganetespib did not improve DRFS in HER2-/Immune+ and/or DRD+ patients compared with control. Conclusion: Our data suggest a promising role for ganetespib in improving long-term outcomes for patients with HER2-/Immune-/DRD- tumors, which account for 38% of all molecularly high-risk breast cancers, even in the absence of a pathologic complete response. Continuous measures of RCB and FTV may serve as early predictors of ganetespib’s long-term survival benefit in HER2-/Immune/-DRD- patients.
Presentation numberPS3-13-16
Cancer systems immunology unravels complexity of reversing immune suppression and predicts beyond RECIST in metastatic breast cancer
Evanthia T Roussos Torres, University of Southern California, Los Angeles, CA
E. T. Roussos Torres1, E. Gonzalez1, J. Kreger1, Y. Liu1, X. Wu1, A. Barbetta1, A. G. Baugh1, B. Al-Zubeidy1, J. Jang1, S. M. Shin2, Z. M. Zhang3, V. Stearns4, R. M. Connolly5, W. Jin Ho2, J. Emamaullee1, A. L. MacLean1; 1University of Southern California, Los Angeles, CA, 2Johns Hopkins University, Baltimore, MD, 3Johns Hopkins University, Los Angeles, MD, 4Weill Cornell Medicine, Meyer Cancer Center, New York, NY, 5University College of Cork, Cork, Ireland
Background: Combination therapy aimed at modulating the tumor microenvironment (TME) is a robust strategy to improve response to checkpoint inhibition and broaden indications for use in patients with metastatic breast cancer. The epigenetic modulator, entinostat when combined with dual checkpoint blockade; nivolumab and ipilimumab demonstrated promising results in our Phase Ib trial (NCI-0944) and in our recent meta-analysis (AACR-3225, 2025). Use of a cancer systems immunology approach accelerated discovery of the complex mechanism of response to treatment. Lastly, we demonstrate the potential for math modeling to predict response similar to RECIST, and inform organ specific response, a powerful tool with potential for use toward a more personalized approach for patients. Methods: To identify mechanisms of response to combination therapy in a preclinical model, we used knowledge-guided subclustering of single-cell RNA-sequencing data and cell circuits analysis to predict salient interactions. Multiparametric flow cytometry and ex-vivo immune suppression assays were used to validate preclinical findings. Imaging mass cytometry and bulk RNA sequencing of patient samples were used to validate findings in patients. Derivation of dynamic mathematical models of tumor-immune modulatory responses were then fit through innovative use of both preclinical (sequencing) and clinical (proteomics) data – to predict that a combination of effects on the TME is necessary for response. Adaptation of this mathematical model was then made to determine its potential for use in prediction of site-specific response in metastatic lesions. We introduced methods employing posterior parameter sampling and simulation to create virtual tumor populations, enabling extrapolation beyond the data to predict probabilities of response in metastatic lesions, even when no data exists at that site. Results: In the mouse lung TME, we identified 39 cell states and salient interactions, of which myeloid, T cell, and B cell subpopulations were most affected by treatment in mice bearing metastatic breast cancer treated with combination therapy. Functional immunologic assays verified inhibition of the ICAM pathway in myeloid cells partially recapitulated treatment effects on CD8+ T cells. We also found evidence that treatment increased anti-tumor IgG production. Analysis of patient biopsies via spatial proteomics corroborated preclinical findings: in responders, we observed increased B cell activation, mature tertiary lymphoid structures (TLS), and increased CD8+ T cell—macrophage distances with treatment. We demonstrated clinical utility of the mathematical model via Bayesian parameter inference with clinical responses measured by RECIST. This revealed that only the immunosuppression parameters were predictive of response; parameters controlling cytotoxicity were uninformative. We also show that the model can predict response at sites that have yet to develop disease—a tool which could be considered for future trials to predict overall response rates based on mechanism of drug response. Conclusions: We conclude that epigenetic modulation via HDACi induces a carefully orchestrated set of changes in plasma cells and CD8 T cells with MDSCs and macrophages to sensitize the TME to checkpoint inhibition. Significant changes in TLS formation and macrophage -T cell interactions in biopsies from patient responders validate findings. More broadly we provide a framework for the discovery of cell-cell interactions that control responses in complex TMEs. We also demonstrate how interdisciplinary data integration fuels this new field of cancer systems immunology to accelerate discovery of mechanisms of successful immunotherapeutic response in previously unresponsive solid tumor types.
Presentation numberPS3-13-18
Ultrasound-guided biopsy-based tils assessment at tumor margins as a predictive biomarker of neoadjuvant treatment response in early her2-positive and triple-negative breast cancer: a multicenter study
Yuri Kimura, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
Y. Kimura1, R. Yamaguchi2, K. Fukui3, A. Kanou3, M. Noma4, S. Akashi-Tanaka5, K. Arihiro6, N. Iwamoto7, Y. Ohta8, T. Okuno9, S. Kashiwagi10, Y. Kamei11, Y. Tanada12, S. Nakano13, A. Nagata14, A. Murakami11, Y. Mochitomi15, A. Yamakawa16, M. Yamaguchi17, H. Shima18, A. Emi19; 1The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, JAPAN, 2Breast Center, Department of Diagnostic Pathology, Nagasaki University Hospital, Nagasaki, JAPAN, 3Division of Laboratory Medicine, Hiroshima University Hospital, Hiroshima, JAPAN, 4Department of Gastrointestinal, Breast, and Transplantation Surgery, Hiroshima Prefectural Hospital, Hiroshima, JAPAN, 5Department of Breast Surgery, Tokyo Women’s Medical University, Tokyo, JAPAN, 6Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, JAPAN, 7Department of Breast Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, JAPAN, 8Department of General Surgery, Kawasaki Medical School, Okayama, JAPAN, 9Department of Breast Surgery, Nishi-Kobe Medical Center, Hyogo, JAPAN, 10Department of Breast Surgery, Osaka Metropolitan University Graduate School of Medicine, Osaka, JAPAN, 11Breast Center, Ehime University Hospital, Ehime, JAPAN, 12Department of Surgery, National Hospital Organization Kanazawa Medical Center, Kanazawa, JAPAN, 13Division of Breast and Endocrine Surgery, Aichi Medical University Hospital, Aichi, JAPAN, 14Division of Breast Surgical Oncology, Department of Surgery, Showa Medical University School of Medicine, Tokyo, JAPAN, 15Division of Laboratory Medicine, Sagara Hospital, Kagoshima, JAPAN, 16Department of Surgery, Ogaki Municipal Hospital, Gifu, JAPAN, 17Department of Breast Surgery, Daido Hospital, Nagoya, JAPAN, 18Department of Surgery, Division of Breast Surgery, Sapporo Medical University, Hokkaido, JAPAN, 19Department of Breast Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, Hirosihma, JAPAN.
Background: Tumor-infiltrating lymphocytes (TILs) reflect host immune response and are associated with prognosis and treatment response in HER2-positive and triple-negative breast cancer (TNBC). While the International Immuno-Oncology Biomarker Working Group recommends evaluating stromal TILs in tumor areas, intratumoral heterogeneity and small sample size in core needle biopsy (CNB) specimens limit consistent assessment. It remains unclear whether central tumor (CT) or invasive margin (IM) regions should be prioritized and what cut-off values are clinically relevant. Ultrasonographic (US) features may also reflect the immune microenvironment. We previously proposed a TILs-US score based on three US features that predicts lymphocyte-predominant breast cancer (LPBC). This study evaluated the clinical utility of TILs assessment from CT and IM regions, appropriate cut-off values for predicting response in HER2-positive and TNBC using CNB, and explored optimal biopsy targeting through pathologic and US comparisons. Methods: In this multicenter retrospective study, we analyzed 239 patients (124 HER2-positive, 115 TNBC) with cT1-3/cN0-2 breast cancer who underwent surgery after neoadjuvant chemotherapy (NAC). TILs were evaluated using CNB specimens obtained before NAC and assessed separately in the central tumor (CT) and invasive margin (IM) regions. The IM was defined as a 1-mm-wide area centered on the interface between the tumor and surrounding stroma, while the CT was defined as the remaining intratumoral area within the IM boundary. TILs were analyzed using binary cut-offs of 30% and 50%, as well as a three-tier classification: low (0-10%), intermediate (11-49%), and high (≥50%). Pre-treatment ultrasound (US) findings included tumor shape (scored 0-2), internal echo (0-2), and posterior echo (0-3), which were used to calculate a composite TILs-US score ranging from 0 to 7. Associations between TIL levels, US features, and pathological complete response (pCR) were analyzed. Results: Overall, 104 patients achieved pCR (HER2-positive: 39; TNBC: 65). Using a 30% cut-off, TILs were evenly distributed both CT and IM, while ≥50% cases were relatively rare especially CT (CT: 10%; IM: 20%). CT TILs ≥30% was significantly associated with pCR (p=0.001), whereas the ≥50% cut-off was not (p=0.128). In contrast, both 30% and 50% cut-offs in the IM region were predictive of pCR (p<0.001 and p=0.009, respectively). Subtype analysis revealed that in TNBC, TILs ≥30% in both CT and IM regions were significantly correlated with pCR. In HER2-positive cases, however, only IM TILs ≥30% was predictive of pCR. Similar trends were observed with the three-tier classification. Among US features, a small lobulated shape was associated with high TILs levels in both CT and IM regions, while an enhanced posterior echo was correlated with high TILs in the IM region. Conclusion: Our findings suggest that TILs assessment should include the IM in addition to the CT, especially in CNB specimens as provide valuable information. Given the limited tissue area available in CNB specimen, using a 30% cut-off for TILs may offer a clinically meaningful threshold to predict treatment response. In cases where US findings suggest central fibrosis, targeting only the tumor center may result in sampling fibrotic areas with sparse immune and tumor cells, potentially hindering proper evaluation. Therefore, selecting the biopsy target based on US imaging is essential. Especially in HER2-positive breast cancer, as well as TNBC, it is important for operators to consider the optimal needle insertion angle and sampling site to ensure appropriate sampling of viable tumor tissue and immune microenvironment for accurate pathological and immunological evaluation.
Presentation numberPS3-13-20
Fibroblastic mutant trp53 in the stroma promotes mammary tumor development through enhanced paracrine factor secretion
Carenza Johnson, Texas Southern University, Department of Pharmaceutical Sciences, Houston, TX
B. Liu1, G. Chau2, G. Lozano2, S. Xiong2, J. McDaniel2, D. Chachad3, C. Johnson4, A. Williams-Villalobo5, H. Chen4, Y. Zhang4, Y. Qi6, X. Su6, A. K. El-Naggar7; 1MD Anderson Cancer Center Department of Epigenetics and Molecular Carcinogenesis, Division of Discovery Science, Houston, TX, 2MD Anderson Cancer Center, Department of Genetics, Houston, TX, 3MD Anderson Cancer Center, Houston, TX, 4Texas Southern University, Department of Pharmaceutical Sciences, Houston, TX, 5Baylor College of Medicine, Houston, TX, 6MD Anderson Cancer Center, Department of Bioinformatics and Computational Biology, Division of Discovery Science, Houston, TX, 7MD Anderson Cancer Center, Department of Anatomical Pathology, Division of Pathology-Lab Medicine Div, Houston, TX.
Tumor progression critically depends on stromal contributions, particularly from fibroblasts that regulate growth factors, angiogenesis, immunity, and ECM remodeling. While TP53 is the most frequently mutated tumor suppressor and its cell-autonomous roles have been extensively studied, emerging evidence shows its stromal functions actively shape tumor-stroma crosstalk. However, the molecular mechanisms by which mutant stromal TP53 influences tumor development remain undefined. This study examined whether fibroblast-specific expression of p53R172H influences HER2/neu-driven mammary tumorigenesis. We generated a female mouse cohort with genotypes MMTV-neu; Fsp-Cre; Trp53wm-R172H/+ (NP) and MMTV-neu; Fsp-Cre or Trp53wm-R172H/+ (N) as controls. In NP mice, Cre recombinase drove recombination of the conditional Trp53wm-R172H/+ allele, coverting wild-type p53 to mutant p53R172H specifically in fibroblasts. NP mice developed mammary tumors significantly earlier, with tumor-free survival of 520 days compared to 620 days in controls (p = 0.0007). Although tumor burden and metastatic frequency were unchanged, RNA sequencing revealed 855 upregulated and 1,927 downregulated genes unique to NP tumors. Gene Set Enrichment Analysis indicated activation of multiple pathways, notably PI3K/AKT/mTOR. In addition, transcriptomic profiling of NP versus N mammary glands showed elevated expression of secretory proteins SAA1, SAA2, and THBS4, previously linked to PI3K/AKT-driven cell growth. In vitro assays using synthetic peptides of these proteins and HER2/neu+ mammary tumor cell lines confirmed that SAA1, SAA2, and THBS4 promote tumor cell proliferation and migration. Analysis of TCGA and METABRIC datasets further demonstrated increased SAA1 and SAA2 expression in TP53-mutant human breast cancers, supporting their relevance in patients. Together, these findings reveal that fibroblastic mutant p53 accelerates breast tumorigenesis through paracrine signaling that activates PI3K/AKT/mTOR. These results highlight stromal mutant p53-mediated tumor-stroma crosstalk as a critical and potential therapeutic target for breast cancer biology.
Presentation numberPS3-13-21
The RXR agonist IRX4204 promotes cytotoxicity of Her2+ breast cancer cells by Her2-targeted CAR-T cells
Martin E Sanders, Io Therapeutics, Inc., The Woodlands, TX
M. E. Sanders, V. Vuligonda; Io Therapeutics, Inc., The Woodlands, TX
Research Objectives and Rationale: IRX4204 is a potent and highly RXR nuclear receptor- selective agonist compound. It is 100-fold more potent as an RXR agonist than bexarotene, the only FDA-approved RXR agonist (approved for treatment of cutaneous T-cell lymphoma). IRX4204 is far more RXR selective than bexarotene, being devoid of RAR, LXR, PPAR gamma nuclear receptor agonism. With our collaborators we have previously reported that: (1) IRX4204 has synergistic cytotoxic effects with Her2-targeted antibodies, Her2-tyrosine kinase inhibitors, and paclitaxel on Her2+ breast cancer cell lines; (2) IRX4204 promotes apoptosis of Her2+ breast cancer cells; (3) IRX4204 promotes in vivo CD8 T-cell infiltration into murine Brca1 mutant breast cancer tumors. (Moyer et. al., Clinical Cancer Research, June 2024; and Moyer et. al., poster presented at SABCS 2023). CAR-T cells targeted against various types of hematologic malignancies have been highly clinically effective. However, progress in developing effective CAR-T cells against solid tumors, such as breast cancers, has been slow. Based on the previously reported combination effects of IRX4204 with anti-Her2-targeted agents on Her2+ breast cancers, we hypothesized that IRX4204 may have combination effects with Her2-targeted CAR-T cells on killing of Her2+ breast cancer. We now report that IRX4204 promotes infiltration and cytotoxicity of cultured spheroids of the Her2+ human breast cancer cell line SKBR3 by Her2-targeted CAR-T cells.Experimental Methods: Initial experiments examined effects of IRX4204 on expression of cell surface Her2 and various adhesion molecules on cultured SKBR3 cells by flow cytometry. To evaluate combination treatment effects with IRX4204 and Her2-targeted CAR-T cells, we obtained Her2-targeted human CAR-T cells from a commercial source (Promab: PM-CAR1024-1M; Her2 scf-v-CD28CD3zeta). SKBR3 cells were stained with cell tracker deep red and cultured till forming spheroids. Her2-targeted CAR-T cells were stained with cell trace violet and added to the SKBR3 cells at effector to target ratios of 0, 2.5, 5, and 10 to 1. Following 72 hours of co-incubation of SKBR3 cells with CAR-T cells, IRX4204 was added to the cultures at 10, 100, and 1000 nM concentrations, for 24 hours. Sytox Green Ready reagent was added to the cultures to quantitate cell death. Images were analyzed using automated systems to quantitate T-cell infiltration and cytotoxicity. Results: IRX4204 did not affect expression of cell surface Her2 on SKBR3 cells. IRX4204 substantially increased expression of the adhesion molecule ICAM-1 on SKBR3 cells, an effect which should promote T-cell adhesion, infiltration, and cytotoxicity by CAR-T cells. IRX4204 increased infiltration of CAR-T cells into SKBR3 spheroids in a dose dependent manner. IRX4204 alone had minimal cytotoxic effect on SKBR3 cells under these test conditions. However, IRX4204 synergistically promoted Her-2 targeted CAR-T cytotoxicity of SKBR3 Her2+ breast cancer cells.Conclusions: Combination of the RXR agonist IRX4204 with Her2-targeted CAR-T cells showed a dose dependent increase in CAR-T infiltration and death of spheroids of Her2+ SKBR3 breast cancer cells in vitro. These promising results support further development of Her2-targeted CAR-T cells, and combination of such cells with the RXR agonist IRX4204 as a therapeutic option for patients with advanced Her2+ breast cancer.
Presentation numberPS3-13-22
Epithelial Podoplanin Co-expression in Postpartum Breast Cancer: An Exploratory TME Analysis
Jasmine A McDonald, Mailman School of Public Health, New York, NY
J. A. McDonald1, A. J. Kociolek1, S. Tun Hein Aung1, Y. Liao1, B. Vasikj Bjelogrlic1, C. Loomis2, J. M. Genkinger1, M. Terry1, K. L. Gardner3; 1Mailman School of Public Health, New York, NY, 2NYU Grossman School of Medicine, New York, NY, 3Columbia University Irving Medical Center, New York, NY
Background: Postpartum breast cancer (PPBC) – diagnosis within 10 years postpartum – shows more aggressive and ~3-fold higher metastasis risk than nulliparous or ≥10 years postpartum cases. Human data on the PPBC tumor microenvironment (TME) are limited. We evaluated epithelial, macrophage, and endothelial wound-healing phenotypes in the PPBC TME. Objective: Identify TME phenotype(s) associated with PPBC status. Study Population: From the international Breast Cancer Family Registry (BCFR), we included 180 invasive ductal carcinoma cases diagnosed ≤50 years within the New York subset who had a H&E and an unstained slide; 31, 40, and 58 cases diagnosed <5, ≥5 – <10, ≥10 years postpartum, respectively, and 51 nulliparous cases. Methods: Multiplex immunofluorescence panel design, staining, and multispectral imaging (Opal/Vectra Polaris) were conducted blinded to patient clinicopathologic data for Podoplanin (PDPN), Macrophages (CD68), Endothelial Cells (CD31/CD34 antibody mixture), panCK, SEMA7A, PD-L1, and DAPI. Slides were scanned with a 10X objective. Tumor containing regions were annotated by pathologists; cell phenotyping, cell counts, and signal intensities were quantified in HALO (full magnification), with fluorescent intensities normalized to 0-255. Phenotypes were defined by co-/anti-coincidence logic. Within tumor regions, we computed densities (cells/mm2) for markers and 20 colocalized phenotypes, normalized each by DAPI+ nuclear density to yield unitless ratios, and log-transformed these ratios to approximate normality. Phenotypes with ≥25% zero values were dichotomized as present/absent. Continuous predictors were z-scored within cross validation (CV) folds. We defined PPBC cases as <10 years postpartum; nulliparous or ≥10 years postpartum were considered non-PPBC cases. Statistical Analysis: We examined clinicopathologic, epidemiologic, and the tumor phenotype predictor variables stratified by PPBC status (chi2, t-tests). We then fit a LASSO-penalized logistic regression; λ was tuned by repeated 5-fold CV with 5 repeats, optimizing for AUC (λ_min). Continuous predictors were standardized (z-scored) within CV folds to prevent information leakage. Binary indicators for categorical predictors remained 0/1 (not standardized). Predictors with non-zero coefficients at the selected λ comprised the active set. For interpretability, we refit an unpenalized logistic regression model on the active set (‘post-LASSO’) and reported odds ratios (OR) and 95% CIs per 1-SD increase. Sensitivity analyses (i) restricted the sample to parous participants and including parity-related predictors (e.g., breastfeeding), and (ii) redefined PPBC cases to <5 years postpartum in the post-LASSO model. Results: PPBC and non-PPBC cases were similar across predictors, except PPBC cases were younger at diagnosis (p<0.001), and among parous women, PPBC cases were older at first FTP prior to diagnosis (p<0.001). At λ_min, only age at diagnosis and PanCK+PDPN+ were retained. Macrophage- and endothelial/lymphatic-centric phenotypes shrank to zero. In post-LASSO models, age at diagnosis was inversely associated with PPBC (ORper 1-SD 0.39, 95% CI (0.26, 0.58) and PanCK+PDPN+ was positively associated (ORper 1-SD 1.72, 95% CI (1.21, 2.52)). When restricted to the parous subsample, PanCK+PDPN+ was retained by LASSO (post-Lasso ORper 1-SD 2.04, 95% CI (1.11, 4.16)); however, when redefining PPBC cases to <5 years postpartum, PanCK+PDPN+ was null (post-Lasso ORper 1-SD 0.93, 95% CI (0.47, 2.04)). Conclusion: PPBC tumors showed enrichment of PanCK⁺PDPN⁺ epithelial cells, consistent with epithelial PDPN upregulation seen in wound-healing-like programs. PanCK⁺PDPN⁺ was retained among parous women, but not within a more conservative definition of PPBC. Findings are exploratory and warrant validation.
Presentation numberPS3-01-29
The Impact of Oophorectomy on Breast Cancer Survival in Patient with a BRCA1 Mutation
Steven A Narod, Women’s College Hospital, Toronto, ON, Canada
S. Narod1, J. Lubinski2, S. Wong3, J. Soukupova4, R. Bernstein5, R. Fruscio6, J. Plichta7, M. Lee8, A. Irmejs9, G. Evans10, J. Ngeow11, N. Tung12, K. Metcalfe1, P. Sun1; 1Women’s College Hospital, Toronto, ON, Canada, 2Pomeranian Medical University, Szczecin, Poland, 3McGill University, Montreal, QC, Canada, 4Charles University, Prague, Czech Republic, 5Sheba Medical Center, Tel-Hashomer, Israel, 6University of Milan, Monza, Italy, 7Duke University, Durham, NC, 8Memorial Sloan Kettering, New York, NY, 9Riga Stradins University, Riga, Latvia, 10University of Manchester, Manchester, United Kingdom, 11National Cancer Centre, Singapore, Singapore, 12Beth Israel Deaconess, Boston, MA
It has been established that preventive bilateral salpingo-oophorectomy (BSO) provides a survival advantage for women with breast cancer and a BRCA1 mutation, if done before or after diagnosis. It is of interest to see if the benefit of BSO varies according to the age at breast cancer diagnosis, age at oophorectomy and ER-status of the tumour. We are following a cohort of 2904 women with breast cancer and a BRCA1 mutation for survival. We excluded 256 women who had an oophorectomy before breast cancer. We also excluded women with metastatic cancer, with cancer diagnosed after age 70, and with cancers for greater than 10cm in size, leaving 2541 women for the present analysis. Of these, 1498 had an oophorectomy after breast cancer (mean time from diagnosis to BSO 2.3 years). We followed the women from diagnosis to death for a mean of 9.5 years (range 0.1 to 27.5 years). 289 women died of breast cancer. We conducted a survival analysis (Cox model) including BSO as a time-dependent variable, adjusting for tumour size (<2cm, 2-5cm, 5-10 cm) and lymph node status (positive/negative). Hazard ratios were for breast cancer survival. Of those women diagnosed with breast cancer before age 50, BSO was associated with a hazard ratio of 0.53 (95%CI 0.40 to 0.69). Of those women diagnosed with breast cancer after age 50, BSO was associated with a hazard ratio of 0.30 (95%CI 0.17 to 0.53). Of all women diagnosed with ER-positive breast cancer, BSO was associated with a hazard ratio of 0.57 (95%CI 0.35 to 0.90). Of those women diagnosed with ER-positive breast cancer before age 50, BSO was associated with a hazard ratio of 0.74 (95%CI 0.42 to 1.30). Of those women diagnosed with ER-positive breast cancer after age 50, BSO was associated with a hazard ratio of 0.26 (95%CI 0.10 to 0.65). Of all women diagnosed with ER-negative breast cancer, BSO was associated with a hazard ratio of 0.45 (95%CI 0.33 to 0.60). Of those women diagnosed with ER-negative breast cancer after age 50, BSO was associated with a hazard ratio of 0.32 (95%CI 0.15 to 0.69). In summary BSO after breast cancer is associated with improved survival across all age groups, with the possible exception of young women with ER+ breast cancer.


