1
|
Perry LM, Sevilimedu V, Polidorio N, Abuhadra N, Morrow M, Plitas G, Downs-Canner S. Predictors of Pathologic Complete Response with Neoadjuvant Chemo-Immunotherapy in Early-Stage Triple-Negative Breast Cancer. Ann Surg Oncol 2025; 32:3991-4001. [PMID: 40025324 PMCID: PMC12055475 DOI: 10.1245/s10434-025-17081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/07/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The combination of pembrolizumab with neoadjuvant chemotherapy (NAC) for triple-negative breast cancer (TNBC) improves pathologic complete response (pCR) rates and event-free survival. Yet it is unclear which patients benefit most from the addition of immunotherapy. This study aims to identify predictive factors for pCR in patients with TNBC receiving chemo-immunotherapy (chemo-IO). PATIENTS AND METHODS This single-institution retrospective analysis included 283 consecutive patients with TNBC treated with neoadjuvant chemo-IO from 1 June 2021 to 20 January 2023. The primary outcome was overall pCR; secondary outcomes were breast pCR and nodal pCR. Univariate and multivariable logistic regression models assessed for characteristics associated with overall, breast, or nodal pCR. RESULTS Most patients presented with cT2 (71%) cN0 (54%) disease. The overall pCR rate was 57%, breast pCR was 58%, and axillary pCR was 67% among biopsy-proven cN+ patients. Race, pathogenic BRCA mutations, backbone chemotherapy regimen, immune-related adverse events, and disruptions in immunotherapy were not associated with pCR. Univariate associations with overall pCR were younger age (p = 0.04), lower clinical T stage (p = 0.01), ductal histology (p < 0.001), poor differentiation (p < 0.001), and unifocality (p < 0.001). Breast and axillary pCR had similar associations. Nodal pCR also had univariate associations with normal body mass index (BMI) (p = 0.04) and absence of lymphovascular invasion (LVI) (p = 0.04). On multivariable analyses, ductal histology and unifocality remained independently associated with overall and breast pCR. CONCLUSIONS This analysis showed few clinical variables to be independently associated with pCR after neoadjuvant chemo-IO for TNBC. Thus, predicting chemo-IO response to personalize treatment and minimize morbidity may instead lie in ongoing basic and translational research to assess for useful biomarkers.
Collapse
Affiliation(s)
- Lauren M Perry
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Varadan Sevilimedu
- Biostatistics Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natalia Polidorio
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nour Abuhadra
- Breast Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Monica Morrow
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - George Plitas
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephanie Downs-Canner
- Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
2
|
Lafci O, Resch D, Santonocito A, Clauser P, Helbich T, Baltzer PAT. Role of imaging based response assesment for adapting neoadjuvant systemic therapy for breast cancer: A systematic review. Eur J Radiol 2025; 187:112105. [PMID: 40252279 DOI: 10.1016/j.ejrad.2025.112105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
PURPOSE The objective of this systematic review is to investigate the role of imaging in response monitoring during neoadjuvant systemic therapy (NST) for breast cancer and assess whether treatment modifications based on imaging response are implemented in clinical practice. METHODS A systematic review was conducted, analyzing five clinical practice guidelines and 147 clinical trial publications involving NST for breast cancer. The snowballing technique was employed, using a "start set" of clinical guidelines to trace relevant trials. Additionally, a PubMed search was conducted to identify trials published between 2023-2024. The review analyzed the use of imaging modalities, timing, and response criteria, and whether escalation, de-escalation, or change of treatment occurred based on imaging response. RESULTS Imaging was utilized in 81 % (119/147) of the trials, with ultrasound, MRI, and mammography being the most frequently employed modalities. Mid-treatment imaging was applied in 56 % (83/147) of the trials. However, only 15 % (22/147) of the trials implemented treatment modifications based on imaging response, highlighting the limited application of imaging response-guided therapy. No standardized imaging protocols or consistent response-guided treatment strategies were identified across the trials or clinical practice guidelines, with considerable variability in imaging methods, timing, and response criteria. CONCLUSION This systematic review underscores the critical need for standardized imaging protocols, response assessment criteria and image-guided treatment decisions. It is therefore evident that imaging for response monitoring during treatment should preferably be performed within clinical trials.
Collapse
Affiliation(s)
- Oguz Lafci
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Daphne Resch
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Ambra Santonocito
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Paola Clauser
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Thomas Helbich
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Pascal A T Baltzer
- Division of General and Pediatric Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
3
|
Tiberi E, Parisi A, Pistelli M, Savini A, Galassi F, Reschini C, Quintavalle D, Napoleoni R, Ferrari C, Berardi R. Immunotherapy in Triple-Negative Breast Cancer. Oncol Ther 2025:10.1007/s40487-025-00346-2. [PMID: 40418298 DOI: 10.1007/s40487-025-00346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
Currently, immunotherapy has led to a paradigmatic shift in the treatment of many cancer types, including triple-negative breast cancer. Immunotherapy increases the efficacy of the immune system in treating cancer, with a durable effect due to immunologic memory. The PD-1 inhibitor, pembrolizumab, combined with neoadjuvant chemotherapy, improved event-free survival and is a new standard of care for patients with high-risk, early stage triple-negative breast cancer (TNBC), regardless of tumor PD-L1 expression. For metastatic TNBC, pembrolizumab combined with chemotherapy is a new standard of care for first-line therapy for PD-L1+ metastatic TNBC, and it improves overall survival. The PD-L1 inhibitor, atezolizumab, combined with nab-paclitaxel, is also approved for first-line treatment of metastatic PD-L1+ TNBC. The aim of this review is to examine the existing evidence and ongoing studies on immunotherapy in patients with early stage and metastatic triple-negative breast cancer (TNBC), including new combination strategies with several drugs, such as chemotherapy, targeted therapy, or radiation and to discuss immune checkpoint inhibitor (ICI) applications and the possibility of emerging strategies in different TNBC stages.
Collapse
Affiliation(s)
- Elisa Tiberi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy.
| | - Alessandro Parisi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy.
| | - Mirco Pistelli
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | - Agnese Savini
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| | | | | | | | | | | | - Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, AOU delle Marche, Ancona, Italy
| |
Collapse
|
4
|
Ran R, Chen X, Yang J, Xu B. Immunotherapy in breast cancer: current landscape and emerging trends. Exp Hematol Oncol 2025; 14:77. [PMID: 40405250 PMCID: PMC12096519 DOI: 10.1186/s40164-025-00667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
Breast cancer remains one of the most prevalent malignancies worldwide, underscoring an urgent need for innovative therapeutic strategies. Immunotherapy has emerged as a transformative frontier in this context. In triple-negative breast cancer (TNBC), the combination of immunotherapy based on PD-1/PD-L1 immune checkpoint inhibitors (ICIs) with chemotherapy has proven efficacious in both early and advanced clinical trials. These encouraging results have led to the approval of ICIs for TNBC, opening up new therapeutic avenues for challenging-to-treat patient populations. Furthermore, a multitude of ongoing trials are actively investigating the efficacy of immunotherapy-based combinations, including ICIs in conjunction with chemotherapy, targeted therapy and radiation therapy, as well as other novel strategies such as bispecific antibodies, CAR-T cells and cancer vaccines across all breast cancer subtypes, including HR-positive/HER2-negative and HER2-positive disease. This review provides a comprehensive overview of current immunotherapeutic approaches in breast cancer, highlighting pivotal findings from recent clinical trials and the potential impact of these advancements on patient outcomes.
Collapse
Affiliation(s)
- Ran Ran
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xi Chen
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Yang
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Binghe Xu
- Cancer Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Melissari MT, Papatheodoridi A, Argyriadis A, Maria K, Athanasios DM, Zagouri F. Immunotherapy in primary hormone-receptor positive breast cancer: A systematic review. Crit Rev Oncol Hematol 2025:104768. [PMID: 40404033 DOI: 10.1016/j.critrevonc.2025.104768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/08/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025] Open
Abstract
Hormone-receptor positive (HR+), HER2 negative breast cancer (BC) is considered immunologically silent, thus investigation of immunotherapy in this subtype has evolved slower. This systematic review offers an overview of the clinical trials investigating the safety and efficacy of ICI in primary HR+/HER2- BC. Literature search was conducted up to October 30, 2022 to identify immunotherapy trials with checkpoint inhibitors in non-metastatic HR+/HER2- breast cancer. 39 trials were identified, mainly in early-phase clinical trials. None of the trials investigate ICI monotherapy and only 2 Phase 3 clinical trials are ongoing. Most trials investigate the use of ICI in the neoadjuvant setting in combination with chemotherapy. 18 trials have reported results and 6 of them efficacy results specifically for HR+/HER2- BC. ICI could be a promising therapeutic strategy for HR+/HER2- BC, however clinical benefit is restricted to subgroups of patients, depending on tumor molecular profile.
Collapse
Affiliation(s)
- Maria-Theodora Melissari
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, Athens, Greece
| | - Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, Athens, Greece
| | | | - Kaparelou Maria
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, Athens, Greece
| | - Dimopoulos Meletios Athanasios
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Medical School of National and Kapodistrian University of Athens, "Alexandra" General Hospital of Athens, Athens, Greece.
| |
Collapse
|
6
|
Pop CF, Ortega C, Lecomte M, Kristanto P, Khaled C, De Neubourg F, Desmet A, De Azambuja E, Larsimont D, Veys I. Impact of margin distance on recurrence and survival following breast-conserving surgery after neoadjuvant systemic therapy. NPJ Breast Cancer 2025; 11:45. [PMID: 40389463 PMCID: PMC12089308 DOI: 10.1038/s41523-025-00756-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
Current evidence does not support the application of "no-ink-on-tumor" negative margins following breast-conserving surgery (BCS) in breast cancer (BC) patients who have received neoadjuvant systemic treatment (NST). We compared loco-regional free survival (LRFS), disease-free survival (DFS), and overall survival (OS) based on different tumor margin distance thresholds in a cohort of 235 BC patients treated with NST and subsequent BCS between 01/2015 and 12/2019. The 5-year LRFS was 81.6% in patients with "no-ink-on-tumour", margins and 71.0% in those with positive margins (p = 0.584). Margins >1 mm were associated with superior outcomes, with a 5-year LRFS of 84.0% compared to 69.3% in patients with margins ≤1 mm (p = 0.005). Additionally, margins >1 mm were significantly correlated with longer DFS (p = 0.028) and OS (p = 0.001). These findings suggest that a surgical margin distance >1 mm provides the best LRFS, DFS, and OS outcomes for this group of BC patients.
Collapse
Affiliation(s)
- C Florin Pop
- Department of Surgical Oncology, Breast Clinic, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium.
| | - Clémence Ortega
- Department of Surgical Oncology, Breast Clinic, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Mathilde Lecomte
- Department of Surgical Oncology, Breast Clinic, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Paulus Kristanto
- Data Centre and Statistics Department, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Chirine Khaled
- Department of Pathology, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Filip De Neubourg
- Department of Surgical Oncology, Breast Clinic, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Antoine Desmet
- Department of Radiation Therapy, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Evandro De Azambuja
- Department of Medical Oncology, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Veys
- Department of Surgical Oncology, Breast Clinic, Institut Jules Bordet, The Brussels University Hospital (HUB), Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
7
|
Xie X, Chen L, Kong X, Huo Y, Huang W, Huang J, Zhang L, Jiang H, Gao J. Comparative efficacy and safety of PD-1 versus PD-L1 inhibitors in breast cancer treatment: A systematic review and meta analysis. Int J Cancer 2025; 156:1936-1949. [PMID: 40110878 DOI: 10.1002/ijc.35313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 03/22/2025]
Abstract
The comparative efficacy and safety of programmed death-ligand 1 (PD-L1) inhibitors versus programmed death protein 1 (PD-1) inhibitors in breast cancer treatment remain inconclusive, as no head-to-head randomized controlled trials (RCTs) conducted. This study aims to evaluate the efficacy and safety of PD-1/PD-L1 inhibitors as monotherapy or in combination with chemotherapy for breast cancer. A systematic review and meta-analysis were performed using major databases and oncology conference proceedings. The primary outcomes were overall survival (OS) for advanced breast cancer and pathological complete response (PCR) rate for early breast cancer. Secondary outcomes included progression-free survival (PFS) for advanced breast cancer and incidence of adverse events (AEs). Seventeen studies met the inclusion criteria, consisting of seven RCTs on early-stage and 10 on advanced breast cancer. For advanced breast cancer, PD-1/PD-L1 inhibitors modestly improved OS compared to chemotherapy, with no significant differences between PD-1 and PD-L1 inhibitors. PD-L1 inhibitors showed greater improvement in PFS compared to PD-1 inhibitors. The likelihood of AEs of any grade was higher with PD-L1 inhibitor treatment than with PD-1 inhibitor treatment. In early breast cancer, combining PD-1/PD-L1 inhibitors with chemotherapy inducing higher PCR rates than chemotherapy alone, with PD-1 inhibitors achieving better outcomes than PD-L1 inhibitors. PD-1 inhibitors were linked to slightly higher rates of grade >2 AEs compared to PD-L1 inhibitors. The findings indicate that PD-1 inhibitors may offer advantages for advanced breast cancer due to similar OS and a lower rate of AEs. For early breast cancer, PD-1 inhibitors are recommended given their superior PCR rates.
Collapse
Affiliation(s)
- Xintong Xie
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Lingzhu Chen
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Center of Biomedical Research, Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department 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
| | - Yujia Huo
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Center of Biomedical Research, Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, China
| | - Weiyuan Huang
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Center of Biomedical Research, Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, China
| | - Junjie Huang
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, Chinese University of Hong Kong, Hongkong, China
| | - Lin Zhang
- The School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Center of Biomedical Research, Suzhou Industrial Park Monash Research Institute of Science and Technology, Suzhou, China
| | - Hongnan Jiang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
- Department 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
| |
Collapse
|
8
|
Robinson TJ, Liveringhouse CL, Wilson C, Friedman S, Nakashima J, Mills MN, Purcell JD, Figura NB, Dongliang D, Thapa R, Welsh E, Ahmed KA, Grass GD, Fridley BL, Diaz R. Association between transcriptomic metrics of exogenous antigen presentation and adaptive immunity with locoregional recurrence in localized estrogen receptor negative breast cancer: retrospective review of multi-institutional datasets. Breast Cancer Res 2025; 27:77. [PMID: 40361136 PMCID: PMC12070507 DOI: 10.1186/s13058-025-01987-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 02/26/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Transcriptomic features of breast cancer locoregional recurrence (LRR) remain poorly understood. We therefore sought to investigate transcriptomic features associated with LRR in newly diagnosed invasive breast tumors from our institutional dataset. METHODS Transcriptomic profiling was performed on 632 tumors from consecutive patients treated within our health system for newly diagnosed non-metastatic breast cancer. Univariable Cox models identified genes whose expression was associated with LRR (q-value < 0.05). Up-regulated (UR) genes were defined as hazard ratio (HR) > 1 and down-regulated (DR) genes were defined as HR < 1. Gene set enrichment analyses were performed for UR and DR gene sets and validated within two external cohorts of ER- tumors. RESULTS With a median follow-up of 7.6 years, we observed 38 LRRs: 28/481 (5.8%) in ER + and 10/151 (6.6%) in ER-. There were 43 UR and 7 DR genes associated with LRR in ER + tumors, while 417 UR and 1150 DR genes were associated with LRR in ER- tumors. UR genes in ER + tumors were enriched for roles in cell proliferation (q < 0.05). In contrast, LRR in ER- tumors was most strongly associated with DR genes enriched for MHC-II-mediated antigen presentation and T cell activation (q < 0.05). In external cohorts of ER- tumors, 97 significant DR genes (p < 0.05) were enriched for 18 pathways, including 5 pathways involved in MHC-II signaling, antigen presentation and T-cell activation. CONCLUSIONS Transcriptomic patterns associated with LRR appear distinct between ER + and ER- tumors. In ER + tumors, LRR appears predominantly associated with proliferation, whereas ER- LRR suggests a robust pattern of suppressed antigen presentation via MHC-II.
Collapse
Affiliation(s)
- Timothy J Robinson
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Casey L Liveringhouse
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Christopher Wilson
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sam Friedman
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Justyn Nakashima
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT, USA
| | - Matthew N Mills
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jacob D Purcell
- Michigan State University College of Human Medicine, East Lansing, MI, USA
| | - Nicholas B Figura
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Du Dongliang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ram Thapa
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric Welsh
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kamran A Ahmed
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - G Daniel Grass
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Brooke L Fridley
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Roberto Diaz
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
9
|
Weiss A, Agnese DM, Al-Hilli Z, Cabioglu N, Farr D, Kantor O, Obeng-Gyasi S, Wilke L. An Overview of the Importance of Neoadjuvant Systemic Therapy for Breast Cancer Patients: From the Society of Surgical Oncology and the American Society of Breast Surgeons. Ann Surg Oncol 2025:10.1245/s10434-025-17405-7. [PMID: 40355803 DOI: 10.1245/s10434-025-17405-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Affiliation(s)
- Anna Weiss
- Division of Surgical Oncology, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| | - Doreen M Agnese
- Division of Surgical Oncology, The Ohio State University, Columbus, OH, USA
| | - Zahraa Al-Hilli
- Breast Center, Integrated Surgical Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Deborah Farr
- Department of Surgery at UT Southwestern Medical Center, Dallas, TX, USA
| | - Olga Kantor
- Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Lee Wilke
- UW Health/Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
10
|
Carnevale MG, Colciago RR, De Santis MC, Cortesi L, De Marco C, Marra A, Vingiani A, Nolè F, Curigliano G, Pruneri G, Llombart-Cussac A, Di Cosimo S, Cortes J. Advancing breast cancer therapy in the era of molecular diagnostics. Breast 2025; 82:104488. [PMID: 40424679 DOI: 10.1016/j.breast.2025.104488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/20/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Advances in cancer biology and drug development now enable treatments tailored to individual tumor profile. Targeting specific molecular alterations marked a significant step forward in cancer care, including breast cancer. Access to these therapies is improving thanks to the implementation of molecular tumor boards and efforts to provide molecular diagnostics at sustainable costs for all. In this context, we highlight recent progress in breast cancer therapy, focusing on biomarker-driven approaches, immunotherapy, and precision medicine paving the way for increasingly personalized and effective options.
Collapse
Affiliation(s)
| | | | | | - Laura Cortesi
- Azienda USL IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Cinzia De Marco
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Andrea Vingiani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Franco Nolè
- European Institute of Oncology IRCCS, Milano, Italy
| | - Giuseppe Curigliano
- European Institute of Oncology IRCCS, Milano, Italy; University "La Statale", Milano, Italy
| | - Giancarlo Pruneri
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy; University "La Statale", Milano, Italy
| | | | | | - Javier Cortes
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain; Universidad Europea de Madrid, Madrid, Spain
| |
Collapse
|
11
|
Wang X, Wang L, Liu Y. Current Status of Immune Checkpoint Inhibitors and Treatment Responsive Biomarkers for Triple-Negative Breast Cancer. Thorac Cancer 2025; 16:e70072. [PMID: 40324951 PMCID: PMC12052518 DOI: 10.1111/1759-7714.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for about 10%-20% of all breast cancer cases, is characterized by its aggressive nature, high recurrence rates, and poor prognosis. Unlike other breast cancer subtypes, TNBC lacks hormone receptors and specific molecular targets, limiting therapeutic options. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in treating TNBC by targeting immune evasion mechanisms. Despite these advancements, several issues remain unresolved, including low response rates in programmed cell death ligand 1 (PD-L1) negative TNBC subtypes and the challenge of predicting which patients will benefit from ICIs. Consequently, there is growing interest in identifying reliable biomarkers beyond PD-L1 expression. This review synthesizes recent studies to provide a comprehensive perspective on ICI therapy in TNBC, clarifying the status of single-agent ICI therapies and combination strategies, emphasizing the need for further research into biomarkers. These insights provide clues for more personalized and effective treatment approaches, ultimately aiming to improve clinical outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Xinran Wang
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Lingxia Wang
- Value & Implementation, Global Medical & Scientific AffairsMSD ChinaShanghaiChina
| | - Yueping Liu
- Department of PathologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| |
Collapse
|
12
|
Ahmed KA, Kim Y, Kim S, Wang MH, DeJesus M, Arrington JA, Soyano AE, Armaghani AJ, Costa RLB, Loftus LS, Rosa M, Caudell JJ, Diaz R, Etame AB, Tran ND, Soliman H, Czerniecki BJ, Forsyth PA, Yu HHM, Han HS. Nivolumab and stereotactic radiosurgery for patients with breast cancer brain metastases: long-term results and biomarker analysis from a non-randomized, open-label, phase Ib study. J Immunother Cancer 2025; 13:e011432. [PMID: 40295143 PMCID: PMC12039042 DOI: 10.1136/jitc-2024-011432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND We hypothesized treatment with nivolumab and stereotactic radiosurgery (SRS) would be feasible, well tolerated, and may improve intracranial tumor control over SRS alone for breast cancer brain metastases (BCBM). METHODS The study is a phase Ib trial of nivolumab and SRS for BCBM. CLINICAL TRIAL INFORMATION NCT03807765. Key eligibility criteria include BCBM of all subtypes, age ≥18, Eastern Cooperative Oncology Group Performace Status (ECOG-PS)≤2 with ≤10 brain metastases. Treatment was initiated with a dose of nivolumab (480 mg intravenously) that was repeated every 4 weeks. The initial dose of nivolumab was followed 1 week later by SRS. Blood was collected at baseline and every 4 weeks for flow cytometry and cell-free DNA (cfDNA) assessment. RESULTS A total of 12 patients received SRS to 17 brain metastases. Breast cancer subtypes included triple negative (50%), hormone receptor (HR)+/HER2- (33%), and HR-/HER2+ (17%). Median follow-up from start of protocol therapy is 56 months. No cases of radionecrosis were noted. Two lesions were noted to undergo local failure, both pathologically confirmed, for a 12-month local control of 94%. Median distant intracranial control was 7.4 months with a 12-month control rate of 33%. Median systemic progression-free survival was 7.7 months with a 12-month rate of 42%. Median overall survival (OS) was 24.7 months with a 12-month OS of 75%. Most patients were noted to have an increase in cfDNA throughout study treatment, at week 5 compared with baseline (83%), week 25 compared with baseline (89%), and 100% at first follow-up. Intracranial control was associated with lower levels of CD4 regulatory T cells (Treg) (p=0.03) and higher levels of CD4 T effector memory (p=0.04). CONCLUSIONS Nivolumab and SRS is a safe and feasible treatment option in BCBM. Long-term follow-up revealed no cases of radiation necrosis. TRIAL REGISTRATION NUMBER NCT03807765.
Collapse
Affiliation(s)
- Kamran A Ahmed
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Younchul Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, USA
| | - Sungjune Kim
- Department of Radiation Oncology, Mayo Clinic Jacksonville Campus, Jacksonville, Florida, USA
| | - Min Hsuan Wang
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Michelle DeJesus
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - John A Arrington
- Department of Radiology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Aixa E Soyano
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Avan J Armaghani
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Ricardo L B Costa
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Loretta S Loftus
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Marilin Rosa
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Jimmy J Caudell
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Roberto Diaz
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Arnold B Etame
- Department of Neuro Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Nam D Tran
- Department of Neuro Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Hatem Soliman
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Brian J Czerniecki
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Peter A Forsyth
- Department of Neuro Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Hyo S Han
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
13
|
Tauber N, Amann N, Dannehl D, Deutsch TM, Dimpfl M, Fasching P, Hartkopf A, Heublein S, Hilmer L, Hörner M, Krawczyk N, Krückel A, Krug D, Marmé F, Michel LL, Reinisch M, Rody A, Schäffler H, Schneeweiss A, Utz D, Veselinovic K, Banys-Paluchowski M. Therapy of early breast cancer: current status and perspectives. Arch Gynecol Obstet 2025:10.1007/s00404-025-08028-0. [PMID: 40261372 DOI: 10.1007/s00404-025-08028-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Medical advancements in breast cancer are truly remarkable. Especially in recent years, numerous new therapeutics have been approved and surgical strategies have been de-escalated for specific patient groups. In the therapeutic setting, CDK4/6 inhibitors as oral maintenance therapy in early breast cancer and immune checkpoint inhibitors (Pembrolizumab) for triple-negative breast cancer (BC) are noteworthy. In the surgical field, prospective randomized controlled trials have currently explored the possibility to deescalate axillary surgery by omitting sentinel lymph node excision (INSEMA, SOUND). As a result, there have been significant improvements in prognosis and a reduction in surgical morbidity for patients. Many exciting trials are underway, and it remains to be seen whether antibody-drug conjugates beyond trastuzumab emtansine, will find their way into the treatment lines for early-stage BC. Furthermore, the integration of artificial intelligence in both diagnostics and treatment recommendation evaluation is a promising area with great potential.
Collapse
Affiliation(s)
- Nikolas Tauber
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany.
| | - Niklas Amann
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Dominik Dannehl
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Thomas M Deutsch
- Department of Obstetrics and Gynecology, University Hospital Heidelberg, Heidelberg, Germany
| | - Moritz Dimpfl
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Peter Fasching
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Andreas Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Sabine Heublein
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Lisbeth Hilmer
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Manuel Hörner
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Natalia Krawczyk
- Department of Obstetrics and Gynecology, University Hospital Duesseldorf, 40225, Duesseldorf, Germany
| | - Annika Krückel
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - David Krug
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Laura L Michel
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center Heidelberg, 69120, Heidelberg, Germany
| | - Mattea Reinisch
- Department of Obstetrics and Gynecology, Medical Faculty Mannheim, University Medical Center Mannheim, University of Heidelberg, Mannheim, Deutschland
| | - Achim Rody
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Henning Schäffler
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center Heidelberg, 69120, Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - David Utz
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University Hospital Tuebingen, 72016, Tuebingen, Germany
| | - Kristina Veselinovic
- Department of Obstetrics and Gynecology, University Hospital Ulm, 89075, Ulm, Germany
| | - Maggie Banys-Paluchowski
- Department of Obstetrics and Gynecology, University Hospital Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| |
Collapse
|
14
|
Gao YX, Guo XJ, Lin B, Huang XB, Tu RH, Lin M, Cao LL, Chen QY, Wang JB, Xie JW, Li P, Zheng CH, Yang YH, Huang CM, Lin JX. Targeting LHPP in neoadjuvant chemotherapy resistance of gastric cancer: insights from single-cell and multi-omics data on tumor immune microenvironment and stemness characteristics. Cell Death Dis 2025; 16:306. [PMID: 40240758 PMCID: PMC12003742 DOI: 10.1038/s41419-025-07614-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Gastric cancer (GC) is a highly heterogeneous and complex malignancy, often characterized by tumor stemness and immune evasion mechanisms, which contribute to a poor response to neoadjuvant chemotherapy (NAC) and treatment resistance. In this study, we performed a comprehensive analysis using single-cell and multi-omics approaches on 375 GC samples from The Cancer Genome Atlas database, along with 141 clinical samples from patients who underwent NAC. We identified key gene modules associated with stemness and immune traits, and developed a novel stem cell-immune risk score. This score effectively distinguished responders from non-responders to chemotherapy, and was significantly associated with overall survival. Through multi-omics analysis, we further elucidated the role of phospholysine phosphohistidine inorganic pyrophosphatase (LHPP) in the tumor immune microenvironment. Our findings showed that high LHPP expression was closely linked to the increased infiltration of antitumor immune cells, such as CD8+ T cells, and significantly suppressed the development of stemness characteristics in GC. Additionally, single-cell sequencing data revealed that tumor epithelial cells with low LHPP expression exhibited heightened stemness and demonstrated the strongest communication with CD8+-exhausted T cells. We also observed that LHPP inhibited stemness and chemotherapy resistance in GC cells by regulating the phosphorylation of GSK-3β. In conclusion, LHPP plays a critical regulatory role in the stemness features and tumor immune microenvironment of GC, presenting a promising biomarker and potential therapeutic target for personalized treatment of GC.
Collapse
Affiliation(s)
- You-Xin Gao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Xiao-Jing Guo
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Bin Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Xiao-Bo Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Ru-Hong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China
| | - Ying-Hong Yang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China.
- Gastrointestinal Cancer Institute, Fujian Medical University, Fuzhou, 350001, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China.
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumour Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
15
|
Garrido-Cano I, Tapia M, Carbonell J, Peña C, Torres-Ruiz S, Ágreda-Roca A, Lameirinhas A, Tebar C, Burgués O, Hernando C, Lluch A, Brasó-Maristany F, Prat A, Bermejo B, Martínez MT, Cejalvo JM. Deciphering the transcriptomic landscape of early HR +/HER2 - breast cancer in very young women. Cancer Commun (Lond) 2025. [PMID: 40207642 DOI: 10.1002/cac2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Affiliation(s)
- Iris Garrido-Cano
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Valencia, Spain
| | - Marta Tapia
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | | | - Carlos Peña
- Biomedical Research Institute INCLIVA, Valencia, Spain
| | | | | | | | | | - Octavio Burgués
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
- Pathology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Cristina Hernando
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Ana Lluch
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Department of Medicine, Universitat de València, Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), Valencia, Spain
| | - Fara Brasó-Maristany
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain
- Institute of Cancer and Blood Disorders, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapies in Solid Tumors Group, August Pi I Sunyer Biomedical Research Institute, Barcelona, Spain
- Institute of Cancer and Blood Disorders, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Begoña Bermejo
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
- Department of Medicine, Universitat de València, Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), Valencia, Spain
| | - María Teresa Martínez
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
| | - Juan Miguel Cejalvo
- Biomedical Research Institute INCLIVA, Valencia, Spain
- Medical Oncology Department, Hospital Clínico Universitario of Valencia, Valencia, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), Valencia, Spain
| |
Collapse
|
16
|
Yu L, Liebenberg K, Shen Y, Liu F, Xu Z, Hao X, Wu L, Zhang W, Chan HL, Wei B, Lorenzi PL, Gao Y, Bado I, Becerra-Dominguez L, Rivas CH, Aguirre S, Pingel BC, Wu YH, Ding Y, Liu J, Edwards DG, Eberlin LS, Zhang XHF. Tumor-derived arachidonic acid reprograms neutrophils to promote immune suppression and therapy resistance in triple-negative breast cancer. Immunity 2025; 58:909-925.e7. [PMID: 40157359 PMCID: PMC11981829 DOI: 10.1016/j.immuni.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/08/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
The combination of immune checkpoint blockade and chemotherapies is the standard of care for triple-negative breast cancer (TNBC). However, initially, responsive tumors can still develop recurrences, suggesting acquired resistance mechanisms that remain poorly understood. Herein, we discover that TNBC cells surviving anti-programmed cell death protein-1 (anti-PD-1) and chemotherapy treatment accumulate neutral lipids. Disrupting lipid droplet formation in cancer cells reverses resistance and mitigates the immunosuppressive microenvironment. Single-cell RNA sequencing reveals a subset of neutrophils exhibiting a lipid-laden phenotype similar to adjacent tumor cells. Mechanistically, tumor-derived extracellular vesicles carrying lipids, including arachidonic acid (AA), mediate neutrophil reprogramming. Blocking dietary intake of omega-6 fatty acids or inhibiting fatty acid elongation for AA synthesis restores anti-tumor immunity and re-sensitizes the resistant tumors to anti-PD-1 and chemotherapy treatment. In human patients, AA metabolism-related pathways correlates with neutrophil enrichment. Overall, we demonstrate how lipid accumulation in TNBC cells leads to immune suppression and therapy resistance.
Collapse
Affiliation(s)
- Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Keziah Liebenberg
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Igor Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luis Becerra-Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bradley C Pingel
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Livia S Eberlin
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
17
|
Corti C, Koca B, Rahman T, Mittendorf EA, Tolaney SM. Recent Advances in Immune Checkpoint Inhibitors for Triple-Negative Breast Cancer. Immunotargets Ther 2025; 14:339-357. [PMID: 40196378 PMCID: PMC11974553 DOI: 10.2147/itt.s495751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/09/2025] Open
Abstract
While immunotherapy has transformed treatment across various cancers, its impact on breast cancer is relatively limited. Recent advances have established immunotherapy as an effective approach for triple-negative breast cancer (TNBC), an aggressive subtype with limited therapeutic targets and poor prognosis. Specifically, pembrolizumab, an immune checkpoint inhibitor (ICI), is now approved for both first-line metastatic and early-stage TNBC. In metastatic TNBC, combining ICIs with chemotherapy, particularly pembrolizumab, has demonstrated survival benefits in patients with PD-L1-positive disease. However, extending these benefits to broader populations has proven challenging, highlighting the need for better patient selection and novel strategies. Emerging approaches include combining ICIs with antibody-drug conjugates, PARP inhibitors, dual ICIs, and bispecific antibodies targeting angiogenesis and immune checkpoints. These strategies aim to overcome resistance and expand immunotherapy's efficacy beyond the PD-1/PD-L1 pathway. In early-stage disease, pembrolizumab combined with chemotherapy in the neoadjuvant setting has significantly improved pathologic complete response, event-free survival, and overall survival, establishing a new standard of care. Ongoing research aims to determine the optimal timing for ICI administration, explore less toxic chemotherapy backbones, utilize biomarkers for personalized treatment, and assess whether adding complementary treatments, such as radiation therapy for high-risk cases, can improve outcomes. This review examines the successes and setbacks of ICI use in TNBC, offering a comprehensive overview of current practices and future directions. It emphasizes optimizing ICI timing, leveraging biomarkers, and integrating novel agents to refine treatment approaches for both metastatic and early-stage TNBC. As immunotherapy continues to evolve, future research must address the unmet needs of this challenging breast cancer subtype, offering hope for improved outcomes.
Collapse
Affiliation(s)
- Chiara Corti
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Beyza Koca
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Tasnim Rahman
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Breast Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
18
|
Xiang H, Tu B, Feng X, Chen L, Huang Y. Dual inhibition of TYK2 and PD-L1 boosts immune response in triple negative breast cancer. Anticancer Drugs 2025; 36:280-289. [PMID: 39774369 PMCID: PMC11884794 DOI: 10.1097/cad.0000000000001685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 01/11/2025]
Abstract
Recent studies have shown that Janus Kinase inhibitors can enhance the tumor therapeutic effect of immune checkpoint inhibitors. However, it remains to be studied whether TYK2 selective inhibitors can enhance the therapeutic effect of small molecule PD-L1 inhibitors in triple-negative breast cancer (TNBC). We verified the efficacy of the combination of the selective TYK2 inhibitor Deucravacitinib and the small molecule inhibitor of PD-L1, INCB086550, in two TNBC animal models: a syngeneic mouse model (4T1 with humanized PD-L1) and a peripheral blood mononuclear cell (PBMC)-humanized model (MDA-MB-231). Following that, we explored the regulation of immune cell activity in tumors by the combined treatment using flow cytometry. Finally, we validated the expression of genes related to the regulated immune cells through reverse transcription-PCR. Both animal models demonstrated that the addition of a TYK2 inhibitor to a PD-L1 inhibitor significantly enhanced the antitumor capabilities of mice with good safety profiles. The combined therapy significantly elevated the counts of T, B, and natural killer cells while concurrently diminishing myeloid-derived suppressor cells in the syngeneic model. Similarly, in the PBMC-humanized model, this therapy markedly augmented progenitor-like and proliferative precursor-like CD8 T cells, while effectively diminishing exhausted and terminally differentiated CD8 T cell populations. This enhanced antitumor effect is associated with the modulation of antitumor immune-related gene expression by the combined therapy. The combination of TYK2 inhibitors and immune checkpoint inhibitors is a potentially effective strategy for treating TNBC.
Collapse
Affiliation(s)
- Huali Xiang
- Physical Examination Department, Jiangxi Maternal and Child Health Hospital
| | - Binfeng Tu
- Neurosurgery Department, Jiangxi Cancer Hospital
| | - Xin Feng
- Hospital Health Education Department
| | | | - Yajuan Huang
- Galactophore Department, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
19
|
Ye Y, Zhang Z, Zhao H, Zhao B. A system review of neoadjuvant immune checkpoint blockade for breast cancer. Front Immunol 2025; 16:1537926. [PMID: 40213551 PMCID: PMC11983617 DOI: 10.3389/fimmu.2025.1537926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 02/27/2025] [Indexed: 04/19/2025] Open
Abstract
Background The clinical application of immune checkpoint blockade (ICB)-based neoadjuvant therapy has been approved in breast cancer since 2021. However, no studies have evaluated its efficacy and safety in randomized and non-randomized settings. Additionally, there exists controversy about which specific subpopulation can benefit from this management strategy. Methods We searched MEDLINE and EMBASE databases for prospective clinical trials of ICB-based neoadjuvant therapy in breast cancer. Information regarding pathological complete response (pCR), event-free survival (EFS), overall survival (OS), and treatment-related adverse event (TRAE) were pooled to estimate the efficacy and safety. Hazard ratio, relative risk (RR) and their 95% confidence intervals (CIs) were calculated. Results Among 22 eligible trials including 6134 women with resectable breast cancer, there were 11 randomized studies with 5574 patients. Pooled analysis on pCR (RR, 1.38; 95% CI, 1.20-1.58; P<0.001), EFS (hazard ratio, 0.67; 95% CI, 0.54-0.81; P<0.001), and OS (hazard ratio, 0.56; 95% CI, 0.35-0.91; P=0.01) revealed that ICB-based neoadjuvant therapy was associated with favorable outcomes over conventional treatment. Moreover, the benefits of EFS were independent of PD-L1 expression (Pinteraction =0.57) and pCR (Pinteraction =0.37) in neoadjuvant immunotherapy. However, combining ICB with conventional neoadjuvant treatment significantly increased the risk of high-grade TRAE (RR, 1.06; 95% CI, 1.01-1.12; P=0.03), serious TRAE (RR, 1.57; 95% CI, 1.26-1.94; P<0.001), treatment discontinuation due to TRAE (RR, 1.47; 95% CI, 1.14-1.90; P=0.003), and potentially fatal adverse event (RR, 2.25; 95% CI, 0.80-6.31; P=0.12). Conclusion The combination of ICB with conventional neoadjuvant treatment is associated with favorable clinical outcomes and importantly, increased grade 3+ toxicities. Clinicians should meticulously monitor patients to minimize the risk of treatment discontinuation in individuals with potentially curable breast cancer.
Collapse
Affiliation(s)
- Yanle Ye
- Central Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Zhishan Zhang
- Central Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Hong Zhao
- The Cancer Center of The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Bin Zhao
- Central Laboratory, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
20
|
Baek SH, Lee MJ, Kook Y, Bae SJ, Jeong J, Cha YJ, Ahn SG. Pathological complete response, histologic grade, and level of stromal tumor-infiltrating lymphocytes in ER + HER2- breast cancer. Breast Cancer Res 2025; 27:42. [PMID: 40114292 PMCID: PMC11927358 DOI: 10.1186/s13058-025-01999-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Recent trials have integrated immune checkpoint inhibitors (ICIs) into neoadjuvant chemotherapy (NAC) in patients with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer of histologic grade (HG) III. We assessed the pathological complete response (pCR) rate according to the level of stromal tumor-infiltrating lymphocytes (sTIL) and HG in patients with ER + HER2- breast cancer undergoing NAC. METHODS Between January 2016 and December 2023, we retrospectively identified 376 patients with ER + HER2- breast cancer who underwent NAC followed by surgery. HG and sTIL levels were examined in the biopsied samples before NAC. Multiple sTIL cutoff values as 10%, 20%, and 30% were applied. RESULTS Twenty-seven patients (7.2%) had HG III tumors. The pCR rate in the HG III group was 22.2%, which was significantly higher than that in the HG I/II group (4.0%) (p < 0.001). The HG III group had a higher mean sTIL level than HG I/II group (38.7% vs. 12.9%; p < 0.001). According to the sTIL levels, the pCR rate in the high sTIL group was significantly higher than that in the low sTIL group: i) cutoff of 10%, 2.4% vs. 9.5%; cutoff of 20%, 2.8% vs. 13.7%; and cutoff of 30%, 3.2% vs. 18.3%. In the high sTIL (≥ 30%) group, the pCR rate for HG III was 33.3%, whereas that for HG I/II was 13.3%. CONCLUSIONS High tumor grade and sTIL levels were associated with higher rates of pCR in ER + HER2- breast cancer. Our findings support that the addition to ICIs to NAC increased pCR in high-risk, HG III, ER + HER2- breast cancer and suggest that sTIL levels could be utilized to identify patients with ER + HER2- breast cancer eligible for chemoimmunotherapy.
Collapse
Affiliation(s)
- Seung Ho Baek
- Department of Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Ji Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoonwon Kook
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soong June Bae
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Sung Gwe Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Tinoco JC, Saunders HI, Werner LR, Sun X, Chowanec EI, Heard A, Chalise P, Vahrenkamp JM, Wilson AE, Liu CX, Lei G, Wei J, Cros H, Mohammed H, Troester M, Perou C, Markiewicz MA, Gertz J, Balko JM, Hartman ZC, Hagan CR. Progesterone receptor-dependent downregulation of MHC class I promotes tumor immune evasion and growth in breast cancer. J Immunother Cancer 2025; 13:e010179. [PMID: 40102028 PMCID: PMC11927445 DOI: 10.1136/jitc-2024-010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/16/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Breast cancer (BC) continues to be a major health concern with 250,000 new cases diagnosed annually in the USA, 75% of which are hormone receptor positive (HR+), expressing estrogen receptor alpha (ER) and/or the progesterone receptor (PR). Although ER-targeted therapies are available, 30% of patients will develop resistance, underscoring the need for new non-ER/estrogen-based treatments. Notably, HR+BCs exhibit poor lymphocyte infiltration and contain an immunosuppressive microenvironment, which contributes to the limited efficacy of immunotherapies in HR+BC. In this study, we demonstrate that PR/progesterone signaling reduces major histocompatibility complex (MHC) Class I expression, facilitating immune evasion and escape from immune-based clearance of PR+tumors. METHODS To determine the effect of PR/progesterone on MHC Class I expression, we treated human and mouse mammary tumor cell lines with progesterone and/or interferon (IFN) and measured expression of genes involved in antigen processing and presentation (APP), as well as surface MHC Class I expression. We used the OT-I/SIINFEKL model antigen system to measure the impact of progesterone on immune cell-mediated killing of modified tumor cells. We also analyzed two large BC clinical cohorts to determine how PR expression correlates with APP gene expression and MHC Class I expression in ER-positive tumors. RESULTS In vitro, we show that PR/progesterone signaling reduces APP gene expression and MHC class I expression in human and breast mammary tumor cell lines. PR-mediated attenuation of APP/MHC Class I expression is more pronounced in the presence of IFN. In immune cell killing assays, PR-expressing mammary tumor cells treated with progesterone are protected from immune-mediated cytotoxicity. We demonstrate that PR expression in vivo prevents immune-mediated rejection of xenoantigen-modified mammary tumor cell lines through mechanisms involving MHC Class I expression and CD8 T cells. Data analysis of two large BC cohorts reveals lower APP gene expression and MHC Class I expression in ER/PR-positive tumors compared with ER-positive/PR-negative tumors. These findings show that HR+BCs, specifically PR+tumors, downregulate APP/MHC class I machinery through PR/progesterone signaling. Use of pharmacological PR/progesterone inhibitors may reverse these effects in patients with BC, thereby improving immunosurveillance and response to immunotherapies.
Collapse
Affiliation(s)
- Julio C Tinoco
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Harmony I Saunders
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Lauryn Rose Werner
- Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Xiaopeng Sun
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Eilidh I Chowanec
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Amanda Heard
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Prabhakar Chalise
- Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jeffery M Vahrenkamp
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Andrea E Wilson
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Junping Wei
- Surgery, Duke University, Chapel Hill, North Carolina, USA
| | - Hugo Cros
- Oregon Health & Science University, Portland, Oregon, USA
| | | | - Melissa Troester
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Charles Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mary A Markiewicz
- Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Christy R Hagan
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biochemistry and Molecular Biology, The University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
22
|
He J, Liu N, Zhao L. New progress in imaging diagnosis and immunotherapy of breast cancer. Front Immunol 2025; 16:1560257. [PMID: 40165974 PMCID: PMC11955504 DOI: 10.3389/fimmu.2025.1560257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Breast cancer (BC) is a predominant malignancy among women globally, with its etiology remaining largely elusive. Diagnosis primarily relies on invasive histopathological methods, which are often limited by sample representation and processing time. Consequently, non-invasive imaging techniques such as mammography, ultrasound, and Magnetic Resonance Imaging (MRI) are indispensable for BC screening, diagnosis, staging, and treatment monitoring. Recent advancements in imaging technologies and artificial intelligence-driven radiomics have enhanced precision medicine by enabling early detection, accurate molecular subtyping, and personalized therapeutic strategies. Despite reductions in mortality through traditional treatments, challenges like tumor heterogeneity and therapeutic resistance persist. Immunotherapies, particularly PD-1/PD-L1 inhibitors, have emerged as promising alternatives. This review explores recent developments in BC imaging diagnostics and immunotherapeutic approaches, aiming to inform clinical practices and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Jie He
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Nan Liu
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li Zhao
- Department of Radiology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
23
|
Dent R, Cortés J, Park YH, Muñoz-Couselo E, Kim SB, Sohn J, Im SA, Holgado E, Foukakis T, Kümmel S, Yearley J, Wang A, Nebozhyn M, Huang L, Cristescu R, Jelinic P, Karantza V, Schmid P. Molecular determinants of response to neoadjuvant pembrolizumab plus chemotherapy in patients with high-risk, early-stage, triple-negative breast cancer: exploratory analysis of the open-label, multicohort phase 1b KEYNOTE-173 study. Breast Cancer Res 2025; 27:35. [PMID: 40069763 PMCID: PMC11895130 DOI: 10.1186/s13058-024-01946-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/09/2024] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The multicohort, open-label, phase 1b KEYNOTE-173 study was conducted to investigate pembrolizumab plus chemotherapy as neoadjuvant therapy for triple-negative breast cancer (TNBC). This exploratory analysis evaluated features of the tumor microenvironment that might be predictive of response. METHODS Cell fractions from 20 paired samples collected at baseline and after one cycle of neoadjuvant pembrolizumab prior to chemotherapy initiation were analyzed by spatial localization (tumor compartment, stromal compartment, or sum of tumor and stromal compartments [total tumor]) using three six-plex immunohistochemistry panels with T-cell, myeloid cell, and natural killer cell components. Area under the receiver operating characteristic curve (AUROC) was used to assess associations between immune subsets and gene expression signatures (T-cell-inflamed gene expression profile [TcellinfGEP] and 10 non-TcellinfGEP signatures using RNA sequencing) and pathologic complete response (pCR). RESULTS At baseline, six immune subsets quantitated within the tumor compartment showed AUROC with 95% CIs not crossing 0.5, including CD11c+ cells (macrophage and dendritic cell [DC]: AUROC, 0.85; 95% confidence interval [CI] 0.63-1.00), CD11c+/MHCII+/CD163-/CD68- cells (DC: 0.76; 95% CI, 0.53-0.99), CD11c+/MHCII-/CD163-/CD68- cells (nonactivated/immature DC: 0.80; 95% CI 0.54-1.00), and CD11c+/CD163+ cells (M2 macrophage: 0.77; 95% CI 0.55-0.99). Other associations with pCR included baseline CD11c+/MHCII-/CD163-/CD68- (nonactivated/immature DC) within the total tumor (AUROC, 0.76; 95% CI 0.51-1.00) and the baseline CD11c/CD3 ratio within the tumor compartment (0.75; 95% CI 0.52-0.98). Changes in immune subsets following one cycle of pembrolizumab were not strongly associated with pCR. Although T-cell associations were relatively weak, specific CD8 subsets trended toward association. The AUROC for discriminating pCR based on TcellinfGEP was 0.55 (95% CI 0.25-0.85); when detrended by TcellinfGEP, AUROC varied for the non-TcellinfGEP signatures. TcellinfGEP expression trended higher in responders than in nonresponders when evaluating pCR. CONCLUSIONS Myeloid cell populations within the tumor compartment at baseline and TcellinfGEP show a promising trend toward an association with pCR in a small subgroup of patients with early-stage TNBC treated with neoadjuvant pembrolizumab plus chemotherapy. TRIAL REGISTRATION ClinicalTrials.gov, NCT02622074; registration date, December 2, 2015.
Collapse
Affiliation(s)
- Rebecca Dent
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Blvd, Singapore, 168583, Singapore.
| | - Javier Cortés
- Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- International Breast Cancer Center, Quironsalud Group, Barcelona, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, European University of Madrid, Madrid, Spain
| | - Yeon Hee Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eva Muñoz-Couselo
- Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron Hospital, Barcelona, Spain
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joohyuk Sohn
- Department of Internal Medicine, Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Esther Holgado
- Medical Oncology Service, Ramón y Cajal University Hospital, Madrid, Spain
| | - Theodoros Foukakis
- Department of Oncology-Pathology, Karolinska Comprehensive Cancer Center, Karolinska Institute and Breast Cancer Centre, Cancer Theme, Karolinska University Hospital, Solna, Sweden
| | - Sherko Kümmel
- Interdisciplinary Breast Unit, Essen-Mitte Clinics, Essen, and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| |
Collapse
|
24
|
Sun Z, Liu C, Yao Y, Gao C, Li H, Wang L, Li Y, Sun C. Therapeutic potential of triple-negative breast cancer immune checkpoint blockers: A 21-year bibliometric analysis. Medicine (Baltimore) 2025; 104:e41739. [PMID: 40068043 PMCID: PMC11903016 DOI: 10.1097/md.0000000000041739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive metastatic subtype of BC that frequently develops chemoresistance. Immune checkpoint blockers (ICB) have led to breakthroughs in TNBC treatment. This study aimed to explore research trends and public interest in ICB interventions for TNBC. METHODS We searched the Web of Science Core Collection (WoSCC) database for publications related to ICB for TNBC from 2003 to 2024. VOSviewer, CiteSpace, and R package "bibliometrix" were used to analyze the characteristics of ICB publications in TNBC from a quantitative and qualitative perspective and to visualize the results to comprehensively present the research trends in this field. RESULTS After removing duplicates, 2698 publications were included. The New England Journal of Medicine may be the leading and influential in the field of ICB in TNBC according to data on the total number of publications, number of citations, and impact factors. Its article entitled "Atezolizumab and Nab-Paclitaxel in Advanced TNBC" is 1 of the most cited articles. Keyword analysis showed that current research hotspots in this field are tumor microenvironment, complete pathological response, neoadjuvant chemotherapy, and PARP inhibitors. Future research hotspots may include the PD-L1 inhibitor durvalumab and antibody-drug conjugates (ADC). CONCLUSIONS This study revealed that ICB therapy for TNBC is a rapidly evolving and high-profile topic. Future research should focus on the optimal selection of different targets for ICB in combination with neoadjuvant chemotherapy, ADC, and poly ADP-ribose polymerase inhibitors to treat TNBC.
Collapse
Affiliation(s)
- Zhongli Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Traditional Chinese Medicine, Chongqing Three Gorges Medical College, Chongqing, China
| | - Cun Liu
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Yan Yao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chundi Gao
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Huayao Li
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Longyun Wang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Changgang Sun
- Department of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China,
| |
Collapse
|
25
|
Liang Z, Li S, Pan Z, Duan Y, Ouyang Q, Zhu L, Song E, Chen K. Profiling Multiple CD8+ T-cell Functional Dimensions Enhances Breast Cancer Immune Assessment. Cancer Immunol Res 2025; 13:337-352. [PMID: 39715293 DOI: 10.1158/2326-6066.cir-24-0235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/19/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
CD8+ T-cell abundance is insufficient to assess antitumor immunity and shows poor performance in predicting breast cancer prognosis and immunotherapy response, presumably owing to the complexity of CD8+ T-cell functionalities. Although single-cell RNA sequencing can dissect the multifaceted functions of CD8+ T cells for better immune assessment, its clinical application is limited. In this study, we developed bulk RNA sequencing-based FuncDimen models from integrative analysis of single-cell RNA sequencing and matched bulk RNA sequencing data to evaluate CD8+ T-cell functionalities across five dimensions: tumor reactivity, cytotoxicity, IFNγ secretion, proliferation, and apoptosis. The FuncDimen models quantifying different functional dimensions of CD8+ T cells were validated in our breast cancer cohort and external databases using immunofluorescence and imaging mass cytometry. We calculated the FuncAggre score by weighted aggregation of all five FuncDimen models to encapsulate the overall antitumor immunity. In our breast cancer cohort and external databases, the FuncAggre score demonstrated superior predictive performance for breast cancer prognosis (time-dependent AUC: 0.56-0.70) and immunotherapy response (AUC: 0.71-0.83) over other immune biomarkers, regardless of the breast cancer molecular subtype. Together, the FuncDimen models offer a refined assessment of antitumor immunity mediated by CD8+ T cells in the clinic, enhancing prognostic prediction and aiding personalized immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Zhuozhi Liang
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Zenith Institute of Medical Sciences, Guangzhou, China
| | - Shunrong Li
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhilong Pan
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuanqiang Duan
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qian Ouyang
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Liling Zhu
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Erwei Song
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Zenith Institute of Medical Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kai Chen
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Artificial Intelligence Lab, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
| |
Collapse
|
26
|
Ramalingam K, Woody R, Glencer A, Schwartz CJ, Mori H, Wong J, Hirst G, Rosenbluth J, Onishi N, Gibbs J, Hylton N, Borowsky AD, Campbell M, Esserman LJ. Intratumoral Injection of mRNA-2752 and Pembrolizumab for High-Risk Ductal Carcinoma In Situ: A Phase 1 Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:288-292. [PMID: 39821301 PMCID: PMC11926637 DOI: 10.1001/jamaoncol.2024.5927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/04/2024] [Indexed: 01/19/2025]
Abstract
Importance Intratumoral immunotherapy that leverages the biological characteristics of high-risk ductal carcinoma in situ (DCIS) may be able to reduce the extent of surgical treatment and provide an alternative approach to improve patient outcomes. Objective To determine if combination intratumoral immunotherapy can activate immune cells to shrink or eliminate high-risk DCIS. Design, Setting, and Participants This phase 1 open-label nonrandomized clinical trial at a single academic center tested the safety and efficacy of intratumoral immunotherapy in patients with high-risk DCIS, defined as at least 2 of the following present: younger than 45 years, tumor size greater than 5 cm, high-grade, palpable mass, hormone receptor (HR)-negative, or ERBB2-positive. Patients were enrolled between June 8, 2021, and December 13, 2022. Intervention Pembrolizumab (anti-programmed cell death protein 1), dose ranging from 2 mg to 8 mg, and mRNA-2752 (a combination of interleukin [IL]-23, IL-36γ, and OX40L mRNAs), dose ranging from 1 mg to 4 mg, delivered intratumorally, with 2 to 4 doses given 2 to 3 weeks apart. Main Outcomes and Measures The primary objective was to evaluate the safety and tolerability of intratumoral injections of pembrolizumab and mRNA-2752. The secondary objectives were to assess radiologic and pathological responses and immunological and histological differences in the posttreatment tumor microenvironment. Results Ten female patients with high-risk DCIS (median [range] age, 46 [35-80] years) were enrolled. The median (range) tumor size was 5.3 (1.0-10.0) cm. Five tumors were HR-negative ERBB2-positive; 2 HR-negative ERBB2-negative; 2 HR-positive ERBB2-negative; and 1 HR-positive ERBB2-positive. Of all treated patients, 8 of 10 responded to treatment, and all 8 patients had ERBB2-positive or HR-negative DCIS. Three patients had complete responses. Three patients with negative posttreatment core biopsy results declined surgery and remained disease-free after 1 to 2 years. Multiplex immunofluorescence staining demonstrated that high baseline levels of tumor-infiltrating lymphocytes and programmed cell death ligand 1-positive cells (immune or tumor) were associated with a better treatment response. All patients experienced up to 1 week of fever, malaise, flulike symptoms, axillary adenopathy, erythema, injection site swelling, and swelling in the breast. One patient had intermittent urticaria for 3 months. The dose was serially reduced from 8 mg to 2 mg for pembrolizumab and 4 mg to 1 mg for mRNA-2752 to improve tolerability. The final recommended combination dose is pembrolizumab, 4 mg, with mRNA-2752, 1 mg. Conclusions and Relevance In this phase 1 nonrandomized clinical trial, the results suggest that intratumoral injections of pembrolizumab and mRNA-2752 are safe and may induce rapid regression of high-risk DCIS with high immune infiltrates. These findings warrant additional investigation, and studies are ongoing. Trial Registration ClinicalTrials.gov Identifier: NCT02872025.
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Middle Aged
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/immunology
- Adult
- Carcinoma, Intraductal, Noninfiltrating/drug therapy
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Injections, Intralesional
- Aged
- RNA, Messenger/administration & dosage
- RNA, Messenger/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Rachel Woody
- Department of Surgery, University of California, San Francisco
| | - Alexa Glencer
- Department of Surgery, University of California, San Francisco
| | | | - Hidetoshi Mori
- Department of Pathology and Lab Medicine, University of California, Davis
| | - Jasmine Wong
- Department of Surgery, University of California, San Francisco
| | - Gillian Hirst
- Department of Surgery, University of California, San Francisco
| | | | - Natsuko Onishi
- Dempartment of Radiology, University of California, San Francisco
| | - Jessica Gibbs
- Dempartment of Radiology, University of California, San Francisco
| | - Nola Hylton
- Dempartment of Radiology, University of California, San Francisco
| | | | | | | |
Collapse
|
27
|
Cha YJ, Kim HM, Koo JS. Inherent PD-L1 22C3 Expression in Alveolar Macrophages Impacts the Combined Positive Score Status in Breast Cancer With Pulmonary Metastasis. Thorac Cancer 2025; 16:e70004. [PMID: 40051246 PMCID: PMC11885796 DOI: 10.1111/1759-7714.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 03/10/2025] Open
Abstract
PURPOSE This study aimed to determine the impact of inherent programmed death-ligand 1 (PD-L1)-expressing alveolar macrophages (AMs) on the combined positive score (CPS) of PD-L1 (22C3) in metastatic breast cancer in the lungs. METHODS A total of 87 patients with pulmonary metastases of breast cancer were included in this study. Immunohistochemical staining of various PD-L1 antibodies was performed. The CPSs and CPSs excluding the number of PD-L1 positive AMs [CPS(NAM)s] with PD-L1 (22C3) were determined and compared. RESULTS Among 87 enrolled patients, 22 had luminal A breast cancer, 24 had luminal B breast cancer, 13 had HER-2-positive breast cancer, and 28 had triple-negative breast cancer (TNBC). CPSs ≥ 10 was observed only in luminal B (12.5%) and TNBC (35.7%) subtypes (p < 0.001), whereas CPS(NAM)s ≥ 10 was observed only in TNBC (14.3%) (p = 0.011). Changes from the CPS-positive to the CPS(NAM)-negative status occurred in nine cases (10.3%), with significantly higher proportions being observed in the luminal B (12.5%) and TNBC (21.4%) subtypes (p = 0.007). Tumors showing changes from the CPS-positive to the CPS(NAM)-negative status were larger (p < 0.001); similar findings were observed in the TNBC subgroup (p = 0.001). CONCLUSION The inclusion of PD-L1 expressing AMs leads to differences in CPS positivity, especially in large TNBC subtype tumors.
Collapse
Affiliation(s)
- Yoon Jin Cha
- Department of PathologyYonsei University College of MedicineSeoulSouth Korea
| | - Hye Min Kim
- Department of PathologyYonsei University College of MedicineSeoulSouth Korea
| | - Ja Seung Koo
- Department of PathologyYonsei University College of MedicineSeoulSouth Korea
| |
Collapse
|
28
|
Bear HD, Deng X, Bandyopadhyay D, Idowu M, Jenkins TM, Kmieciak M, Williams M, Archer G, Gwaltney L, Dillon P, Flora D, Stover D, Poklepovic AS, Hackney M, Ross M, Vachhani H, Louie R, McGuire KP, Grover A, Rahman T, Hendrix A. T-cell immune checkpoint inhibition plus hypomethylation for locally advanced HER2-negative breast cancer: a phase 2 neoadjuvant window trial of decitabine and pembrolizumab followed by standard neoadjuvant chemotherapy. J Immunother Cancer 2025; 13:e010294. [PMID: 40021215 PMCID: PMC11873355 DOI: 10.1136/jitc-2024-010294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Higher levels of tumor-infiltrating lymphocytes (TILs) in breast cancers are associated with increased likelihood of pathologic complete response (pCR) to chemotherapy. DNA methyltransferase inhibitors (DNMTi) can augment immune responses to cancers, decreasing myeloid-derived suppressor cells (MDSCs) and increasing T lymphocyte responsiveness. We have shown that the DNMTi decitabine augments the effectiveness of immunotherapy using murine triple-negative breast cancer (TNBC) models. The primary objective was to determine whether DNMTi+immune checkpoint blockade would increase stromal TIL (sTIL) in primary breast cancers before neoadjuvant chemotherapy (NCT). METHODS In a phase 2 study (NCT02957968), patients with human epidermal growth factor receptor 2-negative breast cancer received window immunotherapy-decitabine (15 mg/m2×4 doses over 5 days) followed by 2 doses of pembrolizumab (200 mg, 2 weeks apart)-before starting NCT. Biopsies before and after window immunotherapy quantified TILs and programmed death-ligand 1 (PD-L1) expression. Patients proceeded to NCT and tumor resection per standard of care. Mid-study, results of the KEYNOTE 522 trial led to patients with TNBC receiving additional pembrolizumab concurrently with standard NCT and in the adjuvant setting. RESULTS 46 patients (median age 54.5 years, range 28-72; 71.7% white, 28.3% black; 100% female) were treated. 21 patients had TNBC and received neither neoadjuvant pembrolizumab concurrently with NCT nor adjuvant pembrolizumab (Cohort A), 7 patients had TNBC and did receive concurrent and/or adjuvant pembrolizumab (Cohort A2), and 18 patients were estrogen receptor positive and/or progesterone receptor positive and received neither concurrent nor adjuvant pembrolizumab (Cohort B). Blood samples collected after decitabine administration before pembrolizumab showed a 59% decrease (p<0.01) in monocytic MDSCs compared with baseline. 38 patients had paired biopsies for sTIL and 37 for PD-L1 evaluation. Cohorts A/A2 experienced an sTIL increase of 6.1% (p<0.008); Cohort B experienced an sTIL increase of 8.3% (p=0.006). PD-L1 expression increased by 73.9% (p<0.01). 14 of 43 patients (32.6%) who proceeded to resection achieved pCR (n=11 of 27 (40.1%) in Cohorts A/A2 and n=3 of 16 (18.8%) in Cohort B). The most frequently reported immune-related adverse events were adrenal insufficiency (AI) (n=6, 13.0%), maculopapular rash (n=3, 6.5%), and hypothyroidism (n=3, 6.5%). Five of the six AI instances were at least partially attributable to hypophysitis/pituitary dysfunction, and one remains uncertain. CONCLUSIONS Treatment in the pre-neoadjuvant window with decitabine and pembrolizumab could sensitize breast cancers to standard NCT by recruitment of TILs to the tumor tissue. The treatment was well-tolerated. TRIAL REGISTRATION NUMBER NCT02957968.
Collapse
Affiliation(s)
- Harry D Bear
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Xiaoyan Deng
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Dipankar Bandyopadhyay
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Michael Idowu
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Taylor M Jenkins
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Maciej Kmieciak
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Monique Williams
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Giovanni Archer
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Lindsey Gwaltney
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Patrick Dillon
- University of Virginia Cancer Center, Charlottesville, Virginia, USA
| | - Daniel Flora
- St Elizabeth Healthcare, Edgewood, Kentucky, USA
| | - Daniel Stover
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Andrew S Poklepovic
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mary Hackney
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Masey Ross
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Hetal Vachhani
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Raphael Louie
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Kandace P McGuire
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Amelia Grover
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Tasnim Rahman
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Amber Hendrix
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
29
|
Xiong X, Zheng LW, Ding Y, Chen YF, Cai YW, Wang LP, Huang L, Liu CC, Shao ZM, Yu KD. Breast cancer: pathogenesis and treatments. Signal Transduct Target Ther 2025; 10:49. [PMID: 39966355 PMCID: PMC11836418 DOI: 10.1038/s41392-024-02108-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/27/2024] [Accepted: 12/08/2024] [Indexed: 02/20/2025] Open
Abstract
Breast cancer, characterized by unique epidemiological patterns and significant heterogeneity, remains one of the leading causes of malignancy-related deaths in women. The increasingly nuanced molecular subtypes of breast cancer have enhanced the comprehension and precision treatment of this disease. The mechanisms of tumorigenesis and progression of breast cancer have been central to scientific research, with investigations spanning various perspectives such as tumor stemness, intra-tumoral microbiota, and circadian rhythms. Technological advancements, particularly those integrated with artificial intelligence, have significantly improved the accuracy of breast cancer detection and diagnosis. The emergence of novel therapeutic concepts and drugs represents a paradigm shift towards personalized medicine. Evidence suggests that optimal diagnosis and treatment models tailored to individual patient risk and expected subtypes are crucial, supporting the era of precision oncology for breast cancer. Despite the rapid advancements in oncology and the increasing emphasis on the clinical precision treatment of breast cancer, a comprehensive update and summary of the panoramic knowledge related to this disease are needed. In this review, we provide a thorough overview of the global status of breast cancer, including its epidemiology, risk factors, pathophysiology, and molecular subtyping. Additionally, we elaborate on the latest research into mechanisms contributing to breast cancer progression, emerging treatment strategies, and long-term patient management. This review offers valuable insights into the latest advancements in Breast Cancer Research, thereby facilitating future progress in both basic research and clinical application.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Le-Wei Zheng
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu Ding
- Department of Breast and Thyroid, Guiyang Maternal and Child Health Care Hospital & Guiyang Children's Hospital, Guiyang, P. R. China
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, P. R. China
| | - Yu-Fei Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Yu-Wen Cai
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Lei-Ping Wang
- Department of Breast and Urologic Medical Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Liang Huang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P. R. China.
| |
Collapse
|
30
|
Xu L, Xu P, Wang J, Ji H, Zhang L, Tang Z. Advancements in clinical research and emerging therapies for triple-negative breast cancer treatment. Eur J Pharmacol 2025; 988:177202. [PMID: 39675457 DOI: 10.1016/j.ejphar.2024.177202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) expression, is acknowledged as the most aggressive form of breast cancer (BC), comprising 15%-20% of all primary cases. Despite the prevalence of TNBC, effective and well-tolerated targeted therapies remain limited, with chemotherapy continuing to be the mainstay of treatment. However, the horizon is brightened by recent advancements in immunotherapy and antibody-drug conjugates (ADCs), which have garnered the U.S. Food and Drug Administration (FDA) approval for various stages of TNBC. Poly (ADP-ribose) polymerase inhibitors (PARPi), particularly for TNBC with BRCA mutations, present a promising avenue, albeit with the challenge of resistance that must be addressed. The success of phosphoinositide-3 kinase (PI3K) pathway inhibitors in hormone receptor (HR)-positive BC suggests potential applicability in TNBC, spurring optimism within the research community. This review endeavors to offer a comprehensive synthesis of both established and cutting-edge targeted therapies for TNBC. We delve into the specifics of PARPi, androgen receptor (AR) inhibitors, Cancer stem cells (CSCs), PI3K/Protein Kinase B (AKT)/mammalian target of rapamycin (mTOR), the transforming growth factor-beta (TGF-β), Ntoch, Wnt/β-catenin, hedgehog (Hh) pathway inhibitors, Epigenetic target-mediated drug delivery, ADCs, immune checkpoint inhibitors (ICIs)and novel immunotherapeutic solutions, contextualizing TNBC within current treatment paradigms. By elucidating the mechanisms of these drugs and their prospective clinical applications, we aim to shed light on the challenges and underscore the beacon of hope that translational research and innovative therapies represent for the oncology field.
Collapse
Affiliation(s)
- Lili Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Pengtao Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, China
| | - Hui Ji
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
31
|
Loi S, Salgado R, Curigliano G, Romero Díaz RI, Delaloge S, Rojas García CI, Kok M, Saura C, Harbeck N, Mittendorf EA, Yardley DA, Suárez Zaizar A, Caminos FR, Ungureanu A, Reinoso-Toledo JG, Guarneri V, Egle D, Ades F, Pacius M, Chhibber A, Chandra R, Nathani R, Spires T, Wu JQ, Pusztai L, McArthur H. Neoadjuvant nivolumab and chemotherapy in early estrogen receptor-positive breast cancer: a randomized phase 3 trial. Nat Med 2025; 31:433-441. [PMID: 39838118 PMCID: PMC11835735 DOI: 10.1038/s41591-024-03414-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/12/2024] [Indexed: 01/23/2025]
Abstract
Patients with estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) primary breast cancer (BC) have low pathological complete response (pCR) rates with neoadjuvant chemotherapy. A subset of ER+/HER2- BC contains dense lymphocytic infiltration. We hypothesized that addition of an anti-programmed death 1 agent may increase pCR rates in this BC subtype. We conducted a randomized, multicenter, double-blind phase 3 trial to investigate the benefit of adding nivolumab to neoadjuvant chemotherapy in patients with newly diagnosed, high-risk, grade 3 or 2 (ER 1 to ≤10%) ER+/HER2- primary BC. In total, 510 patients were randomized to receive anthracycline and taxane-based chemotherapy with either intravenous nivolumab or placebo. The primary endpoint of pCR was significantly higher in the nivolumab arm compared with placebo (24.5% versus 13.8%; P = 0.0021), with greater benefit observed in patients with programmed death ligand 1-positive tumors (VENTANA SP142 ≥1%: 44.3% versus 20.2% respectively). There were no new safety signals identified. Of the five deaths that occurred in the nivolumab arm, two were related to study drug toxicity; no deaths occurred in the placebo arm. Adding nivolumab to neoadjuvant chemotherapy significantly increased pCR rates in high-risk, early-stage ER+/HER2- BC, particularly among patients with higher stromal tumor-infiltrating lymphocyte levels or programmed death ligand 1 expression, suggesting a new treatment paradigm that emphasizes the role of immunotherapy and T cell immunosurveillance in luminal disease. Clinical trials.gov identifier: NCT04109066.
Collapse
Affiliation(s)
- Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- University of Melbourne, Parkville, Victoria, Australia.
| | - Roberto Salgado
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- University of Milan, Milan, Italy
| | | | | | | | - Marleen Kok
- Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology and CCC Munich, Ludwig Maximilians University Hospital, Munich, Germany
| | | | | | | | | | | | | | - Valentina Guarneri
- Istituto Oncologico Veneto IOV, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Daniel Egle
- Department of Gynecology, Medical University of Innsbruck, Innsbruck, Austria
- ABCSG - Austrian Breast & Colorectal Cancer Study Group, Vienna, Austria
| | | | | | | | | | | | | | | | | | - Heather McArthur
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
32
|
Connors C, Valente SA, ElSherif A, Escobar P, Chichura A, Kopicky L, Roesch E, Ritner J, McIntire P, Wu Y, Tu C, Lang JE. Real-World Outcomes with the KEYNOTE-522 Regimen in Early-Stage Triple-Negative Breast Cancer. Ann Surg Oncol 2025; 32:912-921. [PMID: 39436619 PMCID: PMC11843215 DOI: 10.1245/s10434-024-16390-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024]
Abstract
BACKGROUND This study aimed to determine if the neoadjuvant (NAT) KEYNOTE-522 regimen was associated with higher rates of pathologic complete response (pCR), corresponding to higher rates of breast conservation therapy (BCT) in early-stage triple-negative breast cancer (TNBC) patients. PATIENTS AND METHODS Stage II-III TNBC patients diagnosed between 2019 and 2022 who underwent NAT were analyzed retrospectively. NAT with KEYNOTE-522 versus control NAT were compared for rates of BCT, axillary node dissection (ALND), pCR, and survival outcomes. The prevalence of immune-related adverse events (irAE) from chemoimmunotherapy was recorded. RESULTS Of 240 patients identified: 86 received KEYNOTE-522 and 154 received control. The frequency of pCR was significantly higher in KEYNOTE versus the control cohort, 59.3% and 33.1%, respectively (p = 0.001). There was no significant difference in the rate of BCT between the control (33.1%) and the KEYNOTE-522 (32.1%) groups (p = 0.47). Rates of ALND were significantly lower with KEYNOTE-522 (25.6%) as compared with control (39.6%); p = 0.03. The rate of development of grade 2 or higher irAEs was 34.9%. At a median follow-up of 2.4 years, there was no difference in survival outcomes. BRCA1 patients had high rates of pCR regardless of treatment group, KEYNOTE-522: 80.0% (4/5) and control: 75% (9/12), (p = 1). CONCLUSION This real-world evidence supports the use of the KEYNOTE-522 regimen in patients with early-stage TNBC given the higher pCR rate and corresponding decrease in the rate of ALND. The majority of patients in both NAT cohorts became BCT eligible, but the rate of BCT did not differ between the two groups.
Collapse
Affiliation(s)
- Casey Connors
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Stephanie A Valente
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ayat ElSherif
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Paula Escobar
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Chichura
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lauren Kopicky
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Erin Roesch
- Department of Medical Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Julie Ritner
- Department of Radiology, Cleveland Clinic, Cleveland, OH, USA
| | | | - Yueqi Wu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chao Tu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Julie E Lang
- Department of Breast Surgery, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
33
|
Mattar A, Antonini M, Amorim AG, Teixeira MD, de Resende CAA, Cavalcante FP, Zerwes F, Arakelian R, Millen EDC, Brenelli FP, Frasson AL, Leite RM, Gebrim LH. Overall Survival and Economic Impact of Triple-Negative Breast Cancer in Brazilian Public Health Care: A Real-World Study. JCO Glob Oncol 2025; 11:e2400340. [PMID: 39977709 DOI: 10.1200/go-24-00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 12/11/2024] [Accepted: 01/09/2025] [Indexed: 02/22/2025] Open
Abstract
PURPOSE Triple-negative breast cancer (TNBC) presents notable treatment difficulties, especially in the public health care systems of low- and middle-income countries where access to advanced therapies is restricted. This study investigates TNBC's clinical, epidemiologic, and economic effects on survival within Brazil's public health care system. METHODS We conducted a retrospective cohort study of patients with TNBC treated between 2010 and 2019. Overall survival (OS) rates by stage were analyzed across various patient groups, including those receiving neoadjuvant or adjuvant treatment, patients with or without complete pathologic response, Black and non-Black patients, and those treated with or without carboplatin-based therapy. Cox proportional hazards models were applied to estimate hazard ratios (HRs) with 95% CIs, and annual treatment costs were calculated per stage. RESULTS Among 1,266 patients with TNBC, 710 met eligibility criteria. Kaplan-Meier analysis indicated stage II patients had a 47% lower mortality risk than stage III (HR, 0.53 [95% CI, 0.33 to 0.85]; P = .009). Patients in the adjuvant treatment group had a reduced risk (HR, 0.48 [95% CI, 0.34 to 0.69]) compared with the neoadjuvant group. Achieving complete pathologic response (pCR) greatly improved OS (HR, 0.21 [95% CI, 0.11 to 0.43]; P < .001). Black patients had better survival rates than non-Black (HR, 0.58 [95% CI, 0.40 to 0.86]; P = .006). Carboplatin use did not significantly affect OS (HR, 0.96 [95% CI, 0.65 to 1.43]; P = .857). The average monthly cost for systemic TNBC treatment increased with disease progression, from $101.87 in US dollars (USD) for stage I to $314.77 USD for stage IV second-line therapy. CONCLUSION This study provides insight into TNBC in Brazil's public health system, showing that OS decreases with disease progression but is higher among Black patients. pCR and adjuvant therapy improve survival, although costs increase significantly at advanced stages, highlighting the economic burden of late-stage TNBC management.
Collapse
Affiliation(s)
- André Mattar
- Centro de Referência da Saúde da Mulher-Hospital da Mulher, São Paulo, Brazil
- Oncoclínicas, São Paulo, Brazil
| | - Marcelo Antonini
- Hospital do Servidor Público Estadual-Francisco Morado de Oliveira, São Paulo, Brazil
| | | | | | | | | | - Felipe Zerwes
- Pontifícia Universidade Católica, Porto Alegre, Brazil
| | - Renata Arakelian
- Centro de Referência da Saúde da Mulher-Hospital da Mulher, São Paulo, Brazil
- Dasa Oncologia, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
34
|
Matossian MD, Shiang C, Dolcen DN, Dreyer M, Hatogai K, Hall K, Saha P, Biernacka A, Sweis RF, Karrison T, Chen N, Nanda R, Conzen SD. High tumor glucocorticoid receptor expression in early-stage, triple-negative breast cancer is associated with increased T-regulatory cell infiltration. Breast Cancer Res Treat 2025; 209:563-572. [PMID: 39579248 PMCID: PMC11785596 DOI: 10.1007/s10549-024-07515-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 11/25/2024]
Abstract
PURPOSE In early-stage, triple-negative breast cancer (TNBC), immune cell infiltration contributes to cancer cell survival, tumor invasion, and metastasis. High TNBC glucocorticoid receptor (GR) expression in early-stage TNBC is associated with poor long-term outcomes; it is unknown if high GR expression is associated with an immunosuppressed tumor microenvironment. We hypothesized that high tumor GR expression would be associated with an immune-suppressed tumor microenvironment, which could thus account for the poor prognosis observed in GR-positive TNBC. METHODS Formalin fixed-paraffin embedded tissue (n = 47) from patients diagnosed with early-stage TNBC from The University of Chicago (2002-2014) were evaluated for both tumor cell anti-GR immunohistochemistry and for infiltrating immune cells by immunofluorescence. Multiplexed antibodies were used to enumerate CD8+, FOXP3+, and BATF3+ immune cells infiltrating within pan-cytokeratin positive tumor cell regions of interest, and nonparametric tests compared absolute counts of each of these tumor-infiltrating immune cell types. RESULTS The average age of patients represented in this study was 52 years, and 63% self-identified as Black. There was no significant association between tumor GR expression and age, race, or clinical stage at diagnosis. Compared to GR-low tumors, high GR expression in early-stage, treatment-naïve TNBC was associated with relatively increased numbers of immunosuppressive FOXP3 + regulatory T cells (p = 0.046) and BATF3+immune cells (p = 0.021). While there was a positive correlation with high GR expression and CD8+ cell infiltration, it was not significant (p = 0.068). The ratio of CD8+/FOXP3+cells was also not significant (p = 0.24). CONCLUSIONS These data support the hypothesis that in early-stage TNBC, high GR expression is significantly associated with infiltration of immunosuppressive regulatory T cells, suggesting a tumor-intrinsic role in shaping the immunosuppressive immune cell milieu. Furthermore, suppression of GR activity may regulate the tumor immune microenvironment and improve long-term outcomes in GR-high TNBC.
Collapse
Affiliation(s)
- Margarite D Matossian
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Christine Shiang
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Deniz Nesli Dolcen
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Marie Dreyer
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Ken Hatogai
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Katie Hall
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Poornima Saha
- Division of Hematology and Oncology, Department of Medicine, Endeavor Health, Evanston, IL, 60201, USA
| | - Anna Biernacka
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA
| | - Randy F Sweis
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Theodore Karrison
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, 60637, USA
| | - Nan Chen
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Rita Nanda
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| | - Suzanne D Conzen
- Section of Hematology and Oncology, Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
35
|
Huppert LA, Wolf D, Yau C, Brown-Swigart L, Hirst GL, Isaacs C, Pusztai L, Pohlmann PR, DeMichele A, Shatsky R, Yee D, Thomas A, Nanda R, Perlmutter J, Heditsian D, Hylton N, Symmans F, Van't Veer LJ, Esserman L, Rugo HS. Pathologic complete response (pCR) rates for patients with HR+/HER2- high-risk, early-stage breast cancer (EBC) by clinical and molecular features in the phase II I-SPY2 clinical trial. Ann Oncol 2025; 36:172-184. [PMID: 39477071 DOI: 10.1016/j.annonc.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/26/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-negative early-stage breast cancer (EBC) is a heterogenous disease. Identification of better clinical and molecular biomarkers is essential to guide optimal therapy for each patient. PATIENTS AND METHODS We analyzed rates of pathologic complete response (pCR) and distant recurrence-free survival (DRFS) for patients with HR+/HER2-negative EBC in eight neoadjuvant arms in the I-SPY2 trial by clinical/molecular features: age, stage, histology, percentage estrogen receptor (ER) positivity, ER/progesterone receptor status, MammaPrint (MP)-High1 (0 to -0.57) versus MP-High2 (<-0.57), BluePrint (BP)-Luminal-type versus BP-Basal-type, and ImPrint immune signature. We quantified the clinical/molecular heterogeneity, assessed overlap among these biomarkers, and evaluated associations with pCR and DRFS. RESULTS Three hundred and seventy-nine patients with HR+/HER2-negative EBC were included in this analysis, with an observed pCR rate of 17% across treatment arms. pCR rates were higher in patients with stage II versus III disease (21% versus 9%, P = 0.0013), ductal versus lobular histology (19% versus 11%, P = 0.049), lower %ER positivity (≤66% versus >66%) (35% versus 9%, P = 3.4E-09), MP-High2 versus MP-High1 disease (31% versus 11%, P = 1.1E-05), BP-Basal-type versus BP-Luminal-type disease (34% versus 10%, P = 1.62E-07), and ImPrint-positive versus -negative disease (38% versus 10%, P = 1.64E-09). Patients with lower %ER were more likely to have MP-High2 and BP-Basal-type disease. At a median follow-up of 4.8 years, patients who achieved pCR had excellent outcomes irrespective of clinical/molecular features. Among patients who did not achieve pCR, DRFS events were more frequent in patients with MP-High2 and BP-Basal-type disease than those with MP-High1 and BP-Luminal-type disease. CONCLUSIONS Among patients with high molecular-risk HR+/HER2-negative EBC, the MP-High2, BP-Basal-type, and ImPrint-positive signatures identified a partially overlapping subset of patients who were more likely to achieve pCR in response to neoadjuvant chemotherapy ± targeted agents or immunotherapy compared to patients with MP-High1, BP-Luminal-type, and ImPrint-negative disease. I-SPY2.2 is incorporating the use of these biomarkers to molecularly define specific patient populations and optimize treatment selection.
Collapse
Affiliation(s)
- L A Huppert
- Department of Medicine, University of California San Francisco, San Francisco, USA.
| | - D Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, USA
| | - C Yau
- Department of Surgery, University of California San Francisco, San Francisco, USA
| | - L Brown-Swigart
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, USA
| | - G L Hirst
- Department of Surgery, University of California San Francisco, San Francisco, USA
| | - C Isaacs
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, USA
| | - L Pusztai
- Yale School of Medicine, Yale University, New Haven, USA
| | - P R Pohlmann
- Department of Breast Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, USA
| | - A DeMichele
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - R Shatsky
- Division of Hematology/Oncology, University of California San Diego, San Diego, USA
| | - D Yee
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, USA
| | - A Thomas
- Division of Hematology/Oncology, Duke Cancer Center, Durham, USA
| | - R Nanda
- Section of Hematology/Oncology, University of Chicago, Chicago, USA
| | | | | | - N Hylton
- Department of Radiology, University of California San Francisco, San Francisco, USA
| | - F Symmans
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - L J Van't Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, USA
| | - L Esserman
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, USA. https://twitter.com/DrLauraEsserman
| | - H S Rugo
- Department of Medicine, University of California San Francisco, San Francisco, USA. https://twitter.com/hoperugo
| |
Collapse
|
36
|
Cardoso F, O'Shaughnessy J, Liu Z, McArthur H, Schmid P, Cortes J, Harbeck N, Telli ML, Cescon DW, Fasching PA, Shao Z, Loirat D, Park YH, Fernandez MG, Rubovszky G, Spring L, Im SA, Hui R, Takano T, André F, Yasojima H, Ding Y, Jia L, Karantza V, Tryfonidis K, Bardia A. Pembrolizumab and chemotherapy in high-risk, early-stage, ER +/HER2 - breast cancer: a randomized phase 3 trial. Nat Med 2025; 31:442-448. [PMID: 39838117 PMCID: PMC11835712 DOI: 10.1038/s41591-024-03415-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/12/2024] [Indexed: 01/23/2025]
Abstract
Addition of pembrolizumab to neoadjuvant chemotherapy followed by adjuvant pembrolizumab improved outcomes in patients with high-risk, early-stage, triple-negative breast cancer. However, whether the addition of neoadjuvant pembrolizumab to chemotherapy would improve outcomes in high-risk, early-stage, estrogen receptor-positive/human epidermal growth factor receptor 2-negative (ER+/HER2-) breast cancer remains unclear. We conducted a double-blind, placebo-controlled phase 3 study (KEYNOTE-756) in which patients with previously untreated ER+/HER2- grade 3 high-risk invasive breast cancer (T1c-2 (≥2 cm), cN1-2 or T3-4, cN0-2) were randomly assigned (1:1) to neoadjuvant pembrolizumab 200 mg or placebo Q3W given with paclitaxel QW for 12 weeks, followed by four cycles of doxorubicin or epirubicin plus cyclophosphamide Q2W or Q3W. After surgery (with/without adjuvant radiation therapy), patients received adjuvant pembrolizumab or placebo for nine cycles plus adjuvant endocrine therapy. Dual primary endpoints were pathological complete response and event-free survival in the intention-to-treat population. In total, 635 patients were assigned to the pembrolizumab-chemotherapy arm and 643 to the placebo-chemotherapy arm. At the study's prespecified first interim analysis, the pathological complete response rate was 24.3% (95% confidence interval (CI), 21.0-27.8%) in the pembrolizumab-chemotherapy arm and 15.6% (95% CI, 12.8-18.6%) in the placebo-chemotherapy arm (estimated treatment difference, 8.5 percentage points; 95% CI, 4.2-12.8; P = 0.00005). Event-free survival was not mature in this analysis. During the neoadjuvant phase, treatment-related adverse events of grade ≥3 were reported in 52.5% and 46.4% of patients in the pembrolizumab-chemotherapy and placebo-chemotherapy arms, respectively. In summary, the addition of pembrolizumab to neoadjuvant chemotherapy significantly improved the pathological complete response rate in patients with high-risk, early-stage ER+/HER2- breast cancer. Safety was consistent with the known profiles of each study treatment. Follow-up continues for event-free survival. ClinicalTrials.gov identifier: NCT03725059 .
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Middle Aged
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Adult
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Aged
- Double-Blind Method
- Paclitaxel/administration & dosage
- Neoadjuvant Therapy
- Neoplasm Staging
- Doxorubicin/administration & dosage
- Cyclophosphamide/administration & dosage
- Epirubicin/administration & dosage
Collapse
Affiliation(s)
- Fatima Cardoso
- Breast Unit, Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal.
| | - Joyce O'Shaughnessy
- Baylor University Medical Center, Texas Oncology, US Oncology Network, Dallas, TX, USA
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Heather McArthur
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University London, London, UK
| | - Javier Cortes
- Medica Scientia Innovation Research, Ridgewood, NJ, USA
- Medica Scientia Innovation Research, Barcelona, Spain
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Universidad Europea de Madrid Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Nadia Harbeck
- Breast Center, Department of OB&GYN, LMU University Hospital, Munich, Germany
| | | | - David W Cescon
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Peter A Fasching
- University Hospital Erlangen, Comprehensive Cancer Center Erlangen-EMN, Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Zhimin Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | | | - Yeon Hee Park
- Samsung Medical Division, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Laura Spring
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Rina Hui
- Westmead Breast Cancer Institute, Westmead Hospital and the University of Sydney, Sydney, New South Wales, Australia
- Centre of Cancer Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | | | - Fabrice André
- Faculté de Médecine, Paris Saclay University, Gustave Roussy, Villejuif, France
| | | | - Yu Ding
- Merck & Co., Inc., Rahway, NJ, USA
| | - Liyi Jia
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | - Aditya Bardia
- University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
37
|
Rios-Hoyo A, Xiong K, Dai J, Yau C, Marczyk M, García-Milian R, Wolf DM, Huppert LA, Nanda R, Hirst GL, Cobain EF, van ‘t Veer LJ, Esserman LJ, Pusztai L. Hormone Receptor-Positive HER2-Negative/MammaPrint High-2 Breast Cancers Closely Resemble Triple-Negative Breast Cancers. Clin Cancer Res 2025; 31:403-413. [PMID: 39561272 PMCID: PMC11747811 DOI: 10.1158/1078-0432.ccr-24-1553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/16/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
PURPOSE The MammaPrint (MP) prognostic assay categorizes breast cancers into high- and low-risk subgroups, and the high-risk group can be further subdivided into high-1 (MP-H1), and very high-risk high-2 (MP-H2). The aim of this analysis was to assess clinical and molecular differences between the hormone receptor-positive (HR+)/HER2-negative MP-H1, -H2, and triple-negative (TN) MP-H1 and -H2 cancers. EXPERIMENTAL DESIGN Pretreatment gene expression data from 742 HER2-negative breast cancers enrolled in the I-SPY2 neoadjuvant trial were used. Prognostic risk categories were assigned using the MP assay. Transcriptional similarities across the four receptor and prognostic groups were assessed using principal component analyses and by identifying differentially expressed genes. We also examined pathologic complete response rates and event-free survivals by risk group. RESULTS Principal component analysis showed that HR+/MP-H2 tumors clustered with TN/MP-H2 cancers. Only 125 genes showed differential expression between the HR+/MP-H2 and TN/MP-H2 cancers, whereas 1,465 genes were differentially expressed between HR+/MP-H2 and -H1. Gene set analysis revealed similarly high expression of cell cycle, DNA repair, and immune infiltration-related pathways in HR+/MP-H2 and TN/MP-H2 cancers. HR+/MP-H2 cancers also showed low estrogen receptor-related gene expression. Pathologic complete response rates were similarly high in TN/MP-H2 and HR+/MP-H2 cancers (42% vs. 30.5%; P = 0.11), and MP-H2 cancers with residual cancer had similarly poor event-free survival regardless of estrogen receptor status. CONCLUSIONS In conclusion, HR+/MP-H2 cancers closely resemble TN breast cancers in transcriptional and clinical features and benefit from similar treatment strategies.
Collapse
Affiliation(s)
- Alejandro Rios-Hoyo
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
- To be considered as first authors
| | - Kaitlyn Xiong
- Yale School of Medicine, New Haven, Connecticut, USA
- To be considered as first authors
| | - Jiawei Dai
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michal Marczyk
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Rolando García-Milian
- Bioinformatics Support Program, Research and Education Services, Cushing/Whitney Medical Library, Yale University, New Haven, CT, United States of America
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Laura A. Huppert
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, Chicago, IL, USA
| | - Gillian L. Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Laura J. van ‘t Veer
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Laura J. Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
38
|
Wei Y, Li Q, Mo H, Qi Y, Ge H, Sun X, Fan Y, Zhang P, Wang J, Luo Y, Wang J, Ma F. Comparative efficacy of anthracycline-free and anthracycline-containing neoadjuvant chemoimmunotherapy regimens for triple-negative breast cancer. Transl Oncol 2025; 51:102171. [PMID: 39488006 PMCID: PMC11567949 DOI: 10.1016/j.tranon.2024.102171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND The selection of appropriate chemotherapy backbone agents in combination with neoadjuvant immunotherapy for triple-negative breast cancer (TNBC) remains unclear. Herein, we aimed to evaluate the efficacy and safety of anthracycline-free and anthracycline-containing regimens coupled with neoadjuvant immunotherapy. METHOD This retrospective study included 87 patients with TBNC who received neoadjuvant immunotherapy combined with various chemotherapy regimens at three research centers from November 2020 to November 2023. The primary objective was pathological complete response (pCR), while secondary objectives included overall response rates, event-free survival (EFS), and the incidence of adverse events. A subgroup analysis was performed to delineate patients who may substantially benefit from distinct therapeutic strategies. RESULTS Coupled with immunotherapy, anthracycline-free regimens achieved comparable pCR rates (55.1 % vs. 51.4 %; Odds ratio, 1.16; 95 % confidence interval [CI], 0.49-2.74; p = 0.73) and EFS (Hazard ratio, 0.66; 95 % CI, 0.18-2.45; p = 0.53) to anthracycline-containing regimens. According to subgroup analyses, the tumor stage (p = 0.017) and lymph node stage (p = 0.011) exhibit contradictory predictive power for the pCR rate of anthracycline-free regimens when compared with that of anthracycline-containing regimens. Specifically, anthracycline-free regimens yielded significantly higher pCR rates in patients without lymph node metastasis than anthracycline-containing regimens (p = 0.021). Pooled analyses further confirmed the results of both total and subgroup analyses. Most adverse events were grades 1-2, and no new adverse reactions were observed. CONCLUSION Anthracycline-free neoadjuvant chemotherapy regimens could serve as an effective and safe alternative immunotherapy partner for patients with TNBC, particularly in those without lymph node metastasis.
Collapse
Affiliation(s)
- Yuhan Wei
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hongnan Mo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yalong Qi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hewei Ge
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing ChaoYang District, Beijing, China.
| | - Ying Fan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Pin Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jiayu Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yang Luo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jing Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
39
|
Li B, Xie S, Han J, Cao H, Lin Z, Hu H, Pan C, Li Q, Li J, Wang L, Chen S, Rao G, Huang G, Tan Y, Chen R, Fan S, Duan X, Li H, Li J. Neoadjuvant chemoimmunotherapy in resectable locally advanced oral squamous cell carcinoma: a single-center retrospective cohort study. Int J Surg 2025; 111:781-790. [PMID: 38935124 PMCID: PMC11745724 DOI: 10.1097/js9.0000000000001891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Surgery and postoperative adjuvant therapy is the standard treatment for locally advanced resectable oral squamous cell carcinoma (OSCC), while neoadjuvant chemoimmunotherapy (NACI) is believed to lead to better outcomes. This study aims to investigate the effectiveness of NACI regimens in treating locally advanced resectable OSCC. MATERIALS AND METHODS Patients diagnosed with locally advanced resectable OSCC who received NACI and non-NACI were reviewed between December 2020 and June 2022 in our single center. The pathologic response was evaluated to the efficacy of NACI treatment. Adverse events apparently related to NACI treatment were graded by Common Terminology Criteria for Adverse Events, version 5.0. The disease-free survival (DFS) and overall survival (OS) rate were assessed. RESULTS Our analysis involved 104 patients who received NACI. Notably, the pathological complete response rate was 47.1%, and the major pathological response (MPR) rate was 65.4%. The top three grade 1-2 treatment-related adverse events (TRAEs) were alopecia (104; 100%), anemia (81; 77.9%), and pruritus (62; 59.6%). Importantly, patients achieving MPR exhibited higher programmed cell death-ligand 1 (PD-L1) combined positive scores (CPS). The diagnostic value of CPS as a biomarker for NACI efficacy was enhanced when combined with total cholesterol level. The 3-year estimated DFS rates were 89.0% in the NACI cohort compared to 60.8% in the non-NACI cohort, while the 3-year estimated OS rates were 91.3 versus 64.0%, respectively. CONCLUSIONS The NACI treatment showed safe and encouragingly efficacious for locally advanced resectable OSCC patients. The high response rates and favorable prognosis suggest this approach as a potential treatment option. Prospective randomized controlled trials are needed to further validate these findings.
Collapse
Affiliation(s)
- Bowen Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Shule Xie
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Jingjing Han
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Haotian Cao
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Zhaoyu Lin
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Huijun Hu
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Chaobin Pan
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
| | - Qunxing Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
| | - Jintao Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
| | - Liansheng Wang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
| | - Suling Chen
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Guangxin Rao
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Guoxin Huang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Yongmei Tan
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Rongxi Chen
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Song Fan
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Haigang Li
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
| | - Jinsong Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| |
Collapse
|
40
|
Corti C, Binboğa Kurt B, Koca B, Rahman T, Conforti F, Pala L, Bianchini G, Criscitiello C, Curigliano G, Garrido-Castro AC, Kabraji SK, Waks AG, Mittendorf EA, Tolaney SM. Estrogen Signaling in Early-Stage Breast Cancer: Impact on Neoadjuvant Chemotherapy and Immunotherapy. Cancer Treat Rev 2025; 132:102852. [PMID: 39571402 DOI: 10.1016/j.ctrv.2024.102852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 01/01/2025]
Abstract
Neoadjuvant chemoimmunotherapy (NACIT) has been shown to improve pathologic complete response (pCR) rates and survival outcomes in stage II-III triple-negative breast cancer (TNBC). Promising pCR rate improvements have also been documented for selected patients with estrogen receptor-positive (ER+) human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC). However, one size does not fit all and predicting which patients will benefit from NACIT remains challenging. Accurate predictions would be useful to minimize immune-related toxicity, which can be severe, irreversible, and potentially impact fertility and quality of life, and to identify patients in need of alternative treatments. This review aims to capitalize on the existing translational and clinical evidence on predictors of treatment response in patients with early-stage BC treated with neoadjuvant chemotherapy (NACT) and NACIT. It summarizes evidence suggesting that NACT/NACIT effectiveness may correlate with pre-treatment tumor characteristics, including mutational profiles, ER expression and signaling, immune cell presence and spatial organization, specific gene signatures, and the levels of proliferating versus quiescent cancer cells. However, the predominantly qualitative and descriptive nature of many studies highlights the challenges in integrating various potential response determinants into a validated, comprehensive, and multimodal predictive model. The potential of novel multi-modal approaches, such as those based on artificial intelligence, to overcome current challenges remains unclear, as these tools are not free from bias and shortcut learning. Despite these limitations, the rapid evolution of these technologies, coupled with further efforts in basic and translational research, holds promise for improving treatment outcome predictions in early HER2- BC.
Collapse
Affiliation(s)
- Chiara Corti
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Busem Binboğa Kurt
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Beyza Koca
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Tasnim Rahman
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Fabio Conforti
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Laura Pala
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, IRCCS, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Ana C Garrido-Castro
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sheheryar K Kabraji
- Department of Medicine, Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adrienne G Waks
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Shaitelman SF, Le-Petross H, Raso MG, Swanson DM, Schalck AP, Contreras A, Yang F, Muruganandham M, Zhao GZ, Sawakuchi GO, Kim LH, Batra H, Smith BD, Stauder MC, Woodward WA, Reddy JP, Litton JK, Thompson A, Bedrosian I, Mittendorf EA. PRECISE: Preoperative Radiation Therapy to Elicit Critical Immune Stimulating Effects-A Phase 2 Clinical Trial. Int J Radiat Oncol Biol Phys 2025; 121:90-96. [PMID: 39147206 DOI: 10.1016/j.ijrobp.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Radiation therapy is an underinvestigated tool for priming the immune system in intact human breast cancers. We sought here to investigate if a preoperative radiation therapy boost delivered was associated with a significant change in tumor-infiltrating lymphocytes (TILs) in the tumor in estrogen receptor positive, HER2Neu nonamplified breast cancers. METHODS AND MATERIALS A total of 20 patients were enrolled in a phase 2 clinical trial and received either 7.5 Gy × 1 fraction or 2 Gy × 5 fractions, completed 6 to 8 days before surgery. Percent stromal TILs were evaluated on hematoxylin and eosin-stained samples. Short-term safety was assessed based on time to surgery, toxicities, and cosmesis up to 6 months after boost. RESULTS Stromal TIL increased 6 to 8 days after completion of boost radiation therapy (median 3.0 [IQR, 1.0-6.5]) before radiation therapy versus median 5.0 (IQR, 1.5-8.0) after radiation therapy, P = .0037. Zero grade ≥3 toxicities up to 6 months after boost were experienced. In all, 94% (16/17) patients with 6-month follow-up cosmetic assessment after breast conservation had good-excellent cosmesis by physician assessment. CONCLUSION In this phase 2 trial, preoperative radiation therapy boost resulted in a short-term increase in stromal TIL with minimal toxicities. Preoperative breast radiation therapy appears to be safe and may be a feasible means for priming the tumor microenvironment.
Collapse
Affiliation(s)
- Simona F Shaitelman
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Huong Le-Petross
- Department of Breast Imaging, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria G Raso
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David M Swanson
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aislyn P Schalck
- Department of Genomics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alejandro Contreras
- Department of Anatomical Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fei Yang
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Manickam Muruganandham
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Z Zhao
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel O Sawakuchi
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Leonard H Kim
- Department of Radiation Oncology, MD Anderson Cancer Center at Cooper, Camden, New Jersey
| | - Harsh Batra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Benjamin D Smith
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael C Stauder
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wendy A Woodward
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jay P Reddy
- Department of Breast Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer K Litton
- Department of Clinical Research, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alastair Thompson
- Section of Breast Surgery, Division of Surgical Oncology, Baylor College of Medicine, Houston, Texas
| | - Isabelle Bedrosian
- Department of Breast Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth A Mittendorf
- Division of Breast Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Hu Z, Chen Y, Ma R, Sun W, Chen L, Cai Z, Wen W, Lei W. Nomogram Prediction of Response to Neoadjuvant Chemotherapy Plus Pembrolizumab in Locally Advanced Hypopharyngeal Squamous Cell Carcinoma. J Otolaryngol Head Neck Surg 2025; 54:19160216251318255. [PMID: 39921555 PMCID: PMC11807280 DOI: 10.1177/19160216251318255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 12/10/2024] [Indexed: 02/10/2025] Open
Abstract
IMPORTANCE The objective response (ORR) rate in patients with locally advanced hypopharyngeal squamous cell-carcinoma (LA-HPSCC) following neoadjuvant chemotherapy (NACT) of albumin-bound paclitaxel plus carboplatin is low. At present, it is unclear whether the addition of pembrolizumab could increase the ORR or not. OBJECTIVE To investigate whether the addition of pembrolizumab could increase the ORR, and to develop a nomogram to predict the response of pembrolizumab addition. DESIGN Retrospective cohort study. SETTING This study was conducted at a single institution. PARTICIPANTS This study included 129 patients who conformed to the inclusion criteria. INTERVENTION OR EXPOSURES NACT with or without pembrolizumab for patients with LA-HPSCC. MAIN OUTCOME MEASURES The ORR was analyzed according to the RECIST 1.1 criteria and a nomogram was developed based on least absolute shrinkage and selection operator and multivariable Cox regression analysis. Predictive accuracy and discriminative ability of the nomogram were evaluated by receiver operating characteristics, precision recall, calibration curves, and decision curve analysis. RESULTS Eighty-two patients received NACT and 47 also received pembrolizumab. ORR was higher in patients receiving additional pembrolizumab (66.0% vs 47.6%, χ2 = 4.074, P = .044). The nomogram identified pretreatment levels of lymphocytes and red blood cells as independent predictors of a high ORR, while basophil levels were an independent predictor of a low ORR. Calibration curve showed that the nomogram-based predictions corresponded well with actual observations. C-index of the nomogram was 0.925 (0.848-1.002) and the area under curve was 0.925. Decision curve analysis affirmed that the nomogram had important clinical value. CONCLUSIONS AND RELEVANCE Pembrolizumab could improve the ORR in LA-HPSCC patients treated with NACT. Furthermore, a risk-prediction nomogram incorporating readily assessable routine pretreatment blood parameters can accurately estimate the response to NACT with pembrolizumab, leading to precise treatment and minimizing the waste of medical resources.
Collapse
Affiliation(s)
- Zhangwei Hu
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yi Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Renqiang Ma
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Wei Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Lin Chen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Zhimou Cai
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Weiping Wen
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Department of Otolaryngology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Wenbin Lei
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
- Otorhinolaryngology Institute, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
43
|
Leggat-Barr K, Yee D, Duralde E, Hodge C, Borges V, Baxter M, Valdez J, Morgan T, Garber J, Esserman L. A roadmap to reduce the incidence and mortality of breast cancer by rethinking our approach to women's health. Breast Cancer Res Treat 2025; 209:1-14. [PMID: 39531132 PMCID: PMC11785669 DOI: 10.1007/s10549-024-07522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Despite progress, breast cancer remains the most feared disease among women. In the USA alone, the incidence is now almost 300,000 new cancers per year, a rate that has nearly doubled in the last 30 years. Most women survive, but over 40,000 women a year still die of their disease [99]. It is the most diagnosed cancer among women and the second leading cause of cancer death. Important disparities exist in breast cancer outcomes among African American women, where women die of breast cancer at higher rates, are diagnosed younger, and at a more advanced stage. We are proposing a radical shift in our thinking about breast cancer prevention with an aspiration to dramatically lower breast cancer incidence. Most breast cancers are driven by steroid hormones. Throughout the life course, women are offered an array of hormonal treatments for menstrual cycle control, family planning, in vitro fertilization, postpartum weaning, and menopausal symptom management. There are mixed data on the extent to which each of these may contribute to increased or decreased risk for breast cancer. These endocrine manipulations could represent a great opportunity to potentially reduce breast cancer incidence and improve quality of life for survivors. To date, they have not been designed to explicitly reduce breast cancer risk. A new holistic approach will require scientists, drug developers, breast oncologists, obstetricians, gynecologists, endocrinologists, radiologists, and family medicine/internists to work together toward the common goal of reducing breast cancer risk while addressing other critical issues in women's health.
Collapse
Affiliation(s)
| | - Douglas Yee
- Masonic Cancer Center Minneapolis, University of Minnesota, Minneapolis, MN, USA
| | | | - Caroline Hodge
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Virginia Borges
- Division of Medical Oncology, University of Colorado Cancer Center, Aurora, CO, USA
| | - Molly Baxter
- Johns Hopkins Medical School, Baltimore, MD, USA
| | - Jessica Valdez
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tamandra Morgan
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Judy Garber
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Alfred A de Lorimier Endowed Chair in General Surgery, 1825 4th St, 3rd Floor, San Francisco, CA, 94158, USA.
| |
Collapse
|
44
|
Guo Z, Zhu Z, Lin X, Wang S, Wen Y, Wang L, Zhi L, Zhou J. Tumor microenvironment and immunotherapy for triple-negative breast cancer. Biomark Res 2024; 12:166. [PMID: 39741315 DOI: 10.1186/s40364-024-00714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer known for its high aggressiveness and poor prognosis. Conventional treatment of TNBC is challenging due to its heterogeneity and lack of clear targets. Recent advancements in immunotherapy have shown promise in treating TNBC, with immune checkpoint therapy playing a significant role in comprehensive treatment plans. The tumor microenvironment (TME), comprising immune cells, stromal cells, and various cytokines, plays a crucial role in TNBC progression and response to immunotherapy. The high presence of tumor-infiltrating lymphocytes and immune checkpoint proteins in TNBC indicates the potential of immunotherapeutic strategies. However, the complexity of the TME, while offering therapeutic targets, requires further exploration of its multiple roles in immunotherapy. In this review, we discuss the interaction mechanism between TME and TNBC immunotherapy based on the characteristics and composition of TME, and elaborate on and analyze the effect of TME on immunotherapy, the potential of TME as an immune target, and the ability of TME as a biomarker. Understanding these dynamics will offer new insights for enhancing therapeutic approaches and investigating stratification and prognostic markers for TNBC patients.
Collapse
Affiliation(s)
- Zijie Guo
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Ziyu Zhu
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Xixi Lin
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Shenkangle Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Yihong Wen
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China
| | - Linbo Wang
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Lili Zhi
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Affiliated Sir Run Shaw Hospital, Zhejiang University School of Medicine, No.3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center, Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
45
|
Heater NK, Warrior S, Lu J. Current and future immunotherapy for breast cancer. J Hematol Oncol 2024; 17:131. [PMID: 39722028 PMCID: PMC11670461 DOI: 10.1186/s13045-024-01649-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Substantial therapeutic advancement has been made in the field of immunotherapy in breast cancer. The immune checkpoint inhibitor pembrolizumab in combination with chemotherapy received FDA approval for both PD-L1 positive metastatic and early-stage triple-negative breast cancer, while ongoing clinical trials seek to expand the current treatment landscape for immune checkpoint inhibitors in hormone receptor positive and HER2 positive breast cancer. Antibody drug conjugates are FDA approved for triple negative and HER2+ disease, and are being studied in combination with immune checkpoint inhibitors. Vaccines and bispecific antibodies are areas of active research. Studies of cellular therapies such as tumor infiltrating lymphocytes, chimeric antigen receptor-T cells and T cell receptor engineered cells are promising and ongoing. This review provides an update of recent major clinical trials of immunotherapy in breast cancer and discusses future directions in the treatment of breast cancer.
Collapse
Affiliation(s)
- Natalie K Heater
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, 60611, USA
| | - Surbhi Warrior
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 676 N St. Clair, Suite 850, Chicago, IL, 60611, USA
| | - Janice Lu
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 676 N St. Clair, Suite 850, Chicago, IL, 60611, USA.
| |
Collapse
|
46
|
Lin S, Fu B, Khan M. Identifying subgroups deriving the most benefit from PD-1 checkpoint inhibition plus chemotherapy in advanced metastatic triple-negative breast cancer: a systematic review and meta-analysis. World J Surg Oncol 2024; 22:346. [PMID: 39709499 DOI: 10.1186/s12957-024-03424-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The combination of immunotherapy and chemotherapy has demonstrated an enhancement in progression-free survival (PFS) for individuals with advanced and metastatic triple-negative breast cancer (TNBC) when compared to the use of chemotherapy alone. Nevertheless, the extent to which different subgroups of metastatic TNBC patients experience this benefit remains uncertain. OBJECTIVES Our objective was to conduct subgroup analyses to more precisely identify the factors influencing these outcomes. MATERIALS AND METHODS The PubMed database was searched until Dec 2023 for studies that compared PD-1 checkpoint inhibitors plus chemotherapy (ICT) with chemotherapy (CT) alone. The primary outcome of interest was progression-free survival (PFS). Review Manager (RevMan) version 5.4. was used for the data analysis. RESULTS Four randomized controlled trials (RCTs) comprising 2468 advanced and metastatic TNBC were included in this systematic review and meta-analysis. PFS surge with combined therapy was observed in White (HR 0.80 [0.70, 0.91], p = 0.0007) and Asian ethnicities (HR 0.73 [0.58, 0.93], p = 0.01) but not in Blacks (HR 0.72 [0.42, 1.24], p = 0.24). Overall, patients with distant metastasis demonstrated to derive the PFS benefit from additional immunotherapy (HR 0.87 [0.77, 0.99], p = 0.03); however, metastasis to individual distant site was associated with failure to achieve any treatment difference (Bone: HR 0.79 [0.41, 1.52], p = 0.49; Lung: HR 0.85 [0.70, 1.04], p = 0.11; Liver: HR 0.80 [0.64, 1.01], p = 0.06). While number of metastases > 3 also showed to impact the PFS advantage (HR 0.89 [0.69, 1.16], p = 0.39). While patients, regardless of prior chemotherapy, experienced a notable enhancement in PFS with ICT (Overall: HR 0.79 [0.71, 0.88], p < 0.0001; Yes: HR 0.87 [0.76, 1.00], p = 0.05; No: HR 0.67 [0.56, 0.80], p < 0.00001), those previously exposed to chemotherapy exhibited a significantly smaller PFS advantage compared to those without prior chemotherapy, as evidenced by a significant subgroup difference (Test for subgroup difference: P = 0.02, I2 = 82.2%). Patients lacking PD-L1 expression also failed to achieve any additional benefit from immunotherapy (PD-L1-: HR 0.95 [0.81, 1.12]; p = 0.54; PD-L1+: HR 0.73 [0.64, 0.85], p < 0.0001). Age, ECOG status, and presentation with de novo metastasis/recurrent shown no impact on IT-associated PFS advantage. CONCLUSIONS Patient- and treatment- related factors such as ethnicity, distant metastases, number of metastases (> 3), previous exposure to chemotherapy and PD-L1 expression, seem to influence or restrict the advantage in progression-free survival associated with the addition of immunotherapy to chemotherapy, as opposed to chemotherapy alone, in patients with advanced and metastatic TNBC. Larger studies are warranted to validate these outcomes.
Collapse
Affiliation(s)
- Shengfa Lin
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Bihe Fu
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou, Guangdong, 510095, People's Republic of China.
| |
Collapse
|
47
|
Cardoso F, Hirshfield KM, Kraynyak KA, Tryfonidis K, Bardia A. Immunotherapy for hormone receptor‒positive HER2-negative breast cancer. NPJ Breast Cancer 2024; 10:104. [PMID: 39643613 PMCID: PMC11624285 DOI: 10.1038/s41523-024-00704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/14/2024] [Indexed: 12/09/2024] Open
Abstract
Additional therapies are needed to improve outcomes in patients with hormone receptor-positive/human epidermal growth factor receptor 2-negative breast cancer. Research on the potential role of immunotherapy, particularly programmed cell death protein 1/programmed cell death ligand 1 inhibitors, is rapidly expanding in both the early and metastatic settings with some preliminary evidence suggesting benefit when used as part of combination therapy. Several ongoing phase 3 studies should help define their future role in treating these patients.
Collapse
Affiliation(s)
- Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal.
| | | | | | | | - Aditya Bardia
- Department of Medicine, Division of Hematology/Oncology, University of California Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
48
|
Massa D, Vernieri C, Nicolè L, Criscitiello C, Boissière-Michot F, Guiu S, Bobrie A, Griguolo G, Miglietta F, Vingiani A, Lobefaro R, Taurelli Salimbeni B, Pinato C, Schiavi F, Brich S, Pescia C, Fusco N, Pruneri G, Fassan M, Curigliano G, Guarneri V, Jacot W, Dieci MV. Immune and gene-expression profiling in estrogen receptor low and negative early breast cancer. J Natl Cancer Inst 2024; 116:1914-1927. [PMID: 39083015 PMCID: PMC11630536 DOI: 10.1093/jnci/djae178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/25/2024] [Accepted: 07/23/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND The cutoff of <1% positive cells to define estrogen receptor (ER) negativity by immunohistochemistry (IHC) in breast cancer (BC) is debated. We explored the tumor immune microenvironment and gene-expression profile of patients with early-stage HER2-negative ER-low (ER 1%-9%) BC, comparing them to ER-negative (ER <1%) and ER-intermediate (ER 10%-50%) tumors. METHODS Among 921 patients with early-stage I-III, ER ≤50%, HER2-negative BCs, tumors were classified as ER-negative (n = 712), ER-low (n = 128), or ER-intermediate (n = 81). Tumor-infiltrating lymphocytes (TILs) were evaluated. CD8+, FOXP3+ cells, and PD-L1 status were assessed by IHC and quantified by digital pathology. We analyzed 776 BC-related genes in 116 samples. All tests were 2-sided at a <.05 significance level. RESULTS ER-low and ER-negative tumors exhibited similar median TILs, statistically significantly higher than ER-intermediate tumors. CD8/FOXP3 ratio and PD-L1 positivity rates were comparable between ER-low and ER-negative groups. These groups showed similar enrichment in basal-like intrinsic subtypes and comparable expression of immune-related genes. ER-low and ER-intermediate tumors showed significant transcriptomic differences. High TILs (≥30%) were associated with improved relapse-free survival (RFS) in ER-low (5-year RFS 78.6% vs 66.2%, log-rank P = .033, hazard ratio [HR] 0.37 [95% CI = 0.15 to 0.96]) and ER-negative patients (5-year RFS 85.2% vs 69.8%, log-rank P < .001, HR 0.41 [95% CI = 0.27 to 0.60]). CONCLUSIONS ER-low and ER-negative tumors are similar biological and molecular entities, supporting their comparable clinical outcomes and treatment responses, including to immunotherapy. Our findings contribute to the growing evidence calling for a reevaluation of ER-positive BC classification and management, aligning ER-low and ER-negative tumors more closely.
Collapse
Affiliation(s)
- Davide Massa
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- IFOM ETS, The AIRC Institute of Molecular Oncology
| | | | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Séverine Guiu
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Angélique Bobrie
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Gaia Griguolo
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Federica Miglietta
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - Andrea Vingiani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Riccardo Lobefaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Pinato
- UOSD Hereditary Tumors, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Francesca Schiavi
- UOSD Hereditary Tumors, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Silvia Brich
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giancarlo Pruneri
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padova, Italy
- Veneto Institute of Oncology IOV—IRCCS, Padova, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development for Innovative Therapy, European Institute of Oncology IRCCS, Milan, Italy
| | - Valentina Guarneri
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| | - William Jacot
- Translational Research Unit, Institut du Cancer de Montpellier, Montpellier, France
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Maria Vittoria Dieci
- Oncology 2, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, Padova, Italy
| |
Collapse
|
49
|
Shatsky RA, Trivedi MS, Yau C, Nanda R, Rugo HS, Davidian M, Tsiatis B, Wallace AM, Chien AJ, Stringer-Reasor E, Boughey JC, Omene C, Rozenblit M, Kalinsky K, Elias AD, Vaklavas C, Beckwith H, Williams N, Arora M, Nangia C, Roussos Torres ET, Thomas B, Albain KS, Clark AS, Falkson C, Hershman DL, Isaacs C, Thomas A, Tseng J, Sanford A, Yeung K, Boles S, Chen YY, Huppert L, Jahan N, Parker C, Giridhar K, Howard FM, Blackwood MM, Sanft T, Li W, Onishi N, Asare AL, Beineke P, Norwood P, Brown-Swigart L, Hirst GL, Matthews JB, Moore B, Symmans WF, Price E, Heditsian D, LeStage B, Perlmutter J, Pohlmann P, DeMichele A, Yee D, van 't Veer LJ, Hylton NM, Esserman LJ. Datopotamab-deruxtecan plus durvalumab in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial. Nat Med 2024; 30:3737-3747. [PMID: 39277672 DOI: 10.1038/s41591-024-03267-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/17/2024]
Abstract
Sequential adaptive trial designs can help accomplish the goals of personalized medicine, optimizing outcomes and avoiding unnecessary toxicity. Here we describe the results of incorporating a promising antibody-drug conjugate, datopotamab-deruxtecan (Dato-DXd) in combination with programmed cell death-ligand 1 inhibitor, durvalumab, as the first sequence of therapy in the I-SPY2.2 phase 2 neoadjuvant sequential multiple assignment randomization trial for high-risk stage 2/3 breast cancer. The trial includes three blocks of treatment, with initial randomization to different experimental agent(s) (block A), followed by a taxane-based regimen tailored to tumor subtype (block B), followed by doxorubicin-cyclophosphamide (block C). Subtype-specific algorithms based on magnetic resonance imaging volume change and core biopsy guide treatment redirection after each block, including the option of early surgical resection in patients predicted to have a high likelihood of pathologic complete response, which is the primary endpoint assessed when resection occurs. There are two primary efficacy analyses: after block A and across all blocks for six prespecified HER2-negative subtypes (defined by hormone receptor status and/or response-predictive subtypes). In total, 106 patients were treated with Dato-DXd/durvalumab in block A. In the immune-positive subtype, Dato-DXd/durvalumab exceeded the prespecified threshold for success (graduated) after block A; and across all blocks, pathologic complete response rates were equivalent to the rate expected for the standard of care (79%), but 54% achieved that result after Dato-DXd/durvalumab alone (block A) and 92% without doxorubicin-cyclophosphamide (after blocks A + B). The treatment strategy across all blocks graduated in the hormone-negative/immune-negative subtype. No new toxicities were observed. Stomatitis was the most common side effect in block A. No patients receiving block A treatment alone had adrenal insufficiency. Dato-DXd/durvalumab is a promising therapy combination that can eliminate standard chemotherapy in many patients, particularly the immune-positive subtype.ClinicalTrials.gov registration: NCT01042379 .
Collapse
Affiliation(s)
| | | | - Christina Yau
- University of California San Francisco, San Francisco, CA, USA
| | | | - Hope S Rugo
- University of California San Francisco, San Francisco, CA, USA
| | | | | | | | - A Jo Chien
- University of California San Francisco, San Francisco, CA, USA
| | | | | | - Coral Omene
- Cooperman Barnabas Medical Center, New Brunswick, NJ, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | | | | | - Christos Vaklavas
- University of Utah Huntsman Cancer Institute, Salt Lake City, UT, USA
| | | | | | - Mili Arora
- University of California Davis, Davis, CA, USA
| | | | | | | | - Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Chicago, IL, USA
| | - Amy S Clark
- University of Pennsylvania, Philadelphia, PA, USA
| | - Carla Falkson
- University of Rochester Medical Center, Rochester, NY, USA
| | | | - Claudine Isaacs
- Lombardi Comprehensive Cancer Center Georgetown University, Washington, DC, USA
| | | | - Jennifer Tseng
- City of Hope Orange County Lennar Foundation Cancer Center, Irvine, CA, USA
| | | | - Kay Yeung
- University of California San Diego, San Diego, CA, USA
| | - Sarah Boles
- University of California San Diego, San Diego, CA, USA
| | - Yunni Yi Chen
- University of California San Francisco, San Francisco, CA, USA
| | - Laura Huppert
- University of California San Francisco, San Francisco, CA, USA
| | - Nusrat Jahan
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | - Wen Li
- University of California San Francisco, San Francisco, CA, USA
| | - Natsuko Onishi
- University of California San Francisco, San Francisco, CA, USA
| | - Adam L Asare
- University of California San Francisco, San Francisco, CA, USA
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | - Philip Beineke
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | - Peter Norwood
- Quantum Leap Healthcare Collaborative, San Francisco, CA, USA
| | | | - Gillian L Hirst
- University of California San Francisco, San Francisco, CA, USA
| | | | - Brian Moore
- Wake Forest University, Winston-Salem, NC, USA
| | | | - Elissa Price
- University of California San Francisco, San Francisco, CA, USA
| | - Diane Heditsian
- University of California San Francisco, San Francisco, CA, USA
| | - Barbara LeStage
- University of California San Francisco, San Francisco, CA, USA
| | | | - Paula Pohlmann
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Douglas Yee
- University of Minnesota, Minneapolis, MN, USA
| | | | - Nola M Hylton
- University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
50
|
Monberg MJ, Keefe S, Karantza V, Tryfonidis K, Toker S, Mejia J, Orlowski R, Haiderali A, Prabhu VS, Aktan G. A Narrative Review of the Clinical, Humanistic, and Economic Value of Pembrolizumab-Based Immunotherapy for the Treatment of Breast and Gynecologic Cancers. Oncol Ther 2024; 12:701-734. [PMID: 39453600 PMCID: PMC11573950 DOI: 10.1007/s40487-024-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024] Open
Abstract
Breast and gynecologic cancers are common across the world and are associated with substantial societal and economic burden. Pembrolizumab was among the first immune checkpoint inhibitors targeting programmed cell death protein 1 to be approved for the treatment of patients with triple-negative breast cancer, cervical cancer, and endometrial cancer. Recent clinical trials have established pembrolizumab regimens as a standard of care treatment for these tumor types. Clinical data are further supported by patient-reported outcome, cost-effectiveness, and real-world evidence. Pembrolizumab monotherapy and combination regimens do not negatively influence health-related quality of life and are cost-effective relative to comparators. Ongoing phase 3 studies with pembrolizumab will expand the current understanding of its use in breast and gynecologic cancers. Several of these studies are in patients with early-stage disease with the hope of curing patients. The main objective of this review is to summarize the clinical, humanistic, and economic value of pembrolizumab in these settings and to describe the future challenges for patients, caregivers, clinicians, and payers.
Collapse
Affiliation(s)
| | - Steve Keefe
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, 07065, USA
| | | | | | - Sarper Toker
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, 07065, USA
| | - Jaime Mejia
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Amin Haiderali
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Gursel Aktan
- Merck & Co., Inc., 2025 E Scott Ave, Rahway, NJ, 07065, USA
| |
Collapse
|