1
|
Tiwari H, Singh S, Sharma S, Gupta P, Verma A, Chattopadhaya A, Kumar B, Agarwal S, Kumar R, Gupta SK, Gautam V. Deciphering the landscape of triple negative breast cancer from microenvironment dynamics and molecular insights to biomarker analysis and therapeutic modalities. Med Res Rev 2025; 45:817-841. [PMID: 39445844 DOI: 10.1002/med.22090] [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: 05/20/2024] [Revised: 09/05/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Triple negative breast cancer (TNBC) displays a notable challenge in clinical oncology due to its invasive nature which is attributed to the absence of progesterone receptor (PR), estrogen receptor (ER), and human epidermal growth factor receptor (HER-2). The heterogenous tumor microenvironment (TME) of TNBC is composed of diverse constituents that intricately interact to evade immune response and facilitate cancer progression and metastasis. Based on molecular gene expression, TNBC is classified into four molecular subtypes: basal-like (BL1 and BL2), luminal androgen receptor (LAR), immunomodulatory (IM), and mesenchymal. TNBC is an aggressive histological variant with adverse prognosis and poor therapeutic response. The lack of response in most of the TNBC patients could be attributed to the heterogeneity of the disease, highlighting the need for more effective treatments and reliable prognostic biomarkers. Targeting certain signaling pathways and their components has emerged as a promising therapeutic strategy for improving patient outcomes. In this review, we have summarized the interactions among various components of the dynamic TME in TNBC and discussed the classification of its molecular subtypes. Moreover, the purpose of this review is to compile and provide an overview of the most recent data about recently discovered novel TNBC biomarkers and targeted therapeutics that have proven successful in treating metastatic TNBC. The emergence of novel therapeutic strategies such as chemoimmunotherapy, chimeric antigen receptor (CAR)-T cells-based immunotherapy, phytometabolites-mediated natural therapy, photodynamic and photothermal approaches have made a significant positive impact and have paved the way for more effective interventions.
Collapse
Affiliation(s)
- Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sonal Sharma
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Amrit Chattopadhaya
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Brijesh Kumar
- Department of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sakshi Agarwal
- Department of Obstetrics and Gynaecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Sanjeev Kumar Gupta
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
2
|
Choi Y, Tan J, Lin D, Lee JS, Yuan Y. Immunotherapy in Breast Cancer: Beyond Immune Checkpoint Inhibitors. Int J Mol Sci 2025; 26:3920. [PMID: 40332761 PMCID: PMC12027891 DOI: 10.3390/ijms26083920] [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: 03/12/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
The systemic treatment of breast cancer has evolved remarkably over the past decades. With the introduction of immune checkpoint inhibitors (ICIs), clinical outcomes for solid tumor malignancies have significantly improved. However, in breast cancer, the indication for ICIs is currently limited to triple-negative breast cancer (TNBC) only. In high-risk luminal B hormone receptor-positive (HR+) breast cancer (BC) and HER2-positive (HER2+) BC, modest efficacy of ICI and chemotherapy combinations were identified in the neoadjuvant setting. To address the unmet need, several novel immunotherapy strategies are being tested in ongoing clinical trials as summarized in the current review: bispecific antibodies, chimeric antigen receptor T-cell therapy (CAR-T), T-cell receptors (TCRs), tumor-infiltrating lymphocytes (TILs), tumor vaccines, and oncolytic virus therapy.
Collapse
Affiliation(s)
| | | | | | | | - Yuan Yuan
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA; (Y.C.); (J.T.); (D.L.); (J.S.L.)
| |
Collapse
|
3
|
Ababneh E, Velez S, Zhao J. Immune evasion and resistance in breast cancer. Am J Cancer Res 2025; 15:1517-1539. [PMID: 40371160 PMCID: PMC12070088 DOI: 10.62347/pngt6996] [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: 11/11/2024] [Accepted: 12/18/2024] [Indexed: 05/16/2025] Open
Abstract
Breast cancer (BC) is the most common malignancy in females with an increasing incidence in the last decade. The previously observed decline in BC mortality rates has also slowed down recently with an increase in the incidence of invasive BC. BC has various molecular subtypes. Among these subtypes, triple-negative breast cancer (TNBC) represents the most aggressive BC, with a poor prognosis. Because lack of the hormonal or human epidermal growth factor receptor 2 (HER2) receptors, TNBC is resistant to hormonal and HER2 targeted therapy effective for other BC subtypes. The good news is that TNBC has recently been considered an immunologically 'hot' tumor. Therefore, immunotherapy, particularly immune checkpoint inhibitor therapy, represents a promising therapeutic approach TNBC. However, a considerable percentage of patients with TNBC do not respond well to immunotherapy, indicating that TNBC seems to adopt several mechanisms to evade immune surveillance. Thus, it is crucial to investigate the mechanisms underlying TNBC immune evasion and resistance to immunotherapy. In this review, we examine and discuss the most recently discovered mechanisms for BC, with a particular focus on TNBC, to evade the immune surveillance via kidnapping the immune checkpoints, suppressing the immune responses in tumor microenvironment and inhibiting the tumor antigen presentation. Evaluation of these mechanisms in BC will hopefully guide future immunotherapeutic research and clinical trials.
Collapse
Affiliation(s)
- Ebaa Ababneh
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Sarah Velez
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| | - Jihe Zhao
- Burnett School of Biomedical Science, Medical College, University of Central Florida Orlando, FL, USA
| |
Collapse
|
4
|
Dai X, Cao B, Liu X, Meng W, Qiu Y, Sun Y, Zhang L, Li N, Liu Z, Li D, Xiao L, Li B, Zhang Q. Tumor vascular normalization by B7-H3 blockade augments T lymphocyte-mediated antitumor immunity. Eur J Pharmacol 2025; 993:177334. [PMID: 39892447 DOI: 10.1016/j.ejphar.2025.177334] [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: 09/28/2024] [Revised: 01/09/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Triple-negative breast cancer (TNBC), defined by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), presents unique clinical challenges and generally predicts a less favorable prognosis. Despite recent advancements in TNBC treatment, a subset of patients remains resistant to immunotherapy. B7-H3, a member of the B7 family of immune checkpoints, is correlated with poor outcomes in various cancers and is distinctively expressed in tumor vasculature, marking it as a potential biomarker for tumor-associated endothelial cells. We found high expression of B7-H3 in the endothelial cells of the postoperative tissue of TNBC patients. Elevated gene expression of CD276 (encoding B7-H3) and PECAM1 (encoding CD31) in TNBC is associated with poor prognosis. Anti-B7-H3 blockade reduces tumor burden and promotes lymphocyte infiltration in a TNBC mouse model. Additionally, anti-B7-H3 blockade promotes tumor vessel normalization and enhances programmed cell death ligand 1 (PD-L1) expression. Synergistic effects were observed when B7-H3 blockade was combined with programmed cell death protein 1 (PD-1) inhibition in the TNBC mouse model. Furthermore, anti-B7-H3 inhibits human umbilical vein endothelial cell (HUVEC) proliferation by suppression of the nuclear factor kappa-B (NF-κB) signaling pathway. Downregulation of B7-H3 expression in HUVECs promotes lymphocyte trans-endothelial migration. These findings suggest that B7-H3 represents a promising therapeutic target for TNBC, and the combination of anti-B7-H3 and anti-PD-1 therapies may have synergetic effects in treating TNBC.
Collapse
Affiliation(s)
- Xin Dai
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China; Department of Oncology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boran Cao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinnan Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wangyang Meng
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiran Qiu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
| | - Yidan Sun
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Zhang
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nan Li
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyu Liu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lianbo Xiao
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Bin Li
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
5
|
Jiang Y, Qian Z, Wang C, Wu D, Liu L, Ning X, You Y, Mei J, Zhao X, Zhang Y. Targeting B7-H3 inhibition-induced activation of fatty acid synthesis boosts anti-B7-H3 immunotherapy in triple-negative breast cancer. J Immunother Cancer 2025; 13:e010924. [PMID: 40221152 PMCID: PMC11997833 DOI: 10.1136/jitc-2024-010924] [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: 10/29/2024] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most malignant breast cancer, highlighting the need for effective immunotherapeutic targets. The immune checkpoint molecule B7-H3 has recently gained attention as a promising therapeutic target due to its pivotal role in promoting tumorigenesis and cancer progression. However, the therapeutic impact of B7-H3 inhibitors (B7-H3i) remains unclear. METHODS Transcriptomic and metabolomic analyses were conducted to explore the underlying mechanisms of B7-H3 inhibition in TNBC. The therapeutic efficacy of the combined treatment strategy was substantiated through comprehensive phenotypic assays conducted in vitro and validated in vivo using animal models. RESULTS B7-H3 blockade induces a "primed for death" stress state in cancer cells, leading to distinct alterations in metabolic pathways. Specifically, B7-H3 knockdown activated the AKT signaling pathway and upregulated sterol regulatory element-binding protein 1 (SREBP1), which in turn elevated FASN expression. The simultaneous inhibition of both B7-H3 and FASN more effectively attenuated the malignant progression of TNBC. CONCLUSIONS Our findings propose an "immune attack-metabolic compensation" dynamic model and suggest the feasibility of a dual-targeting strategy that concurrently inhibits both B7-H3 and FASN to enhance therapeutic efficacy in TNBC patients.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhiwen Qian
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cenzhu Wang
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Danping Wu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Lu Liu
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xin Ning
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yilan You
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqian Zhao
- Department of Breast Surgery, Women's Hospital of Jiangnan University, Wuxi, China
| | - Yan Zhang
- Department of Oncology, Women's Hospital of Jiangnan University, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Oncology, Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Zhou S, Hong M, Zhao D, Li W, Yuan X, Wang Y, Li H, Yang Y, Jin T, Pan J. Reprogramming the Tumor Immune Microenvironment with ICAM-1-Targeted Antibody‒Drug Conjugates and B7-H3-CD3 Bispecific Antibodies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415577. [PMID: 39996528 PMCID: PMC12021053 DOI: 10.1002/advs.202415577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/27/2025] [Indexed: 02/26/2025]
Abstract
Reprogramming the tumor immune microenvironment (TIM) plays an important role in promoting the reversal of immune "cold" tumors into "hot" inflammatory tumors. Improving drug targeting, blocking immune checkpoints, and promoting the activation of immune cells are crucial for reprogramming the TIM. Here, an intercellular adhesion molecule 1-targeted antibody‒drug conjugate in combination with a B7-H3-CD3 bispecific antibody is selected for TIM reprogramming, which improved the efficacy of triple-negative breast cancer immunotherapy. This combination therapy improves drug targeting, blocks immune checkpoint pathways, and activates effector T cells to release cytokines, leading to immunogenic cell death and the release of tumor-associated antigens. This effect promotes the maturation of dendritic cells, infiltration and activation of cytotoxic CD8+ T cells, repolarization of M1-type macrophages, and reduction of M2-type macrophages, immune suppressor Tregs, and MDS cells, thereby reprogramming the TIM. In addition, this innovative strategy promotes the accumulation of immune cells at metastasis sites and significantly impedes the progression of lung metastatic lesions. Overall, this study provides novel insights for reprogramming the TIM using novel immunotherapeutic strategies that leverage the synergistic effects of antibody-drug conjugates and bispecific antibodies.
Collapse
Affiliation(s)
- Shoubing Zhou
- Department of Breast OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230031China
- Department of Breast OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| | - Mengyu Hong
- Laboratory of Structural ImmunologyCAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Dan Zhao
- Laboratory of Structural ImmunologyCAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Wenyu Li
- Department of Breast OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230031China
- Department of Breast OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| | - Xiaolong Yuan
- Department of Breast OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230031China
- Department of Breast OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| | - Yinghong Wang
- Department of Breast OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230031China
- Department of Breast OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| | - Hualong Li
- Department of Tumor RadiotherapyPeople Hospital of Fengyang CountyChuzhouAnhui233100China
| | - Yang Yang
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of OncologySuzhou Hospital of Anhui Medical UniversitySuzhou234000China
| | - Tengchuan Jin
- Laboratory of Structural ImmunologyCAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230027China
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineCenter for Advanced Interdisciplinary Science and Biomedicine of IHMUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Institute of Health and Medicine, Hefei Comprehensive National Science CenterHefeiAnhuiChina
- Biomedical Sciences and Health Laboratory of Anhui ProvinceUniversity of Science & Technology of ChinaHefei230027China
- Clinical Research Hospital of the Chinese Academy of Sciences (Hefei)University of Science and Technology of ChinaHefei230001China
| | - Jing Pan
- Department of Breast OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230031China
- Department of Breast OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| |
Collapse
|
7
|
Mei J, Yang K, Zhang X, Luo Z, Tian M, Fan H, Chu J, Zhang Y, Ding J, Xu J, Cai Y, Yin Y. Intratumoral Collagen Deposition Supports Angiogenesis Suggesting Anti-angiogenic Therapy in Armored and Cold Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409147. [PMID: 39823457 PMCID: PMC11904994 DOI: 10.1002/advs.202409147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/05/2025] [Indexed: 01/19/2025]
Abstract
A previous study classifies solid tumors based on collagen deposition and immune infiltration abundance, identifying a refractory subtype termed armored & cold tumors, characterized by elevated collagen deposition and diminished immune infiltration. Beyond its impact on immune infiltration, collagen deposition also influences tumor angiogenesis. This study systematically analyzes the association between immuno-collagenic subtypes and angiogenesis across diverse cancer types. As a result, armored & cold tumors exhibit the highest angiogenic activity in lung adenocarcinoma (LUAD). Single-cell and spatial transcriptomics reveal close interactions and spatial co-localization of fibroblasts and endothelial cells. In vitro experiments demonstrate that collagen stimulates tumor cells to express vascular endothelial growth factor A (VEGFA) and directly enhances vessel formation and endothelial cell proliferation through sex determining region Y box 18 (SOX18) upregulation. Collagen inhibition via multiple approaches effectively suppresses tumor angiogenesis in vivo. In addition, armored & cold tumors display superior responsiveness to anti-angiogenic therapy in advanced LUAD cohorts. Post-immunotherapy resistance, the transformation into armored & cold tumors emerges as a potential biomarker for selecting anti-angiogenic therapy. In summary, collagen deposition is shown to drive angiogenesis across various cancers, providing a novel and actionable framework to refine therapeutic strategies combining chemotherapy with anti-angiogenic treatments.
Collapse
Affiliation(s)
- Jie Mei
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Kai Yang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Xinkang Zhang
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Zhiwen Luo
- Department of Sports MedicineHuashan Hospital Affiliated to Fudan UniversityShanghai200040P. R. China
| | - Min Tian
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Hanfang Fan
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Jiahui Chu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsu211166P. R. China
| | - Yan Zhang
- Departments of GynecologyThe Women's Hospital Affiliated to Jiangnan UniversityWuxi214023China
| | - Junli Ding
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Junying Xu
- Departments of OncologyWuxi People's HospitalThe Affiliated Wuxi People's Hospital of Nanjing Medical UniversityWuxi Medical CenterNanjing Medical UniversityWuxiJiangsu214023P. R. China
| | - Yun Cai
- Central LaboratoryChangzhou Jintan First People's HospitalThe Affiliated Jintan Hospital of Jiangsu UniversityChangzhouJiangsu213200P. R. China
| | - Yongmei Yin
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu211166P. R. China
- Jiangsu Key Lab of Cancer BiomarkersPrevention and TreatmentCollaborative Innovation Center for Personalized Cancer MedicineNanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
8
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [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: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
9
|
Mei J, Luo Z, Cai Y, Wan R, Qian Z, Chu J, Sun Y, Shi Y, Jiang Y, Zhang Y, Yin Y, Chen S. Altered Atlas of Exercise-Responsive MicroRNAs Revealing miR-29a-3p Attacks Armored and Cold Tumors and Boosts Anti-B7-H3 Therapy. RESEARCH (WASHINGTON, D.C.) 2025; 8:0590. [PMID: 39845707 PMCID: PMC11751204 DOI: 10.34133/research.0590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/03/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
Increasing evidence has shown that physical exercise remarkably inhibits oncogenesis and progression of numerous cancers and exercise-responsive microRNAs (miRNAs) exert a marked role in exercise-mediated tumor suppression. In this research, expression and prognostic values of exercise-responsive miRNAs were examined in breast cancer (BRCA) and further pan-cancer types. In addition, multiple independent public and in-house cohorts, in vitro assays involving multiple, macrophages, fibroblasts, and tumor cells, and in vivo models were utilized to uncover the tumor-suppressive roles of miR-29a-3p in cancers. Here, we reported that miR-29a-3p was the exercise-responsive miRNA, which was lowly expressed in tumor tissues and associated with unfavorable prognosis in BRCA. Mechanistically, miR-29a-3p targeted macrophages, fibroblasts, and tumor cells to down-regulate B7 homolog 3 (B7-H3) expression. Single-cell RNA sequencing (scRNA-seq) and cytometry by time-of-flight (CyTOF) demonstrated that miR-29a-3p attacked the armored and cold tumors, thereby shaping an immuno-hot tumor microenvironment (TME). Translationally, liposomes were developed and loaded with miR-29a-3p (lipo@miR-29a-3p), and lipo@miR-29a-3p exhibited promising antitumor effects in a mouse model with great biocompatibility. In conclusion, we uncovered that miR-29a-3p is a critical exercise-responsive miRNA, which attacked armored and cold tumors by inhibiting B7-H3 expression. Thus, miR-29a-3p restoration could be an alternative strategy for antitumor therapy.
Collapse
Affiliation(s)
- Jie Mei
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Yun Cai
- Department of Central Laboratory, Changzhou Jintan First People’s Hospital, Jiangsu University, Changzhou 213200, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Zhiwen Qian
- Departments of Gynecology, Wuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China
| | - Yaying Sun
- Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yuxin Shi
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Ying Jiang
- Department of Gynecology, The Obstetrics and Gynecology Hospital Affiliated to Jiangnan University, Wuxi 214023, China
| | - Yan Zhang
- Departments of Gynecology, Wuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China
- Department of Gynecology, The Obstetrics and Gynecology Hospital Affiliated to Jiangnan University, Wuxi 214023, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 211166, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai 200040, China
| |
Collapse
|
10
|
Sardela de Miranda F, Martinez-Marin D, Babcock RL, Castro M, Boligala GP, Khan SY, Furr KL, Castro-Piedras I, Wagner N, Robison DE, Daniele K, Singh SP, Pruitt K, Melkus MW, Layeequr Rahman R. Cryoablation of primary breast cancer tumors induces a systemic abscopal effect altering TIME (Tumor Immune Microenvironment) in distant tumors. Front Immunol 2024; 15:1498942. [PMID: 39703517 PMCID: PMC11657241 DOI: 10.3389/fimmu.2024.1498942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Despite recent advances, triple-negative breast cancer (TNBC) patients remain at high risk for recurrence and metastasis, which creates the need for innovative therapeutic approaches to improve patient outcomes. Cryoablation is a promising, less invasive alternative to surgical resection, capable of inducing tumor necrosis via freeze/thaw cycles. Necrotic cell death results in increased inflammatory signals and release of preserved tumor antigens, which have the potential to boost the local and systemic anti-tumor immune response. Thus, compared to surgery, cryoablation enhances the activation of T cells leading to an improved abscopal effect, defined as the occurrence of a systemic response after local treatment. We previously showed with a bilateral-tumor mouse model of TNBC that cryoablation of the primary tumor leads to increased infiltration of distant (abscopal) tumors by tumor infiltrating lymphocytes (TILs) and decreased rates of recurrence and metastasis. However, the early drivers of the cryoablation generated abscopal effect are still unknown and knowledge of the mechanism could provide insight into improving the anti-tumor immune response through pharmacologic immune modulation in addition to cryoablation. Methods One million 4T1-12B-luciferase expressing cells were transplanted into the mammary fat pad of BALB/c mice. Two weeks later, left (primary) tumors were either resected or cryoablated. A week after the procedure, right (abscopal) and left tumors, along with spleen, tumor-draining lymph node and blood were collected and processed for flow cytometry and/or RNA-sequencing and immunofluorescence. Results Here we show that cryoablation of mouse mammary carcinomas results in smaller abscopal tumors that harbor increased frequencies of anti-tumor cells [such as natural killer (NK) cells], accompanied by a systemic increase in the frequency of migratory conventional type 1 dendritic cells (cDC1; CD103+ XCR1+), compared to resection. The changes in cell frequencies are mirrored by the immune gene signature of the abscopal tumors, with cryoablation inducing genes involved with NK cell activation and leukocyte-mediated toxicity, including IL11ra1 and Pfr1. Conclusions These results better define the early mechanisms through which cryoablation improves tumor elimination, which is mediated by enhanced frequencies of anti-tumoral cells such as NK and cDC1s at the abscopal tumor and in the spleen of mice treated with cryoablation, respectively.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Dalia Martinez-Marin
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachel L. Babcock
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Maribel Castro
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Geetha P. Boligala
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sonia Y. Khan
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Surgery, The University of Texas Rio Grande Valley (UTRGV) Rio Grande Valley, Harlingen, TX, United States
| | - Kathryn L. Furr
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Isabel Castro-Piedras
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Nicholas Wagner
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Dakota E. Robison
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Karla Daniele
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sharda P. Singh
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael W. Melkus
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Cancer Institute, MetroHealth System, Cleveland, OH, United States
| |
Collapse
|
11
|
Özalp FR, Yörükoğlu K, Yıldırım EÇ, Uzun M, Semiz HS. Prognostic value of B7-H3 expression in metastatic renal cell carcinoma and its impact on immunotherapy response. BMC Cancer 2024; 24:1471. [PMID: 39614178 DOI: 10.1186/s12885-024-13238-x] [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: 06/05/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) is characterised by its immunogenic and proangiogenic nature and its resistance to conventional therapies. The advent of immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs) has significantly improved patient survival, but resistance to these treatments remains a challenge. B7-H3, a potential immune checkpoint, has been implicated in modulating the tumour microenvironment and immune escape mechanisms in RCC. METHODS Immunohistochemical analysis of B7-H3 expression was performed in 84 metastatic RCC patients. Tissue microarrays and separate sections of formalin-fixed paraffin-embedded tissue were used for immunohistochemical staining. Membranous staining of the tumor cells was scored and statistical analyses were performed to assess the correlation between B7-H3 expression and treatment outcome. RESULTS B7-H3 expression was absent in 31% of patients, while 33.3% had a score of 1+, 31% had 2+, and 4.8% had 3+. High B7-H3 expression correlated with poorer OS (20 months vs. 45 months, p = 0.012). In patients receiving nivolumab, those with high B7-H3 expression had shorter PFS (2 months vs. 8 months, p = 0.037) and OS (17 months vs. 51 months, p = 0.01). B7-H3 expression was the only factor significantly affecting PFS and OS in multivariate analysis. CONCLUSION High B7-H3 expression is associated with poorer survival outcomes and reduced response to nivolumab in metastatic RCC patients. B7-H3 may serve as a predictive biomarker for immunotherapy response. Future studies should explore targeting B7-H3 in combination with existing therapies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Faruk Recep Özalp
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| | - Kutsal Yörükoğlu
- Department of Pathology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Eda Çalışkan Yıldırım
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Mehmet Uzun
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Hüseyin Salih Semiz
- Department of Medical Oncology, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
12
|
Hagelstein I, Wessling L, Rochwarger A, Zekri L, Klimovich B, Tegeler CM, Jung G, Schürch CM, Salih HR, Lutz MS. Targeting CD276 for T cell-based immunotherapy of breast cancer. J Transl Med 2024; 22:902. [PMID: 39367484 PMCID: PMC11452943 DOI: 10.1186/s12967-024-05689-4] [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: 06/14/2024] [Accepted: 09/17/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in women. Immunotherapy has revolutionized treatment options in many malignancies, and the introduction of immune checkpoint inhibition yielded beneficial results also in BC. However, many BC patients are ineligible for this T cell-based therapy, others do not respond or only briefly. Thus, there remains a high medical need for new therapies, particularly for triple-negative BC. CD276 (B7-H3) is overexpressed in several tumors on both tumor cells and tumor vessels, constituting a promising target for immunotherapy. METHODS We analyzed tumor samples of 25 patients using immunohistochemistry to assess CD276 levels. The potential of CC-3, a novel bispecific CD276xCD3 antibody, for BC treatment was evaluated using various functional in vitro assays. RESULTS Pronounced expression of CD276 was observed in all analyzed tumor samples including triple negative BC. In analyses with BC cells, CC-3 induced profound T cell activation, proliferation, and T cell memory subset formation. Moreover, treatment with CC-3 induced cytokine secretion and potent tumor cell lysis. CONCLUSION Our findings characterize CD276 as promising target and preclinically document the therapeutic potential of CC-3 for BC treatment, providing a strong rationale for evaluation of CC-3 in BC patients in a clinical trial for which the recruitment has recently started.
Collapse
Affiliation(s)
- Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Laura Wessling
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department for Immunology and German Cancer Consortium (DKTK), Eberhard Karls University, Tübingen, Germany
| | - Boris Klimovich
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Christian M Tegeler
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Department of Obstetrics and Gynecology, University Hospital Tübingen, Tübingen, Germany
- Department of Peptide-Based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Gundram Jung
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department for Immunology and German Cancer Consortium (DKTK), Eberhard Karls University, Tübingen, Germany
| | - Christian M Schürch
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| | - Martina S Lutz
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, German Cancer Consortium (DKTK), University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| |
Collapse
|
13
|
Curcean S, Hendea RM, Buiga R, Tipcu A, Curcean A, Vlad C, Fekete Z, Muntean AS, Martin D, Irimie A. B7H3 Immune Checkpoint Overexpression Is Associated with Decreased Complete Response Rates to Neoadjuvant Therapy in Locally Advanced Rectal Cancer. Diagnostics (Basel) 2024; 14:2023. [PMID: 39335702 PMCID: PMC11431099 DOI: 10.3390/diagnostics14182023] [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: 08/20/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Rectal cancer accounts for approximately one-third of colorectal cancers, with over 340,000 deaths globally in 2022. Despite advancements in treatment, the five-year overall survival for locally advanced rectal cancer (LARC) remains at 74%, with significant morbidity. B7H3 (CD276), an immune checkpoint protein, plays a role in tumor progression and resistance to therapy, and correlates with poor prognosis in various cancers, including colorectal cancer. This study aims to evaluate the expression of B7H3 in LARC and its impact on overall complete response (oCR) rates to neoadjuvant therapy. METHODS A retrospective study was conducted on 60 patients with LARC who received neoadjuvant chemoradiation (nCRT) followed by total mesorectal excision (TME). B7H3 expression was assessed using immunohistochemistry on surgical specimens. Expression levels were categorized as high or low based on a composite score, and their association with oCR rates was analyzed. RESULTS High B7H3 expression was observed in 60% of patients, with 73.5% showing expression in more than 50% of tumor cells. Patients who achieved oCR had significantly lower B7H3 expression compared to those with residual disease (p < 0.001). No nuclear expression of B7H3 was detected. No significant correlation was found between B7H3 expression and other clinicopathological variables, except for a higher likelihood of non-restorative surgery in patients with elevated B7H3 levels (p = 0.049). Mucinous adenocarcinoma had high expression of B7H3. CONCLUSIONS Elevated B7H3 expression is associated with reduced oCR rates in LARC, highlighting its potential role as a prognostic biomarker. Further studies with larger cohorts are warranted to validate these findings and explore B7H3-targeted therapies as a treatment strategy for LARC.
Collapse
Affiliation(s)
- Sebastian Curcean
- Department of Radiation Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Radiation Oncology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Raluca Maria Hendea
- Department of Pathology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Rares Buiga
- Department of Pathology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Alexandru Tipcu
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andra Curcean
- Department of Imaging, Affidea Center, 400487 Cluj-Napoca, Romania
| | - Catalin Vlad
- Department of Oncological Surgery and Gynecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Oncological Surgery, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Zsolt Fekete
- Department of Radiation Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Radiation Oncology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Alina-Simona Muntean
- Department of Radiation Oncology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Daniela Martin
- Department of Radiation Oncology, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Oncological Surgery and Gynecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Oncological Surgery, “Prof. Dr. Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
14
|
Mei J, Chu J, Yang K, Luo Z, Yang J, Xu J, Li Q, Zhang Y, Zhang Q, Wan M, Xue N, Ding J, Zhu Y, Cai Y, Yin Y. Angiotensin receptor blocker attacks armored and cold tumors and boosts immune checkpoint blockade. J Immunother Cancer 2024; 12:e009327. [PMID: 39244215 PMCID: PMC11418576 DOI: 10.1136/jitc-2024-009327] [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] [Accepted: 08/23/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has made remarkable achievements, but newly identified armored and cold tumors cannot respond to ICB therapy. The high prevalence of concomitant medications has huge impact on immunotherapeutic responses, but the clinical effects on the therapeutic outcome of armored and cold tumors are still unclear. METHODS In this research, using large-scale transcriptomics datasets, the expression and potential biological functions of angiotensin II receptor 1 (AGTR1), the target of angiotensin receptor blocker (ARB), were investigated. Next, the roles of ARB in tumor cells and tumor microenvironment cells were defined by a series of in vitro and in vivo assays. In addition, the clinical impacts of ARB on ICB therapy were assessed by multicenter cohorts and meta-analysis. RESULTS AGTR1 was overexpressed in armored and cold tumors and associated with poor response to ICB therapy. ARB, the inhibitor for AGTR1, only suppressed the aggressiveness of tumor cells with high AGTR1 expression, which accounted for a very small proportion. Further analysis revealed that AGTR1 was always highly expressed in cancer-associated fibroblasts (CAFs) and ARB inhibited type I collagen expression in CAFs by suppressing the RhoA-YAP axis. Moreover, ARB could also drastically reverse the phenotype of armored and cold to soft and hot in vivo, leading to a higher response to ICB therapy. In addition, both our in-house cohorts and meta-analysis further supported the idea that ARB can significantly enhance ICB efficacy. CONCLUSION Overall, we identify AGTR1 as a novel target in armored and cold tumors and demonstrate the improved therapeutic efficacy of ICB in combination with ARB. These findings could provide novel clinical insight into how to treat patients with refractory armored and cold tumors.
Collapse
Affiliation(s)
- Jie Mei
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiahui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kai Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jiayue Yang
- Departments of Endocrinology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Junying Xu
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Qing Li
- Departments of Oncology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, Jiangsu, China
| | - Yan Zhang
- Departments of Gynecology, The Obstetrics and Gynecology Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu, China
- Departments of Gynecology, Wuxi Maternal and Child Health Care Hospital, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Qinglin Zhang
- Departments of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Mengyun Wan
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningyi Xue
- The First Clinical Medicine College, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junli Ding
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yichao Zhu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Cai
- Department of Central Laboratory, The First People's Hospital of Jintan, Jintan Affiliated Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
15
|
Mei J, Cai Y, Xu R, Li Q, Chu J, Luo Z, Sun Y, Shi Y, Xu J, Li D, Liang S, Jiang Y, Liu J, Qian Z, Zhou J, Wan M, Yang Y, Zhu Y, Zhang Y, Yin Y. Conserved immuno-collagenic subtypes predict response to immune checkpoint blockade. Cancer Commun (Lond) 2024; 44:554-575. [PMID: 38507505 PMCID: PMC11110954 DOI: 10.1002/cac2.12538] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/06/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has revolutionized the treatment of various cancer types. Despite significant preclinical advancements in understanding mechanisms, identifying the molecular basis and predictive biomarkers for clinical ICB responses remains challenging. Recent evidence, both preclinical and clinical, underscores the pivotal role of the extracellular matrix (ECM) in modulating immune cell infiltration and behaviors. This study aimed to create an innovative classifier that leverages ECM characteristics to enhance the effectiveness of ICB therapy. METHODS We analyzed transcriptomic collagen activity and immune signatures in 649 patients with cancer undergoing ICB therapy. This analysis led to the identification of three distinct immuno-collagenic subtypes predictive of ICB responses. We validated these subtypes using the transcriptome data from 9,363 cancer patients from The Cancer Genome Atlas (TCGA) dataset and 1,084 in-house samples. Additionally, novel therapeutic targets were identified based on these established immuno-collagenic subtypes. RESULTS Our categorization divided tumors into three subtypes: "soft & hot" (low collagen activity and high immune infiltration), "armored & cold" (high collagen activity and low immune infiltration), and "quiescent" (low collagen activity and immune infiltration). Notably, "soft & hot" tumors exhibited the most robust response to ICB therapy across various cancer types. Mechanistically, inhibiting collagen augmented the response to ICB in preclinical models. Furthermore, these subtypes demonstrated associations with immune activity and prognostic predictive potential across multiple cancer types. Additionally, an unbiased approach identified B7 homolog 3 (B7-H3), an available drug target, as strongly expressed in "armored & cold" tumors, relating with poor prognosis. CONCLUSION This study introduces histopathology-based universal immuno-collagenic subtypes capable of predicting ICB responses across diverse cancer types. These findings offer insights that could contribute to tailoring personalized immunotherapeutic strategies for patients with cancer.
Collapse
Affiliation(s)
- Jie Mei
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yun Cai
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Rui Xu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Qing Li
- Departments of OncologyXuzhou Central HospitalThe Xuzhou School of Clinical Medicine of Nanjing Medical UniversityXuzhouJiangsuP. R. China
| | - Jiahui Chu
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- The First Clinical Medicine CollegeNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Zhiwen Luo
- Department of Sports MedicineHuashan Hospital Affiliated to Fudan UniversityShanghaiP. R. China
| | - Yaying Sun
- Department of Sports MedicineShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiP. R. China
| | - Yuxin Shi
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Junying Xu
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Di Li
- Shanghai Outdo Biotech Co., Ltd., National Engineering Center for BiochipShanghaiP. R. China
| | - Shuai Liang
- Departments of OncologyThe Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Ying Jiang
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Jiayu Liu
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Zhiwen Qian
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
| | - Jiaofeng Zhou
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Mengyun Wan
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yunlong Yang
- Department of Cellular and Genetic MedicineSchool of Basic Medical Sciences, Fudan UniversityShanghaiP. R. China
| | - Yichao Zhu
- Department of PhysiologySchool of Basic Medical SciencesNanjing Medical UniversityNanjingJiangsuP. R. China
| | - Yan Zhang
- Departments of GynecologyWuxi Maternal and Child Health Care Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuP. R. China
- Departments of GynecologyWuxi Maternity and Child Health Care HospitalAffiliated Women's Hospital of Jiangnan UniversityWuxiJiangsuP. R. China
| | - Yongmei Yin
- Department of OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsuP. R. China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical UniversityNanjingJiangsuP. R. China
| |
Collapse
|
16
|
Shen B, Mei J, Xu R, Cai Y, Wan M, Zhou J, Ding J, Zhu Y. B7-H3 is associated with the armored-cold phenotype and predicts poor immune checkpoint blockade response in melanoma. Pathol Res Pract 2024; 256:155267. [PMID: 38520953 DOI: 10.1016/j.prp.2024.155267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
Melanoma is the most suitable tumor type for immunotherapy, but not all melanoma patients could respond to immunotherapy. B7 homolog 3 (B7-H3) belongs to the B7 family and is overexpressed in a number of malignant tumors, but the expression pattern of B7-H3 in melanoma has not been well summarized. The expression of B7-H3 was investigated in melanoma and its correlations with features of the tumor microenvironment (TME) by using various public databases, including the Cancer Genome Atlas (TCGA), the GEPIA, and the Human Protein Atlas databases. In addition, the in-house melanoma tissue microarray was applied to validate the results from public databases. Based on the public and in-house cohorts, we found that B7-H3 was overexpressed in melanoma tumor tissues and high B7-H3 expression was related to poor clinical outcome. Moreover, B7-H3 was negatively correlated with levels of tumor-infiltrating lymphocytes (TILs) and positively correlated with collagen infiltration. With clinical translational value, the predictive value of B7-H3 for conventional immunotherapy was detected using the Kaplan-Meier plotter tool, and the results showed that melanoma patients with high B7-H3 expression were insensitive to anti-PD-1 and anti-CTLA-4 immunotherapy. In conclusion, we first investigate the expression of B7-H3 in melanoma and its correlations with the TME features, and indicate B7-H3 as a promising therapeutic target in melanoma patients that are insensitive to conventional immunotherapy.
Collapse
Affiliation(s)
- Bozhi Shen
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China; The Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Jie Mei
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China
| | - Rui Xu
- The First Clinical Medicine College, Nanjing Medical University, Nanjing 211166, China
| | - Yun Cai
- Department of Laboratory Medicine, Changzhou Jintan First People's Hospital, Changzhou 213200, China
| | - Mengyun Wan
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Ji Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China
| | - Junli Ding
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi 214023, China.
| | - Yichao Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
17
|
Jiang Y, Liu J, Chen L, Qian Z, Zhang Y. A promising target for breast cancer: B7-H3. BMC Cancer 2024; 24:182. [PMID: 38326735 PMCID: PMC10848367 DOI: 10.1186/s12885-024-11933-3] [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: 11/22/2023] [Accepted: 01/29/2024] [Indexed: 02/09/2024] Open
Abstract
Breast cancer (BC) is the second-leading factor of mortality for women globally and is brought on by a variety of genetic and environmental causes. The conventional treatments for this disease have limitations, making it difficult to improve the lifespan of breast cancer patients. As a result, extensive research has been conducted over the past decade to find innovative solutions to these challenges. Targeting of the antitumor immune response through the immunomodulatory checkpoint protein B7 family has revolutionized cancer treatment and led to intermittent patient responses. B7-H3 has recently received attention because of its significant demodulation and its immunomodulatory effects in many cancers. Uncontrolled B7-H3 expression and a bad outlook are strongly associated, according to a substantial body of cancer research. Numerous studies have shown that BC has significant B7-H3 expression, and B7-H3 induces an immune evasion phenotype, consequently enhancing the survival, proliferation, metastasis, and drug resistance of BC cells. Thus, an innovative target for immunotherapy against BC may be the B7-H3 checkpoint.In this review, we discuss the structure and regulation of B7-H3 and its double costimulatory/coinhibitory function within the framework of cancer and normal physiology. Then we expound the malignant behavior of B7-H3 in BC and its role in the tumor microenvironment (TME) and finally focus on targeted drugs against B7-H3 that have opened new therapeutic opportunities in BC.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China
| | - Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Wuxi, 214000, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Wuxi, 214000, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China.
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Wuxi, 214000, China.
| |
Collapse
|