NPJ Breast CancerPub Date : 2025-07-30DOI: 10.1038/s41523-025-00800-4
Pooja Patel, Antonio Giordano, Sara Giordano, Ilana Schlam, Sara M Tolaney, Paolo Tarantino
{"title":"Antibody-drug conjugates for treating early-stage breast cancer: current use, anticipated evolutions.","authors":"Pooja Patel, Antonio Giordano, Sara Giordano, Ilana Schlam, Sara M Tolaney, Paolo Tarantino","doi":"10.1038/s41523-025-00800-4","DOIUrl":"10.1038/s41523-025-00800-4","url":null,"abstract":"<p><p>Antibody-drug conjugates (ADCs) combine the specificity of monoclonal antibodies with potent cytotoxic payloads, leading to improved tumor delivery of the latter. After improving outcomes for patients with metastatic disease, ADC testing moved to the curative setting, where already one agent (trastuzumab emtansine) has been approved, after demonstrating to improve overall survival. Herein, we review the rationale, evidence, and challenges with the use of ADCs in the early-stage setting.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"81"},"PeriodicalIF":7.6,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311120/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144753916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-30DOI: 10.1038/s41523-025-00795-y
Silvia Steinbauer, Jamie D Cowles, Mohammad Ali Sabbaghi, Marle Poppelaars, Azaz Hussain, Marina Wagesreither, Daniela Laimer-Gruber, Jozsef Tovari, Gergely Szakacs, Agnes Csiszar
{"title":"Enhanced bioluminescence imaging of tumor cells surviving chemotherapy in a murine model of triple-negative breast cancer.","authors":"Silvia Steinbauer, Jamie D Cowles, Mohammad Ali Sabbaghi, Marle Poppelaars, Azaz Hussain, Marina Wagesreither, Daniela Laimer-Gruber, Jozsef Tovari, Gergely Szakacs, Agnes Csiszar","doi":"10.1038/s41523-025-00795-y","DOIUrl":"10.1038/s41523-025-00795-y","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is associated with poor prognosis and high recurrence, driven by residual tumor cells that survive chemotherapy. To monitor therapy response in vivo, we established a clinically relevant TAC regimen (docetaxel, doxorubicin, cyclophosphamide) in mice bearing mammary tumors derived from K14cre;Brca1<sup>F/F</sup>;Trp53<sup>F/F</sup> (KB1P) organoids expressing an mCherry-AkaLuc dual reporter (mCA-KB1P). AkaLuc bioluminescence imaging (AkaBLI) enabled non-invasive detection of minimal residual disease (MRD) with a sensitivity of approximately 1000 cells. As AkaLuc elicited an immune response, we generated Histon2B-mCherry-expressing KB1P organoids (HmC-KB1P) to study tumor cell survival in immunocompetent hosts. Flow cytometry and histological analysis revealed that MRD in immunocompetent mice is characterized by few residual cells with transient loss of epithelial markers, in contrast to immunodeficient hosts, which retains more epithelial-like cells. These findings validate AkaBLI for sensitive MRD detection and highlight the immune system's critical role in modulating residual tumor cell fate following chemotherapy.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"80"},"PeriodicalIF":7.6,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311029/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144753917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-26DOI: 10.1038/s41523-025-00790-3
Humaira Noor, Yuanning Zheng, Adam B Mantz, Ryle Zhou, Andrew Kozlov, Wendy B DeMartini, Shu-Tian Chen, Satoko Okamoto, Debra M Ikeda, Melinda L Telli, Allison W Kurian, James M Ford, Shaveta Vinayak, Mina Satoyoshi, Vishal Joshi, Sarah A Mattonen, Kevin Lee, Olivier Gevaert, George W Sledge, Haruka Itakura
{"title":"A 20-feature radiomic signature of triple-negative breast cancer identifies patients at high risk of death.","authors":"Humaira Noor, Yuanning Zheng, Adam B Mantz, Ryle Zhou, Andrew Kozlov, Wendy B DeMartini, Shu-Tian Chen, Satoko Okamoto, Debra M Ikeda, Melinda L Telli, Allison W Kurian, James M Ford, Shaveta Vinayak, Mina Satoyoshi, Vishal Joshi, Sarah A Mattonen, Kevin Lee, Olivier Gevaert, George W Sledge, Haruka Itakura","doi":"10.1038/s41523-025-00790-3","DOIUrl":"10.1038/s41523-025-00790-3","url":null,"abstract":"<p><p>A substantial proportion of patients with non-metastatic triple-negative breast cancer (TNBC) experience disease progression and death despite treatment. However, no tool currently exists to discriminate those at higher risk of death. To identify high-risk TNBC, we conducted a retrospective analysis of 749 patients from two independent cohorts. We built a prediction model that leverages breast magnetic resonance imaging (MRI) features to predict risk groups based on a 50-gene Transcriptomics Signature (TS). The TS distinguished patients with high-risk for death in multivariate survival analysis (Transcriptomic cohort: [HR] = 13.6, 95% confidence interval [CI] = 1.56-1, p = 0.02; SCAN-B cohort: HR = 1.45, CI 1.04-2.03, p = 0.02). The model identified a 20-feature radiomic signature derived from breast MRI that predicted the TS-based risk groups. This imaging-based classifier was applied to a validation cohort (log rank p = 0.013, accuracy 0.72, AUC 0.71, F1 0.74, precision 0.67, and recall 0.82), detecting a 25% absolute survival difference between high- and low-risk groups after 5 years.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"79"},"PeriodicalIF":7.6,"publicationDate":"2025-07-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12297616/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144718233","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-24DOI: 10.1038/s41523-025-00799-8
Amod Sharma, Sarabjeet Kour Sudan, Kunwar Somesh Vikramdeo, Mohammad Aslam Khan, Muhammad Tahir, James E Carter, Todd Kendall, Cindy Nelson, Ajay P Singh, Seema Singh
{"title":"Subtype- and race-specific variations in the immune landscape of breast cancer: therapeutic implications.","authors":"Amod Sharma, Sarabjeet Kour Sudan, Kunwar Somesh Vikramdeo, Mohammad Aslam Khan, Muhammad Tahir, James E Carter, Todd Kendall, Cindy Nelson, Ajay P Singh, Seema Singh","doi":"10.1038/s41523-025-00799-8","DOIUrl":"10.1038/s41523-025-00799-8","url":null,"abstract":"<p><p>Breast cancer is a heterogeneous disease with distinct molecular subtypes that disproportionately affects Black women. Immune cells are a key component of the tumor microenvironment, influencing tumor growth and treatment outcomes. Here, we explored immune landscape differences between TNBC and non-TNBC subtypes, assessing any race-specific patterns. TNBC showed higher infiltration of B-cells, Treg cells, Th1 cells, and CD8+ cells, and fewer mast cells than non-TNBC. Race-wise comparisons revealed that White TNBC had more Th1 cells than Black TNBC, while Black non-TNBC exhibited higher NK and Treg cells but lower DCs. KEGG pathway analysis identified immunosuppression in TNBC, with Black patients exhibiting the same regardless of molecular subtype. Higher TAM and lower T-cell infiltration were linked to metastatic disease. In White patients, lower immune cells (particularly T-cells, DCs, and NK cells) correlated with more metastasis, but not in Black patients. These race- and subtype-specific immune differences may guide tailored immunotherapies.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"78"},"PeriodicalIF":7.6,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12289971/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144708298","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-24DOI: 10.1038/s41523-025-00796-x
Matthew Fenton, Miki Yoneyama, Erik Wennerberg, Tom Lund, Andrew Tutt, Alan Melcher, Sandra Demaria, Navita Somaiah
{"title":"The untapped potential of radiation and immunotherapy for hormone receptor-positive breast cancer.","authors":"Matthew Fenton, Miki Yoneyama, Erik Wennerberg, Tom Lund, Andrew Tutt, Alan Melcher, Sandra Demaria, Navita Somaiah","doi":"10.1038/s41523-025-00796-x","DOIUrl":"10.1038/s41523-025-00796-x","url":null,"abstract":"<p><p>Hormone receptor-positive breast cancers are a diverse group of tumours, with only some responding well to immunotherapy. Alternative combination strategies such as radiotherapy present an exciting opportunity to improve immunotherapy responses. We review an intriguing overlap between the impact of oestrogen signalling and radiation on multiple signalling pathways and immune cells that may be exploited for therapeutic gains in breast cancer. This is synthesised with the pre-clinical data and clinical trial landscape supporting the use of combined radiation and immunotherapy to derive insights for future neo-adjuvant trial design.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"77"},"PeriodicalIF":7.6,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12289982/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144708299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-22DOI: 10.1038/s41523-025-00794-z
Christopher J Schwartz, Iskender Genco, Matteo Repetto, Daniel Muldoon, Andrea Gazzo, Panieh Terraf, Anne Grabenstetter, Dara Ross, Hong Zhang, Diana Mandelker, Simon Powell, Britta Weigelt, Chaitanya Bandlamudi, Edi Brogi, Fresia Pareja, Hannah Y Wen
{"title":"Breast carcinomas associated with microglandular adenosis are linked to germline alterations in homologous recombination-deficiency genes.","authors":"Christopher J Schwartz, Iskender Genco, Matteo Repetto, Daniel Muldoon, Andrea Gazzo, Panieh Terraf, Anne Grabenstetter, Dara Ross, Hong Zhang, Diana Mandelker, Simon Powell, Britta Weigelt, Chaitanya Bandlamudi, Edi Brogi, Fresia Pareja, Hannah Y Wen","doi":"10.1038/s41523-025-00794-z","DOIUrl":"10.1038/s41523-025-00794-z","url":null,"abstract":"<p><p>Invasive breast carcinomas associated with microglandular adenosis (IBC-MGA) represent a rare and poorly characterized form of triple-negative breast cancer (TNBC). We analyzed clinical, pathological, and germline genetic data from 38 patients, including 34 IBC-MGAs and 4 in situ cases. Germline pathogenic or likely pathogenic variants in homologous recombination-deficiency (HRD) genes were found in 42% (16/38) of patients, predominantly in BRCA1 (81%, 13/16). Most tumors were grade 3 invasive ductal or metaplastic carcinomas with limited tumor-infiltrating lymphocytes. No significant clinicopathologic differences were observed between germline HRD-associated and sporadic cases. Paired tumor-normal targeted sequencing revealed frequent TP53 mutations and high HRD scores. These findings underscore the relationship of breast carcinomas associated with MGA with HRD-related germline variants and highlight the potential for targeted therapeutic strategies and the importance of genetic testing in this rare subset of TNBC.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"76"},"PeriodicalIF":7.6,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12280099/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144682827","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-21DOI: 10.1038/s41523-025-00786-z
Filipa Lynce, Xianchen Liu, Benjamin Li, Lynn McRoy, Connie Chen, Raymond Liu, Hope S Rugo
{"title":"Real-world effectiveness of palbociclib plus an aromatase inhibitor in HR+/HER2- MBC patients living in disadvantaged neighborhoods.","authors":"Filipa Lynce, Xianchen Liu, Benjamin Li, Lynn McRoy, Connie Chen, Raymond Liu, Hope S Rugo","doi":"10.1038/s41523-025-00786-z","DOIUrl":"10.1038/s41523-025-00786-z","url":null,"abstract":"<p><p>Palbociclib (PAL) combined with endocrine therapy (ET) is approved for hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2- ) advanced/metastatic breast cancer (MBC). However, there is limited data on the effectiveness of PAL + ET in patients with MBC and low socioeconomic status. This retrospective study of the Flatiron Health database compared overall survival (OS) and real-world progression-free survival (rwPFS) in patients with MBC living in disadvantaged neighborhoods who received either first-line PAL with an aromatase inhibitor (AI) or an AI alone. Of 723 patients, 394 received PAL + AI and 329 received AI alone. After stabilized inverse probability of treatment weighting, median OS was 57.1 months versus 38.2 months (hazard ratio, 0.70, P = 0.0053) and median rwPFS was 19.1 months versus 14.0 months (hazard ratio, 0.66, P = 0.0007) for PAL + AI versus AI alone, respectively. This real-world data analysis demonstrated that first-line PAL + AI versus AI alone was associated with survival benefit in patients with HR+/HER2- MBC living in disadvantaged neighborhoods. Trial registration number: NCT06495164.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"75"},"PeriodicalIF":6.5,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12280009/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144682828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-19DOI: 10.1038/s41523-025-00783-2
Jesús Fuentes-Antrás, Mitchell J Elliott, Sasha C Main, Philippe Echelard, Aaron Dou, Philippe L Bedard, Eitan Amir, Michelle B Nadler, Nicholas Meti, Nancy Gregorio, Elizabeth Shah, Emily Van de Laar, Celeste Yu, Yangqing Deng, Lisa Gates, Clodagh Murray, Christopher G Smith, Amber Chevalier, Scott V Bratman, Lillian L Siu, Hal K Berman, David W Cescon
{"title":"Personalized ctDNA monitoring in metastatic HR+/HER2- breast cancer patients during endocrine and CDK4/6 inhibitor therapy.","authors":"Jesús Fuentes-Antrás, Mitchell J Elliott, Sasha C Main, Philippe Echelard, Aaron Dou, Philippe L Bedard, Eitan Amir, Michelle B Nadler, Nicholas Meti, Nancy Gregorio, Elizabeth Shah, Emily Van de Laar, Celeste Yu, Yangqing Deng, Lisa Gates, Clodagh Murray, Christopher G Smith, Amber Chevalier, Scott V Bratman, Lillian L Siu, Hal K Berman, David W Cescon","doi":"10.1038/s41523-025-00783-2","DOIUrl":"10.1038/s41523-025-00783-2","url":null,"abstract":"<p><p>Improved methods to monitor treatment response may enhance patient management and clinical outcomes. This study assessed the feasibility and performance of a tumor-informed circulating tumor DNA (ctDNA) assay in metastatic HR+/HER2- breast cancer patients receiving endocrine and CDK4/6 inhibitor therapy. By conducting whole exome sequencing on archival tumors, highly sensitive personalized ctDNA panels were designed for blood monitoring. The assay showed high detection sensitivity (91% baseline, 70% all timepoints) and associations between higher baseline estimated variant allele fractions, liver metastases, and shorter time to treatment failure (TTF) and overall survival (OS). Complete molecular response, defined as ctDNA clearance, was observed in 28% of patients and correlated with improved TTF (HR 0.07) and OS (HR 0.07). The last cleared timepoint predated treatment failure by a median 14.3 months. ctDNA rises or limited decreases preceded radiographic progression. Molecular metrics may facilitate plasma-first monitoring and innovative strategies for clinical practice and trial design.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"74"},"PeriodicalIF":6.5,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12276344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144668097","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-17DOI: 10.1038/s41523-025-00792-1
Chuang Zhao, Xin-Nan Zheng, Han-Ying Huang, Lin Tian
{"title":"Modeling tumor relapse using proliferation tracing and ablation transgenic mouse.","authors":"Chuang Zhao, Xin-Nan Zheng, Han-Ying Huang, Lin Tian","doi":"10.1038/s41523-025-00792-1","DOIUrl":"10.1038/s41523-025-00792-1","url":null,"abstract":"<p><p>Tumor relapse remains a significant obstacle to successful therapy. Preclinical animal models that accurately reflect tumor relapse in patients are urgently needed. Here, we employed a dual recombinase-mediated genetic system to genetically trace and ablate proliferating cells in a polyomavirus middle T antigen (PyMT)-induced spontaneous murine breast cancer model. This system enabled the acute ablation of cells that had undergone proliferation within a defined time window, resulting in a drastic tumor shrinkage, followed by a gradual tumor relapse due to the presence of residual low-cycling cells. We then applied single-cell RNA sequencing (scRNA-seq) to unbiasedly compare the tumor ecosystems of the primary and relapsed PyMT tumors. Compared with the primary tumors, the relapsed tumors exhibited a higher proportion of cancer stem cells and pro-tumor γδ T cells, as well as co-expression of Spp1 and Vegfa in multiple myeloid cell populations - features that predict poor therapeutic response and unfavorable outcomes in human breast cancer patients. Collectively, this proliferation tracing and ablation model emulates chemotherapies that preferentially eliminate proliferating cancer cells, serving as a robust tool and a valuable resource for testing novel therapeutic strategies in relapsed tumors.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"73"},"PeriodicalIF":6.5,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12271533/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144659713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
NPJ Breast CancerPub Date : 2025-07-17DOI: 10.1038/s41523-025-00791-2
Yee Hoon Foong, Qi Wang, Matthew Kearney, Hortense Le, Hua Guo, Diane Chen, Michelle Garlin Politis, Courtney F Connelly, Kevin Kalinsky, Rami S Vanguri, Eileen P Connolly
{"title":"Spatial biomarkers of response to eribulin plus pembrolizumab in patients with metastatic triple negative breast cancer in the ENHANCE-1 trial.","authors":"Yee Hoon Foong, Qi Wang, Matthew Kearney, Hortense Le, Hua Guo, Diane Chen, Michelle Garlin Politis, Courtney F Connelly, Kevin Kalinsky, Rami S Vanguri, Eileen P Connolly","doi":"10.1038/s41523-025-00791-2","DOIUrl":"10.1038/s41523-025-00791-2","url":null,"abstract":"<p><p>ENHANCE-1 was a phase Ib/II study which evaluated eribulin plus pembrolizumab as a treatment for metastatic triple negative breast cancer (mTNBC). All patients had measurable disease and up to 2 prior systemic treatments. We identified 142 patients with available samples and evaluated associations between the pre-treatment tumor-immune microenvironment and response using diagnostic H&Es and multiplexed immunofluorescence with 2 antibody panels. Markers were chosen to evaluate lymphocytes and myeloid cells including: CD4, CD8, FoxP3, CD56, CD20, CD68, CD163, Vimentin, and HLA-DR. While H&E-assessed computational and manual assessments of stromal tumor infiltrating lymphocytes (sTILs) did not associate with response, multiplex immunofluorescence revealed several significant associations. These include enrichment of stromal CD56+ in non-responders and higher stromal CD4 + CD8+ in non-responders in breast samples. Responders exhibited higher stromal macrophage populations CD68 + , CD68+Vimentin + , CD68 + CD163+Vimentin+ as well as higher stromal HLA-DR + . Our results suggest that further studies on immune cell populations other than T-cells as predictive biomarkers for combination therapies in mTNBC are warranted.</p>","PeriodicalId":19247,"journal":{"name":"NPJ Breast Cancer","volume":"11 1","pages":"72"},"PeriodicalIF":6.5,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12271393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144659714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}